WO2018149978A1 - ALTERNATIVE ACTIVATION OF MACROPHAGES (M2 CELLS) THROUGH THE POLYAMINE-eIF5a-HYPUSINE AXIS - Google Patents
ALTERNATIVE ACTIVATION OF MACROPHAGES (M2 CELLS) THROUGH THE POLYAMINE-eIF5a-HYPUSINE AXIS Download PDFInfo
- Publication number
- WO2018149978A1 WO2018149978A1 PCT/EP2018/053913 EP2018053913W WO2018149978A1 WO 2018149978 A1 WO2018149978 A1 WO 2018149978A1 EP 2018053913 W EP2018053913 W EP 2018053913W WO 2018149978 A1 WO2018149978 A1 WO 2018149978A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- elf5a
- compound
- activation
- macrophages
- Prior art date
Links
- 210000004027 cell Anatomy 0.000 title claims abstract description 221
- 230000004913 activation Effects 0.000 title claims abstract description 105
- 210000002540 macrophage Anatomy 0.000 title claims abstract description 68
- 150000001875 compounds Chemical class 0.000 claims abstract description 125
- 238000000034 method Methods 0.000 claims abstract description 63
- 230000002401 inhibitory effect Effects 0.000 claims abstract description 57
- 210000004322 M2 macrophage Anatomy 0.000 claims abstract description 48
- 239000000523 sample Substances 0.000 claims abstract description 44
- 230000024245 cell differentiation Effects 0.000 claims abstract description 13
- 230000009467 reduction Effects 0.000 claims abstract description 9
- 239000013068 control sample Substances 0.000 claims abstract description 8
- 108090000623 proteins and genes Proteins 0.000 claims description 94
- 102000004169 proteins and genes Human genes 0.000 claims description 72
- -1 CCL4 Proteins 0.000 claims description 51
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 49
- 239000004055 small Interfering RNA Substances 0.000 claims description 45
- 108020004459 Small interfering RNA Proteins 0.000 claims description 40
- 108091027967 Small hairpin RNA Proteins 0.000 claims description 33
- 108010028753 deoxyhypusine hydroxylase Proteins 0.000 claims description 31
- 208000035475 disorder Diseases 0.000 claims description 31
- 206010028980 Neoplasm Diseases 0.000 claims description 30
- 102000006735 deoxyhypusine hydroxylase Human genes 0.000 claims description 30
- 102000048231 Deoxyhypusine synthases Human genes 0.000 claims description 28
- 108700023218 Deoxyhypusine synthases Proteins 0.000 claims description 28
- 230000000692 anti-sense effect Effects 0.000 claims description 25
- 238000004458 analytical method Methods 0.000 claims description 23
- SCKYRAXSEDYPSA-UHFFFAOYSA-N ciclopirox Chemical compound ON1C(=O)C=C(C)C=C1C1CCCCC1 SCKYRAXSEDYPSA-UHFFFAOYSA-N 0.000 claims description 22
- 238000011282 treatment Methods 0.000 claims description 22
- 101000832225 Homo sapiens Stabilin-1 Proteins 0.000 claims description 21
- 102100024471 Stabilin-1 Human genes 0.000 claims description 21
- 229960003749 ciclopirox Drugs 0.000 claims description 21
- 102000004060 Transforming Growth Factor-beta Type II Receptor Human genes 0.000 claims description 20
- 108010082684 Transforming Growth Factor-beta Type II Receptor Proteins 0.000 claims description 20
- 239000003814 drug Substances 0.000 claims description 20
- 102100025354 Macrophage mannose receptor 1 Human genes 0.000 claims description 19
- 229940079593 drug Drugs 0.000 claims description 19
- 230000003176 fibrotic effect Effects 0.000 claims description 19
- 230000003463 hyperproliferative effect Effects 0.000 claims description 19
- 108091023037 Aptamer Proteins 0.000 claims description 18
- 201000010099 disease Diseases 0.000 claims description 18
- 208000032672 Histiocytosis haematophagic Diseases 0.000 claims description 17
- 239000003550 marker Substances 0.000 claims description 17
- 210000001519 tissue Anatomy 0.000 claims description 17
- 102000053642 Catalytic RNA Human genes 0.000 claims description 16
- 108090000994 Catalytic RNA Proteins 0.000 claims description 16
- 108091092562 ribozyme Proteins 0.000 claims description 16
- 108010009992 CD163 antigen Proteins 0.000 claims description 15
- 102100025831 Scavenger receptor cysteine-rich type 1 protein M130 Human genes 0.000 claims description 15
- 201000011510 cancer Diseases 0.000 claims description 15
- 101001018258 Homo sapiens Macrophage receptor MARCO Proteins 0.000 claims description 14
- 208000004987 Macrophage activation syndrome Diseases 0.000 claims description 14
- 102100033272 Macrophage receptor MARCO Human genes 0.000 claims description 14
- 102000052812 Ornithine decarboxylases Human genes 0.000 claims description 14
- 108700005126 Ornithine decarboxylases Proteins 0.000 claims description 14
- RPTUSVTUFVMDQK-UHFFFAOYSA-N Hidralazin Chemical compound C1=CC=C2C(NN)=NN=CC2=C1 RPTUSVTUFVMDQK-UHFFFAOYSA-N 0.000 claims description 13
- WZNJWVWKTVETCG-YFKPBYRVSA-N L-mimosine Chemical compound OC(=O)[C@@H](N)CN1C=CC(=O)C(O)=C1 WZNJWVWKTVETCG-YFKPBYRVSA-N 0.000 claims description 13
- 238000001262 western blot Methods 0.000 claims description 13
- 210000004369 blood Anatomy 0.000 claims description 12
- 239000008280 blood Substances 0.000 claims description 12
- 102100023701 C-C motif chemokine 18 Human genes 0.000 claims description 11
- 102100029057 Coagulation factor XIII A chain Human genes 0.000 claims description 11
- 101000978371 Homo sapiens C-C motif chemokine 18 Proteins 0.000 claims description 11
- 101000918352 Homo sapiens Coagulation factor XIII A chain Proteins 0.000 claims description 11
- 101000577881 Homo sapiens Macrophage metalloelastase Proteins 0.000 claims description 11
- 101000635938 Homo sapiens Transforming growth factor beta-1 proprotein Proteins 0.000 claims description 11
- 102100027998 Macrophage metalloelastase Human genes 0.000 claims description 11
- 108010031099 Mannose Receptor Proteins 0.000 claims description 11
- 102100030742 Transforming growth factor beta-1 proprotein Human genes 0.000 claims description 11
- 102100036006 Adenosine receptor A3 Human genes 0.000 claims description 10
- 102100023702 C-C motif chemokine 13 Human genes 0.000 claims description 10
- 102100021396 Cell surface glycoprotein CD200 receptor 1 Human genes 0.000 claims description 10
- 102100039436 DNA-binding protein inhibitor ID-3 Human genes 0.000 claims description 10
- 101000783645 Homo sapiens Adenosine receptor A3 Proteins 0.000 claims description 10
- 101000978379 Homo sapiens C-C motif chemokine 13 Proteins 0.000 claims description 10
- 101000969553 Homo sapiens Cell surface glycoprotein CD200 receptor 1 Proteins 0.000 claims description 10
- 101001036287 Homo sapiens DNA-binding protein inhibitor ID-3 Proteins 0.000 claims description 10
- 101001011441 Homo sapiens Interferon regulatory factor 4 Proteins 0.000 claims description 10
- 101001019600 Homo sapiens Interleukin-17 receptor B Proteins 0.000 claims description 10
- 101001064853 Homo sapiens Polyunsaturated fatty acid lipoxygenase ALOX15 Proteins 0.000 claims description 10
- 101000666171 Homo sapiens Protein-glutamine gamma-glutamyltransferase 2 Proteins 0.000 claims description 10
- 102100030126 Interferon regulatory factor 4 Human genes 0.000 claims description 10
- 102100035014 Interleukin-17 receptor B Human genes 0.000 claims description 10
- 102100031950 Polyunsaturated fatty acid lipoxygenase ALOX15 Human genes 0.000 claims description 10
- 102100038095 Protein-glutamine gamma-glutamyltransferase 2 Human genes 0.000 claims description 10
- 102100021269 Regulator of G-protein signaling 1 Human genes 0.000 claims description 10
- 101710140408 Regulator of G-protein signaling 1 Proteins 0.000 claims description 10
- 101150045565 Socs1 gene Proteins 0.000 claims description 10
- 101150043341 Socs3 gene Proteins 0.000 claims description 10
- 102000058018 Suppressor of Cytokine Signaling 1 Human genes 0.000 claims description 10
- 108700027336 Suppressor of Cytokine Signaling 1 Proteins 0.000 claims description 10
- 102000058015 Suppressor of Cytokine Signaling 3 Human genes 0.000 claims description 10
- 108700027337 Suppressor of Cytokine Signaling 3 Proteins 0.000 claims description 10
- 238000003753 real-time PCR Methods 0.000 claims description 10
- 150000003384 small molecules Chemical class 0.000 claims description 10
- 102100022278 Arachidonate 5-lipoxygenase-activating protein Human genes 0.000 claims description 9
- 101000755875 Homo sapiens Arachidonate 5-lipoxygenase-activating protein Proteins 0.000 claims description 9
- 108010051753 Spermidine Synthase Proteins 0.000 claims description 9
- 102100030413 Spermidine synthase Human genes 0.000 claims description 9
- 101000576894 Homo sapiens Macrophage mannose receptor 1 Proteins 0.000 claims description 8
- 108010071698 Spermine synthase Proteins 0.000 claims description 8
- 102100037616 Spermine synthase Human genes 0.000 claims description 8
- 230000001404 mediated effect Effects 0.000 claims description 8
- MWZMHLBLVDPOJE-UHFFFAOYSA-N 1-[4-[[n'-(4,6-dimethylpyrimidin-2-yl)carbamimidoyl]amino]phenyl]sulfonyl-3-phenylurea Chemical compound CC1=CC(C)=NC(N=C(N)NC=2C=CC(=CC=2)S(=O)(=O)NC(=O)NC=2C=CC=CC=2)=N1 MWZMHLBLVDPOJE-UHFFFAOYSA-N 0.000 claims description 7
- TZXKOCQBRNJULO-UHFFFAOYSA-N Ferriprox Chemical compound CC1=C(O)C(=O)C=CN1C TZXKOCQBRNJULO-UHFFFAOYSA-N 0.000 claims description 7
- 229960003266 deferiprone Drugs 0.000 claims description 7
- 210000003734 kidney Anatomy 0.000 claims description 7
- WZNJWVWKTVETCG-UHFFFAOYSA-N kojic acid Natural products OC(=O)C(N)CN1C=CC(=O)C(O)=C1 WZNJWVWKTVETCG-UHFFFAOYSA-N 0.000 claims description 7
- 229950002289 mimosine Drugs 0.000 claims description 7
- PWDYHMBTPGXCSN-VCBMUGGBSA-N n,n'-bis[3,5-bis[(e)-n-(diaminomethylideneamino)-c-methylcarbonimidoyl]phenyl]decanediamide Chemical compound NC(N)=N/N=C(\C)C1=CC(C(=N/N=C(N)N)/C)=CC(NC(=O)CCCCCCCCC(=O)NC=2C=C(C=C(C=2)C(\C)=N\N=C(N)N)C(\C)=N\N=C(N)N)=C1 PWDYHMBTPGXCSN-VCBMUGGBSA-N 0.000 claims description 7
- HUPNLWZFGMGNKB-UHFFFAOYSA-N 3-chloro-4,4-dimethyl-1,3-oxazolidin-2-one Chemical compound CC1(C)COC(=O)N1Cl HUPNLWZFGMGNKB-UHFFFAOYSA-N 0.000 claims description 6
- 206010016654 Fibrosis Diseases 0.000 claims description 6
- 230000004761 fibrosis Effects 0.000 claims description 6
- 229960002474 hydralazine Drugs 0.000 claims description 6
- 230000002265 prevention Effects 0.000 claims description 5
- 210000000056 organ Anatomy 0.000 claims description 4
- 208000026310 Breast neoplasm Diseases 0.000 claims description 3
- 208000035269 cancer or benign tumor Diseases 0.000 claims description 3
- 210000004185 liver Anatomy 0.000 claims description 3
- 210000004072 lung Anatomy 0.000 claims description 3
- 210000001672 ovary Anatomy 0.000 claims description 3
- 210000003491 skin Anatomy 0.000 claims description 3
- 201000003883 Cystic fibrosis Diseases 0.000 claims description 2
- 201000009794 Idiopathic Pulmonary Fibrosis Diseases 0.000 claims description 2
- 208000024934 IgG4-related mediastinitis Diseases 0.000 claims description 2
- 208000014919 IgG4-related retroperitoneal fibrosis Diseases 0.000 claims description 2
- 208000002260 Keloid Diseases 0.000 claims description 2
- 206010023330 Keloid scar Diseases 0.000 claims description 2
- 208000002805 Mediastinal fibrosis Diseases 0.000 claims description 2
- 208000003510 Nephrogenic Fibrosing Dermopathy Diseases 0.000 claims description 2
- 206010067467 Nephrogenic systemic fibrosis Diseases 0.000 claims description 2
- 206010038748 Restrictive cardiomyopathy Diseases 0.000 claims description 2
- 206010038979 Retroperitoneal fibrosis Diseases 0.000 claims description 2
- 206010039710 Scleroderma Diseases 0.000 claims description 2
- 230000001746 atrial effect Effects 0.000 claims description 2
- 230000009787 cardiac fibrosis Effects 0.000 claims description 2
- 208000020832 chronic kidney disease Diseases 0.000 claims description 2
- 208000019425 cirrhosis of liver Diseases 0.000 claims description 2
- 230000002124 endocrine Effects 0.000 claims description 2
- 201000010048 endomyocardial fibrosis Diseases 0.000 claims description 2
- 230000002496 gastric effect Effects 0.000 claims description 2
- 210000000777 hematopoietic system Anatomy 0.000 claims description 2
- 230000001969 hypertrophic effect Effects 0.000 claims description 2
- 208000036971 interstitial lung disease 2 Diseases 0.000 claims description 2
- 230000000968 intestinal effect Effects 0.000 claims description 2
- 208000028867 ischemia Diseases 0.000 claims description 2
- 210000001117 keloid Anatomy 0.000 claims description 2
- 230000001926 lymphatic effect Effects 0.000 claims description 2
- 206010028537 myelofibrosis Diseases 0.000 claims description 2
- 230000001537 neural effect Effects 0.000 claims description 2
- 210000000496 pancreas Anatomy 0.000 claims description 2
- 210000002307 prostate Anatomy 0.000 claims description 2
- 208000005069 pulmonary fibrosis Diseases 0.000 claims description 2
- 201000002793 renal fibrosis Diseases 0.000 claims description 2
- 201000000306 sarcoidosis Diseases 0.000 claims description 2
- 230000037390 scarring Effects 0.000 claims description 2
- 210000004291 uterus Anatomy 0.000 claims description 2
- 125000003729 nucleotide group Chemical group 0.000 description 114
- 239000002773 nucleotide Substances 0.000 description 112
- YAOAMZOGXBMLFQ-UHFFFAOYSA-N 2-(7-aminoheptyl)guanidine Chemical compound NCCCCCCCN=C(N)N YAOAMZOGXBMLFQ-UHFFFAOYSA-N 0.000 description 79
- 235000018102 proteins Nutrition 0.000 description 67
- 102000004388 Interleukin-4 Human genes 0.000 description 66
- 108090000978 Interleukin-4 Proteins 0.000 description 66
- 230000014509 gene expression Effects 0.000 description 61
- 102000004190 Enzymes Human genes 0.000 description 53
- 108090000790 Enzymes Proteins 0.000 description 53
- 108020004999 messenger RNA Proteins 0.000 description 48
- 238000002474 experimental method Methods 0.000 description 41
- 150000007523 nucleic acids Chemical class 0.000 description 41
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 40
- ATHGHQPFGPMSJY-UHFFFAOYSA-N spermidine Chemical compound NCCCCNCCCN ATHGHQPFGPMSJY-UHFFFAOYSA-N 0.000 description 39
- 229920000768 polyamine Polymers 0.000 description 31
- 230000015572 biosynthetic process Effects 0.000 description 28
- 230000001419 dependent effect Effects 0.000 description 27
- 239000003862 glucocorticoid Substances 0.000 description 27
- 229940037128 systemic glucocorticoids Drugs 0.000 description 27
- 239000002924 silencing RNA Substances 0.000 description 26
- 230000004102 tricarboxylic acid cycle Effects 0.000 description 26
- 210000001744 T-lymphocyte Anatomy 0.000 description 25
- 150000001413 amino acids Chemical class 0.000 description 25
- 230000037361 pathway Effects 0.000 description 25
- 230000000295 complement effect Effects 0.000 description 24
- 239000002243 precursor Substances 0.000 description 24
- 239000002207 metabolite Substances 0.000 description 22
- 108091028043 Nucleic acid sequence Proteins 0.000 description 21
- 108091034117 Oligonucleotide Proteins 0.000 description 20
- 241000282414 Homo sapiens Species 0.000 description 19
- 230000010627 oxidative phosphorylation Effects 0.000 description 19
- 229940063673 spermidine Drugs 0.000 description 19
- 230000000694 effects Effects 0.000 description 17
- 230000001965 increasing effect Effects 0.000 description 17
- 239000003112 inhibitor Substances 0.000 description 17
- 230000004060 metabolic process Effects 0.000 description 17
- 230000014616 translation Effects 0.000 description 17
- 229940024606 amino acid Drugs 0.000 description 16
- 235000001014 amino acid Nutrition 0.000 description 16
- 239000000074 antisense oligonucleotide Substances 0.000 description 16
- 238000012230 antisense oligonucleotides Methods 0.000 description 16
- 230000006870 function Effects 0.000 description 16
- BZUIJMCJNWUGKQ-BDAKNGLRSA-N hypusine Chemical compound NCC[C@@H](O)CNCCCC[C@H](N)C(O)=O BZUIJMCJNWUGKQ-BDAKNGLRSA-N 0.000 description 16
- 102000039446 nucleic acids Human genes 0.000 description 16
- 108020004707 nucleic acids Proteins 0.000 description 16
- 238000013519 translation Methods 0.000 description 16
- 239000000203 mixture Substances 0.000 description 15
- 108020004414 DNA Proteins 0.000 description 14
- 101000934372 Homo sapiens Macrosialin Proteins 0.000 description 14
- 102000003814 Interleukin-10 Human genes 0.000 description 14
- 108090000174 Interleukin-10 Proteins 0.000 description 14
- 102100025136 Macrosialin Human genes 0.000 description 14
- 238000005516 engineering process Methods 0.000 description 14
- 230000004048 modification Effects 0.000 description 14
- 238000012986 modification Methods 0.000 description 14
- 238000012360 testing method Methods 0.000 description 14
- 230000004069 differentiation Effects 0.000 description 13
- 239000013598 vector Substances 0.000 description 13
- 239000013604 expression vector Substances 0.000 description 12
- 238000001727 in vivo Methods 0.000 description 12
- 230000005764 inhibitory process Effects 0.000 description 12
- 108090000765 processed proteins & peptides Proteins 0.000 description 12
- KIDHWZJUCRJVML-UHFFFAOYSA-N putrescine Chemical compound NCCCCN KIDHWZJUCRJVML-UHFFFAOYSA-N 0.000 description 12
- 230000029058 respiratory gaseous exchange Effects 0.000 description 12
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 11
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 11
- 230000003247 decreasing effect Effects 0.000 description 11
- 238000000684 flow cytometry Methods 0.000 description 11
- 230000002503 metabolic effect Effects 0.000 description 11
- 210000001616 monocyte Anatomy 0.000 description 11
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 11
- 238000013518 transcription Methods 0.000 description 11
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 10
- 108010002350 Interleukin-2 Proteins 0.000 description 10
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 10
- 238000006243 chemical reaction Methods 0.000 description 10
- 238000000338 in vitro Methods 0.000 description 10
- UMJJGDUYVQCBMC-UHFFFAOYSA-N n-ethyl-n'-[3-[3-(ethylamino)propylamino]propyl]propane-1,3-diamine Chemical compound CCNCCCNCCCNCCCNCC UMJJGDUYVQCBMC-UHFFFAOYSA-N 0.000 description 10
- PFNFFQXMRSDOHW-UHFFFAOYSA-N spermine Chemical compound NCCCNCCCCNCCCN PFNFFQXMRSDOHW-UHFFFAOYSA-N 0.000 description 10
- 238000003786 synthesis reaction Methods 0.000 description 10
- 230000035897 transcription Effects 0.000 description 10
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 9
- 230000027721 electron transport chain Effects 0.000 description 9
- 239000008103 glucose Substances 0.000 description 9
- 230000002829 reductive effect Effects 0.000 description 9
- 230000001105 regulatory effect Effects 0.000 description 9
- 241001559542 Hippocampus hippocampus Species 0.000 description 8
- 102000019010 Methylmalonyl-CoA Mutase Human genes 0.000 description 8
- 108010051862 Methylmalonyl-CoA mutase Proteins 0.000 description 8
- 241000699670 Mus sp. Species 0.000 description 8
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 8
- UTJLXEIPEHZYQJ-UHFFFAOYSA-N Ornithine Natural products OC(=O)C(C)CCCN UTJLXEIPEHZYQJ-UHFFFAOYSA-N 0.000 description 8
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 8
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 8
- VLCYCQAOQCDTCN-UHFFFAOYSA-N eflornithine Chemical compound NCCCC(N)(C(F)F)C(O)=O VLCYCQAOQCDTCN-UHFFFAOYSA-N 0.000 description 8
- 229910052731 fluorine Inorganic materials 0.000 description 8
- 230000009368 gene silencing by RNA Effects 0.000 description 8
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 8
- 239000002502 liposome Substances 0.000 description 8
- 229960003104 ornithine Drugs 0.000 description 8
- 239000008194 pharmaceutical composition Substances 0.000 description 8
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 8
- 101001124991 Homo sapiens Nitric oxide synthase, inducible Proteins 0.000 description 7
- 102000013462 Interleukin-12 Human genes 0.000 description 7
- 108010065805 Interleukin-12 Proteins 0.000 description 7
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 7
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 7
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 7
- 102100029438 Nitric oxide synthase, inducible Human genes 0.000 description 7
- 229910052739 hydrogen Inorganic materials 0.000 description 7
- 210000002865 immune cell Anatomy 0.000 description 7
- 208000015181 infectious disease Diseases 0.000 description 7
- 230000002452 interceptive effect Effects 0.000 description 7
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 230000035755 proliferation Effects 0.000 description 7
- 238000011002 quantification Methods 0.000 description 7
- 230000004044 response Effects 0.000 description 7
- VRYALKFFQXWPIH-PBXRRBTRSA-N (3r,4s,5r)-3,4,5,6-tetrahydroxyhexanal Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)CC=O VRYALKFFQXWPIH-PBXRRBTRSA-N 0.000 description 6
- 241000894006 Bacteria Species 0.000 description 6
- 241000712899 Lymphocytic choriomeningitis mammarenavirus Species 0.000 description 6
- 229920000037 Polyproline Polymers 0.000 description 6
- 239000005700 Putrescine Substances 0.000 description 6
- 102000019259 Succinate Dehydrogenase Human genes 0.000 description 6
- 108010012901 Succinate Dehydrogenase Proteins 0.000 description 6
- PMMURAAUARKVCB-UHFFFAOYSA-N alpha-D-ara-dHexp Natural products OCC1OC(O)CC(O)C1O PMMURAAUARKVCB-UHFFFAOYSA-N 0.000 description 6
- 230000015556 catabolic process Effects 0.000 description 6
- 238000001514 detection method Methods 0.000 description 6
- 239000000975 dye Substances 0.000 description 6
- 239000012634 fragment Substances 0.000 description 6
- 230000030279 gene silencing Effects 0.000 description 6
- 230000034659 glycolysis Effects 0.000 description 6
- 238000011068 loading method Methods 0.000 description 6
- 210000004962 mammalian cell Anatomy 0.000 description 6
- 230000002438 mitochondrial effect Effects 0.000 description 6
- 230000006540 mitochondrial respiration Effects 0.000 description 6
- 108010026466 polyproline Proteins 0.000 description 6
- 230000008685 targeting Effects 0.000 description 6
- 241001430294 unidentified retrovirus Species 0.000 description 6
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 5
- 108090000193 Interleukin-1 beta Proteins 0.000 description 5
- 102000003777 Interleukin-1 beta Human genes 0.000 description 5
- 238000003559 RNA-seq method Methods 0.000 description 5
- 241000251131 Sphyrna Species 0.000 description 5
- 230000009471 action Effects 0.000 description 5
- 230000033228 biological regulation Effects 0.000 description 5
- 229910052799 carbon Inorganic materials 0.000 description 5
- 230000003197 catalytic effect Effects 0.000 description 5
- 230000001413 cellular effect Effects 0.000 description 5
- 238000009826 distribution Methods 0.000 description 5
- 101150074736 eif5a gene Proteins 0.000 description 5
- 239000003623 enhancer Substances 0.000 description 5
- 230000004907 flux Effects 0.000 description 5
- 238000003119 immunoblot Methods 0.000 description 5
- 230000001939 inductive effect Effects 0.000 description 5
- 210000003470 mitochondria Anatomy 0.000 description 5
- 229940063675 spermine Drugs 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- 230000001225 therapeutic effect Effects 0.000 description 5
- 238000010361 transduction Methods 0.000 description 5
- 230000026683 transduction Effects 0.000 description 5
- 230000003612 virological effect Effects 0.000 description 5
- BNYAARSFGNYTGE-UHFFFAOYSA-N 1-cyano-N-phenylmethanehydrazonoyl fluoride Chemical compound N#CC(F)=NNC1=CC=CC=C1 BNYAARSFGNYTGE-UHFFFAOYSA-N 0.000 description 4
- 238000011740 C57BL/6 mouse Methods 0.000 description 4
- 102000004127 Cytokines Human genes 0.000 description 4
- 108090000695 Cytokines Proteins 0.000 description 4
- 101150048270 DHPS gene Proteins 0.000 description 4
- 241000588724 Escherichia coli Species 0.000 description 4
- 101000963371 Homo sapiens Deoxyhypusine hydroxylase Proteins 0.000 description 4
- 101001041245 Homo sapiens Ornithine decarboxylase Proteins 0.000 description 4
- 101000701815 Homo sapiens Spermidine synthase Proteins 0.000 description 4
- 101000881206 Homo sapiens Spermine synthase Proteins 0.000 description 4
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 4
- 101000819111 Homo sapiens Trans-acting T-cell-specific transcription factor GATA-3 Proteins 0.000 description 4
- 102000012011 Isocitrate Dehydrogenase Human genes 0.000 description 4
- 108010075869 Isocitrate Dehydrogenase Proteins 0.000 description 4
- 102000043131 MHC class II family Human genes 0.000 description 4
- 108091054438 MHC class II family Proteins 0.000 description 4
- 241001529936 Murinae Species 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 108010058846 Ovalbumin Proteins 0.000 description 4
- 229930182555 Penicillin Natural products 0.000 description 4
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 4
- 108010026552 Proteome Proteins 0.000 description 4
- 239000012980 RPMI-1640 medium Substances 0.000 description 4
- 102000011929 Succinate-CoA Ligases Human genes 0.000 description 4
- 108010075728 Succinate-CoA Ligases Proteins 0.000 description 4
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 4
- 102100021386 Trans-acting T-cell-specific transcription factor GATA-3 Human genes 0.000 description 4
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 4
- 230000008901 benefit Effects 0.000 description 4
- 238000001574 biopsy Methods 0.000 description 4
- 230000037396 body weight Effects 0.000 description 4
- 210000004979 bone marrow derived macrophage Anatomy 0.000 description 4
- 239000004202 carbamide Substances 0.000 description 4
- 125000002091 cationic group Chemical group 0.000 description 4
- 238000005119 centrifugation Methods 0.000 description 4
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical group C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 4
- 210000003690 classically activated macrophage Anatomy 0.000 description 4
- 238000003776 cleavage reaction Methods 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- 238000006731 degradation reaction Methods 0.000 description 4
- 210000003527 eukaryotic cell Anatomy 0.000 description 4
- 239000012530 fluid Substances 0.000 description 4
- 239000007850 fluorescent dye Substances 0.000 description 4
- 239000000499 gel Substances 0.000 description 4
- 238000001114 immunoprecipitation Methods 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 238000003368 label free method Methods 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- 239000012139 lysis buffer Substances 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- 210000003071 memory t lymphocyte Anatomy 0.000 description 4
- 108091070501 miRNA Proteins 0.000 description 4
- 239000002679 microRNA Substances 0.000 description 4
- 229940049954 penicillin Drugs 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 239000013612 plasmid Substances 0.000 description 4
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 4
- 230000010076 replication Effects 0.000 description 4
- 238000011160 research Methods 0.000 description 4
- 238000012216 screening Methods 0.000 description 4
- 229960005322 streptomycin Drugs 0.000 description 4
- 230000008093 supporting effect Effects 0.000 description 4
- 230000002103 transcriptional effect Effects 0.000 description 4
- 230000009261 transgenic effect Effects 0.000 description 4
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 3
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 3
- 102000000452 Acetyl-CoA carboxylase Human genes 0.000 description 3
- 108010016219 Acetyl-CoA carboxylase Proteins 0.000 description 3
- 108010085238 Actins Proteins 0.000 description 3
- 108010018763 Biotin carboxylase Proteins 0.000 description 3
- 102100039868 Cytoplasmic aconitate hydratase Human genes 0.000 description 3
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 3
- 241000196324 Embryophyta Species 0.000 description 3
- 108091062183 EsiRNA Proteins 0.000 description 3
- 241000206602 Eukaryota Species 0.000 description 3
- 108700039691 Genetic Promoter Regions Proteins 0.000 description 3
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 3
- 241000238631 Hexapoda Species 0.000 description 3
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 3
- 206010061218 Inflammation Diseases 0.000 description 3
- 102100022338 Integrin alpha-M Human genes 0.000 description 3
- 102100037850 Interferon gamma Human genes 0.000 description 3
- 108010074328 Interferon-gamma Proteins 0.000 description 3
- 229930182816 L-glutamine Natural products 0.000 description 3
- 241001599018 Melanogaster Species 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 102000006404 Mitochondrial Proteins Human genes 0.000 description 3
- 108010058682 Mitochondrial Proteins Proteins 0.000 description 3
- 108010044843 Peptide Initiation Factors Proteins 0.000 description 3
- 102000005877 Peptide Initiation Factors Human genes 0.000 description 3
- 229920001213 Polysorbate 20 Polymers 0.000 description 3
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 3
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 3
- 230000006044 T cell activation Effects 0.000 description 3
- 230000006536 aerobic glycolysis Effects 0.000 description 3
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 230000005754 cellular signaling Effects 0.000 description 3
- 230000008859 change Effects 0.000 description 3
- 238000010367 cloning Methods 0.000 description 3
- 238000004163 cytometry Methods 0.000 description 3
- 238000000326 densiometry Methods 0.000 description 3
- PGPFBXMCOQNMJO-VIFPVBQESA-N deoxyhypusine Chemical compound NCCCCNCCCC[C@H](N)C(O)=O PGPFBXMCOQNMJO-VIFPVBQESA-N 0.000 description 3
- 230000003828 downregulation Effects 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- 210000003162 effector t lymphocyte Anatomy 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 238000009396 hybridization Methods 0.000 description 3
- 230000036039 immunity Effects 0.000 description 3
- 230000004054 inflammatory process Effects 0.000 description 3
- 239000000543 intermediate Substances 0.000 description 3
- PGHMRUGBZOYCAA-UHFFFAOYSA-N ionomycin Natural products O1C(CC(O)C(C)C(O)C(C)C=CCC(C)CC(C)C(O)=CC(=O)C(C)CC(C)CC(CCC(O)=O)C)CCC1(C)C1OC(C)(C(C)O)CC1 PGHMRUGBZOYCAA-UHFFFAOYSA-N 0.000 description 3
- PGHMRUGBZOYCAA-ADZNBVRBSA-N ionomycin Chemical compound O1[C@H](C[C@H](O)[C@H](C)[C@H](O)[C@H](C)/C=C/C[C@@H](C)C[C@@H](C)C(/O)=C/C(=O)[C@@H](C)C[C@@H](C)C[C@@H](CCC(O)=O)C)CC[C@@]1(C)[C@@H]1O[C@](C)([C@@H](C)O)CC1 PGHMRUGBZOYCAA-ADZNBVRBSA-N 0.000 description 3
- 210000003292 kidney cell Anatomy 0.000 description 3
- 239000006166 lysate Substances 0.000 description 3
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 210000004379 membrane Anatomy 0.000 description 3
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 3
- 239000011325 microbead Substances 0.000 description 3
- 230000006677 mitochondrial metabolism Effects 0.000 description 3
- 239000002105 nanoparticle Substances 0.000 description 3
- 229940092253 ovalbumin Drugs 0.000 description 3
- 230000036284 oxygen consumption Effects 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 3
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 102000004196 processed proteins & peptides Human genes 0.000 description 3
- 210000001236 prokaryotic cell Anatomy 0.000 description 3
- 230000001177 retroviral effect Effects 0.000 description 3
- 230000007017 scission Effects 0.000 description 3
- 210000002966 serum Anatomy 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 210000004988 splenocyte Anatomy 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 238000007619 statistical method Methods 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 238000012546 transfer Methods 0.000 description 3
- MNULEGDCPYONBU-WMBHJXFZSA-N (1r,4s,5e,5'r,6'r,7e,10s,11r,12s,14r,15s,16s,18r,19s,20r,21e,25s,26r,27s,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trio Polymers O([C@@H]1CC[C@@H](/C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)[C@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)O[C@H]([C@H]2C)[C@H]1C)CC)[C@]12CC[C@@H](C)[C@@H](C[C@H](C)O)O1 MNULEGDCPYONBU-WMBHJXFZSA-N 0.000 description 2
- MNULEGDCPYONBU-DJRUDOHVSA-N (1s,4r,5z,5'r,6'r,7e,10s,11r,12s,14r,15s,18r,19r,20s,21e,26r,27s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers O([C@H]1CC[C@H](\C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)C(C)C(=O)[C@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)OC([C@H]2C)C1C)CC)[C@]12CC[C@@H](C)[C@@H](CC(C)O)O1 MNULEGDCPYONBU-DJRUDOHVSA-N 0.000 description 2
- MNULEGDCPYONBU-YNZHUHFTSA-N (4Z,18Z,20Z)-22-ethyl-7,11,14,15-tetrahydroxy-6'-(2-hydroxypropyl)-5',6,8,10,12,14,16,28,29-nonamethylspiro[2,26-dioxabicyclo[23.3.1]nonacosa-4,18,20-triene-27,2'-oxane]-3,9,13-trione Polymers CC1C(C2C)OC(=O)\C=C/C(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)C\C=C/C=C\C(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-YNZHUHFTSA-N 0.000 description 2
- MNULEGDCPYONBU-VVXVDZGXSA-N (5e,5'r,7e,10s,11r,12s,14s,15r,16r,18r,19s,20r,21e,26r,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers C([C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)OC([C@H]1C)[C@H]2C)\C=C\C=C\C(CC)CCC2OC21CC[C@@H](C)C(C[C@H](C)O)O2 MNULEGDCPYONBU-VVXVDZGXSA-N 0.000 description 2
- KZKAYEGOIJEWQB-UHFFFAOYSA-N 1,3-dibromopropane;n,n,n',n'-tetramethylhexane-1,6-diamine Chemical compound BrCCCBr.CN(C)CCCCCCN(C)C KZKAYEGOIJEWQB-UHFFFAOYSA-N 0.000 description 2
- 108010030844 2-methylcitrate synthase Proteins 0.000 description 2
- 102100035923 4-aminobutyrate aminotransferase, mitochondrial Human genes 0.000 description 2
- MNULEGDCPYONBU-UHFFFAOYSA-N 4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers CC1C(C2C)OC(=O)C=CC(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)CC=CC=CC(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-UHFFFAOYSA-N 0.000 description 2
- 102100031126 6-phosphogluconolactonase Human genes 0.000 description 2
- 108010029731 6-phosphogluconolactonase Proteins 0.000 description 2
- 108010009924 Aconitate hydratase Proteins 0.000 description 2
- 102100022900 Actin, cytoplasmic 1 Human genes 0.000 description 2
- 102100022089 Acyl-[acyl-carrier-protein] hydrolase Human genes 0.000 description 2
- 101710191958 Amino-acid acetyltransferase Proteins 0.000 description 2
- 241000372033 Andromeda Species 0.000 description 2
- UIFFUZWRFRDZJC-UHFFFAOYSA-N Antimycin A1 Natural products CC1OC(=O)C(CCCCCC)C(OC(=O)CC(C)C)C(C)OC(=O)C1NC(=O)C1=CC=CC(NC=O)=C1O UIFFUZWRFRDZJC-UHFFFAOYSA-N 0.000 description 2
- NQWZLRAORXLWDN-UHFFFAOYSA-N Antimycin-A Natural products CCCCCCC(=O)OC1C(C)OC(=O)C(NC(=O)c2ccc(NC=O)cc2O)C(C)OC(=O)C1CCCC NQWZLRAORXLWDN-UHFFFAOYSA-N 0.000 description 2
- 102000009042 Argininosuccinate Lyase Human genes 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 206010006187 Breast cancer Diseases 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 2
- 108010051109 Cell-Penetrating Peptides Proteins 0.000 description 2
- 102000020313 Cell-Penetrating Peptides Human genes 0.000 description 2
- 108010071536 Citrate (Si)-synthase Proteins 0.000 description 2
- 102000006732 Citrate synthase Human genes 0.000 description 2
- 108020004394 Complementary RNA Proteins 0.000 description 2
- 241000701022 Cytomegalovirus Species 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 230000004544 DNA amplification Effects 0.000 description 2
- 102000028526 Dihydrolipoamide Dehydrogenase Human genes 0.000 description 2
- 108010028127 Dihydrolipoamide Dehydrogenase Proteins 0.000 description 2
- MUXOBHXGJLMRAB-UHFFFAOYSA-N Dimethyl succinate Chemical compound COC(=O)CCC(=O)OC MUXOBHXGJLMRAB-UHFFFAOYSA-N 0.000 description 2
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 2
- 108010039731 Fatty Acid Synthases Proteins 0.000 description 2
- 208000033962 Fontaine progeroid syndrome Diseases 0.000 description 2
- 108010018962 Glucosephosphate Dehydrogenase Proteins 0.000 description 2
- DHCLVCXQIBBOPH-UHFFFAOYSA-N Glycerol 2-phosphate Chemical compound OCC(CO)OP(O)(O)=O DHCLVCXQIBBOPH-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 208000036066 Hemophagocytic Lymphohistiocytosis Diseases 0.000 description 2
- 229920000209 Hexadimethrine bromide Polymers 0.000 description 2
- 101100116826 Homo sapiens DHPS gene Proteins 0.000 description 2
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 2
- 108090000144 Human Proteins Proteins 0.000 description 2
- 102000003839 Human Proteins Human genes 0.000 description 2
- 206010059176 Juvenile idiopathic arthritis Diseases 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 239000004472 Lysine Substances 0.000 description 2
- 101710199877 Malate dehydrogenase 2 Proteins 0.000 description 2
- 102100039742 Malate dehydrogenase, mitochondrial Human genes 0.000 description 2
- 101100260702 Mus musculus Tinagl1 gene Proteins 0.000 description 2
- 229930193140 Neomycin Natural products 0.000 description 2
- 101001083189 Neosartorya fumigata (strain ATCC MYA-4609 / Af293 / CBS 101355 / FGSC A1100) Hexokinase-1 Proteins 0.000 description 2
- 101710163270 Nuclease Proteins 0.000 description 2
- 101000840634 Oryza sativa subsp. japonica Hexokinase-5 Proteins 0.000 description 2
- 102000002508 Peptide Elongation Factors Human genes 0.000 description 2
- 108010068204 Peptide Elongation Factors Proteins 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 2
- 239000013614 RNA sample Substances 0.000 description 2
- 102000000574 RNA-Induced Silencing Complex Human genes 0.000 description 2
- 108010016790 RNA-Induced Silencing Complex Proteins 0.000 description 2
- 208000025747 Rheumatic disease Diseases 0.000 description 2
- 102000003661 Ribonuclease III Human genes 0.000 description 2
- 108010057163 Ribonuclease III Proteins 0.000 description 2
- 102000013968 STAT6 Transcription Factor Human genes 0.000 description 2
- 108010011005 STAT6 Transcription Factor Proteins 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 2
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 2
- 108010022394 Threonine synthase Proteins 0.000 description 2
- 108700029229 Transcriptional Regulatory Elements Proteins 0.000 description 2
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 2
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 230000004075 alteration Effects 0.000 description 2
- UIFFUZWRFRDZJC-SBOOETFBSA-N antimycin A Chemical compound C[C@H]1OC(=O)[C@H](CCCCCC)[C@@H](OC(=O)CC(C)C)[C@H](C)OC(=O)[C@H]1NC(=O)C1=CC=CC(NC=O)=C1O UIFFUZWRFRDZJC-SBOOETFBSA-N 0.000 description 2
- PVEVXUMVNWSNIG-UHFFFAOYSA-N antimycin A3 Natural products CC1OC(=O)C(CCCC)C(OC(=O)CC(C)C)C(C)OC(=O)C1NC(=O)C1=CC=CC(NC=O)=C1O PVEVXUMVNWSNIG-UHFFFAOYSA-N 0.000 description 2
- 101150088826 arg1 gene Proteins 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- 238000002869 basic local alignment search tool Methods 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 230000006696 biosynthetic metabolic pathway Effects 0.000 description 2
- 210000002798 bone marrow cell Anatomy 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 229960001714 calcium phosphate Drugs 0.000 description 2
- 229910000389 calcium phosphate Inorganic materials 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- 238000004422 calculation algorithm Methods 0.000 description 2
- 230000006860 carbon metabolism Effects 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 230000003915 cell function Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 210000002421 cell wall Anatomy 0.000 description 2
- 230000033077 cellular process Effects 0.000 description 2
- 238000012512 characterization method Methods 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 235000012000 cholesterol Nutrition 0.000 description 2
- 239000003184 complementary RNA Substances 0.000 description 2
- 238000010668 complexation reaction Methods 0.000 description 2
- 238000004624 confocal microscopy Methods 0.000 description 2
- 210000002808 connective tissue Anatomy 0.000 description 2
- 239000000356 contaminant Substances 0.000 description 2
- 210000004748 cultured cell Anatomy 0.000 description 2
- 238000012258 culturing Methods 0.000 description 2
- 230000007547 defect Effects 0.000 description 2
- 230000009699 differential effect Effects 0.000 description 2
- 102000004419 dihydrofolate reductase Human genes 0.000 description 2
- 230000003292 diminished effect Effects 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 230000006539 extracellular acidification Effects 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 230000002538 fungal effect Effects 0.000 description 2
- 238000002290 gas chromatography-mass spectrometry Methods 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 2
- 208000014752 hemophagocytic syndrome Diseases 0.000 description 2
- IPCSVZSSVZVIGE-UHFFFAOYSA-M hexadecanoate Chemical compound CCCCCCCCCCCCCCCC([O-])=O IPCSVZSSVZVIGE-UHFFFAOYSA-M 0.000 description 2
- 229950007870 hexadimethrine bromide Drugs 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 210000004408 hybridoma Anatomy 0.000 description 2
- 238000002649 immunization Methods 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 230000002757 inflammatory effect Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- BPHPUYQFMNQIOC-NXRLNHOXSA-N isopropyl beta-D-thiogalactopyranoside Chemical compound CC(C)S[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O BPHPUYQFMNQIOC-NXRLNHOXSA-N 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- 238000004949 mass spectrometry Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000037353 metabolic pathway Effects 0.000 description 2
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 2
- 238000010208 microarray analysis Methods 0.000 description 2
- 235000013336 milk Nutrition 0.000 description 2
- 239000008267 milk Substances 0.000 description 2
- 210000004080 milk Anatomy 0.000 description 2
- 210000003643 myeloid progenitor cell Anatomy 0.000 description 2
- 229960004927 neomycin Drugs 0.000 description 2
- 210000004940 nucleus Anatomy 0.000 description 2
- 230000009437 off-target effect Effects 0.000 description 2
- 229930191479 oligomycin Natural products 0.000 description 2
- MNULEGDCPYONBU-AWJDAWNUSA-N oligomycin A Polymers O([C@H]1CC[C@H](/C=C/C=C/C[C@@H](C)[C@H](O)[C@@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)O[C@@H]([C@@H]2C)[C@@H]1C)CC)[C@@]12CC[C@H](C)[C@H](C[C@@H](C)O)O1 MNULEGDCPYONBU-AWJDAWNUSA-N 0.000 description 2
- 150000002894 organic compounds Chemical class 0.000 description 2
- 230000008520 organization Effects 0.000 description 2
- 230000001590 oxidative effect Effects 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 244000052769 pathogen Species 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 210000002381 plasma Anatomy 0.000 description 2
- 239000013600 plasmid vector Substances 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 230000001124 posttranscriptional effect Effects 0.000 description 2
- 230000001323 posttranslational effect Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 230000000770 proinflammatory effect Effects 0.000 description 2
- 230000001737 promoting effect Effects 0.000 description 2
- 229950010131 puromycin Drugs 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 210000003705 ribosome Anatomy 0.000 description 2
- 229940080817 rotenone Drugs 0.000 description 2
- JUVIOZPCNVVQFO-UHFFFAOYSA-N rotenone Natural products O1C2=C3CC(C(C)=C)OC3=CC=C2C(=O)C2C1COC1=C2C=C(OC)C(OC)=C1 JUVIOZPCNVVQFO-UHFFFAOYSA-N 0.000 description 2
- 238000007493 shaping process Methods 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 210000000130 stem cell Anatomy 0.000 description 2
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 2
- 208000020408 systemic-onset juvenile idiopathic arthritis Diseases 0.000 description 2
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 2
- 238000004454 trace mineral analysis Methods 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 2
- 238000011144 upstream manufacturing Methods 0.000 description 2
- CALDMMCNNFPJSI-CRCLSJGQSA-N (3r,5s)-5-(hydroxymethyl)pyrrolidin-3-ol Chemical compound OC[C@@H]1C[C@@H](O)CN1 CALDMMCNNFPJSI-CRCLSJGQSA-N 0.000 description 1
- OEVXWWFLUUMMHL-UHFFFAOYSA-N 2-(2-hydroxy-5-methylphenyl)-4,5-dihydro-1,3-thiazole-4-carboxylic acid Chemical compound Cc1ccc(O)c(c1)C1=NC(CS1)C(O)=O OEVXWWFLUUMMHL-UHFFFAOYSA-N 0.000 description 1
- BHNQPLPANNDEGL-UHFFFAOYSA-N 2-(4-octylphenoxy)ethanol Chemical compound CCCCCCCCC1=CC=C(OCCO)C=C1 BHNQPLPANNDEGL-UHFFFAOYSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 1
- 101710094518 4-aminobutyrate aminotransferase Proteins 0.000 description 1
- 101150038502 ALDH2 gene Proteins 0.000 description 1
- 101150061635 ALDH6A1 gene Proteins 0.000 description 1
- 101150067361 Aars1 gene Proteins 0.000 description 1
- 101150040074 Aco2 gene Proteins 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 208000026326 Adult-onset Still disease Diseases 0.000 description 1
- 108091093088 Amplicon Proteins 0.000 description 1
- 101000678861 Arabidopsis thaliana Aconitate hydratase 1 Proteins 0.000 description 1
- 241000203069 Archaea Species 0.000 description 1
- 102000004452 Arginase Human genes 0.000 description 1
- 108700024123 Arginases Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 101100080292 Aspergillus oryzae (strain ATCC 42149 / RIB 40) pltp gene Proteins 0.000 description 1
- 101150096470 Atp5f1e gene Proteins 0.000 description 1
- 101150069123 Atp5mk gene Proteins 0.000 description 1
- 101150022891 BCKDHA gene Proteins 0.000 description 1
- 101150077604 BLMH gene Proteins 0.000 description 1
- 101000950981 Bacillus subtilis (strain 168) Catabolic NAD-specific glutamate dehydrogenase RocG Proteins 0.000 description 1
- 108091032955 Bacterial small RNA Proteins 0.000 description 1
- 206010060999 Benign neoplasm Diseases 0.000 description 1
- 108090000342 C-Type Lectins Proteins 0.000 description 1
- 102000003930 C-Type Lectins Human genes 0.000 description 1
- 101150065475 C1QA gene Proteins 0.000 description 1
- 101150084780 C1qb gene Proteins 0.000 description 1
- 101150104394 C1qc gene Proteins 0.000 description 1
- 101150107998 COQ9 gene Proteins 0.000 description 1
- 101150044301 CRYL1 gene Proteins 0.000 description 1
- 101150016254 CYP51A1 gene Proteins 0.000 description 1
- 101100094495 Caenorhabditis elegans mrps-23 gene Proteins 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 1
- 101150004620 Cebpb gene Proteins 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- 101150055030 Clybl gene Proteins 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- 102000013701 Cyclin-Dependent Kinase 4 Human genes 0.000 description 1
- 108010025464 Cyclin-Dependent Kinase 4 Proteins 0.000 description 1
- 102100038417 Cytoplasmic FMR1-interacting protein 1 Human genes 0.000 description 1
- KDXKERNSBIXSRK-RXMQYKEDSA-N D-lysine Chemical compound NCCCC[C@@H](N)C(O)=O KDXKERNSBIXSRK-RXMQYKEDSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 101150074155 DHFR gene Proteins 0.000 description 1
- 101150082833 DLGAP3 gene Proteins 0.000 description 1
- 108091008102 DNA aptamers Proteins 0.000 description 1
- 230000007067 DNA methylation Effects 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 101150092565 DOHH gene Proteins 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- 102100039487 Deoxyhypusine hydroxylase Human genes 0.000 description 1
- 102100031242 Deoxyhypusine synthase Human genes 0.000 description 1
- 101710086762 Diamine acetyltransferase 1 Proteins 0.000 description 1
- 102100034274 Diamine acetyltransferase 1 Human genes 0.000 description 1
- 102100024746 Dihydrofolate reductase Human genes 0.000 description 1
- 206010059866 Drug resistance Diseases 0.000 description 1
- ZGTMUACCHSMWAC-UHFFFAOYSA-L EDTA disodium salt (anhydrous) Chemical compound [Na+].[Na+].OC(=O)CN(CC([O-])=O)CCN(CC(O)=O)CC([O-])=O ZGTMUACCHSMWAC-UHFFFAOYSA-L 0.000 description 1
- 101150010120 EIF2B5 gene Proteins 0.000 description 1
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 1
- 101150027628 Eloa gene Proteins 0.000 description 1
- 108010067770 Endopeptidase K Proteins 0.000 description 1
- 102000002494 Endoribonucleases Human genes 0.000 description 1
- 108010093099 Endoribonucleases Proteins 0.000 description 1
- 101800001466 Envelope glycoprotein E1 Proteins 0.000 description 1
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- 102100026761 Eukaryotic translation initiation factor 5A-1 Human genes 0.000 description 1
- 101710126270 Eukaryotic translation initiation factor 5A-1 Proteins 0.000 description 1
- 102000008946 Fibrinogen Human genes 0.000 description 1
- 108010049003 Fibrinogen Proteins 0.000 description 1
- 229930091371 Fructose Natural products 0.000 description 1
- RFSUNEUAIZKAJO-ARQDHWQXSA-N Fructose Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O RFSUNEUAIZKAJO-ARQDHWQXSA-N 0.000 description 1
- 239000005715 Fructose Substances 0.000 description 1
- 101150019273 GATM gene Proteins 0.000 description 1
- 101150038604 GFM2 gene Proteins 0.000 description 1
- 101150037962 GOT1 gene Proteins 0.000 description 1
- 101150113130 Gdi1 gene Proteins 0.000 description 1
- 102000016901 Glutamate dehydrogenase Human genes 0.000 description 1
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 101150031411 H1-1 gene Proteins 0.000 description 1
- HVLSXIKZNLPZJJ-TXZCQADKSA-N HA peptide Chemical group C([C@@H](C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](C)C(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=C(O)C=C1 HVLSXIKZNLPZJJ-TXZCQADKSA-N 0.000 description 1
- 101150021598 HDHD2 gene Proteins 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 101150100579 Hadhb gene Proteins 0.000 description 1
- 241000724709 Hepatitis delta virus Species 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101001000686 Homo sapiens 4-aminobutyrate aminotransferase, mitochondrial Proteins 0.000 description 1
- 101000690301 Homo sapiens Aldo-keto reductase family 1 member C4 Proteins 0.000 description 1
- 101000745370 Homo sapiens Cytoplasmic aconitate hydratase Proteins 0.000 description 1
- 101000844963 Homo sapiens Deoxyhypusine synthase Proteins 0.000 description 1
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 1
- 101000976075 Homo sapiens Insulin Proteins 0.000 description 1
- 101000603420 Homo sapiens Nuclear pore complex-interacting protein family member A1 Proteins 0.000 description 1
- 101001116548 Homo sapiens Protein CBFA2T1 Proteins 0.000 description 1
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 1
- 208000028958 Hyperferritinemia Diseases 0.000 description 1
- 206010020648 Hyperkeratoses Diseases 0.000 description 1
- 206010051125 Hypofibrinogenaemia Diseases 0.000 description 1
- KDYRQBCJBKCHIQ-UHFFFAOYSA-N Hypusin Natural products NCC(O)CNCCCCC(N)C(=O)O KDYRQBCJBKCHIQ-UHFFFAOYSA-N 0.000 description 1
- 101150044523 ITGB3 gene Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 101710203526 Integrase Proteins 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 208000011200 Kawasaki disease Diseases 0.000 description 1
- 241000235058 Komagataella pastoris Species 0.000 description 1
- SNDPXSYFESPGGJ-BYPYZUCNSA-N L-2-aminopentanoic acid Chemical compound CCC[C@H](N)C(O)=O SNDPXSYFESPGGJ-BYPYZUCNSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- SNDPXSYFESPGGJ-UHFFFAOYSA-N L-norVal-OH Natural products CCCC(N)C(O)=O SNDPXSYFESPGGJ-UHFFFAOYSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- 101150065910 LGALS3BP gene Proteins 0.000 description 1
- 101150086890 LRPPRC gene Proteins 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- GDBQQVLCIARPGH-UHFFFAOYSA-N Leupeptin Natural products CC(C)CC(NC(C)=O)C(=O)NC(CC(C)C)C(=O)NC(C=O)CCCN=C(N)N GDBQQVLCIARPGH-UHFFFAOYSA-N 0.000 description 1
- 102000019298 Lipocalin Human genes 0.000 description 1
- 108050006654 Lipocalin Proteins 0.000 description 1
- 241000186779 Listeria monocytogenes Species 0.000 description 1
- 208000008771 Lymphadenopathy Diseases 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 102000043129 MHC class I family Human genes 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 101150004048 MIPEP gene Proteins 0.000 description 1
- 101150072835 MPST gene Proteins 0.000 description 1
- 102100028123 Macrophage colony-stimulating factor 1 Human genes 0.000 description 1
- 101710127797 Macrophage colony-stimulating factor 1 Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 101150039305 Mccc2 gene Proteins 0.000 description 1
- 101150061546 Mcrip1 gene Proteins 0.000 description 1
- GMPKIPWJBDOURN-UHFFFAOYSA-N Methoxyamine Chemical compound CON GMPKIPWJBDOURN-UHFFFAOYSA-N 0.000 description 1
- 101150101095 Mmp12 gene Proteins 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 101100121615 Mus musculus Arhgdia gene Proteins 0.000 description 1
- 101100058903 Mus musculus Ca13 gene Proteins 0.000 description 1
- 101100166612 Mus musculus Cd72 gene Proteins 0.000 description 1
- 101100276469 Mus musculus Cyfip1 gene Proteins 0.000 description 1
- 101100452019 Mus musculus Icam2 gene Proteins 0.000 description 1
- 101100111631 Mus musculus Naip2 gene Proteins 0.000 description 1
- 101001041236 Mus musculus Ornithine decarboxylase Proteins 0.000 description 1
- 101100242031 Mus musculus Pdha2 gene Proteins 0.000 description 1
- 101100190814 Mus musculus Pltp gene Proteins 0.000 description 1
- 101100533959 Mus musculus Serpinb6 gene Proteins 0.000 description 1
- 101150015113 Myl12b gene Proteins 0.000 description 1
- QRKUHYFDBWGLHJ-UHFFFAOYSA-N N-(tert-butyldimethylsilyl)-N-methyltrifluoroacetamide Chemical compound FC(F)(F)C(=O)N(C)[Si](C)(C)C(C)(C)C QRKUHYFDBWGLHJ-UHFFFAOYSA-N 0.000 description 1
- 101150115061 NDUFV2 gene Proteins 0.000 description 1
- 101100360525 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) mrpl-4 gene Proteins 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 101150106321 Nrp2 gene Proteins 0.000 description 1
- 101150113610 Nt5c3b gene Proteins 0.000 description 1
- 102000008297 Nuclear Matrix-Associated Proteins Human genes 0.000 description 1
- 108010035916 Nuclear Matrix-Associated Proteins Proteins 0.000 description 1
- 102100038845 Nuclear pore complex-interacting protein family member A1 Human genes 0.000 description 1
- 108091005461 Nucleic proteins Proteins 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 101150080351 P4ha1 gene Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 101150002582 PSPH gene Proteins 0.000 description 1
- 101150089545 PYCR3 gene Proteins 0.000 description 1
- 206010033661 Pancytopenia Diseases 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- 108010030544 Peptidyl-Lys metalloendopeptidase Proteins 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 241000286209 Phasianidae Species 0.000 description 1
- 241000233805 Phoenix Species 0.000 description 1
- 241000255969 Pieris brassicae Species 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- XBDQKXXYIPTUBI-UHFFFAOYSA-N Propionic acid Chemical compound CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 201000004681 Psoriasis Diseases 0.000 description 1
- 201000001263 Psoriatic Arthritis Diseases 0.000 description 1
- 208000036824 Psoriatic arthropathy Diseases 0.000 description 1
- LCTONWCANYUPML-UHFFFAOYSA-M Pyruvate Chemical compound CC(=O)C([O-])=O LCTONWCANYUPML-UHFFFAOYSA-M 0.000 description 1
- 108091008103 RNA aptamers Proteins 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 101100378744 Rattus norvegicus Aldh1a7 gene Proteins 0.000 description 1
- 101100237460 Rattus norvegicus Mgll gene Proteins 0.000 description 1
- 101100061105 Rattus norvegicus Mtco3 gene Proteins 0.000 description 1
- 101150003895 Rbm5 gene Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 101150042366 Rrp1 gene Proteins 0.000 description 1
- 101150051644 SDF4 gene Proteins 0.000 description 1
- 102000000395 SH3 domains Human genes 0.000 description 1
- 108050008861 SH3 domains Proteins 0.000 description 1
- 101150077666 SRA1 gene Proteins 0.000 description 1
- 108010044012 STAT1 Transcription Factor Proteins 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 241000293869 Salmonella enterica subsp. enterica serovar Typhimurium Species 0.000 description 1
- 101000833181 Schizosaccharomyces pombe (strain 972 / ATCC 24843) Glycerol dehydrogenase 1 Proteins 0.000 description 1
- 241000239226 Scorpiones Species 0.000 description 1
- 241000252141 Semionotiformes Species 0.000 description 1
- 102100023085 Serine/threonine-protein kinase mTOR Human genes 0.000 description 1
- 102100029904 Signal transducer and activator of transcription 1-alpha/beta Human genes 0.000 description 1
- 108091027568 Single-stranded nucleotide Proteins 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 101710181456 Spermidine N(1)-acetyltransferase Proteins 0.000 description 1
- 108091027076 Spiegelmer Proteins 0.000 description 1
- 241000256248 Spodoptera Species 0.000 description 1
- 101150017664 Spryd4 gene Proteins 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- 241000187747 Streptomyces Species 0.000 description 1
- 101150008632 Sucla2 gene Proteins 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 101150001932 TARS2 gene Proteins 0.000 description 1
- 101150002170 TBRG4 gene Proteins 0.000 description 1
- PZBFGYYEXUXCOF-UHFFFAOYSA-N TCEP Chemical compound OC(=O)CCP(CCC(O)=O)CCC(O)=O PZBFGYYEXUXCOF-UHFFFAOYSA-N 0.000 description 1
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 1
- 101150026786 TUFM gene Proteins 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- 102000002689 Toll-like receptor Human genes 0.000 description 1
- 108020000411 Toll-like receptor Proteins 0.000 description 1
- 101150107801 Top2a gene Proteins 0.000 description 1
- 108090000340 Transaminases Proteins 0.000 description 1
- 102000004338 Transferrin Human genes 0.000 description 1
- 108090000901 Transferrin Proteins 0.000 description 1
- 101710105083 Translation initiation factor 5A Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 101150002177 Txnip gene Proteins 0.000 description 1
- 101150014930 UFM1 gene Proteins 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 230000006682 Warburg effect Effects 0.000 description 1
- 101150060786 Wbp2 gene Proteins 0.000 description 1
- 238000002679 ablation Methods 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- TXUZVZSFRXZGTL-QPLCGJKRSA-N afimoxifene Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=C(O)C=C1 TXUZVZSFRXZGTL-QPLCGJKRSA-N 0.000 description 1
- 101150004762 aldh7a1 gene Proteins 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 230000030741 antigen processing and presentation Effects 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 101150014732 asnS gene Proteins 0.000 description 1
- 229940009098 aspartate Drugs 0.000 description 1
- 230000003190 augmentative effect Effects 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 230000033590 base-excision repair Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000001851 biosynthetic effect Effects 0.000 description 1
- OWMVSZAMULFTJU-UHFFFAOYSA-N bis-tris Chemical compound OCCN(CCO)C(CO)(CO)CO OWMVSZAMULFTJU-UHFFFAOYSA-N 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 201000008274 breast adenocarcinoma Diseases 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 235000011089 carbon dioxide Nutrition 0.000 description 1
- 230000021523 carboxylation Effects 0.000 description 1
- 238000006473 carboxylation reaction Methods 0.000 description 1
- 238000010366 cell biology technique Methods 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 238000001023 centrifugal evaporation Methods 0.000 description 1
- OEUUFNIKLCFNLN-LLVKDONJSA-N chembl432481 Chemical compound OC(=O)[C@@]1(C)CSC(C=2C(=CC(O)=CC=2)O)=N1 OEUUFNIKLCFNLN-LLVKDONJSA-N 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 238000013375 chromatographic separation Methods 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 101150116749 chuk gene Proteins 0.000 description 1
- 238000012761 co-transfection Methods 0.000 description 1
- 239000005515 coenzyme Substances 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 230000001143 conditioned effect Effects 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 101150060251 coq8b gene Proteins 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 238000012350 deep sequencing Methods 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 230000000994 depressogenic effect Effects 0.000 description 1
- 238000011033 desalting Methods 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 239000000747 designer drug Substances 0.000 description 1
- WQZGKKKJIJFFOK-UKLRSMCWSA-N dextrose-2-13c Chemical compound OC[C@H]1OC(O)[13C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-UKLRSMCWSA-N 0.000 description 1
- 238000002059 diagnostic imaging Methods 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 230000007120 differential activation Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 108020001096 dihydrofolate reductase Proteins 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 208000009190 disseminated intravascular coagulation Diseases 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 238000006911 enzymatic reaction Methods 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 1
- 108010085279 eukaryotic translation initiation factor 5A Proteins 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 230000004133 fatty acid degradation Effects 0.000 description 1
- 230000004129 fatty acid metabolism Effects 0.000 description 1
- 230000004136 fatty acid synthesis Effects 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 229940012952 fibrinogen Drugs 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 229940050411 fumarate Drugs 0.000 description 1
- VZCYOOQTPOCHFL-OWOJBTEDSA-L fumarate(2-) Chemical compound [O-]C(=O)\C=C\C([O-])=O VZCYOOQTPOCHFL-OWOJBTEDSA-L 0.000 description 1
- 239000005350 fused silica glass Substances 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 108091008053 gene clusters Proteins 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 230000004110 gluconeogenesis Effects 0.000 description 1
- 229930195712 glutamate Natural products 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- 150000002337 glycosamines Chemical class 0.000 description 1
- HHLFWLYXYJOTON-UHFFFAOYSA-N glyoxylic acid Chemical compound OC(=O)C=O HHLFWLYXYJOTON-UHFFFAOYSA-N 0.000 description 1
- 238000011194 good manufacturing practice Methods 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 206010019847 hepatosplenomegaly Diseases 0.000 description 1
- 239000008241 heterogeneous mixture Substances 0.000 description 1
- 208000021760 high fever Diseases 0.000 description 1
- 238000012203 high throughput assay Methods 0.000 description 1
- 238000010842 high-capacity cDNA reverse transcription kit Methods 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 102000054751 human RUNX1T1 Human genes 0.000 description 1
- 238000002013 hydrophilic interaction chromatography Methods 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 230000033444 hydroxylation Effects 0.000 description 1
- 238000005805 hydroxylation reaction Methods 0.000 description 1
- 208000006575 hypertriglyceridemia Diseases 0.000 description 1
- 230000003832 immune regulation Effects 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000012212 insulator Substances 0.000 description 1
- PBGKTOXHQIOBKM-FHFVDXKLSA-N insulin (human) Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 PBGKTOXHQIOBKM-FHFVDXKLSA-N 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000009830 intercalation Methods 0.000 description 1
- 210000000936 intestine Anatomy 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- 229930027917 kanamycin Natural products 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- SBUJHOSQTJFQJX-NOAMYHISSA-N kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 1
- 229930182823 kanamycin A Natural products 0.000 description 1
- 150000002597 lactoses Chemical class 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- GDBQQVLCIARPGH-ULQDDVLXSA-N leupeptin Chemical compound CC(C)C[C@H](NC(C)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C=O)CCCN=C(N)N GDBQQVLCIARPGH-ULQDDVLXSA-N 0.000 description 1
- 108010052968 leupeptin Proteins 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000008206 lipophilic material Substances 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 208000019423 liver disease Diseases 0.000 description 1
- 230000005976 liver dysfunction Effects 0.000 description 1
- 239000012160 loading buffer Substances 0.000 description 1
- 208000018555 lymphatic system disease Diseases 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 230000002132 lysosomal effect Effects 0.000 description 1
- 238000007403 mPCR Methods 0.000 description 1
- 229940049920 malate Drugs 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-L malate(2-) Chemical compound [O-]C(=O)C(O)CC([O-])=O BJEPYKJPYRNKOW-UHFFFAOYSA-L 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 238000002493 microarray Methods 0.000 description 1
- 230000003641 microbiacidal effect Effects 0.000 description 1
- 239000013586 microbial product Substances 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 230000005787 mitochondrial ATP synthesis coupled electron transport Effects 0.000 description 1
- 230000025608 mitochondrion localization Effects 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 101150075875 mrpl24 gene Proteins 0.000 description 1
- 101150118594 mrps-5 gene Proteins 0.000 description 1
- 208000001725 mucocutaneous lymph node syndrome Diseases 0.000 description 1
- 210000001167 myeloblast Anatomy 0.000 description 1
- 229940105132 myristate Drugs 0.000 description 1
- 238000010844 nanoflow liquid chromatography Methods 0.000 description 1
- 101150025238 ndufa9 gene Proteins 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 230000007959 normoxia Effects 0.000 description 1
- 210000000299 nuclear matrix Anatomy 0.000 description 1
- 108091008104 nucleic acid aptamers Proteins 0.000 description 1
- 230000030648 nucleus localization Effects 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 230000000242 pagocytic effect Effects 0.000 description 1
- 244000045947 parasite Species 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 238000003068 pathway analysis Methods 0.000 description 1
- 239000013610 patient sample Substances 0.000 description 1
- 101150028177 pdhB gene Proteins 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 210000004976 peripheral blood cell Anatomy 0.000 description 1
- 210000001322 periplasm Anatomy 0.000 description 1
- 210000003200 peritoneal cavity Anatomy 0.000 description 1
- 210000003024 peritoneal macrophage Anatomy 0.000 description 1
- 210000004303 peritoneum Anatomy 0.000 description 1
- 210000002824 peroxisome Anatomy 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 230000009038 pharmacological inhibition Effects 0.000 description 1
- CPJSUEIXXCENMM-UHFFFAOYSA-N phenacetin Chemical compound CCOC1=CC=C(NC(C)=O)C=C1 CPJSUEIXXCENMM-UHFFFAOYSA-N 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 125000002467 phosphate group Chemical group [H]OP(=O)(O[H])O[*] 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 210000004986 primary T-cell Anatomy 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000002206 pro-fibrotic effect Effects 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 239000003531 protein hydrolysate Substances 0.000 description 1
- 238000000575 proteomic method Methods 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 description 1
- 150000003230 pyrimidines Chemical class 0.000 description 1
- 229940076788 pyruvate Drugs 0.000 description 1
- 239000003642 reactive oxygen metabolite Substances 0.000 description 1
- 238000003259 recombinant expression Methods 0.000 description 1
- 238000002271 resection Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000004202 respiratory function Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 238000002473 ribonucleic acid immunoprecipitation Methods 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 125000000548 ribosyl group Chemical group C1([C@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- 101150108347 sdhB gene Proteins 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 238000004062 sedimentation Methods 0.000 description 1
- 239000006152 selective media Substances 0.000 description 1
- 229950011005 semapimod Drugs 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- FQENQNTWSFEDLI-UHFFFAOYSA-J sodium diphosphate Chemical compound [Na+].[Na+].[Na+].[Na+].[O-]P([O-])(=O)OP([O-])([O-])=O FQENQNTWSFEDLI-UHFFFAOYSA-J 0.000 description 1
- 229940048086 sodium pyrophosphate Drugs 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- 238000009987 spinning Methods 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 238000011105 stabilization Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 230000004960 subcellular localization Effects 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-L succinate(2-) Chemical compound [O-]C(=O)CCC([O-])=O KDYFGRWQOYBRFD-UHFFFAOYSA-L 0.000 description 1
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- WWJZWCUNLNYYAU-UHFFFAOYSA-N temephos Chemical compound C1=CC(OP(=S)(OC)OC)=CC=C1SC1=CC=C(OP(=S)(OC)OC)C=C1 WWJZWCUNLNYYAU-UHFFFAOYSA-N 0.000 description 1
- BCNZYOJHNLTNEZ-UHFFFAOYSA-N tert-butyldimethylsilyl chloride Chemical compound CC(C)(C)[Si](C)(C)Cl BCNZYOJHNLTNEZ-UHFFFAOYSA-N 0.000 description 1
- 210000001550 testis Anatomy 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 235000019818 tetrasodium diphosphate Nutrition 0.000 description 1
- 239000001577 tetrasodium phosphonato phosphate Substances 0.000 description 1
- 230000007838 tissue remodeling Effects 0.000 description 1
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 1
- 102000014898 transaminase activity proteins Human genes 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 239000012581 transferrin Substances 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 230000014621 translational initiation Effects 0.000 description 1
- 150000003628 tricarboxylic acids Chemical class 0.000 description 1
- 238000009966 trimming Methods 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 238000001195 ultra high performance liquid chromatography Methods 0.000 description 1
- 238000004704 ultra performance liquid chromatography Methods 0.000 description 1
- 241000712461 unidentified influenza virus Species 0.000 description 1
- 150000004670 unsaturated fatty acids Chemical class 0.000 description 1
- 235000021122 unsaturated fatty acids Nutrition 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 239000000277 virosome Substances 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 101150024173 wdr44 gene Proteins 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
- VLCYCQAOQCDTCN-ZCFIWIBFSA-N α-difluoromethylornithine Chemical compound NCCC[C@@](N)(C(F)F)C(O)=O VLCYCQAOQCDTCN-ZCFIWIBFSA-N 0.000 description 1
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5044—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
- G01N33/5047—Cells of the immune system
- G01N33/5055—Cells of the immune system involving macrophages
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5044—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
- G01N33/5073—Stem cells
Definitions
- the present invention relates to a method for determining whether a compound has the capability of inhibiting elF5a activation, comprising (a) contacting a sample comprising cells being capable of differentiating into M2 macrophages with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and (b) quantifying in the sample the cells having a M2-phenotype, wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting el F5a activation.
- the polyamine spermidine is needed to hypusinate the eukaryotic translation elongation factor 5A (elF5A) 1 .
- the polyamine family of molecules is comprised of small polycationic metabolites, the most well described being putrescine, spermidine and spermine. They are formed by a series of enzymatic reactions that begin with the conversion of the amino acid ornithine to putrescine by ornithine decarboxylase (ODC) (figure 1 ).
- ODC ornithine decarboxylase
- a notable feature of the polyamine pathway is its role in the activation of the translation elongation factor elF5a.
- elF5a is an essential cellular protein required for the production of a certain subset of proteins. Its function is dependent on the two enzymes deoxyhypusine hydroxylase (DOHH) and deoxyhypusine synthase (DHPS) that use the metabolite spermidine to mediate the activation of elF5a.
- DOHH deoxyhypusine hydroxylase
- DHPS deoxyhypusine synthase
- the present invention aims at providing novel screening methods and therapeutic uses encompassing to interfere with the activation of elF5a.
- the present invention therefore relates in a first aspect to a method for determining whether a compound has the capability of inhibiting elF5a activation, comprising (a) contacting a sample comprising cells being capable of differentiating into M2 macrophages with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and (b) quantifying in the sample the cells having a M2-phenotype, wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting elF5a activation
- elF5a refers to the protein eukaryotic translation initiation factor 5A-1 that is encoded by the EIF5A gene.
- the compound may interfere with any step in the polyamine pathway and the subsequent hypusine-elF5a formation as shown in Figures 1 and 2 as all these steps are required for the final activation of elF5a.
- activated elF5a is hypusine-elF5a.
- Hypusine is the amino acid N (e)-(4-amino-2-hydroxybutyl)-lysine.
- the compound having the capability of inhibiting elF5a activation may interfere with any one of the precursors of hypusine-elF5a, e.g. spermidine or deoxyhypusine-elF5a, so that hypusine-elF5a cannot be generated as one or more of the required precursors are absent or not processible.
- the compound having the capability of inhibiting elF5a activation may also interfere with one or more of the enzymes in the pathway, e.g.
- the method of the invention may be performed in high through-put format for testing several compounds at the same time.
- High-throughput methods are capable of screening up to several thousand compounds in parallel.
- High-throughput assays independently of being biochemical, cellular or other assays, generally may be performed in wells of microtiter plates, wherein each plate may contain 96, 384 or 1536 wells. Handling of the plates, including incubation at temperatures other than ambient temperature, and bringing into contact of test compounds with the assay mixture is preferably effected by one or more computer-controlled robotic systems including pipetting devices.
- mixtures of, for example 10, 20, 30, 40, 50 or 100 test compounds may be added to each well.
- said mixture of test compounds may be deconvolved to identify the one or more test compounds in said mixture giving rise to the inhibition of elF5a activation.
- the compound having the capability of inhibiting elF5a activation may either act on the DNA level (e.g. a siRNA against the mRNA encoding DHS or DOHH) or on the protein or precursor level (e.g. an antibody against the DHS or DOHH enzyme or an antibody against spermidine). If follows that the compound having the capability of inhibiting elF5a activation may be (i) a compound inhibiting the expression of an mRNA encoding an enzyme being required for the activation of elF5a, or (ii) a compound inhibiting an enzyme or a precursor being required for the activation of elF5a.
- Compounds under option (i) include compounds interfering with the transcriptional machinery and/or its interaction with the promoter of said gene and/or with expression control elements remote from the promoter such as enhancers.
- Compounds under option (i) also include compounds interfering with the translational machinery.
- the compound inhibiting the expression of a mRNA encoding an enzyme being required for the activation of elF5a inhibits the expression and/or translation of said mRNA, for example, by specifically interfering with the promoter region controlling the expression of the mRNA or leading to the silencing of the mRNA.
- the transcription and/or translation of the mRNA is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98% and most preferred by about 100% (e.g., as compared to the same experimental set up in the absence of the compound).
- the compound preferably specifically inhibits the activity of the enzyme or precursor.
- the activity of the enzyme is its capability of catalyzing a chemical reaction being involved in the pathway leading to hypsuine-elF5a.
- the activity of the precursor is that it can be processed into another precursor further upstream in the pathway of elF5a activation or hypsuine-elF5a itself.
- the activity of the enzyme or precursor is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98%, and most preferably about 100% (e.g., as compared to the same experimental set up in the absence of the compound).
- the capability of the compound of inhibiting elF5a is determined by comparing the amount of cells having an M2-phenotype in the presence of the compound to that in the absence of the compound. In case the amount of cells having an M2-phenotype is reduced in the presence of the compound the compound is capable of inhibiting elF5a activation.
- a compound is preferably qualified as being capable of inhibiting elF5a activation in case the amount of cells having a M2-phenotype is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98% and most preferred by about 100% (e.g., as compared to the same experimental set up in the absence of the compound).
- a sample comprising cells being capable of differentiating into M2 macrophages is contacted with the test compound.
- a cell being capable of differentiating into M2 macrophages may be a precursor cell of a M2 macrophage.
- Preferred examples are hemoblasts, common myeloid progenitor cells, myeloblasts and monocytes.
- the most preferred precursor cells of macrophages are monocytes or other progenitor cells derived from the bone marrow, such as the common myeloid progenitor, that can differentiate into macrophages under the control of factors such as colony stimulating factor 1 (CFS-1 , otherwise known as macrophage-colony stimulating factor 1 ).
- CFS-1 colony stimulating factor 1
- Monocytes are the direct precursor cells of macrophages in the hematopoietic cell lineage.
- unactivated macrophages i.e. M0 cells
- M2 macrophages e.g. when cultured in the presence of IL-4.
- M1 and M2 macrophage activation represent two possible, distinct fates that unactivated macrophages can differentiate into.
- the sample comprising cells being capable of differentiating into M2 macrophages preferably comprises unactivated macrophages (MO macropahges) and/or monocytes.
- the sample can be a body sample or in vitro cultured cells.
- Preferred body samples are a tissue sample (e.g. bone marrow derived macrophages or organ biopsy), including a tumor sample derived from a biopsy or resection, a blood sample (e.g. whole blood, plasma or serum), spinal fluid, and fluid from bronchoalveolar lavage.
- Non-limiting examples of in vitro cultured cells are the human macrophage or monocytic cell lines THP-1 (ATCC TIB-202), U937 (ATCC CRL-1593.2), KG-1 (KG-1 ATCC CCL-246), MV-4-11 (ATCC CRL-959), SC (ATCC CRL-9855), MD (ATCC CRL-9850), Mono Mac 6 (DSM ACC 124) or hMo-PB cells (commercially available human "untouched" monocytes isolated from fresh peripheral blood, Cat.No. CSC-C1671 ).
- the M1 macrophage phenotype is characterized by the production of high levels of pro-inflammatory cytokines, an ability to mediate resistance to pathogens, strong microbicidal properties, high production of reactive nitrogen and oxygen intermediates, and promotion of Th1 responses.
- M2 macrophages are characterized by their involvement in parasite control, tissue remodeling, immune regulation, tumor promotion and efficient phagocytic activity.
- the sample comprising cells being capable of differentiating into M2 macrophages is cultured under conditions that allow for the differentiation of the precursor cells into M2- type macrophages in the absence of the compound. Conditions that can be used in order to differentiate monocytes and/or macrophages into M2 macrophages are known in the art.
- Preferred examples are the differentiation of monocytes and/or unactivated macrophages (MO cells) into M2 macrophages with (i) IL-4, (ii) IL-10, (iii) glucocorticoids (GC), or (iv) GC plus TGF-beta plus.
- a more preferred example is the differentiation of monocytes and/or unactivated macrophages (MO cells) into M2 macrophages with IL- 4, preferably in conjunction with M-CSF.
- IL-4 at a concentration of about 10ng/mL and, if present, M-CSF at a concentration of about 20ng/m.
- Macrophages designates a cell of myeloid origin. Macrophages are large white blood cells, occurring principally in connective tissue and in the bloodstream. They ingest foreign particles and infectious microorganisms by phagocytosis andhave the capacity for antigen presentation. Macrophages are classified in the art into MO, M1 and M2 macrophages.
- MO macrophages are also termed unactivated macrophages. They express the classical macrophage markers CD11b and CD68 but exhibit low expression of the activation markers MHC class II molecules, CD80, and CD86. They are moreover negative for cytokine expression in the form of IL-12, IL-10 and IL-1 beta.
- a MO cell is preferably a cell expressing CD11b and CD68 but not expressing IL-12, IL-10 and IL-1 beta. More preferably, a MO cell is a cell expressing CD11b and CD68 but not expressing IL-12, IL-10 and IL-1 beta, and expressing low amounts of MHC class II molecules, CD80, and CD86.
- a M1 macrophage is defined herein as a macrophage that was activated in the presence of a bacterium, virus, or a bacterial or viral compound (such as and preferably LPS) and optionally also in the presence of IFN-gamma. Accordingly, these cells can be denoted as M(LPS) or M(LPS+IFN) macrophages. M(LPS) and M(LPS+IFN) macrophages express the classical macrophage markers CD68 and CD11 b. Moreover these cells express - in contrast to MO cells - high amounts of MHC class II molecules, CD80 and CD86 expression. These cells also express the cytokines IL-1 beta, IL-12, and TNFalpha.
- a M1 cell is preferably a cell expressing CD11 b and CD68, IL-1beta, IL-12, and TNFalpha. More preferably, a M1 cell is a cell expressing CD11 b, CD68, IL-12, IL-10 and IL-1 beta and expressing high amounts of MHC class II molecules, CD80, and CD86.
- a M2 cell is also designated as alternatively activated macrophage in the art and hence as a macrophage that was activated in the absence of a bacterium, a virus, or a bacterial or viral compound (such as and preferably LPS) and IFN-gamma.
- a M2 macrophage as used herein refers to a macrophage cell that was activated in the presence of a compound selected from IL-4, IL-10, glucocorticoids (GC),and GC plus TGF-beta plus, or any combination of these compounds.
- M2 cells are also designated M(IL-4), M(IL-10), M(GC) or M(GC+TGF-beta) cells.
- the M2 cells herein are preferably M(IL-4), M(IL-10), M(GC) or M(GC+TGF-beta) cells and are most preferably M(IL- 4) cells.
- M(IL-4) cells express CD68, GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, but do not express MARCO and CD163.
- M(IL-10) cells express CD68, SOCS3 and IL-4Ra.
- M(GC) cells express CD68, CD163, STAB1 , MARCO, TGFBR2, ADORA3.
- M(GC+TGF-beta) cells express CD68, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, IL17RB.
- M(IL-4), M(IL-10), M(GC) and M(GC+TGF-beta) cells are preferably identified by the presence or absence of at least three of the respective markers listed in Table 1.
- M(IL-10) cells are preferably identified by the presence of CD68, SOCS3 and IL- 4Ra.
- M(IL-4)) cells are, for example, preferably identified by the presence of CCL17 and CCL18, and the absence of MARCO.
- M(IL-4), M(GC) and M(GC+TGF-beta) cells are more preferably identified by the presence or absence of at least six of the markers listed in Table 1.
- (M(GC)) cells are more preferably identified by the presence of CD68, CD163, STAB1 , MARCO, TGFBR2, ADORA3.
- (M(IL-4)) cells are, for example, preferably identified by the presence of MRC1 , STAB1 , F13A1 , TGFB1 and MMP12, and the absence of CD163.
- (M(GC+TGF)) cells are most preferably identified by the presence of CD68, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, IL17RB.
- M(GC+TGF) cells are most preferably identified by the presence of CD68, GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15 and CD200R, and the absence of CD163 and MARCO.
- CD206 is a general marker of M2 cells in vivo.
- Hypusination of elF5A is a conserved process critical for TCA (i.e. tricarboxylic acid) cycle integrity and mitochondrial respiration.
- Hypusinated elF5A enables translation of a subset of TCA cycle and mitochondrial electron transport chain (ETC) enzymes that mediate stable TCA cycle flux and oxidative phosphorylation (OXPHOS).
- ETC mitochondrial electron transport chain
- OXPHOS oxidative phosphorylation
- Inhibition of deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH) the enzymes that hypusinate elF5A 2,3 , leads to a break in the TCA cycle that is defined by the loss of elF5AH-regulated enzymes and associated metabolic flux into the TCA cycle.
- DHPS deoxyhypusine synthase
- DOHH deoxyhypusine hydroxylase
- elF5AH facilitates efficient production of proteins with difficult to translate mitochondrial targeting sequences (MTS), an activity that is consistent with a known function of elF5AH in overcoming ribosome stalling 4,5 .
- MMS mitochondrial targeting sequences
- elF5a regulates the differential activation of macrophages (M2 cells), a cell type whose divergent functional fates are defined by a metabolic switch between respiration and glycolysis 6 .
- quantifying the cells having a M2-phenotype comprises measuring the amounts of at least one marker molecule selected from the group consisting of GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB.
- marker molecule selected from the group consisting of GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGF
- M2 macrophages are phenotypically characterized by the expression of a specific set of markers.
- markers are GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB.
- One or more of these markers can be used in order to quantify the cells having a M2-phenotype is step (b) of the method of the invention.
- Using more than one cell surface markers may increase the reliability of determining the cells having a M2-phenotype. With increasing preference at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 and at least 10 markers are measured.
- the specific set of cell surface markers on M2 macrophages depends on the conditions used to allow for the differentiation into M2 macrophages.
- IL-4 GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15 and CD200R are expressed as markers.
- step (b) quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15 and CD200R.
- at least 2 at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 and at least 10 of these markers are measured. Most preferably all of these markers are measured.
- step (a) of the claimed method comprise IL-10 quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of SOCS3 and IL-4Ra. More preferably both of these markers are measured.
- step (a) of the claimed method comprise GC quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of CD163, STAB1 , MARCO, TGFBR2 and ADORA3. With increasing preference at least 2, at least 3 and at least 4 of these markers are measured. Most preferably all five of these markers are measured.
- step (a) of the claimed method comprise GC plus TGF-beta quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP and IL17RB. With increasing preference at least 2, at least 3, at least 4 and at least 5 of these markers are measured. Most preferably all six of these markers are measured.
- the absence of a particular marker can further help to define cells having a M2-phenotype.
- Non-limiting examples are phosphorylated STAT1 and IL-12.
- IL-4 is used in the conditions allowing for the differentiation into M2 macrophages furthermore the absence of CD163 and MARCO can further help to define cells having a M2-phenotype.
- quantifying the cells having a M2-phenotype comprises flow cytometry analysis, quantitative PCR, and/or western blot analysis.
- the skilled person is aware of several methods being suitable for quantifying cells having a M2-phenotype. Non-limiting preferred examples of such methods are cytometry analysis, quantitative PCR, and/or western blot analysis. These methods are employed depending on whether nucleic acid or protein is measured.
- Quantitative PCR qPCR or real-time PCR or RT-PCR uses the linearity of DNA amplification to determine absolute or relative amounts of a known sequence in a sample. By using a fluorescent reporter in the reaction, it is possible to measure DNA generation.
- Quantitative PCR DNA amplification may be monitored at each cycle of PCR. Quantitative PCR also lends itself to relative studies. A reaction may be performed using primers unique to each region to be amplified and tagged with different fluorescent dyes. Several commercially available quantitative thermal cyclers include multiple detection channels. In this multiplex system, the amount of target DNA/cDNA can be compared to the amount of a housekeeping sequence e.g. GAPDH or ⁇ -actin. Two types of detection chemistries are most commonly used for quantitative PCR. The first uses an intercalating dye that incorporates into double-stranded DNA. Of these fluorescent dyes, SYBR ® Green I dye is the most common one used.
- the second detection method uses a primer or oligonucleotide specific to the target of interest, as in TaqMan ® probes, Molecular BeaconsTM, or Scorpion primers.
- the oligonucleotide is labeled with a fluorescent dye and quencher.
- the oligonucleotide itself has no significant fluorescence, but fluoresces either when annealed to the template (as in molecular beacons) or when the dye is clipped from the oligonucleotide during extension (as in TaqMan probes). Multiplex PCR is possible by using dyes with different fluorescent emissions for each primer.
- a gene encoding a marker selected from the group consisting of GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB is amplified.
- Western blotting is an important technique used in cell and molecular biology. By using a western blot, researchers are able to identify specific proteins from a complex mixture of proteins extracted from cells. The technique uses three elements to accomplish this task: (1 ) separation by size, (2) transfer to a solid support, and (3) marking target protein using a proper primary and secondary antibody to visualize. Western blots can be carried out in a quantitative manner. The quantification may be based on the differential densitometry of the associated chemiluminescent and/or fluorescent signals from the blots (see, for example, Tyloer and Posh, BioMed Research International, Volume 2014 (2014), Article ID 361590).
- Flow cytometry is an analytical cell-biology technique that utilizes light to count and profile cells in a heterogenous fluid mixture. Flow cytometry is a particularly and preferred powerful method because it allows a researcher to rapidly, accurately, and simply collect data related to many parameters from a heterogeneous fluid mixture containing live cells.
- flow cytometry is used to identify, separate, and characterize various immune cell subtypes by virtue of their size and morphology.
- antibodies tagged with fluorescent dyes, and raised against highly specific cell surface antigens e.g. clusters of differentiation or CD markers
- the compound is an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, protein drug or small molecule.
- the aptamer, ribozyme, antibody, small molecule, protein drug, siRNA, a shRNA or an antisense oligonucleotide of this embodiment specifically binds to / interacts with a compound being involved in the activation of elF5a thereby inhibiting the compound.
- the compounds are the precursor and enzymes (including the mRNA encoding the enzymes) as shown in Figures 1 and 2 and the respective nucleic acid molecules encoding them.
- the enzymes ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH) are involved in the activation of elF5a.
- the mRNA of human ornithine decarboxylase is shown in SEQ ID NO: 1 , of human spermidine synthase in SEQ ID NO: 3, of human spermine synthase in SEQ ID NO: 5, of human DOHH in SEQ ID NO: 7 and of human DDHPS in SEQ ID NO: 9.
- amino acid sequence of human ornithine decarboxylase is shown in SEQ ID NO: 2, of human spermidine synthase in SEQ ID NO: 4, of human spermine synthase in SEQ ID NO: 6, of human DOHH in SEQ ID NO: 8 and of human DHPS in SEQ ID NO: 10.
- SEQ ID NOs 7 and 9 and within the amino acid sequences SEQ ID NOs 8 and 10 are preferred.
- the mRNAs of SEQ ID NOs 7 and 9 encode the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a and the amino acid sequences of SEQ ID NOs 8 and 10 are the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a.
- These enzymes catalyze the deoxyhypusination (i.e. the addition of deoxyhypusine) and the hypusination (i.e. the addition of hypusin) of elF5a.
- a compound targeting the mRNA of SEQ ID NOs 7 or 9 or the amino acid sequence of SEQ ID NOs 8 of 10 is expected to prevent unwanted off target effects.
- the term "aptamer” in accordance with the present invention refers to DNA or RNA molecules being either in the natural D-conformation or in the L-conformation ("aptamer") that usually have been selected from random pools based on their ability to bind other molecules. Aptamers have been selected which bind nucleic acid, proteins, small organic compounds, and even entire organisms. Hence, apatamer can be designed against the precursors, enzymes and the mRNA encoding the enzymes.
- aptamers can be classified as DNA or RNA aptamers or peptide aptamers. Whereas the former consist of (usually short) strands of oligonucleotides, the latter consist of a short variable peptide domain, attached at both ends to a protein scaffold.
- Nucleic acid aptamers are nucleic acid species that have been engineered through repeated rounds of in vitro selection or equivalently, SELEX (systematic evolution of ligands by exponential enrichment) to bind to various molecular targets such as small molecules, proteins, nucleic acids, and even cells, tissues and organisms.
- Aptamers offer the utility for biotechnological and therapeutic applications as they offer molecular recognition properties that rival those of the commonly used biomolecules, in particular antibodies.
- aptamers offer advantages over antibodies as they can be engineered completely in a test tube, are readily produced by chemical synthesis, possess desirable storage properties, and elicit little or no immunogenicity in therapeutic applications.
- Non-modified aptamers are cleared rapidly from the bloodstream, with a half-life of minutes to hours, mainly due to nuclease degradation and clearance from the body by the kidneys, a result of the aptamer's inherently low molecular weight.
- the rapid clearance of aptamers can be an advantage in applications such as in vivo diagnostic imaging.
- ribozymes refers to RNA molecules that act as enzymes in the absence of proteins. These RNA molecules act catalyticly or autocatalyticly and are capable of cleaving e.g. other RNAs at specific target sites but they have also been found to catalyze the aminotransferase activity of the ribosome. Hence, the ribozymes may act on the mRNA encoding the enzymes being involved in elF5a activation. Selection of appropriate target sites and corresponding ribozymes can be done as described for example in Zaher and Unrau (2007), RNA, 13 (7): 1017-1026.
- RNAs examples include the hammerhead, hairpin, hepatitis delta virus, and in vitro-selected lead- dependent ribozymes.
- the principle of catalytic self-cleavage has become well established in the last 10 years.
- the hammerhead ribozymes are characterized best among the RNA molecules with ribozyme activity. Since it was shown that hammerhead structures can be integrated into heterologous RNA sequences and that ribozyme activity can thereby be transferred to these molecules, it appears that catalytic sequences for almost any target sequence can be created, provided the target sequence contains a potential matching cleavage site.
- RNA which contains the GUC (or CUC) triplet
- GUC GUC
- CUC CUC
- Molecules of this type were synthesized for numerous target sequences. They showed catalytic activity in vitro and in some cases also in vivo. The best results are usually obtained with short ribozymes and target sequences.
- the aptamers and ribozymes may comprise modified nucleotides, such as locked nucleic acids (LNAs).
- LNAs locked nucleic acids
- antibody as used in accordance with the present invention comprises, for example, polyclonal or monoclonal antibodies. Furthermore, also derivatives or fragments thereof, which still retain the binding specificity, are comprised in the term "antibody”. Antibody fragments or derivatives comprise, inter alia, Fab or Fab' fragments, Fd, F(ab') 2 , Fv or scFv fragments, single domain V H or V-like domains, such as VhH or V-NAR-domains, as well as multimeric formats such as minibodies, diabodies, tribodies, tetrabodies or chemically conjugated Fab'-multimers (see, for example, Altshuler et al., Biochemistry (Mosc).
- antibody also includes embodiments such as chimeric (human constant domain, non- human variable domain), single chain and humanized (human antibody with the exception of non-human CDRs) antibodies.
- chimeric human constant domain, non- human variable domain
- single chain humanized antibodies.
- Various techniques for the production of antibodies and fragments thereof are well known in the art and described, e.g. in Altshuler et al., Biochemistry (Mosc). 2010 Dec; 75(13): 1584-605.
- polyclonal antibodies can be obtained from the blood of an animal following immunisation with an antigen in mixture with additives and adjuvans and monoclonal antibodies can be produced by any technique which provides antibodies produced by continuous cell line cultures. Examples for such techniques are described, e.g. Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1988 and Harlow and Lane, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1999 and include the hybridoma technique originally described by Kohler and Milstein, Nature 256 (1975), 495-497, the trioma technique, the human B-cell hybridoma technique (see e.g.
- recombinant antibodies may be obtained from monoclonal antibodies or can be prepared de novo using various display methods such as phage, ribosomal, mRNA, or cell display.
- a suitable system for the expression of the recombinant (humanized) antibodies or fragments thereof may be selected from, for example, bacteria, yeast, insects, mammalian cell lines or transgenic animals or plants (see, e.g., US patent 6,080,560; Holliger and Hudson, Nat Biotechnol., 2005; 23(9):1126-36). Further, techniques described for the production of single chain antibodies (see, inter alia, US Patent 4,946,778) can be adapted to produce single chain antibodies specific for the target of this invention. Surface plasmon resonance as employed in the BIAcore system can be used to increase the efficiency of phage antibodies. Antibodies may be directed to proteins or small organic molecules.
- the antibody used by the present invention may act on one of the enzymes or precursors being involved in elF5a activation.
- the antibody thus preferably specifically binds to an amino acid sequence being selected from SEQ ID NOs 2, 4, 6, 8 and 10, preferably from SEQ ID NOs 8 and 10, or a precursor being selected from ornithine, spermidine, spermine, and elF5a-deoxyhypusine.
- the antibody inhibits the activity of elF5a by inhibiting an enzyme or a precursor being required for the activation of elF5a.
- the antibody more preferably directly targets elF5a- hypusine thereby inhibiting its activity.
- protein drug designates designer drugs that are derivatives of human proteins. These proteins are used as scaffold to create a protein drug by well-established screening procedures (see Tomlinson et al (2004), Nature Biotechnology, 22(5): 521-522).
- Non-limiting examples of human proteins which serve as a scaffold for designing protein drugs are transferrin, C-type lectins, trinectins, domain antibodies, kunitz domains, lipocalins and the Fyn SH3 domain.
- the protein drug used by the present invention may act on one of the enzymes or precursors being involved in elF5a activation.
- small molecule designates a low molecular weight organic compound that may have or has the capability to inhibit elF5a activation.
- the upper molecular weight limit for a small molecule is preferably about 900 Daltons. Small molecule of this molecular weight can in general rapidly diffuse across cell membranes, so that they can reach intracellular sites of action. As lower molecular weight a cut-off of 500 Daltons is preferred. This is because clinical attrition rates may be significantly reduced if the molecular weight is kept is below 500 Daltons.
- the antisense technology for the downregulation of RNA is well-established and widely used in the art to treat various diseases.
- the basic idea of the antisense technology is the use of oligonucleotides for silencing a selected target mRNA through the extraordinar specificity of complementary-based pairing (Re, Ochsner J. 2000 Oct; 2(4): 233-236).
- oligonucleotides for silencing a selected target mRNA through the extraordinar specificity of complementary-based pairing (Re, Ochsner J. 2000 Oct; 2(4): 233-236).
- compound classes of siRNAs, shRNAs and antisense oligonucleotides will be provided.
- antisense oligonucleotides are single stranded antisense constructs while siRNAs and shRNAs are double stranded antisense constructs with one strand comprising an antisense oligonucleotide sequence (i.e. the so-called antisense strand). All these compound classes may be used to achieve downregulation or inhibition of a target RNA.
- the target of antisense constructs in general as well as all the specific classes of antisense constructs being described herein is preferably an mRNA of an enzyme being involved in the activation of elF5a.
- the target is preferably a nucleic acid sequence selected from one or more of SEQ ID NOs 1 , 3, 5, 7 and 9. Within SEQ ID NOs 1 , 3, 5, 7 and 9, SEQ ID NOs are 7 and 9 are preferred. It is a matter of routine in the field of antisense technology to design an antisense construct with a sufficient number of nucleotide mismatches to any off-target in order to ensure that no off- targets become down-regulated.
- siRNA in accordance with the present invention refers to small interfering RNA, also known as short interfering RNA or silencing RNA.
- siRNAs are a class of 12 to 30, preferably 18 to 30, more preferably 20 to 25, and most preferred 21 to 23 or 21 nucleotide- long double-stranded RNA molecules that play a variety of roles in biology. Most notably, siRNA is involved in the RNA interference (RNAi) pathway where the siRNA interferes with the expression of a specific gene. In addition to their role in the RNAi pathway, siRNAs also act in RNAi-related pathways, e.g. as an antiviral mechanism or in shaping the chromatin structure of a genome.
- RNAi RNA interference
- siRNAs have a well defined structure: a short double-strand of RNA (dsRNA), advantageously with at least one RNA strand having a 5' or 3' overhang. Each strand typically has a 5' phosphate group and a 3' hydroxyl (-OH) group.
- dsRNA short double-strand of RNA
- Each strand typically has a 5' phosphate group and a 3' hydroxyl (-OH) group.
- This structure is the result of processing by dicer, an enzyme that converts either long dsRNAs or small hairpin RNAs into siRNAs.
- siRNAs can also be exogenously (artificially) introduced into cells to bring about the specific knockdown of a gene of interest. Thus, any gene of which the sequence is known can in principle be targeted based on sequence complementarity with an appropriately tailored siRNA.
- the double-stranded RNA molecule or a metabolic processing product thereof is capable of mediating target-specific nucleic acid modifications, particularly RNA interference and/or DNA methylation.
- one or both ends of the double-strand have a 3'-overhang from 1-5 nucleotides, more preferably from 1-3 nucleotides and most preferably 2 nucleotides.
- the most efficient silencing was so far obtained with siRNA duplexes composed of 21 -nt sense and 21 -nt antisense strands, paired in a manner to have 2-nt 3'- overhangs.
- the sequence of the 2-nt 3' overhang makes a small contribution to the specificity of target recognition restricted to the unpaired nucleotide adjacent to the first base pair (Elbashir et al. Nature. 2001 May 24; 41 1 (6836):494-8).
- 2'-deoxynucleotides in the 3' overhangs are as efficient as ribonucleotides, but are often cheaper to synthesize and probably more nuclease resistant.
- the siRNA used in the invention preferably comprises an antisense strand which comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides of one or more sequences selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably SEQ ID NOs 7 and 9.
- a preferred example of a siRNA is an endoribonuclease-prepared siRNA (esiRNA).
- esiRNA is a mixture of siRNA oligos resulting from cleavage of a long double-stranded RNA (dsRNA) with an endoribonuclease such as Escherichia coli RNase III or dicer.
- esiRNAs are an alternative concept to the usage of chemically synthesized siRNA for RNA interference (RNAi).
- RNAi RNA interference
- a cDNA of an mRNA template may be amplified by PCR and tagged with two bacteriophage-promotor sequences. RNA polymerase is then used to generate long double-stranded RNA that is complentary to the target-gene cDNA.
- This complementary RNA may be subsequently digested with RNase III from Escherichia coli to generate short overlapping fragments of siRNAs with a length between 18-25 base pairs.
- This complex mixture of short double-stranded RNAs is similar to the mixture generated by dicer cleavage in vivo and is therefore called endoribonuclease-prepared siRNA or short esiRNA.
- esiRNA are a heterogeneous mixture of siRNAs that all target the same mRNA sequence. esiRNAs lead to highly specific and effective gene silencing.
- a “shRNA” in accordance with the present invention is a short hairpin RNA, which is a sequence of RNA that makes a (tight) hairpin turn that can also be used to silence gene expression via RNA interference.
- shRNA preferably utilizes the U6 promoter for its expression.
- the shRNA hairpin structure is cleaved by the cellular machinery into siRNA, which is then bound to the RNA-induced silencing complex (RISC). This complex binds to and cleaves mRNAs which match the shRNA that is bound to it.
- RISC RNA-induced silencing complex
- the shRNA used in the invention preferably comprises an antisense strand which comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably SEQ ID NOs 7 and 9.
- antisense oligonucleotide in accordance with the present invention preferably refers to a single-stranded nucleotide sequence being complementary by virtue of Watson- Crick base pair hybridization to an mRNA selected from SEQ ID NOs 1 , 3, 5, 7 and 9 whereby the respective mRNA is blocked and cannot be translated into protein.
- the antisense oligonucleotides may be unmodified or chemically modified. In general, they are relatively short (preferably between 13 and 25 nucleotides). Moreover, they are specific for one of SEQ ID NOs 1 , 3, 5, 7 and 9, i.e. they hybridize to a unique sequence in the total pool of targets present in the target cells/organism.
- the antisense oligonucleotide according to the invention comprises or consists a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, or at least 25 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9.
- At least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, or at least 25 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO.
- the antisense oligonucleotide is preferably a LNA-GapmeR, an Antagomir, or an antimiR.
- GapmeRs LNA-GapmeRs or simply GapmeRs are potent antisense oligonucleotides used for highly efficient inhibition of mRNA function. GapmeRs function by RNase H dependent degradation of complementary RNA targets. They are an excellent alternative to siRNA for knockdown of mRNA. They are advantageously taken up by cell without transfection reagents. GapmeRs contain a central stretch of DNA monomers flanked by blocks of LNAs. The GapmeRs are preferably 14-16 nucleotides in length and are optionally fully phosphorothioated. The DNA gap activates the RNAse H-mediated degradation of targeted RNAs and is also suitable to target transcripts directly in the nucleus.
- the LNA-GapmeR used in the invention preferably comprises a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, or at least 15 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9.
- These at least 13 nucleotides, at least 14 nucleotides, or at least 15 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO.
- LNA-GapmeRs are routinely designed using established algorithms. LNA-GapmeRs to a selected target are commercially available including positive and negative controls, for example, from Exiqon.
- AntimiRs are oligonucleotide inhibitors that were initially designed to be complementary to a miRNA. AntimiRs against miRNAs have been used extensively as tools to gain understanding of specific miRNA functions and as potential therapeutics. As used herein, the AntimiRs are preferably designed to be complementary to a sequence selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. AntimiRs are preferably 14 to 23 nucleotides in length.
- An AntimiR according to the invention more preferably comprises or consists a sequence which is with increasing preference complementary to at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or at least 23 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9.
- At least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or at least 23 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO.
- AntimiRs are preferably AntagomiRs.
- AntagomiRs are synthetic 2-O-methyl RNA oligonucleotides, preferably of 21 to 23 nucleotides which are preferably fully complementary to the selected target RNA. While AntagomiRs were initially designed against miRNAs they may also be designed against mRNAs.
- the AntagomiRs used according to the invention therefore preferably comprise a sequence being complementary to 21 to 23 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably within SEQ ID NOs 7 and 9.
- nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO.
- AntagomiRs are preferably synthesized with 2'-OMe modified bases (2'-hydroxyl of the ribose is replaced with a methoxy group), phosphorothioate (phosphodiester linkages are changed to phosphorothioates) on the first two and last four bases, and an addition of cholesterol motif at 3' end through a hydroxyprolinol modified linkage.
- the addition of 2'-OMe and phosphorothioate modifications improves the bio-stability whereas cholesterol conjugation enhances distribution and cell permeation of the AntagomiRs.
- Antisense molecules are preferably chemically synthesized using a conventional nucleic acid synthesizer.
- Suppliers of nucleic acid sequence synthesis reagents include Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, CO, USA), Pierce Chemical (part of Perbio Science, Rockford, IL, USA), Glen Research (Sterling, VA, USA), ChemGenes (Ashland, MA, USA), and Cruachem (Glasgow, UK).
- antisense molecules including antisense oligonucleotides, such as LNA- GapmeR, an Antagomir, an antimiR
- siRNA and shRNA to potently, but reversibly, silence or inhibit a target mRNA in vivo makes these molecules particularly well suited for use in the medical applications of the invention being further described herein below.
- Ways of administering siRNA to humans are described in De Fougerolles et al., Current Opinion in Pharmacology, 2008, 8:280-285. Such ways are also suitable for administering other small RNA molecules like antisense oligonucleotides or shRNAs.
- compositions may be administered directly formulated as a saline, via liposome based and polymer-based nanoparticle approaches, as conjugated or complexation pharmaceutical compositions, or via viral delivery systems.
- Direct administration comprises injection into tissue, intranasal and intratracheal administration.
- Liposome based and polymer-based nanoparticle approaches comprise the cationic lipid Genzyme Lipid (GL) 67, cationic liposomes, chitosan nanoparticles and cationic cell penetrating peptides (CPPs).
- Conjugated or complexation pharmaceutical compositions comprise PEI-complexed antisense molecules (including antisense oligonucleotides), siRNA, or shRNA.
- viral delivery systems comprise influenza virus envelopes and virosomes.
- the antisense molecules may comprise modified nucleotides such as locked nucleic acids (LNAs).
- LNAs locked nucleic acids
- the ribose moiety of an LNA nucleotide is modified with an extra bridge connecting the 2' oxygen and 4' carbon. The bridge "locks" the ribose in the 3'-endo (North) conformation, which is often found in the A-form duplexes.
- LNA nucleotides can be mixed with DNA or RNA residues in the oligonucleotide whenever desired.
- nucleic acid sequences as defined in items (a) to (c) of this preferred embodiment comprise or consist of sequences being complementary to nucleotides of the amino acids encoding the enzymes required for the activation of elF5a as defined by SEQ ID NOs 1 , 3, 5, 7 and 9, preferably by SEQ ID NOs 7 and 9.
- the nucleic acid sequences as defined in items (a) to (c) comprise or are antisense nucleic acid sequences.
- the nucleic acid sequence according to item (a) of this further preferred embodiment of the invention comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides of one or more sequences selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9.
- At least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9.
- the format of the nucleic acid sequence according to item (a) is not particularly limited as long as it comprises or consists of at least 12 continuous nucleotides being complementary to a nucleic acid sequence selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9.
- the nucleic acid sequence according to item (a) reflects the above- mentioned basic principle of the antisense technology which is the use of an oligonucleotide for silencing a selected target RNA through the extraordinar specificity of complementary-based pairing. Therefore, it is to be understood that the nucleic acid sequence according to item (a) is preferably in the format of an siRNA, shRNA or an antisense oligonucleotide as defined herein above.
- the antisense oligonucleotides are preferably LNA-GapmeRs, AntagomiRs, or antimiRs as defined herein above.
- a nucleic acid sequence according to item (b) of the above preferred embodiment of the invention is capable of interacting with, more specifically hybridizing with the target mRNA being selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. By formation of the hybrid the function of the mRNA being selected from SEQ ID NOs 1 , 3, 5, 7 and 9 is reduced or blocked.
- sequence identity of the molecule according to item (b) in connection with a sequence selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9 is with increasing preference at least 75%, at least 80%, at least 85%, at least 90%, at least 92.5%, at least 95%, at least 98%, at least 99% and 100%.
- sequence identity in connection with each of SEQ ID NOs 1 , 3, 5, 7 and 9 can be individually selected. For instance, a non-limiting example is at least 85% in connection with SEQ ID NO: 7 and at least 90% in connection with SEQ ID NO: 9. Means and methods for determining sequence identity are known in the art.
- the BLAST (Basic Local Alignment Search Tool) program is used for determining the sequence identity with regard to one or more of SEQ ID NOs 1 , 3, 5, 7 and 9.
- nucleic acid sequences which comprise a nucleotide sequence which is at least 70% identical to the complementary strand of one or more of SEQ ID NOs 1 , 3, 5, 7 and 9 are the complementary strands of SEQ ID NOs 1 , 3, 5, 7 and 9.
- the nucleotide sequences may be RNA or DNA.
- RNA or DNA encompasses chemically modified RNA nucleotides or DNA nucleotides.
- RNA comprises the nucleotide U while DNA comprises the nucleotide T.
- the inhibitor may also be an expression vector or host, respectively being capable of producing an nucleic acid sequence as defined in any one of items (a) to (c).
- An expression vector may be a plasmid that is used to introduce a specific transcript into a target cell. Once the expression vector is inside the cell, the inhibitor of the activation of elF5a being encoded by the expression vector is produced by the cellular-transcription.
- the plasmid is in general engineered to contain regulatory sequences that act as enhancer and/or promoter regions and lead to efficient transcription of the transcript.
- the expression vector preferably contains a macrophage-specific promoter. Macrophage-specific promoters are known in the art, for example, from Levin et a., Gene Ther., 2012; 19(11 ):1041-7. Using a macrophage-specific promoter ensures that the nucleic acid sequence is only expressed in macrophages and may avoid potential undesired side effects by expression in other cell types.
- Non-limiting examples of expression vectors include prokaryotic plasmid vectors, such as the pUC-series, pBluescript (Stratagene), the pET-series of expression vectors (Novagen) or pCRTOPO (Invitrogen) and vectors compatible with an expression in mammalian cells like pREP (Invitrogen), pcDNA3 (Invitrogen), pCEP4 (Invitrogen), pMCI neo (Stratagene), pXT1 (Stratagene), pSG5 (Stratagene), EBO-pSV2neo, pBPV-1 , pdBPVMMTneo, pRSVgpt, pRSVneo, pSV2-dhfr, plZD35, pLXIN, pSIR (Clontech), pIRES-EGFP (Clontech), pEAK-10 (Edge Biosystems) pTriEx-H
- Examples for plasmid vectors suitable for Pichia pastoris comprise e.g. the plasmids pA0815, pPIC9K and pPIC3.5K (all Intvitrogen).
- a suitable vector is selected in accordance with good manufacturing practice.
- Such vectors are known in the art, for example, from Ausubel et al, Hum Gene Ther. 201 1 Apr; 22(4):489-97 or Allay et al., Hum Gene Ther. May 2011 ; 22(5): 595-604.
- a typical mammalian expression vector contains the promoter element, which mediates the initiation of transcription of mRNA, the protein coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Moreover, elements such as origin of replication, drug resistance gene, regulators (as part of an inducible promoter) may also be included.
- the lac promoter is a typical inducible promoter, useful for prokaryotic cells, which can be induced using the lactose analogue isopropylthiol-b-D-galactoside ("IPTG").
- IPTG lactose analogue isopropylthiol-b-D-galactoside
- the polynucleotide of interest may be ligated between e.g.
- PelB leader signal which directs the recombinant protein in the periplasm and the gene III in a phagemid called pHEN4 (described in Ghahroudi et al, 1997, FEBS Letters 414:521 -526). Additional elements might include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing. Highly efficient transcription can be achieved with the early and late promoters from SV40, the long terminal repeats (LTRs) from retroviruses, e.g., RSV, HTLVI, HIVI, and the early promoter of the cytomegalovirus (CMV).
- LTRs long terminal repeats
- CMV cytomegalovirus
- cellular elements can also be used (e.g., the human actin promoter).
- Suitable expression vectors for use in practicing the present invention include, for example, vectors such as pSVL and pMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146) and pBC12MI (ATCC 67109).
- the inhibitor can be expressed in stable cell lines that contain the gene construct integrated into a chromosome. The co-transfection with a selectable marker such as dhfr, gpt, neomycin, hygromycin allows the identification and isolation of the transfected cells.
- the transfected nucleic acid can also be amplified to express large amounts of the encoded (poly)peptide.
- the DHFR (dihydrofolate reductase) marker is useful to develop cell lines that carry several hundred or even several thousand copies of the nucleic acid molecule encoding the inhibitor.
- Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy et al.1991 , Biochem J. 227:277-279; Bebbington et al. 1992, Bio/Technology 70:169-175). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected.
- the expression vectors will preferably include at least one selectable marker.
- vectors can contain one or more origins of replication (ori) and inheritance systems for cloning or expression, one or more markers for selection in the host, e.g., antibiotic resistance, and one or more expression cassettes.
- origins of replication include, for example, the Col E1 , the SV40 viral and the M 13 origins of replication.
- the sequences to be inserted into the vector can e.g. be synthesized by standard methods, or isolated from natural sources. Ligation of the coding sequences to transcriptional regulatory elements and/or to other amino acid encoding sequences can be carried out using established methods.
- Transcriptional regulatory elements parts of an expression cassette
- These elements comprise regulatory sequences ensuring the initiation of the transcription (e.g., translation initiation codon, promoters, enhancers, and/or insulators), internal ribosomal entry sites (IRES) (Owens, Proc. Natl. Acad. Sci.
- nucleotide sequence as defined in item (a) of the above preferred embodiment of the invention is operatively linked to such expression control sequences allowing expression in prokaryotic or eukaryotic cells.
- the host may be a prokaryotic or eukaryotic cell.
- a suitable eukaryotic host may be a mammalian cell, an amphibian cell, a fish cell, an insect cell, a fungal cell or a plant cell.
- Representative examples of bacterial cells are E. coli, Streptomyces and Salmonella typhimurium cells; of fungal cells are yeast cells; and of insect cells are Drosophila S2 and Spodoptera Sf9 cells. It is preferred that the cell is a mammalian cell such as a human cell.
- Mammalian host cells that could be used include, human Hela, 293, H9 and Jurkat cells, mouse NIH3T3 and C127 cells, Cos 1 , Cos 7 and CV1 , quail QC1-3 cells, mouse L cells and Chinese hamster ovary (CHO) cells.
- the cell may be a part of a cell line, preferably a human cell line. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
- the host is preferably a host cell and more preferably an isolated host cell.
- the host is also preferably a non-human host.
- the compound is directed against ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH) and is preferably directed against DHPS or DOHH.
- ornithine decarboxylase, spermidine synthase, spermine synthase, DHPS and DOHH are enzymes being required for the activation of elF5a.
- the compound is directed against the mRNA encoding one of the enzymes or one of the enzymes in their protein from.
- siRNA or shRNA it may be directed against the mRNA encoding one of the enzymes.
- the compound is an antibody it may be directed against one of the enzymes in their protein form.
- the compounds are preferably directed against the human mRNAs and enzymes.
- the mRNA of human ornithine decarboxylase is shown in SEQ ID NO: 1 , of human spermidine synthase in SEQ ID NO: 3, of human spermine synthase in SEQ ID NO: 5, of human DOHH in SEQ ID NO: 7 and of human DDHPS in SEQ ID NO: 9.
- the amino acid sequence of human ornithine decarboxylase is shown in SEQ ID NO: 2, of human spermidine synthase in SEQ ID NO: 4, of human spermine synthase in SEQ ID NO: 6, of human DOHH in SEQ ID NO: 8 and of human DHPS in SEQ ID NO: 10.
- mRNAs SEQ ID NOs 7 and 9 and within the amino acid sequences SEQ ID NOs 8 and 10 are the preferred targets. This is because in the polyamine synthesis pathway the mRNAs of SEQ ID NOs 7 and 9 encode the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a and the amino acid sequences of SEQ ID NOs 8 and 10 are the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a. These enzymes catalyze the deoxyhypusiantion and the hypusination of elF5a.
- the efficacy of a compound for inhibiting elF5a activation in a patient is determined, wherein the sample of step (a) and the control sample are samples that have been obtained from the patient, and wherein a reduction of the number of cells having a M2-phenotype in the sample as compared to the control sample not contacted with the compound indicates that the compound is effective for inhibiting elF5a activation in the patient.
- the compound to be tested for its capability of inhibiting elF5a activation may also be a compound for which it is already known that it has the capability of inhibiting elF5a activation and in this case the method can determine how much inhibition is achieved by the compound.
- This option of performing the claimed method is particularly suitable for determining the efficacy of a compound for inhibiting elF5a activation in a patient.
- the first aspect of the present invention also encompasses a method for determining the efficacy of a compound for inhibiting elF5a activation in a patient, comprising (a) obtaining a sample comprising cells being capable of differentiating into M2 macrophages from a patient, (a) contacting the obtained sample with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and (b) quantifying in the sample the cells having a M2-phenotype, wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting elF5a activation.
- a compound for inhibiting elF5a activation can be used to treat several diseases in a patient.
- determining the efficacy of a compound for inhibiting elF5a activation in a patient allows to predict whether this compound will be effective to treat or prevent a disease in the patient, in particular a disease being mediated by M2 macrophages, or that can be treated by inhibiting cellular differentiation into M2 macrophages.
- the patient is afflicted with a hyperproliferative disorder, a fibrotic disorder or the macrophage activation syndrome.
- a hyperproliferative disorder is any disease comprising an abnormally high rate of proliferation of cells by rapid cell division.
- Non-limiting examples are psoriasis, psoriatic arthritis, rheumatoid arthritis, cutaneous hyperkeratoses, inflammatory bowel disease tumors and cancers.
- a fibrotic disorder is any disorder comprising the formation of excess fibrous connective tissue, e.g. in an organ or tissue in a reparative or reactive process.
- Non-limiting examples of fibrotic disorders will be provided herein below.
- MAS macrophage activation syndrome
- SoJIA systemic-onset juvenile idiopathic arthritis
- MAS has been described in association with systemic lupus erythematosus (SLE), Kawasaki disease, and adult-onset Still's disease. It is thought to be closely related and pathophysiologically very similar to reactive (secondary) hemophagocytic lymphohistiocytosis (HLH).
- the hallmark clinical and laboratory features include high fever, hepatosplenomegaly, lymphadenopathy, pancytopenia, liver dysfunction, disseminated intravascular coagulation, hypofibrinogenemia, hyperferritinemia, and hypertriglyceridemia.
- ESR erythrocyte sedimentation rate
- the low ESR helps to distinguish the disorder from a flare of the underlying rheumatic disorder, in which case the ESR is usually elevated.
- a bone marrow biopsy or aspirate usually shows hemophagocytosis.
- M2 macrophages are known to secrete large amounts of pro-fibrotic factors, such as TGF- ⁇ and Galactin-3 (Braga et I. (2015), Front Immunol; 6:602 and Wermuth and Jimenez (2015), Clin Transl Med; 4: 2.). M2 macrophages are also known to promote cell proliferation (Mills (2012), Crit Rev Immunol.; 32(6):463-88.). It follows that M2 macrophages play an important role in promoting the formation or hyperproliferative or fibrotic disorders and interfering with the formation of M2 macrophages is a suitable means for treating or preventing a hyperproliferative or fibrotic disorders. In connection with the method of the invention it is particularly useful to determine the efficacy of a compound for inhibiting elF5a activation in a patient being afflicted with a hyperproliferative or fibrotic disorder.
- pro-fibrotic factors such as TGF- ⁇ and Galactin-3
- the sample is a tissue sample or a blood sample.
- the sample has to be a sample that has been obtained from the body of the patient and has to comprise cells being M2 macrophages or capable of differentiating into M2 macrophages.
- a sample is preferably a tissue sample or a blood sample. Macrophages are known to be distributed in tissues throughout the body, e.g. in the brain, liver, lung, spleen, intestine, skin, heart, kidney and peritoneum (Epelman et al.
- tissue sample is preferably obtained from such a patient.
- Macrophages are likewise found in the blood, including the peripheral blood.
- the blood sample may be a whole blood sample, serum or plasma.
- the compound is guanyl-1 ,7-diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I or CNI-1493.
- GC7 guanyl-1 ,7-diaminoheptane
- L-mimosine L-mimosine
- ciclopirox deferiprone
- hydralazine agent I or CNI-1493.
- GC7 Guanyl-1 ,7-diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I and CNI-1493 are non-limiting examples of compounds interfering with an enzyme being involved in elF5a activation.
- GC-7 Guanyl-1 ,7-diaminoheptane (CAS 150333-69-0) is an efficient inhibitor of deoxyhypusine synthase. As a competitive inhibitor it binds to the active site of deoxyhypusine synthase and prevents the first step in the post-translational conversion of a single lysine to hypusine in elF5A. GC-7 is used in the examples of the application.
- L-mimosine (CAS 500-44-7) is a plant non-protein amino acid, chemically similar to tyrosine. The synthesis of the amino acid hypusine by deoxyhypusine hydroxylase is blocked by L- mimosine, further noting that hypusine is necessary for the activation of elF-5A.
- the drugs ciclopirox (CAS 29342-05-0) and deferiprone (CAS 30652-1 1-0) are inhibitors of deoxyhypusine hydroxylase and hence the posttranslational modification by hypusine being requiring for the activation of elF-5A (Memin et al al. (2014), Cancer Res. 2014 Jan 15;74(2):552-62).
- Hydralazine (CAS 86-54-4) inhibits the posttranslational hydroxylation of deoxyhypusine (Paz et al. (1984), Biochem Pharmacol, 33: 779-785).
- Agent I Hoechst 768159, [2-(4-hydroxytoluene-3-yl)-4,5-dihydro-4- carboxythiazole] is an iron chelator that blocks the function of deoxyhypusine hydroxylase and hence the hypusination of elF5a; see Hanauske-Abel et al. (1994) Biochimica et Biophysica Acta 1221 115-124.
- CNI-1493 or semapimod; CAS 164301-51 -3) is an inhibitor of deoxyhypusine synthase (Specht et al. (2008), Parasitol Res; 102(6): 1 177-84).
- the present invention relates to a compound inhibiting elF5a activation for use in the treatment or prevention of a disease being mediated by M2 macrophages, wherein the disease is preferably a hyperproliferative, a fibrotic disorder or the macrophage activation syndrome.
- the present invention likewise relates to a method for treating or preventing a disease being mediated by M2 macrophages, wherein the disease is preferably a hyperproliferative, a fibrotic disorder or the or the macrophage activation syndrome comprising administering a therapeutically effective amount of a compound inhibiting elF5a activation to a subject in need thereof.
- the present invention relates to a compound inhibiting elF5a activation for use in the treatment or prevention of a hyperproliferative, a fibrotic disorder or the macrophage activation syndrome by inhibiting cellular differentiation into M2 macrophages.
- the present invention also relates to a method for treating or preventing a hyperproliferative disorder, a fibrotic disorder or the macrophage activation syndrome by inhibiting cellular differentiation into M2 macrophages comprising administering a therapeutically effective amount of a compound inhibiting elF5a activation to a subject in need thereof, thereby inhibiting the cellular differentiation into M2 macrophages in the subject.
- the subject to be treated in accordance with the second and third aspect of the invention is preferably a mammal, more preferably a primate and most preferably a human.
- the compounds inhibiting elF5a activation are preferably admixed with a pharmaceutically acceptable carrier or excipient to form a pharmaceutical composition.
- a pharmaceutically acceptable carrier or excipient is meant a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type (see also Handbook of Pharmaceutical Excipients 6ed. 2010, Published by the Pharmaceutical Press).
- the compounds inhibiting elF5a activation or the pharmaceutical composition may be administered, for example, orally, parenterally, such as subcutaneously, intravenously, intramuscularly, intraperitoneally, intrathecally, transdermally, transmucosally, subdurally, locally or topically via iontopheresis, sublingually, by inhalation spray, aerosol or rectally and the like in dosage unit formulations optionally comprising conventional pharmaceutically acceptable carriers or excipients.
- parenterally such as subcutaneously, intravenously, intramuscularly, intraperitoneally, intrathecally, transdermally, transmucosally, subdurally, locally or topically via iontopheresis, sublingually, by inhalation spray, aerosol or rectally and the like in dosage unit formulations optionally comprising conventional pharmaceutically acceptable carriers or excipients.
- the compounds inhibiting elF5a activation may be formulated as vesicles, such as liposomes.
- Liposomes have attracted great interest because of their specificity and the duration of action they offer from the standpoint of drug delivery. Liposomal delivery systems have been used to effectively deliver nucleic acids, such as siRNA in vivo into cells (Zimmermann et al. (2006) Nature, 441 :11 1-114). Liposomes are unilamellar or multilamellar vesicles which have a membrane formed from a lipophilic material and an aqueous interior. The aqueous portion contains the composition to be delivered. Cationic liposomes possess the advantage of being able to fuse to the cell wall.
- Non-cationic liposomes although not able to fuse as efficiently with the cell wall, are phagocytosed by macrophages and other cells in vivo.
- the compounds inhibiting elF5a activation can be administered to the subject at a suitable dose.
- the dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. The therapeutically effective amount for a given situation will readily be determined by routine experimentation and is within the skills and judgement of the ordinary clinician or physician.
- the regimen as a regular administration of the pharmaceutical composition should be in the range of 1 g to 5 g units per day.
- a more preferred dosage is in the range of 0.01 mg to 100 mg, even more preferably 0.01 mg to 50 mg and most preferably 0.01 mg to 10 mg per day.
- said compound comprises or is an nucleic acid molecule, such as an siRNA
- the total pharmaceutically effective amount of pharmaceutical composition administered will typically be less than about 75 mg per kg of body weight, such as for example less than about 70, 60, 50, 40, 30, 20, 10, 5, 2, 1 , 0.5, 0.1 , 0.05, 0.01 , 0.005, 0.001 , or 0.0005 mg per kg of body weight.
- the amount will be less than 2000 nmol of nucleic acid molecule per kg of body weight, such as for example less than 1500, 750, 300, 150, 75, 15, 7.5, 1.5, 0.75, 0.15, 0.075, 0.015, 0.0075, 0.0015, 0.00075 or 0.00015 nmol per kg of body weight.
- the length of treatment needed to observe changes and the interval following treatment for responses to occur vary depending on the desired effect.
- the particular amounts may be determined by conventional tests which are well known to the person skilled in the art. Suitable tests are, for example, described in Tamhane and Logan (2002), Journal of the American statistical association, 97(457):1-9.
- M2 macrophages play an important role in promoting the formation or hyperproliferative or fibrotic disorders as well as the macrophage activation syndrome and therefore these diseases can be treated or prevented by interfering with the formation of M2 macrophages.
- a compound inhibiting elF5a activation can be used to treat or prevent a disease being mediated by M2 macrophages or a hyperproliferative or fibrotic disorder or the macrophage activation syndrome by inhibiting cellular differentiation into M2 macrophages.
- the finding that the formation of M2 macrophages can be inhibited by inhibiting the activation of elF5a also revealed that diseases that are mediated by M2 macrophages can be treated or prevented by inhibiting the activation of elF5a.
- the compound targets ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH) and preferably DHPS or DOHH.
- ornithine decarboxylase, spermidine synthase, spermine synthase, DHPS and DOHH are enzymes being involved in the activation of elF5a. Further details on how the compounds of the invention target these enzymes are provided herein above in connection with the corresponding embodiment of the first aspect of the invention. These details apply mutatis mutandis to the second and third aspect of the invention.
- the compound is an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, or protein drug, small molecule.
- the compound inhibiting elF5a activation may be any one of an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, protein drug and small molecule as defined herein above in connection with the first aspect of the invention.
- the details on the compounds of the invention as provided herein above in connection with the corresponding embodiment of the first aspect of the invention apply mutatis mutandis to the second and third aspect of the invention.
- the compound is guanyl-1 ,7-diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I or CNI-1493.
- GC7 guanyl-1 ,7-diaminoheptane
- L-mimosine L-mimosine
- ciclopirox deferiprone
- hydralazine agent I or CNI-1493.
- the hyperproliferative disorder is a neoplasm, tumor or cancer and is preferably selected from cancer of the breast, lung, prostate, kidney, skin, neural, ovary, uterus, liver, pancreas, epithelial, gastric, intestinal, exocrine, endocrine, lymphatic, hematopoietic system or a head and neck tissue; and/or (ii) the fibrotic disorder is selected from sarcoidosis, renal fibrosis, pulmonary fibrosis, idiopathic pulmonary fibrosis, cystic fibrosis, liver fibrosis, cardiac fibrosis, endomyocardial fibrosis, atrial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, chronic kidney disease, nephrogenic systemic fibrosis, Chron's disease, hypertrophic scarring, keloid
- a neoplasm or tumor is an abnormal benign or malignant new growth of tissue that possesses no physiological function and arises from uncontrolled usually rapid cellular proliferation.
- the WHO classifies neoplasms into four main groups: benign neoplasms, in situ neoplasms, malignant neoplasms, and neoplasms of uncertain or unknown behavior.
- a malignant neoplasm is also referred to as cancer.
- each embodiment mentioned in a dependent claim is combined with each embodiment of each claim (independent or dependent) said dependent claim depends from.
- a dependent claim 2 reciting 3 alternatives D, E and F and a claim 3 depending from claims 1 and 2 and reciting 3 alternatives G, H and I
- the specification unambiguously discloses embodiments corresponding to combinations A, D, G; A, D, H; A, D, I; A, E, G; A, E, H; A, E, I; A, F, G; A, F, H; A, F, I; B, D, G; B, D, H; B, D, I; B, E, G; B, E, H; B, E, I; B, F, G; B, F, H; B, F, I; C, D, G; C, D, H; C, D, I; C,
- FIG. 1 The polyamine synthesis pathway. Polyamine synthesis begins with the production of putrescine from the amino acid ornithine, catalysed by ornithine decarboxylase. Spermidine and spermine are then synthesised from putrescine from spermidine synthase and spermine synthase, respectively.
- DHS deoxyhypusine synthase
- DOHH deoxyhypusine hydroxylase
- FIG. 3 The deoxyhypusine synthase inhibitor GC7 blocks M2 macrophage differentiation
- BMDMs Bone marrow-derived macrophages
- IL-4 IL-4 in the presence or absence of 10 uM GC7.
- M2 phenotype was assessed by flow cytometry analysis of CD301 and RELMa expression, two prototypic markers of M2 cells
- BMDMs were differentiated into M1 cells with LPS and IFN-y in the presence or absence of 10 uM GC7. 18 hours later, the expression of nitric oxide synthase 2 (NOS2), a hallmark of M1 cells, was assessed by flow cytometry.
- NOS2 nitric oxide synthase 2
- GC7 disrupts oxidative phosphorylation in M2 macrophages.
- BMDCs were polarised toward a M2 phenotype, as described above, in the presence or absence of 10 uM GC7.
- oxygen consumption rate OCR - an indicator of OXPHOS was measured using a Seahorse Analyser.
- the polyamine biosynthesis pathway regulates OXPHOS via hypusinated elF5A.
- the polyamine pathway comprises the cationic metabolites putrescine, spermidine, and spermine that are synthesized downstream of the amino acid ornithine.
- Spermidine acts as a substrate for the hypusination of elF5A, catalyzed by DHPS and DOHH.
- DFMO inhibits ODC
- DENSPM induces polyamine catabolism.
- Both GC7 and CPX act as inhibitors of hypusination.
- Relative ECAR of (c) GC7- and (d) CPX-treated MEFs (e) MEFs transduced with an IPTG-inducible E/ ' f5a-inducible shRNA; (f) Dhps itoxinox MEFs expressing a 4-OHT-inducible Cre-ER;
- S2 cells D. melanogaster
- MDCK C. familiaris
- MCF-7 H. sapiens
- FIG. 8 (a) ECAR of E/f5a-shRNA transduced MEFs at baseline, or in response to 2- deoxyglucose (2-DG), Oligo, FCCP, and R+A. (b) Fold increase in cell number of Eif5a- shRNA-expressing MEFs over 5 days of culture. Relative ECAR of (c) GC7- and (d) CPX- treated M(IL-4); (e-g) and M(IL-4) transduced with indicated shRNA. (h) Immunoblot analysis of specified proteins in M(IL-4) expressing control, Eif5a, Dhps or EG7-OVA cells expressing control or Dohh-shRNA.
- Hypusinated elF5A maintains TCA cycle and ETC integrity in macrophages.
- FIG. 10 (a) 13 C-palmitate, (b) D- 13 C-glucose, and (c) 3 C-glutamine GCMS trace analysis of specified metabolites in M(IL-4) ⁇ GC7 for 24 hours, (c-e) Isotopologue evaluation of indicated metabolites in 13 C-glutamine-labelled M(IL-4) ⁇ GC7, where d,e indicate oxidative and reductive metabolism, respectively. Red X indicates TCA cycle breaks from proteomics data. All data are mean ⁇ SEM (p* ⁇ 0.05, p** ⁇ 0.005, p** * ⁇ 0.0005). (a) Represents one experiment, (b-f) representative of two experiments.
- FIG. 11 Immunoblot analysis of specified proteins in (a, b) M(IL-4) ⁇ 10 ⁇ GC7 for 22 hours; (c) M0, M(L/y), M(IL-4) (left panel) and M(IL-4) (right panel) ⁇ GC7; (d) EG7-OVA cells ⁇ DFMO/DENSPM for 48 hours; (e) M(IL-4) ⁇ DFMO for 48 hours; (f) M(IL-4) ⁇ 20 ⁇ CPX for 24 hours; (g) M(IL-4) expressing control, Dhps, or Eif5a shRNA. (h) OCR in M(IL-4) treated for 24 hours with GC7 ⁇ dimethylsuccinate.
- FIG. 13 Expression of markers associated with alternative activation in M(IL-4) (a) ⁇ CPX, (b) ⁇ GC7, (c) expressing control or £/r ⁇ 5a-shRNA, (d) expressing control or Dhps-s R A, (e) expressing control or Dohft-shRNA. (f, g) Western blots of indicated proteins in M0, M(LJy), M(IL-4) ⁇ GC7 and (g) analysis of NOS2 expression by flow cytometry in M0, M(L/y) ⁇ GC7 (right panel).
- Figure 14 (a) Activated and control OT-I CD8 + T cells ⁇ the indicated concentration of GC7 were assessed for proliferation on day 3 of culture, (b) CFSE proliferation analysis of OT-I CD8 + T cells treated with GC7 for the indicated time periods, (a-b) representative of one to three experiments.
- FIG. 15 Mitochondrial metabolism is controlled by hypusinated elF5A-dependent translation of metabolic machinery, (a) Relative mRNA expression of indicated genes in GC7-treated M(IL-4). (b) HA-tagged elF5A construct was retrovirally transduced into MEFs and bound mRNAs were immunoprecipitated and submitted for microarray analysis against total RNA.
- FIG. 16 (a) IDH2 MTS and MCM MTS target sequences fused to mCherry. (b) Representative histograms of indicated constructs ⁇ GC7, representative of six independent experiments.
- elF5a and the polyamine pathway could be important for the alternative activation of macrophages (M2 cells) given that they express arginase, the enzyme that converts the amino acid arginine into ornithine, the precursor to polyamines.
- M2 cells macrophages
- arginase the enzyme that converts the amino acid arginine into ornithine, the precursor to polyamines.
- GC7 N1-Guanyl-1 ,7-diaminoheptane
- M2 macrophage differentiation is impaired in the absence of functional elF5a, unlike M1 cells which are unaffected (figure 3).
- the action of GC7 also had profound effects on macrophage metabolism.
- elF5a M2 cells engage oxidative phosphorylation (OXPHOS) to help drive their differentiation
- OXPHOS oxidative phosphorylation
- GC7 treatment significantly decreased OXPHOS in M2 cells (figure 4).
- M1 metabolism was unaffected in the presence of GC7 (data not shown).
- hypusinated elF5a was found to be an important factor in regulating macrophage differentiation and respiration.
- the ability of compounds to inhibit elF5a hypusination can be determined by analyzing the quantity of elF5a-hypusine by western blot in untreated control cells versus treated cells.
- figure 5 an example of this is shown - here, kidney cells were incubated with the deoxyhypusine synthase inhibitor GC7 and the quantity of elF5a-hypusine determined by western blot. GC7-treated cells showed a significant reduction in elF5a-hypusine levels.
- Example 3 Exposure of murine embryonic fibroblasts (MEFs) to 2-difluromethylornithine (DFMO) and to diethylnorspermine (DENSPM)
- Polyamines are synthesized in metabolically active cells 7 (Fig. 6a). To investigate how this pathway contributes to metabolism murine embryonic fibroblasts (MEFs) were exposed to 2- difluromethylornithine (DFMO), an ornithine decarboxylase (ODC) inhibitor 8 , and to diethylnorspermine (DENSPM), which activates the spermidine catabolising enzyme SSAT 9 .
- DFMO 2- difluromethylornithine
- ODC ornithine decarboxylase
- DESPM diethylnorspermine
- OCR oxygen consumption rates
- ECAR an indicator of aerobic glycolysis
- LCMS analysis of treated cells confirmed a drop in spermidine levels, whereas the upstream metabolite ornithine was unaffected (Fig. 6c).
- An important cellular function of spermidine is to provide substrate for DHPS, the rate-limiting enzyme during elF5A H formation (Fig 6a).
- M(IL-4) and LPS/IFN-y-activated macrophages [M(LPS/IFN-y)] do not proliferate to any appreciable extent in v/ ' fro 13,14 , it was possible to assess the role of polyamine biosynthesis on respiration dissociated from its known ability to regulate proliferation (Fig. 8b) 15 .
- Inhibiting elF5A H either pharmacologically or genetically, blocked respiration in M(IL-4) (Fig. 9b-f, Fig. 8h), while again having differential effects on ECAR (Fig. 8c-g).
- metabolites were analyzed by LCMS.
- GC7-treated M(IL-4) (Fig. 9g, h) and £/75a-shRNA-expressing MEFs (Fig. 8i) displayed decreased metabolites associated with the first half of the TCA cycle, as well as additional alterations in other metabolic pathways (Fig. 8j, k).
- M(IL-4) were cultured with 13 C-labeled glucose, glutamine, or palmitate and carbons were traced from these substrates into metabolites.
- GC7-treated M(IL-4) incorporated significantly less carbon from 13 C-glucose into TCA cycle metabolites compared to control cells (Fig. 9i), indicating that DHPS function, and thus elF5A H , regulates TCA cycle engagement.
- Subgroup column6 M IL4 is 2
- TCA cycle enzymes in GC7-treated M(IL-4), as well as in resting macrophages (MO) and M(LPS/IFN-y) were assessed, by western blot.
- TCA cycle enzymes in GC7-treated M(IL-4), as well as in resting macrophages (MO) and M(LPS/IFN-y) were assessed, by western blot.
- succinyl-CoA synthetase Suclgl
- SDH succinate dehydrogenase
- CS citrate synthase
- IDH isocitrate dehydrogenase
- MCM methylmalonyl-CoA mutase
- FIG. 11 b Other enzymes identified by proteomics analysis that feed substrates into the TCA cycle, such as methylmalonyl-CoA mutase (MCM), were also diminished after GC7 treatment (Fig. 11 b), while expression of many enzymes in glycolysis, fatty acid synthesis, and the aspartate-arginosuccinate shunt remained stable (Fig. 11c). Exposing M(IL-4) or lymphoma cells to polyamine synthesis inhibitors also resulted in TCA cycle enzyme deficiencies, as did treatment with CPX or expression of Eif5a and Dhps-s RNA (Fig. 11d- g)-
- Proteomics data of GC7-treated M(IL-4) confirmed the down-regulation of many proteins associated with alternative activation, including CD301 (Table 2).
- GC7 also diminished accumulation in vivo of IL-4 complex (IL-4c)-elicited macrophages in the peritoneal cavity (Fig. 12d).
- Proteins in the polyamine-elF5A H axis were enriched in M(IL-4) compared to M(LPS/IFN-y) (Fig. 12e), supporting the notion that this pathway is dynamically regulated in these cells.
- GC7 in vitro generated memory T cells cultured with GC7 augmented ECAR in response to restimulation, but could not increase OXPHOS (Fig. 12i, Fig. 13k).
- GC7 also blocked T cell proliferation in a dose- dependent manner when present during naive T cell activation (Fig. 14a), but this effect was mitigated when GC7 was introduced days later (Fig. 14b), consistent with an initial requirement for OXPHOS to induce clonal expansion during T cell activation 17,18 .
- LCMV-specific P14 T cells (gp33 + ) were transduced with a retrovirus expressing £/T5a-shRNA, which was introduced after activation, and adoptively transferred these cells into LCMV-infected recipients.
- Donor cells after infection were tracked and it was found that while both control and £/75a-shRNA-transduced cells participated in the primary effector T cell response 8 days after infection, the frequency of £/75a-shRNA-transduced donor cells was decreased in the weeks after infection compared to control cells, indicating that the cells expressing £/T5a-shRNA were less able to form memory T cells (Fig 3j).
- elF5A H regulates the phenotype of immune cells that rely on mitochondrial metabolism.
- RNA-IP immunoprecipitated bound mRNAs
- elF5A H facilitates translation elongation of difficult to translate proteins or motifs, such as those with proline, glycine, or charged amino acids, which can lead to ribosome stalling 4,5 .
- mice specific for LCMV were purchased from Jackson Laboratories. All mice were bred and maintained under specific pathogen free conditions under protools approved by the Animal Welfare Committee of the Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany. Mice used in all in vitro and in vivo experiments were 6-10 weeks of age and were age/sex matched. For lymphocytic choriomeningitis virus (LCMV) infections, mice were immunised with 2x10 5 plaque forming units (PFU) LCMV Armstrong strain by intraperitoneal injection.
- LCMV lymphocytic choriomeningitis virus
- Bone marrow cells were differentiated for 7 days into bone marrow macrophages ( ⁇ ) by culturing in complete medium (RPMI 1640 media supplemented with 10% FCS, 2mM L- glutamine, 100 U/mL penicillin/streptomycin) with 20 ng/mL macrophage colony-stimulating factor (M-CSF; PeproTech).
- M(IL-4) were generated with 20 ng/mL IL-4 overnight from day 7 of culture;
- M(LPS/IFNy) were generated with 20 ng/mL LPS (Sigma) and 50 ng/mL IFN- ⁇ (R&D Systems) overnight from day 7 of culture. All drug treatments on ⁇ began from day 7 of culture.
- N1-guanyl-1 7-diaminoheptane (GC7; Enzo Life Sciences) and ciclopirox (Sigma) were typically used at 10 ⁇ and 20 ⁇ , respectively, unless otherwise stated.
- DHPS FIOXROX Cre-ER MEFs generated as previously described 31 , were cultured in complete DMEM (DMEM supplemented with 10% FCS, 2mM L-glutamine, 100 U/mL penicillin/streptomycin) and generated as previously described 31 , Cre-ER expression was induced with 1 ⁇ 4-OHT (Sigma) for the indicated time period.
- NIH3T3 MEFs purchased from ATCC
- E/ " f5a-shRNA expression was induced with 100 ⁇ isopropyl ⁇ -D-1 thiogalactopyranoside (IPTG, Sigma) for the indicated period of time.
- Madin-Darby Kidney Canine (MDCK) cells were grown in complete DMEM, as was the human breast adenocarcinoma line MCF-7 but with 0.01mg/ml recombinant human insulin.
- D. melanogaster Schneider 2 (S2) cells were cultured without C0 2 at 28°C in complete Schneider's Drosophila medium (Gibco; supplemented with 10% FCS, 50 U/mL penicillin/streptomycin, 25% conditioned complete Schneider's medium).
- OTI splenocytes were activated with OVA peptide (SINFEKL, New England Peptide), or with anti- CD3 and anti-CD28 antibodies, with IL-2 (100 U/mL) in T cell media (RPMI 1640 media supplemented with 10% FCS, 2mM L-glutamine, 100 U/mL penicillin/streptomycin and 55 ⁇ ⁇ -mercaptoethanol) for the indicated length of time.
- OVA peptide SINFEKL, New England Peptide
- IL-2 100 U/mL
- T cell media RPMI 1640 media supplemented with 10% FCS, 2mM L-glutamine, 100 U/mL penicillin/streptomycin and 55 ⁇ ⁇ -mercaptoethanol
- OT-I splenocytes were activated with OVA-peptide and IL-2 (100 U/mL) for 3 days and subsequently cultured in the presence of either IL-2 or IL-15 (10 ng/mL), respectively, for an additional 3 days in TCM.
- Extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) were measured using the Seahorse XFe Bioanalyser (Seahorse Bioscience). 8x10 4 ⁇ were added to seahorse 96 well plates on day 7 of culture and analysed in XF media (non-buffered RPMI 1640 containing 25 mM glucose, 2 mM L-glutamine, and 1 mM sodium pyruvate) the following day after cytokine and drug treatment. For T cells, 2x10 5 were spun down on a poly-D-lysine-coated seahorse 96 well plate.
- MEFs were plated at 4x10 4 cells per well of a 96 well seahorse plate in XF. Prior to analysis, cells were incubated for a minimum of 45 minutes at 37°C in the absence of C0 2 .
- OCR and ECAR were measured under basal conditions, after restimulation with 50 ng/mL phorbal 12-myristate 13-acetate (PMA) and 0.5 ⁇ g/mL ionomycin, or after the addition of the following drugs: 1 ⁇ oligomycin, 1.5 ⁇ fluoro- carbonyl cyanide phenylhydrazone (FCCP), 100 nM rotenone, and 1 ⁇ antimycin A (all Sigma). Measurements were taken using a 96 well Extracellular Flux Analyser (Seahorse Bioscience).
- Isotopomer distributions were measured using a DB5-MS GC column in a 7890 GC system (Agilent Technologies) combined with a 5977 MS system (Agilent Technologies). Correction for natural isotope abundance and calculation of fractional contribution was performed as described elsewhere 32 .
- Metabolites were quantified by LC-MS using HILIC Chromatography on an Acquity UPLC BEH Amide column 1.7 ⁇ , 2.1x100 mm (polyamines) or a Luna NH2 column (all other metabolites) on a 1290 Infinity II UHPLC system (Agilent Technologies) combined with targeted detection in a 6495 MS system (Agilent Technologies). Peak areas were normalized to 13 C labelled internal standard (ISOtopic Solutions).
- cells were washed with ice cold PBS and lysed in 1 x Cell Signaling lysis buffer (20 mM Tris-HCI, [pH 7.5], 150 mM NaCI, 1 mM Na 2 EDTA, 1 mM EGTA, 1% Triton X-100, 2.5 mM sodium pyrophosphate, 1 mM ⁇ - glycerophosphate, 1 mM Na 3 V0 4 , 1 g/mL leupeptin (Cell Signaling Technologies), supplemented with 1 mM PMSF. Samples were frozen and thawed 3 times followed by centrifugation at 20,000 x g for 10 min at 4°C.
- 1 x Cell Signaling lysis buffer 20 mM Tris-HCI, [pH 7.5], 150 mM NaCI, 1 mM Na 2 EDTA, 1 mM EGTA, 1% Triton X-100, 2.5 mM sodium pyrophosphate, 1 mM ⁇ -
- All antibodies were from Cell Signaling Technologies except for anti-ABAT, Anti-ACC, anti- Aconitase 1 , anti-ASL, anti-DLD, anti-DOHH, anti-DHPS, anti-MCM, anti-ODC (Abeam), anti- elF5A (BD Bioscience), anti-hypusine (Merck-Millipore). Electron transport chain complexes were probed with the Total OXPHOS Rodent WB Antibody Cocktail (Abeam).
- activated P14 splenocytes were transduced with luciferase (empty vector) or £ 5a-expressing retrovirus by centrifugation for 90 minutes in media containing hexadimethrine bromide (8 pg/mL; Sigma) and IL-2 (100 U/mL).
- GFP was used as a marker for retroviral expression.
- bone marrow cells exposed to M-CSF were transduced with luciferase (empty vector) or Eif5a-,Dhps-, or ⁇ -expressing retrovirus by centrifugation for 90 minutes in media containing hexadimethrine bromide on day 2 of culture.
- Transduced cells were subsequently drug or cytokine-treated on day 7 of culture and assayed on day 8, sometimes following sorting on day 6.
- GFP was used as a marker for transduction in these cells.
- a G-block construct (IDT) containing mCherry fused to the degron from ODC 33 (HGFPPEVEEQDDGTLPMSCAQESGMDRH*) (mCherry* 39 ) was constructed to reduce half- life of the mCherry fusion protein. Between the cloning sites and mCherry a Gly-Ser-Gly-Ser- Gly flexible linker was included, to allow correct and independent folding of the introduced sequences and mCherry.
- the mCherry deg was cloned into MSCV-I-GFP using Xhol and EcoRI.
- MTS, NLS, or control sequences were ordered as G-blocks (IDT) or as phosphorylated oligos containg Xhol and BamHI compatible overhangs and cloned into MSCV-mCherry de9 -l-GFP using Xhol and BamHI.
- the targeted sequences were as follows: MTS-IDH
- MPKKKRKV (SEQ ID NO: 4) PP (MPPPP) (SEQ ID NO: 5)
- IPTG-inducible MISSION shRNA lentiviral vector pl_KO-puro-IPTG-3xl_acO was purchased either with a shRNA against the 3 " -UTR of the murine elF5A mRNA sequence (custom-made from #SHCLND-NM181582-TRCN0000125229; Sigma) or a corresponding non-target shRNA control (#SHC332-1 EA; Sigma).
- Stable transduction of the lentiviral was performed as previously described 34 using HEK293T cells, the packaging plasmids: pMDLg/pRRE (Gag/Pol), pRSV-Rev (Rev) and phCMV-VSV-G (envelope) as well as the ProFection Mammalian Transfection System Calcium Phosphate Kit (Promega). Positive cells were selected using puromycin.
- MS raw files were analyzed by MaxQuant software and peak lists were searched against the mouse Uniprot FASTA database (concatenated with a database containing common contaminants) by the Andromeda search engine embedded in MaxQuant 38,39 .
- MS1 -based label free quantification (LFQ) was done using maxLFQ algorithm 40 .
- Perseus platform 41 was used to perform data filtering and statistical testing.
- step 1 contaminant hits, reverse identification hits, and proteins "only identified by site” were removed from the dataset.
- LFQ intensities were log 2 transformed.
- Step 3 involved removal of missing quantitative data points to minimize the number of missing values in the dataset and this was followed by missing value data imputation using a normal distribution simulating the distribution of low abundant proteins in the dataset.
- Student's T-test was utilized to define differentially expressed proteins employing a two-fold change as a cut-off at a 5% FDR.
- Flow cytometric staining was performed as previously described 42 . All fluorochrome- conjugated monoclonal antibodies were from (eBioscience), except for anti-CD301 (BioRad). Both NOS2 and RELMa protein levels were quantified after fixation and permeabilisation using the transcription buffer staining set (eBioscience) and monoclonal antibodies against NOS2 (Santa Cruz) and RELMa (Peprotech). Cells were stained with Live/Dead viability dye (Thermo) prior to antibody staining. Cells were labelled with CFSE as described 43 .
- P14 TCR transgenic T cells were identified in vivo and ex vivo using the congenic marker CD45.1 and H-2D b GP 33 -4i MHC Class I tetramer.
- Cells were collected on LSR II and Fortessa flow cytometers (BD Biosciences) and analysed using FlowJo (TreeStar) software. Cells were sorted using a FACS Aria II. Cells were imaged using a Zeiss spinning disk confocal microscope with an Evolve (EMCCD) camera. Cells were kept in a humidified incubation chamber at 37°C with 5% C0 2 during image collection. Images were deconvolved and analysed using ImageJ (NIH).
- EMCD Evolve
- RNA isolations were done by using the RNeasy kit (Qiagen) and single-strand cDNA was synthesized using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems). All RT-PCR was performed with Taqman primers using an Applied Biosystems 7000 sequence detection system. The expression levels of mRNA were normalized to the expression of a housekeeping gene ( ⁇ -actin). For RNA-Seq, total RNA samples were extracted using RNeasy isolation kit (Qiagen).
- Murine elF5A-1 open reading frame were PCR-amplified, fused with a C-terminal HA-tag and cloned into the pMSCV-puro vector (Clontech, pMSCV-elF-5A1-HA-puro).
- the following primers were used for cloning:
- NIH3T3 cells Ecotropic retroviruses for the transduction of NIH3T3 cells were obtained by transient calcium-phosphate-mediated transfection of the retroviral vectors into the packaging cell line Phoenix eco.
- NIH-3T3 cells were transduced by adding the filtered retrovirus-containing supernatant and selected with puromycin.
- 3.5x10 6 transduced cells were seeded in triplicates on a 100-mm 3 culture dish the day before.
- Cells were homogenized in pre-cooled lysis buffer containing 100 mM KCI, 5 mM MgCI 2 , 10 mM HEPES, 0.5 % Nonidet-P40, 1 mM DTT, 100 U/mL Ribolock (Invitrogen) and 25 ⁇ /mL Protease Inhibitor Cocktail (Sigma). Lysates were incubated for 15 minutes, cellular debris was pelleted at 20,000 g for 15 minutes and the supernatants were incubated with 50 pL magnetic anit-HA-MicroBeads (Miltenyi Biotec) for 30 minutes on ice in the dark.
- lysis buffer containing 100 mM KCI, 5 mM MgCI 2 , 10 mM HEPES, 0.5 % Nonidet-P40, 1 mM DTT, 100 U/mL Ribolock (Invitrogen) and 25 ⁇ /mL Protease Inhibitor Cocktail (Sigma). Lysates were incubated
- RNA-protein- anti-HA-MicroBead complexes were eluted with lysis buffer containing 0.1 % SDS and 0.3 pg/pL proteinase K and incubated for 35 minutes at 50 C. Then the anti-HA-MicroBeads were removed by applying the solution to the ⁇ Columns and eluting the RNA containing fraction with DEPC-treated water. The RNA was cleaned and concentrated with the NucleoSpin RNA XS Kit (Macherey-Nagel).
- RNA from the same cells were isolated as back ground control. RNA quantity and quality were evaluated by Nanodrop ND1000 Spectrophotometer and Agilent 2100 Bioanalyzer measurement. Procedures for cDNA synthesis, labelling and hybridisation were carried out according to the manufacturer's protocol (Affymetrix 3'-IVT Express Kit) starting with 100 ng of each RNA sample (three samples from immunoprecipitation and three samples of total RNA without immunoprecipitation). The experiments were performed using Affymetrix Mouse Genome 430 2.0 GeneChip. All reactions were performed in triplicates. The signals were processed with a target value of 300 using Affymetrix GeneChip Operating Software 1.4.
- P14 TCR transgenic CD45.1 T cells were activated in vitro with gp33-41 peptide and transduced with E/ " f5a-shRNA.
- a day later 5x10 5 T cells were transferred into CD45.2 C57BL/6 congenic recipient mice on day one of infection. Blood samples were collected at the indicated time points and analysed by flow cytometry.
- MaxLFQ normalization and maximal peptide ratio extraction
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Biomedical Technology (AREA)
- Hematology (AREA)
- Chemical & Material Sciences (AREA)
- Cell Biology (AREA)
- Urology & Nephrology (AREA)
- Molecular Biology (AREA)
- Microbiology (AREA)
- Medicinal Chemistry (AREA)
- Tropical Medicine & Parasitology (AREA)
- Toxicology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pathology (AREA)
- Food Science & Technology (AREA)
- Biotechnology (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- General Health & Medical Sciences (AREA)
- General Physics & Mathematics (AREA)
- Developmental Biology & Embryology (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
The present invention relates to a method for determining whether a compound has the capability of inhibiting elF5a activation, comprising (a) contacting a sample comprising cells being capable of differentiating into M2 macrophages with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and (b) quantifying in the sample the cells having a M2-phenotype, wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting elF5a activation.
Description
ALTERNATIVE ACTIVATION OF MACROPHAGES (M2 CELLS) THROUGH THE
POLYAMINE-eIF5a-HYPUSINE AXIS
The present invention relates to a method for determining whether a compound has the capability of inhibiting elF5a activation, comprising (a) contacting a sample comprising cells being capable of differentiating into M2 macrophages with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and (b) quantifying in the sample the cells having a M2-phenotype, wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting el F5a activation.
In this specification, a number of documents including patent applications and manufacturer's manuals are cited. The disclosure of these documents, while not considered relevant for the patentability of this invention, is herewith incorporated by reference in its entirety. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference.
Much has been undertaken in recent years to understand the role of metabolism in immune cell function and differentiation. Metabolic switches in mammalian cells have been the focus of attention since Otto Warburg discovered that cancer cells use aerobic glycolysis even during normoxia. Since then many studies have investigated mechanisms that underlie the Warburg effect, however, the precise mechanisms cells utilize to modulate mitochondrial respiration remains unclear. However, the polyamine synthesis pathway represents an overlooked metabolic process that might be important for immune cell fate. Polyamine biosynthesis is a highly regulated pathway that occurs in the cytoplasm of all cells. In eukaryotes, amongst other wide-ranging functions the polyamine spermidine is needed to hypusinate the eukaryotic translation elongation factor 5A (elF5A)1.The polyamine family of molecules is comprised of small polycationic metabolites, the most well described being putrescine, spermidine and spermine. They are formed by a series of enzymatic reactions that begin with the conversion of the amino acid ornithine to putrescine by ornithine decarboxylase (ODC) (figure 1 ).
A notable feature of the polyamine pathway is its role in the activation of the translation elongation factor elF5a. This factor is activated through the unique modification of one of its lysine residues, which is converted into the unusual amino acid hypusine in the presence of spermidine to form elF5a-hypusine (elF5aHyp) (figure 2). elF5a, the true function of which has only recently been described (Gutierrez E et al., elF5A promotes translation of polyproline motifs, Molecular Cell 51 : 35-45 (2013) and Nakanishi, S. and Cleveland, J.L. (2016), Targeting the polyamine-hypusine circuit for the prevention and treatment of cancer, Amino Acids 1-10), is thought to be the only protein to contain the hypusine modification. elF5a is an essential cellular protein required for the production of a certain subset of proteins. Its function is dependent on the two enzymes deoxyhypusine hydroxylase (DOHH) and deoxyhypusine synthase (DHPS) that use the metabolite spermidine to mediate the activation of elF5a.
Although some knowledge on the polyamine pathway and the action of elF5a has been collected in the prior art, a full picture of all downstream cellular processes being governed by the polyamine pathway and the action of elF5a is not available. It is thus also not fully known which cellular processes can be controlled by interfering with the activation of elF5a.
The present invention aims at providing novel screening methods and therapeutic uses encompassing to interfere with the activation of elF5a.
The present invention therefore relates in a first aspect to a method for determining whether a compound has the capability of inhibiting elF5a activation, comprising (a) contacting a sample comprising cells being capable of differentiating into M2 macrophages with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and (b) quantifying in the sample the cells having a M2-phenotype, wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting elF5a activation
The nature of the compound to be tested for its capability of inhibiting elF5a activation is not particularly limited and preferred examples thereof will be described herein below. As used herein "elF5a" refers to the protein eukaryotic translation initiation factor 5A-1 that is encoded by the EIF5A gene. The compound may interfere with any step in the polyamine pathway and the subsequent hypusine-elF5a formation as shown in Figures 1 and 2 as all these steps are required for the final activation of elF5a. As shown in Figures 1 and 2, activated elF5a is hypusine-elF5a. Hypusine is the amino acid N (e)-(4-amino-2-hydroxybutyl)-lysine. It is
synthesized on elF5A at a specific lysine residue from the polyamine spermidine by the two catalytic steps shown in Figure 2. The compound having the capability of inhibiting elF5a activation may interfere with any one of the precursors of hypusine-elF5a, e.g. spermidine or deoxyhypusine-elF5a, so that hypusine-elF5a cannot be generated as one or more of the required precursors are absent or not processible. The compound having the capability of inhibiting elF5a activation may also interfere with one or more of the enzymes in the pathway, e.g. spermidine synthase, DHS or DOHH, so that the precursors no longer can be processed into hypusine-elF5a as the required enzymes are absent or inhibited. The method of the invention may be performed in high through-put format for testing several compounds at the same time. High-throughput methods are capable of screening up to several thousand compounds in parallel. High-throughput assays, independently of being biochemical, cellular or other assays, generally may be performed in wells of microtiter plates, wherein each plate may contain 96, 384 or 1536 wells. Handling of the plates, including incubation at temperatures other than ambient temperature, and bringing into contact of test compounds with the assay mixture is preferably effected by one or more computer-controlled robotic systems including pipetting devices. In case large libraries of test compounds are to be screened and/or screening is to be effected within short time, mixtures of, for example 10, 20, 30, 40, 50 or 100 test compounds may be added to each well. In case a well exhibits inhibition of elF5a activation, said mixture of test compounds may be deconvolved to identify the one or more test compounds in said mixture giving rise to the inhibition of elF5a activation.
The compound having the capability of inhibiting elF5a activation may either act on the DNA level (e.g. a siRNA against the mRNA encoding DHS or DOHH) or on the protein or precursor level (e.g. an antibody against the DHS or DOHH enzyme or an antibody against spermidine). If follows that the compound having the capability of inhibiting elF5a activation may be (i) a compound inhibiting the expression of an mRNA encoding an enzyme being required for the activation of elF5a, or (ii) a compound inhibiting an enzyme or a precursor being required for the activation of elF5a.
Compounds under option (i) include compounds interfering with the transcriptional machinery and/or its interaction with the promoter of said gene and/or with expression control elements remote from the promoter such as enhancers. Compounds under option (i) also include compounds interfering with the translational machinery. The compound inhibiting the expression of a mRNA encoding an enzyme being required for the activation of elF5a inhibits the expression and/or translation of said mRNA, for example, by specifically interfering with
the promoter region controlling the expression of the mRNA or leading to the silencing of the mRNA. Preferably, the transcription and/or translation of the mRNA is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98% and most preferred by about 100% (e.g., as compared to the same experimental set up in the absence of the compound).
Compounds under option (ii) cause said protein or precursor to perform its function with lowered efficiency. The compound preferably specifically inhibits the activity of the enzyme or precursor. The activity of the enzyme is its capability of catalyzing a chemical reaction being involved in the pathway leading to hypsuine-elF5a. The activity of the precursor is that it can be processed into another precursor further upstream in the pathway of elF5a activation or hypsuine-elF5a itself. Preferably, the activity of the enzyme or precursor is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98%, and most preferably about 100% (e.g., as compared to the same experimental set up in the absence of the compound).
In accordance with the method of the invention the capability of the compound of inhibiting elF5a is determined by comparing the amount of cells having an M2-phenotype in the presence of the compound to that in the absence of the compound. In case the amount of cells having an M2-phenotype is reduced in the presence of the compound the compound is capable of inhibiting elF5a activation. A compound is preferably qualified as being capable of inhibiting elF5a activation in case the amount of cells having a M2-phenotype is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98% and most preferred by about 100% (e.g., as compared to the same experimental set up in the absence of the compound).
In this connection a sample comprising cells being capable of differentiating into M2 macrophages is contacted with the test compound. A cell being capable of differentiating into M2 macrophages may be a precursor cell of a M2 macrophage. Preferred examples are hemoblasts, common myeloid progenitor cells, myeloblasts and monocytes. The most preferred precursor cells of macrophages (including M2 macrophages) are monocytes or other progenitor cells derived from the bone marrow, such as the common myeloid progenitor, that can differentiate into macrophages under the control of factors such as colony stimulating factor 1 (CFS-1 , otherwise known as macrophage-colony stimulating factor 1 ). Monocytes are the direct precursor cells of macrophages in the hematopoietic cell lineage. Moreover and as shown in the examples, unactivated macrophages (i.e. M0 cells) can be polarised into M2 macrophages, e.g. when cultured in the presence of IL-4. As will be
further detailed herein below, M1 and M2 macrophage activation represent two possible, distinct fates that unactivated macrophages can differentiate into. Hence, the sample comprising cells being capable of differentiating into M2 macrophages preferably comprises unactivated macrophages (MO macropahges) and/or monocytes.
The sample can be a body sample or in vitro cultured cells. Preferred body samples are a tissue sample (e.g. bone marrow derived macrophages or organ biopsy), including a tumor sample derived from a biopsy or resection, a blood sample (e.g. whole blood, plasma or serum), spinal fluid, and fluid from bronchoalveolar lavage. Non-limiting examples of in vitro cultured cells are the human macrophage or monocytic cell lines THP-1 (ATCC TIB-202), U937 (ATCC CRL-1593.2), KG-1 (KG-1 ATCC CCL-246), MV-4-11 (ATCC CRL-959), SC (ATCC CRL-9855), MD (ATCC CRL-9850), Mono Mac 6 (DSM ACC 124) or hMo-PB cells (commercially available human "untouched" monocytes isolated from fresh peripheral blood, Cat.No. CSC-C1671 ). Isolated monocytes that can be used in the context of the present invention are, for example, available from Lonza Group AG (see http://www.lonza.com/products-services/bio-research/primary-cells/hematopoietic- cells/peripheral-blood- cells/monocytes.aspx?gclid=CjwKEAiAvs7CBRC24rao6bGCoiASJABaCt5DHiBzsi1 aEKCK8t 5vXYK356kw4IWEefSGUUhyUIAsNhoCNDLw_wcB) or stem cell technologies (see www.stemcell.com/human-peripheral-blood-monocytes-frozen.html).
Unactivated macrophages derived from precursors undergo specific differentiation depending on the local tissue environment. They respond to environmental cues within tissues such as damaged cells, activated lymphocytes, or microbial products, to differentiate into distinct functional phenotypes. For instance, monocytes in the blood can enter the tissue during inflammation or insult and are, depending on the local microenvironment, polarised towards an M1 or M2 phenotype.The M1 macrophage phenotype is characterized by the production of high levels of pro-inflammatory cytokines, an ability to mediate resistance to pathogens, strong microbicidal properties, high production of reactive nitrogen and oxygen intermediates, and promotion of Th1 responses. In contrast, M2 macrophages are characterized by their involvement in parasite control, tissue remodeling, immune regulation, tumor promotion and efficient phagocytic activity. In accordance with the method of the invention the sample comprising cells being capable of differentiating into M2 macrophages is cultured under conditions that allow for the differentiation of the precursor cells into M2- type macrophages in the absence of the compound. Conditions that can be used in order to differentiate monocytes and/or macrophages into M2 macrophages are known in the art. Preferred examples are the differentiation of monocytes and/or unactivated macrophages
(MO cells) into M2 macrophages with (i) IL-4, (ii) IL-10, (iii) glucocorticoids (GC), or (iv) GC plus TGF-beta plus. A more preferred example is the differentiation of monocytes and/or unactivated macrophages (MO cells) into M2 macrophages with IL- 4, preferably in conjunction with M-CSF. In this respect it is further preferred to use IL-4 at a concentration of about 10ng/mL and, if present, M-CSF at a concentration of about 20ng/m.
As used herein the term "macrophage" designates a cell of myeloid origin. Macrophages are large white blood cells, occurring principally in connective tissue and in the bloodstream. They ingest foreign particles and infectious microorganisms by phagocytosis andhave the capacity for antigen presentation. Macrophages are classified in the art into MO, M1 and M2 macrophages.
MO macrophages are also termed unactivated macrophages. They express the classical macrophage markers CD11b and CD68 but exhibit low expression of the activation markers MHC class II molecules, CD80, and CD86. They are moreover negative for cytokine expression in the form of IL-12, IL-10 and IL-1 beta. Hence, a MO cell is preferably a cell expressing CD11b and CD68 but not expressing IL-12, IL-10 and IL-1 beta. More preferably, a MO cell is a cell expressing CD11b and CD68 but not expressing IL-12, IL-10 and IL-1 beta, and expressing low amounts of MHC class II molecules, CD80, and CD86.
A M1 macrophage is defined herein as a macrophage that was activated in the presence of a bacterium, virus, or a bacterial or viral compound (such as and preferably LPS) and optionally also in the presence of IFN-gamma. Accordingly, these cells can be denoted as M(LPS) or M(LPS+IFN) macrophages. M(LPS) and M(LPS+IFN) macrophages express the classical macrophage markers CD68 and CD11 b. Moreover these cells express - in contrast to MO cells - high amounts of MHC class II molecules, CD80 and CD86 expression. These cells also express the cytokines IL-1 beta, IL-12, and TNFalpha. Hence, a M1 cell is preferably a cell expressing CD11 b and CD68, IL-1beta, IL-12, and TNFalpha. More preferably, a M1 cell is a cell expressing CD11 b, CD68, IL-12, IL-10 and IL-1 beta and expressing high amounts of MHC class II molecules, CD80, and CD86.
A M2 cell is also designated as alternatively activated macrophage in the art and hence as a macrophage that was activated in the absence of a bacterium, a virus, or a bacterial or viral compound (such as and preferably LPS) and IFN-gamma. Preferably, a M2 macrophage as used herein refers to a macrophage cell that was activated in the presence of a compound selected from IL-4, IL-10, glucocorticoids (GC),and GC plus TGF-beta plus, or any combination of these compounds. Based on the mode of activation M2 cells are also
designated M(IL-4), M(IL-10), M(GC) or M(GC+TGF-beta) cells. The M2 cells herein are preferably M(IL-4), M(IL-10), M(GC) or M(GC+TGF-beta) cells and are most preferably M(IL- 4) cells. M(IL-4) cells express CD68, GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, but do not express MARCO and CD163. M(IL-10) cells express CD68, SOCS3 and IL-4Ra. M(GC) cells express CD68, CD163, STAB1 , MARCO, TGFBR2, ADORA3. Finally, M(GC+TGF-beta) cells express CD68, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, IL17RB. An overview of the expressed markers being characteristic for M2 cells that were activated by IL-4, IL-10, (GC), and GC plus TGF-beta, respectively, is provided in Table 1.
Table 1
Herein M(IL-4), M(IL-10), M(GC) and M(GC+TGF-beta) cells are preferably identified by the presence or absence of at least three of the respective markers listed in Table 1. For instance, M(IL-10) cells are preferably identified by the presence of CD68, SOCS3 and IL- 4Ra. (M(IL-4)) cells are, for example, preferably identified by the presence of CCL17 and CCL18, and the absence of MARCO. M(IL-4), M(GC) and M(GC+TGF-beta) cells are more preferably identified by the presence or absence of at least six of the markers listed in Table 1. For example, (M(GC)) cells are more preferably identified by the presence of CD68, CD163, STAB1 , MARCO, TGFBR2, ADORA3. (M(IL-4)) cells are, for example, preferably identified by the presence of MRC1 , STAB1 , F13A1 , TGFB1 and MMP12, and the absence of CD163. (M(GC+TGF)) cells are most preferably identified by the presence of CD68, ID3,
RGS1 , pSMAD2, TGFBR2, ALOX5AP, IL17RB. (M(GC+TGF)) cells are most preferably identified by the presence of CD68, GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15 and CD200R, and the absence of CD163 and MARCO.
In addition, CD206 is a general marker of M2 cells in vivo.
Hypusination of elF5A is a conserved process critical for TCA (i.e. tricarboxylic acid) cycle integrity and mitochondrial respiration. Hypusinated elF5A (elF5AH) enables translation of a subset of TCA cycle and mitochondrial electron transport chain (ETC) enzymes that mediate stable TCA cycle flux and oxidative phosphorylation (OXPHOS). Inhibition of deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH), the enzymes that hypusinate elF5A2,3, leads to a break in the TCA cycle that is defined by the loss of elF5AH-regulated enzymes and associated metabolic flux into the TCA cycle. As can be taken from the examples herein the experimental results show that elF5AH facilitates efficient production of proteins with difficult to translate mitochondrial targeting sequences (MTS), an activity that is consistent with a known function of elF5AH in overcoming ribosome stalling4,5. In particular, it was surprisingly found that the activation of elF5a along the polyamine pathway is important for the alternative activation of M2 cells. It is shown in the examples that elF5AH regulates the differential activation of macrophages (M2 cells), a cell type whose divergent functional fates are defined by a metabolic switch between respiration and glycolysis6. The inhibition of hypusination prevents OXPHOS-dependent alternative activation of these cells, while leaving glycolysis-dependent classical activation intact. As is exemplarily demonstrated in the examples the activation of elF5a (i.e. elF5aHyp formation) was blocked by N1-Guanyl-1 ,7- diaminoheptane (GC7) and it was observed that thereby M2 macrophage differentiation is prevented. It was not known before that the activation of elF5a is required for the differentiation of precursor cells into M2 cells. Based on this surprising finding it is now possible to test whether a compound is capable of inhibiting the activation of elF5a based on a read-out relying on quantifying the frequency of M2-phenotype cells in samples, such as tissue biopsies or peripheral blood. These results also have implications for modulating metabolism in immune cells to therapeutically regulate inflammation.
In accordance with a preferred embodiment of the method of the invention, quantifying the cells having a M2-phenotype comprises measuring the amounts of at least one marker molecule selected from the group consisting of GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB.
As detailed herein above and as shown in Table 1 , M2 macrophages are phenotypically characterized by the expression of a specific set of markers. Non-limiting examples of such markers are GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB. One or more of these markers can be used in order to quantify the cells having a M2-phenotype is step (b) of the method of the invention. Using more than one cell surface markers may increase the reliability of determining the cells having a M2-phenotype. With increasing preference at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 and at least 10 markers are measured.
As also detailed herein above, the specific set of cell surface markers on M2 macrophages depends on the conditions used to allow for the differentiation into M2 macrophages.
In case IL-4 is used GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15 and CD200R are expressed as markers. Accordingly, in case the conditions in step (a) of the claimed method comprise IL-4 quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15 and CD200R. With increasing preference at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 and at least 10 of these markers are measured. Most preferably all of these markers are measured.
In case IL-10 is used SOCS3 and IL-4Ra are expressed as markers. Accordingly, in case the conditions in step (a) of the claimed method comprise IL-10 quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of SOCS3 and IL-4Ra. More preferably both of these markers are measured.
In case GC is used CD163, STAB1 , MARCO, TGFBR2 and ADORA3 are expressed as markers. Accordingly, in case the conditions in step (a) of the claimed method comprise GC quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of CD163, STAB1 , MARCO, TGFBR2 and ADORA3. With increasing preference at least 2, at least 3 and at least 4 of these markers are measured. Most preferably all five of these markers are
measured.
In case GC plus TGF-beta is used ID3, RGS1 , pSMAD2, TGFBR2, AL0X5AP and IL17RB are expressed as markers. Accordingly, in case the conditions in step (a) of the claimed method comprise GC plus TGF-beta quantifying the cells having a M2-phenotype in step (b) preferably comprises measuring the amounts of at least one marker molecule selected from the group consisting of ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP and IL17RB. With increasing preference at least 2, at least 3, at least 4 and at least 5 of these markers are measured. Most preferably all six of these markers are measured.
The absence of a particular marker can further help to define cells having a M2-phenotype. Non-limiting examples are phosphorylated STAT1 and IL-12. In case IL-4 is used in the conditions allowing for the differentiation into M2 macrophages furthermore the absence of CD163 and MARCO can further help to define cells having a M2-phenotype.
In accordance with a further preferred embodiment of the method of the invention, quantifying the cells having a M2-phenotype comprises flow cytometry analysis, quantitative PCR, and/or western blot analysis. The skilled person is aware of several methods being suitable for quantifying cells having a M2-phenotype. Non-limiting preferred examples of such methods are cytometry analysis, quantitative PCR, and/or western blot analysis. These methods are employed depending on whether nucleic acid or protein is measured. Quantitative PCR (qPCR or real-time PCR or RT-PCR) uses the linearity of DNA amplification to determine absolute or relative amounts of a known sequence in a sample. By using a fluorescent reporter in the reaction, it is possible to measure DNA generation. In quantitative PCR, DNA amplification may be monitored at each cycle of PCR. Quantitative PCR also lends itself to relative studies. A reaction may be performed using primers unique to each region to be amplified and tagged with different fluorescent dyes. Several commercially available quantitative thermal cyclers include multiple detection channels. In this multiplex system, the amount of target DNA/cDNA can be compared to the amount of a housekeeping sequence e.g. GAPDH or β-actin. Two types of detection chemistries are most commonly used for quantitative PCR. The first uses an intercalating dye that incorporates into double-stranded DNA. Of these fluorescent dyes, SYBR® Green I dye is the most common one used. This detection method is suitable when a single amplicon is being studied, as the dye will intercalate into any double-stranded DNA generated. The second
detection method uses a primer or oligonucleotide specific to the target of interest, as in TaqMan® probes, Molecular Beacons™, or Scorpion primers. The oligonucleotide is labeled with a fluorescent dye and quencher. The oligonucleotide itself has no significant fluorescence, but fluoresces either when annealed to the template (as in molecular beacons) or when the dye is clipped from the oligonucleotide during extension (as in TaqMan probes). Multiplex PCR is possible by using dyes with different fluorescent emissions for each primer. In order to quantify cells having a M2-phenotype by quantitative PCR preferably a gene encoding a marker selected from the group consisting of GATA3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB is amplified.
Western blotting is an important technique used in cell and molecular biology. By using a western blot, researchers are able to identify specific proteins from a complex mixture of proteins extracted from cells. The technique uses three elements to accomplish this task: (1 ) separation by size, (2) transfer to a solid support, and (3) marking target protein using a proper primary and secondary antibody to visualize. Western blots can be carried out in a quantitative manner. The quantification may be based on the differential densitometry of the associated chemiluminescent and/or fluorescent signals from the blots (see, for example, Tyloer and Posh, BioMed Research International, Volume 2014 (2014), Article ID 361590). In order to quantify cells having a M2-phenotype by quantitative western blot analysis preferably a protein selected from the group consisting of GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB is analysed.
The most preferred way to quantify cells having a M2-phenotype is by cytometry analysis. Flow cytometry is an analytical cell-biology technique that utilizes light to count and profile cells in a heterogenous fluid mixture. Flow cytometry is a particularly and preferred powerful method because it allows a researcher to rapidly, accurately, and simply collect data related to many parameters from a heterogeneous fluid mixture containing live cells. In immunology flow cytometry is used to identify, separate, and characterize various immune cell subtypes by virtue of their size and morphology. When additional information is required, antibodies tagged with fluorescent dyes, and raised against highly specific cell surface antigens (e.g. clusters of differentiation or CD markers) can be used to identify, segregate and quantify specific sub-populations within a larger group. In order to quantify cells having a M2- phenotype by cytometry analysis preferably a protein selected from the group consisting of
CD163, GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB is analysed.
In accordance with another preferred embodiment of the method of the invention, the compound is an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, protein drug or small molecule. The aptamer, ribozyme, antibody, small molecule, protein drug, siRNA, a shRNA or an antisense oligonucleotide of this embodiment specifically binds to / interacts with a compound being involved in the activation of elF5a thereby inhibiting the compound. The compounds are the precursor and enzymes (including the mRNA encoding the enzymes) as shown in Figures 1 and 2 and the respective nucleic acid molecules encoding them.
As shown in Figures 1 and 2, the enzymes ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH) are involved in the activation of elF5a. The mRNA of human ornithine decarboxylase is shown in SEQ ID NO: 1 , of human spermidine synthase in SEQ ID NO: 3, of human spermine synthase in SEQ ID NO: 5, of human DOHH in SEQ ID NO: 7 and of human DDHPS in SEQ ID NO: 9. The amino acid sequence of human ornithine decarboxylase is shown in SEQ ID NO: 2, of human spermidine synthase in SEQ ID NO: 4, of human spermine synthase in SEQ ID NO: 6, of human DOHH in SEQ ID NO: 8 and of human DHPS in SEQ ID NO: 10. Within the mRNAs SEQ ID NOs 7 and 9 and within the amino acid sequences SEQ ID NOs 8 and 10 are preferred. This is because in the polyamine synthesis pathway the mRNAs of SEQ ID NOs 7 and 9 encode the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a and the amino acid sequences of SEQ ID NOs 8 and 10 are the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a. These enzymes catalyze the deoxyhypusination (i.e. the addition of deoxyhypusine) and the hypusination (i.e. the addition of hypusin) of elF5a. As elF5a is currently thought to be the only protein containing the hypusine modification, a compound targeting the mRNA of SEQ ID NOs 7 or 9 or the amino acid sequence of SEQ ID NOs 8 of 10 is expected to prevent unwanted off target effects. The term "aptamer" in accordance with the present invention refers to DNA or RNA molecules being either in the natural D-conformation or in the L-conformation ("spiegelmer") that usually have been selected from random pools based on their ability to bind other
molecules. Aptamers have been selected which bind nucleic acid, proteins, small organic compounds, and even entire organisms. Hence, apatamer can be designed against the precursors, enzymes and the mRNA encoding the enzymes. A database of aptamers is maintained at http://aptamer.icmb.utexas.edu/. More specifically, aptamers can be classified as DNA or RNA aptamers or peptide aptamers. Whereas the former consist of (usually short) strands of oligonucleotides, the latter consist of a short variable peptide domain, attached at both ends to a protein scaffold. Nucleic acid aptamers are nucleic acid species that have been engineered through repeated rounds of in vitro selection or equivalently, SELEX (systematic evolution of ligands by exponential enrichment) to bind to various molecular targets such as small molecules, proteins, nucleic acids, and even cells, tissues and organisms. Aptamers offer the utility for biotechnological and therapeutic applications as they offer molecular recognition properties that rival those of the commonly used biomolecules, in particular antibodies. In addition to their discriminate recognition, aptamers offer advantages over antibodies as they can be engineered completely in a test tube, are readily produced by chemical synthesis, possess desirable storage properties, and elicit little or no immunogenicity in therapeutic applications. Non-modified aptamers are cleared rapidly from the bloodstream, with a half-life of minutes to hours, mainly due to nuclease degradation and clearance from the body by the kidneys, a result of the aptamer's inherently low molecular weight. The rapid clearance of aptamers can be an advantage in applications such as in vivo diagnostic imaging. Several modifications, such as 2'-fluorine-substituted pyrimidines, polyethylene glycol (PEG) linkage, etc. are available to scientists with which the half-life of aptamers easily can be increased to the day or even week time scale.
The term "ribozymes" refers to RNA molecules that act as enzymes in the absence of proteins. These RNA molecules act catalyticly or autocatalyticly and are capable of cleaving e.g. other RNAs at specific target sites but they have also been found to catalyze the aminotransferase activity of the ribosome. Hence, the ribozymes may act on the mRNA encoding the enzymes being involved in elF5a activation. Selection of appropriate target sites and corresponding ribozymes can be done as described for example in Zaher and Unrau (2007), RNA, 13 (7): 1017-1026. Examples of well-characterized small self-cleaving RNAs are the hammerhead, hairpin, hepatitis delta virus, and in vitro-selected lead- dependent ribozymes. The principle of catalytic self-cleavage has become well established in the last 10 years. The hammerhead ribozymes are characterized best among the RNA molecules with ribozyme activity. Since it was shown that hammerhead structures can be integrated into heterologous RNA sequences and that ribozyme activity can thereby be transferred to these molecules, it appears that catalytic sequences for almost any target sequence can be created, provided the target sequence contains a potential matching
cleavage site. The basic principle of constructing hammerhead ribozymes is as follows: An interesting region of the RNA, which contains the GUC (or CUC) triplet, is selected. Two oligonucleotide strands, each with 6 to 8 nucleotides, are taken and the catalytic hammerhead sequence is inserted between them. Molecules of this type were synthesized for numerous target sequences. They showed catalytic activity in vitro and in some cases also in vivo. The best results are usually obtained with short ribozymes and target sequences.
The aptamers and ribozymes may comprise modified nucleotides, such as locked nucleic acids (LNAs).
The term "antibody" as used in accordance with the present invention comprises, for example, polyclonal or monoclonal antibodies. Furthermore, also derivatives or fragments thereof, which still retain the binding specificity, are comprised in the term "antibody". Antibody fragments or derivatives comprise, inter alia, Fab or Fab' fragments, Fd, F(ab')2, Fv or scFv fragments, single domain VH or V-like domains, such as VhH or V-NAR-domains, as well as multimeric formats such as minibodies, diabodies, tribodies, tetrabodies or chemically conjugated Fab'-multimers (see, for example, Altshuler et al., Biochemistry (Mosc). 2010 Dec; 75(13):1584-605, Holliger and Hudson, Nat Biotechnol., 2005; 23(9): 1 126-36). The term "antibody" also includes embodiments such as chimeric (human constant domain, non- human variable domain), single chain and humanized (human antibody with the exception of non-human CDRs) antibodies. Various techniques for the production of antibodies and fragments thereof are well known in the art and described, e.g. in Altshuler et al., Biochemistry (Mosc). 2010 Dec; 75(13): 1584-605. Thus, polyclonal antibodies can be obtained from the blood of an animal following immunisation with an antigen in mixture with additives and adjuvans and monoclonal antibodies can be produced by any technique which provides antibodies produced by continuous cell line cultures. Examples for such techniques are described, e.g. Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1988 and Harlow and Lane, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1999 and include the hybridoma technique originally described by Kohler and Milstein, Nature 256 (1975), 495-497, the trioma technique, the human B-cell hybridoma technique (see e.g. Kozbor, Immunology Today 4 (1983), 72; Milstein, C (1999), BioEssays 21 (1 1 ): 966-73.) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. (1985), 77-96). Furthermore, recombinant antibodies may be obtained from monoclonal antibodies or can be prepared de novo using various display methods such as phage, ribosomal, mRNA, or cell display. A suitable system for the
expression of the recombinant (humanized) antibodies or fragments thereof may be selected from, for example, bacteria, yeast, insects, mammalian cell lines or transgenic animals or plants (see, e.g., US patent 6,080,560; Holliger and Hudson, Nat Biotechnol., 2005; 23(9):1126-36). Further, techniques described for the production of single chain antibodies (see, inter alia, US Patent 4,946,778) can be adapted to produce single chain antibodies specific for the target of this invention. Surface plasmon resonance as employed in the BIAcore system can be used to increase the efficiency of phage antibodies. Antibodies may be directed to proteins or small organic molecules. Hence, the antibody used by the present invention may act on one of the enzymes or precursors being involved in elF5a activation. The antibody thus preferably specifically binds to an amino acid sequence being selected from SEQ ID NOs 2, 4, 6, 8 and 10, preferably from SEQ ID NOs 8 and 10, or a precursor being selected from ornithine, spermidine, spermine, and elF5a-deoxyhypusine. In these cases the antibody inhibits the activity of elF5a by inhibiting an enzyme or a precursor being required for the activation of elF5a. The antibody more preferably directly targets elF5a- hypusine thereby inhibiting its activity.
The term "protein drug" designates designer drugs that are derivatives of human proteins. These proteins are used as scaffold to create a protein drug by well-established screening procedures (see Tomlinson et al (2004), Nature Biotechnology, 22(5): 521-522). Non-limiting examples of human proteins which serve as a scaffold for designing protein drugs are transferrin, C-type lectins, trinectins, domain antibodies, kunitz domains, lipocalins and the Fyn SH3 domain. Also the protein drug used by the present invention may act on one of the enzymes or precursors being involved in elF5a activation. The term "small molecule" as used herein designates a low molecular weight organic compound that may have or has the capability to inhibit elF5a activation. The upper molecular weight limit for a small molecule is preferably about 900 Daltons. Small molecule of this molecular weight can in general rapidly diffuse across cell membranes, so that they can reach intracellular sites of action. As lower molecular weight a cut-off of 500 Daltons is preferred. This is because clinical attrition rates may be significantly reduced if the molecular weight is kept is below 500 Daltons.
The antisense technology for the downregulation of RNA is well-established and widely used in the art to treat various diseases. The basic idea of the antisense technology is the use of oligonucleotides for silencing a selected target mRNA through the exquisite specificity of complementary-based pairing (Re, Ochsner J. 2000 Oct; 2(4): 233-236). Herein below details on the antisense construct compound classes of siRNAs, shRNAs and antisense
oligonucleotides will be provided. As will be further detailed herein below, antisense oligonucleotides are single stranded antisense constructs while siRNAs and shRNAs are double stranded antisense constructs with one strand comprising an antisense oligonucleotide sequence (i.e. the so-called antisense strand). All these compound classes may be used to achieve downregulation or inhibition of a target RNA.
In accordance with the present invention the target of antisense constructs in general as well as all the specific classes of antisense constructs being described herein is preferably an mRNA of an enzyme being involved in the activation of elF5a. Accordingly, the target is preferably a nucleic acid sequence selected from one or more of SEQ ID NOs 1 , 3, 5, 7 and 9. Within SEQ ID NOs 1 , 3, 5, 7 and 9, SEQ ID NOs are 7 and 9 are preferred. It is a matter of routine in the field of antisense technology to design an antisense construct with a sufficient number of nucleotide mismatches to any off-target in order to ensure that no off- targets become down-regulated.
The term "siRNA" in accordance with the present invention refers to small interfering RNA, also known as short interfering RNA or silencing RNA. siRNAs are a class of 12 to 30, preferably 18 to 30, more preferably 20 to 25, and most preferred 21 to 23 or 21 nucleotide- long double-stranded RNA molecules that play a variety of roles in biology. Most notably, siRNA is involved in the RNA interference (RNAi) pathway where the siRNA interferes with the expression of a specific gene. In addition to their role in the RNAi pathway, siRNAs also act in RNAi-related pathways, e.g. as an antiviral mechanism or in shaping the chromatin structure of a genome. siRNAs have a well defined structure: a short double-strand of RNA (dsRNA), advantageously with at least one RNA strand having a 5' or 3' overhang. Each strand typically has a 5' phosphate group and a 3' hydroxyl (-OH) group. This structure is the result of processing by dicer, an enzyme that converts either long dsRNAs or small hairpin RNAs into siRNAs. siRNAs can also be exogenously (artificially) introduced into cells to bring about the specific knockdown of a gene of interest. Thus, any gene of which the sequence is known can in principle be targeted based on sequence complementarity with an appropriately tailored siRNA. The double-stranded RNA molecule or a metabolic processing product thereof is capable of mediating target-specific nucleic acid modifications, particularly RNA interference and/or DNA methylation. Preferably, one or both ends of the double-strand have a 3'-overhang from 1-5 nucleotides, more preferably from 1-3 nucleotides and most preferably 2 nucleotides. The most efficient silencing was so far obtained with siRNA duplexes composed of 21 -nt sense and 21 -nt antisense strands, paired in a manner to have 2-nt 3'- overhangs. The sequence of the 2-nt 3' overhang makes a small contribution to the specificity of target recognition restricted to the unpaired nucleotide adjacent to the first base
pair (Elbashir et al. Nature. 2001 May 24; 41 1 (6836):494-8). 2'-deoxynucleotides in the 3' overhangs are as efficient as ribonucleotides, but are often cheaper to synthesize and probably more nuclease resistant. The siRNA used in the invention preferably comprises an antisense strand which comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides of one or more sequences selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably SEQ ID NOs 7 and 9. A preferred example of a siRNA is an endoribonuclease-prepared siRNA (esiRNA). An esiRNA is a mixture of siRNA oligos resulting from cleavage of a long double-stranded RNA (dsRNA) with an endoribonuclease such as Escherichia coli RNase III or dicer. esiRNAs are an alternative concept to the usage of chemically synthesized siRNA for RNA interference (RNAi). For the generation of esiRNAs a cDNA of an mRNA template may be amplified by PCR and tagged with two bacteriophage-promotor sequences. RNA polymerase is then used to generate long double-stranded RNA that is complentary to the target-gene cDNA. This complementary RNA may be subsequently digested with RNase III from Escherichia coli to generate short overlapping fragments of siRNAs with a length between 18-25 base pairs. This complex mixture of short double-stranded RNAs is similar to the mixture generated by dicer cleavage in vivo and is therefore called endoribonuclease-prepared siRNA or short esiRNA. Hence, esiRNA are a heterogeneous mixture of siRNAs that all target the same mRNA sequence. esiRNAs lead to highly specific and effective gene silencing.
A "shRNA" in accordance with the present invention is a short hairpin RNA, which is a sequence of RNA that makes a (tight) hairpin turn that can also be used to silence gene expression via RNA interference. shRNA preferably utilizes the U6 promoter for its expression. The shRNA hairpin structure is cleaved by the cellular machinery into siRNA, which is then bound to the RNA-induced silencing complex (RISC). This complex binds to and cleaves mRNAs which match the shRNA that is bound to it. The shRNA used in the invention preferably comprises an antisense strand which comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably SEQ ID NOs 7 and 9.
The term "antisense oligonucleotide" in accordance with the present invention preferably
refers to a single-stranded nucleotide sequence being complementary by virtue of Watson- Crick base pair hybridization to an mRNA selected from SEQ ID NOs 1 , 3, 5, 7 and 9 whereby the respective mRNA is blocked and cannot be translated into protein. The antisense oligonucleotides may be unmodified or chemically modified. In general, they are relatively short (preferably between 13 and 25 nucleotides). Moreover, they are specific for one of SEQ ID NOs 1 , 3, 5, 7 and 9, i.e. they hybridize to a unique sequence in the total pool of targets present in the target cells/organism. The antisense oligonucleotide according to the invention comprises or consists a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, or at least 25 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, or at least 25 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO.
The antisense oligonucleotide is preferably a LNA-GapmeR, an Antagomir, or an antimiR.
LNA-GapmeRs or simply GapmeRs are potent antisense oligonucleotides used for highly efficient inhibition of mRNA function. GapmeRs function by RNase H dependent degradation of complementary RNA targets. They are an excellent alternative to siRNA for knockdown of mRNA. They are advantageously taken up by cell without transfection reagents. GapmeRs contain a central stretch of DNA monomers flanked by blocks of LNAs. The GapmeRs are preferably 14-16 nucleotides in length and are optionally fully phosphorothioated. The DNA gap activates the RNAse H-mediated degradation of targeted RNAs and is also suitable to target transcripts directly in the nucleus. The LNA-GapmeR used in the invention preferably comprises a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, or at least 15 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. These at least 13 nucleotides, at least 14 nucleotides, or at least 15 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO. LNA-GapmeRs are routinely designed using established algorithms. LNA-GapmeRs to a
selected target are commercially available including positive and negative controls, for example, from Exiqon.
As mentioned, AntimiRs are oligonucleotide inhibitors that were initially designed to be complementary to a miRNA. AntimiRs against miRNAs have been used extensively as tools to gain understanding of specific miRNA functions and as potential therapeutics. As used herein, the AntimiRs are preferably designed to be complementary to a sequence selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. AntimiRs are preferably 14 to 23 nucleotides in length. An AntimiR according to the invention more preferably comprises or consists a sequence which is with increasing preference complementary to at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or at least 23 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. These at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or at least 23 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO.
AntimiRs are preferably AntagomiRs. AntagomiRs are synthetic 2-O-methyl RNA oligonucleotides, preferably of 21 to 23 nucleotides which are preferably fully complementary to the selected target RNA. While AntagomiRs were initially designed against miRNAs they may also be designed against mRNAs. The AntagomiRs used according to the invention therefore preferably comprise a sequence being complementary to 21 to 23 nucleotides of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably within SEQ ID NOs 7 and 9. These 21 to 23 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, i.e. the nucleotides are consecutive in the respective SEQ ID NO. AntagomiRs are preferably synthesized with 2'-OMe modified bases (2'-hydroxyl of the ribose is replaced with a methoxy group), phosphorothioate (phosphodiester linkages are changed to phosphorothioates) on the first two and last four bases, and an addition of cholesterol motif at 3' end through a hydroxyprolinol modified linkage. The addition of 2'-OMe and phosphorothioate modifications improves the bio-stability whereas cholesterol conjugation enhances distribution and cell permeation of the AntagomiRs.
Antisense molecules (including antisense oligonucleotides, such as LNA-GapmeR,
Antagomir, an antimiR), siRNAs and shRNAs of the present invention are preferably chemically synthesized using a conventional nucleic acid synthesizer. Suppliers of nucleic acid sequence synthesis reagents include Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, CO, USA), Pierce Chemical (part of Perbio Science, Rockford, IL, USA), Glen Research (Sterling, VA, USA), ChemGenes (Ashland, MA, USA), and Cruachem (Glasgow, UK).
The ability of antisense molecules (including antisense oligonucleotides, such as LNA- GapmeR, an Antagomir, an antimiR), siRNA, and shRNA to potently, but reversibly, silence or inhibit a target mRNA in vivo makes these molecules particularly well suited for use in the medical applications of the invention being further described herein below. Ways of administering siRNA to humans are described in De Fougerolles et al., Current Opinion in Pharmacology, 2008, 8:280-285. Such ways are also suitable for administering other small RNA molecules like antisense oligonucleotides or shRNAs. Accordingly, such pharmaceutical compositions may be administered directly formulated as a saline, via liposome based and polymer-based nanoparticle approaches, as conjugated or complexation pharmaceutical compositions, or via viral delivery systems. Direct administration comprises injection into tissue, intranasal and intratracheal administration. Liposome based and polymer-based nanoparticle approaches comprise the cationic lipid Genzyme Lipid (GL) 67, cationic liposomes, chitosan nanoparticles and cationic cell penetrating peptides (CPPs). Conjugated or complexation pharmaceutical compositions comprise PEI-complexed antisense molecules (including antisense oligonucleotides), siRNA, or shRNA. Further, viral delivery systems comprise influenza virus envelopes and virosomes. The antisense molecules (including antisense oligonucleotides, such as LNA-GapmeR, an Antagomir, an antimiR), siRNAs, shRNAs may comprise modified nucleotides such as locked nucleic acids (LNAs). The ribose moiety of an LNA nucleotide is modified with an extra bridge connecting the 2' oxygen and 4' carbon. The bridge "locks" the ribose in the 3'-endo (North) conformation, which is often found in the A-form duplexes. LNA nucleotides can be mixed with DNA or RNA residues in the oligonucleotide whenever desired. Such oligomers are synthesized chemically and are commercially available. The locked ribose conformation enhances base stacking and backbone pre-organization. This significantly increases the hybridization properties (melting temperature) of oligonucleotides. In accordance with a more preferred embodiment of the first aspect of the invention the compound comprises (a) a nucleic acid sequence which comprises or consists of a nucleic acid sequence being complementary to at least 12 continuous nucleotides of a nucleic acid
sequence selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, (b) a nucleic acid sequence which comprises or consists of a nucleic acid sequence which is at least 70% identical to the complementary strand of one or more nucleic acid sequences selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9, (c) a nucleic acid sequence which comprises or consists of a nucleic acid sequence according to (a) or (b), wherein the nucleic acid sequence is DNA or RNA, (d) an expression vector expressing the nucleic acid sequence as defined in any one of (a) to (c), preferably under the control of a macrophage-specific promoter, or (e) a host comprising the expression vector of (d).
The nucleic acid sequences as defined in items (a) to (c) of this preferred embodiment comprise or consist of sequences being complementary to nucleotides of the amino acids encoding the enzymes required for the activation of elF5a as defined by SEQ ID NOs 1 , 3, 5, 7 and 9, preferably by SEQ ID NOs 7 and 9. Hence, the nucleic acid sequences as defined in items (a) to (c) comprise or are antisense nucleic acid sequences.
The nucleic acid sequence according to item (a) of this further preferred embodiment of the invention comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides of one or more sequences selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides are preferably complementary to a contiguous part of one or more selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. The format of the nucleic acid sequence according to item (a) is not particularly limited as long as it comprises or consists of at least 12 continuous nucleotides being complementary to a nucleic acid sequence selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. Hence, the nucleic acid sequence according to item (a) reflects the above- mentioned basic principle of the antisense technology which is the use of an oligonucleotide for silencing a selected target RNA through the exquisite specificity of complementary-based pairing. Therefore, it is to be understood that the nucleic acid sequence according to item (a) is preferably in the format of an siRNA, shRNA or an antisense oligonucleotide as defined herein above. The antisense oligonucleotides are preferably LNA-GapmeRs, AntagomiRs, or antimiRs as defined herein above.
A nucleic acid sequence according to item (b) of the above preferred embodiment of the invention is capable of interacting with, more specifically hybridizing with the target mRNA being selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9. By formation of the hybrid the function of the mRNA being selected from SEQ ID NOs 1 , 3, 5, 7 and 9 is reduced or blocked.
The sequence identity of the molecule according to item (b) in connection with a sequence selected from SEQ ID NOs 1 , 3, 5, 7 and 9, preferably from SEQ ID NOs 7 and 9 is with increasing preference at least 75%, at least 80%, at least 85%, at least 90%, at least 92.5%, at least 95%, at least 98%, at least 99% and 100%. The sequence identity in connection with each of SEQ ID NOs 1 , 3, 5, 7 and 9 can be individually selected. For instance, a non-limiting example is at least 85% in connection with SEQ ID NO: 7 and at least 90% in connection with SEQ ID NO: 9. Means and methods for determining sequence identity are known in the art. Preferably, the BLAST (Basic Local Alignment Search Tool) program is used for determining the sequence identity with regard to one or more of SEQ ID NOs 1 , 3, 5, 7 and 9. Most preferred examples of nucleic acid sequences which comprise a nucleotide sequence which is at least 70% identical to the complementary strand of one or more of SEQ ID NOs 1 , 3, 5, 7 and 9 are the complementary strands of SEQ ID NOs 1 , 3, 5, 7 and 9. In the nucleic acid sequence according to item (c) the nucleotide sequences may be RNA or DNA. RNA or DNA encompasses chemically modified RNA nucleotides or DNA nucleotides. As commonly known RNA comprises the nucleotide U while DNA comprises the nucleotide T. In accordance with items (d) and (e) of the above preferred embodiment the inhibitor may also be an expression vector or host, respectively being capable of producing an nucleic acid sequence as defined in any one of items (a) to (c).
An expression vector may be a plasmid that is used to introduce a specific transcript into a target cell. Once the expression vector is inside the cell, the inhibitor of the activation of elF5a being encoded by the expression vector is produced by the cellular-transcription. The plasmid is in general engineered to contain regulatory sequences that act as enhancer and/or promoter regions and lead to efficient transcription of the transcript. In accordance with the present invention the expression vector preferably contains a macrophage-specific promoter. Macrophage-specific promoters are known in the art, for example, from Levin et a., Gene Ther., 2012; 19(11 ):1041-7. Using a macrophage-specific promoter ensures that the nucleic acid sequence is only expressed in macrophages and may avoid potential undesired
side effects by expression in other cell types.
Non-limiting examples of expression vectors include prokaryotic plasmid vectors, such as the pUC-series, pBluescript (Stratagene), the pET-series of expression vectors (Novagen) or pCRTOPO (Invitrogen) and vectors compatible with an expression in mammalian cells like pREP (Invitrogen), pcDNA3 (Invitrogen), pCEP4 (Invitrogen), pMCI neo (Stratagene), pXT1 (Stratagene), pSG5 (Stratagene), EBO-pSV2neo, pBPV-1 , pdBPVMMTneo, pRSVgpt, pRSVneo, pSV2-dhfr, plZD35, pLXIN, pSIR (Clontech), pIRES-EGFP (Clontech), pEAK-10 (Edge Biosystems) pTriEx-Hygro (Novagen) and pCINeo (Promega). Examples for plasmid vectors suitable for Pichia pastoris comprise e.g. the plasmids pA0815, pPIC9K and pPIC3.5K (all Intvitrogen). For the formulation of a pharmaceutical composition a suitable vector is selected in accordance with good manufacturing practice. Such vectors are known in the art, for example, from Ausubel et al, Hum Gene Ther. 201 1 Apr; 22(4):489-97 or Allay et al., Hum Gene Ther. May 2011 ; 22(5): 595-604.
A typical mammalian expression vector contains the promoter element, which mediates the initiation of transcription of mRNA, the protein coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Moreover, elements such as origin of replication, drug resistance gene, regulators (as part of an inducible promoter) may also be included. The lac promoter is a typical inducible promoter, useful for prokaryotic cells, which can be induced using the lactose analogue isopropylthiol-b-D-galactoside ("IPTG"). For recombinant expression and secretion, the polynucleotide of interest may be ligated between e.g. the PelB leader signal, which directs the recombinant protein in the periplasm and the gene III in a phagemid called pHEN4 (described in Ghahroudi et al, 1997, FEBS Letters 414:521 -526). Additional elements might include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing. Highly efficient transcription can be achieved with the early and late promoters from SV40, the long terminal repeats (LTRs) from retroviruses, e.g., RSV, HTLVI, HIVI, and the early promoter of the cytomegalovirus (CMV). However, cellular elements can also be used (e.g., the human actin promoter). Suitable expression vectors for use in practicing the present invention include, for example, vectors such as pSVL and pMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146) and pBC12MI (ATCC 67109). Alternatively, the inhibitor can be expressed in stable cell lines that contain the gene construct integrated into a chromosome. The co-transfection with a selectable marker such as dhfr, gpt, neomycin, hygromycin allows the identification and isolation of the transfected cells. The transfected nucleic acid can also be amplified to express large amounts of the encoded (poly)peptide. The DHFR (dihydrofolate reductase) marker is useful to develop cell
lines that carry several hundred or even several thousand copies of the nucleic acid molecule encoding the inhibitor. Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy et al.1991 , Biochem J. 227:277-279; Bebbington et al. 1992, Bio/Technology 70:169-175). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. As indicated above, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, G418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. For vector modification techniques, see Sambrook and Russel (2001 ), Molecular Cloning: A Laboratory Manual, 3 Vol. Generally, vectors can contain one or more origins of replication (ori) and inheritance systems for cloning or expression, one or more markers for selection in the host, e.g., antibiotic resistance, and one or more expression cassettes. Suitable origins of replication (ori) include, for example, the Col E1 , the SV40 viral and the M 13 origins of replication.
The sequences to be inserted into the vector can e.g. be synthesized by standard methods, or isolated from natural sources. Ligation of the coding sequences to transcriptional regulatory elements and/or to other amino acid encoding sequences can be carried out using established methods. Transcriptional regulatory elements (parts of an expression cassette) ensuring expression in prokaryotes or eukaryotic cells are well known to those skilled in the art. These elements comprise regulatory sequences ensuring the initiation of the transcription (e.g., translation initiation codon, promoters, enhancers, and/or insulators), internal ribosomal entry sites (IRES) (Owens, Proc. Natl. Acad. Sci. USA 98 (2001 ), 1471 - 1476) and optionally poly-A signals ensuring termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers, and/or naturally-associated or heterologous promoter regions. Preferably, the nucleotide sequence as defined in item (a) of the above preferred embodiment of the invention is operatively linked to such expression control sequences allowing expression in prokaryotic or eukaryotic cells.
The host may be a prokaryotic or eukaryotic cell. A suitable eukaryotic host may be a mammalian cell, an amphibian cell, a fish cell, an insect cell, a fungal cell or a plant cell. Representative examples of bacterial cells are E. coli, Streptomyces and Salmonella typhimurium cells; of fungal cells are yeast cells; and of insect cells are Drosophila S2 and Spodoptera Sf9 cells. It is preferred that the cell is a mammalian cell such as a human cell. Mammalian host cells that could be used include, human Hela, 293, H9 and Jurkat cells, mouse NIH3T3 and C127 cells, Cos 1 , Cos 7 and CV1 , quail QC1-3 cells, mouse L cells and
Chinese hamster ovary (CHO) cells. The cell may be a part of a cell line, preferably a human cell line. Appropriate culture mediums and conditions for the above-described host cells are known in the art. The host is preferably a host cell and more preferably an isolated host cell. The host is also preferably a non-human host.
In accordance with another preferred embodiment of the method of the invention, the compound is directed against ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH) and is preferably directed against DHPS or DOHH.
As discussed above and as shown in Figures 1 and 2, ornithine decarboxylase, spermidine synthase, spermine synthase, DHPS and DOHH are enzymes being required for the activation of elF5a. Depending on the nature of the compound the compound is directed against the mRNA encoding one of the enzymes or one of the enzymes in their protein from. For instance and as detailed herein above, in case the compound is an antisense molecule, siRNA or shRNA it may be directed against the mRNA encoding one of the enzymes. On the other hand, in case the compound is an antibody it may be directed against one of the enzymes in their protein form. The compounds are preferably directed against the human mRNAs and enzymes. The mRNA of human ornithine decarboxylase is shown in SEQ ID NO: 1 , of human spermidine synthase in SEQ ID NO: 3, of human spermine synthase in SEQ ID NO: 5, of human DOHH in SEQ ID NO: 7 and of human DDHPS in SEQ ID NO: 9. The amino acid sequence of human ornithine decarboxylase is shown in SEQ ID NO: 2, of human spermidine synthase in SEQ ID NO: 4, of human spermine synthase in SEQ ID NO: 6, of human DOHH in SEQ ID NO: 8 and of human DHPS in SEQ ID NO: 10. With the mRNAs SEQ ID NOs 7 and 9 and within the amino acid sequences SEQ ID NOs 8 and 10 are the preferred targets. This is because in the polyamine synthesis pathway the mRNAs of SEQ ID NOs 7 and 9 encode the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a and the amino acid sequences of SEQ ID NOs 8 and 10 are the enzymes catalyzing the last two chemical reactions being required for the activation of elF5a. These enzymes catalyze the deoxyhypusiantion and the hypusination of elF5a. As elF5a is currently thought to be the only protein containing the hypusine modification, a compound targeting the mRNA of SEQ ID NOs 7 or 9 or the amino acid sequence of SEQ ID NOs 8 of 10 is expected to prevent unwanted off target effects.
In accordance with a further preferred embodiment of the method of the invention, the efficacy of a compound for inhibiting elF5a activation in a patient is determined, wherein the sample of step (a) and the control sample are samples that have been obtained from the patient, and wherein a reduction of the number of cells having a M2-phenotype in the sample as compared to the control sample not contacted with the compound indicates that the compound is effective for inhibiting elF5a activation in the patient.
The compound to be tested for its capability of inhibiting elF5a activation may also be a compound for which it is already known that it has the capability of inhibiting elF5a activation and in this case the method can determine how much inhibition is achieved by the compound. This option of performing the claimed method is particularly suitable for determining the efficacy of a compound for inhibiting elF5a activation in a patient.
Hence, the first aspect of the present invention also encompasses a method for determining the efficacy of a compound for inhibiting elF5a activation in a patient, comprising (a) obtaining a sample comprising cells being capable of differentiating into M2 macrophages from a patient, (a) contacting the obtained sample with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and (b) quantifying in the sample the cells having a M2-phenotype, wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting elF5a activation.
As will be discussed in more detail herein below, a compound for inhibiting elF5a activation can be used to treat several diseases in a patient. Hence, determining the efficacy of a compound for inhibiting elF5a activation in a patient allows to predict whether this compound will be effective to treat or prevent a disease in the patient, in particular a disease being mediated by M2 macrophages, or that can be treated by inhibiting cellular differentiation into M2 macrophages.
In accordance with a more preferred embodiment of the method of the invention, the patient is afflicted with a hyperproliferative disorder, a fibrotic disorder or the macrophage activation syndrome. A hyperproliferative disorder is any disease comprising an abnormally high rate of proliferation of cells by rapid cell division. Non-limiting examples are psoriasis, psoriatic
arthritis, rheumatoid arthritis, cutaneous hyperkeratoses, inflammatory bowel disease tumors and cancers.
A fibrotic disorder is any disorder comprising the formation of excess fibrous connective tissue, e.g. in an organ or tissue in a reparative or reactive process. Non-limiting examples of fibrotic disorders will be provided herein below.
The macrophage activation syndrome (MAS) is a severe, potentially life-threatening, complication of several chronic rheumatic diseases of childhood. It occurs most commonly with systemic-onset juvenile idiopathic arthritis (SoJIA). In addition, MAS has been described in association with systemic lupus erythematosus (SLE), Kawasaki disease, and adult-onset Still's disease. It is thought to be closely related and pathophysiologically very similar to reactive (secondary) hemophagocytic lymphohistiocytosis (HLH). The hallmark clinical and laboratory features include high fever, hepatosplenomegaly, lymphadenopathy, pancytopenia, liver dysfunction, disseminated intravascular coagulation, hypofibrinogenemia, hyperferritinemia, and hypertriglyceridemia. Despite marked systemic inflammation, the erythrocyte sedimentation rate (ESR) is paradoxically depressed, caused by low fibrinogen levels. The low ESR helps to distinguish the disorder from a flare of the underlying rheumatic disorder, in which case the ESR is usually elevated. A bone marrow biopsy or aspirate usually shows hemophagocytosis.
M2 macrophages are known to secrete large amounts of pro-fibrotic factors, such as TGF-β and Galactin-3 (Braga et I. (2015), Front Immunol; 6:602 and Wermuth and Jimenez (2015), Clin Transl Med; 4: 2.). M2 macrophages are also known to promote cell proliferation (Mills (2012), Crit Rev Immunol.; 32(6):463-88.). It follows that M2 macrophages play an important role in promoting the formation or hyperproliferative or fibrotic disorders and interfering with the formation of M2 macrophages is a suitable means for treating or preventing a hyperproliferative or fibrotic disorders. In connection with the method of the invention it is particularly useful to determine the efficacy of a compound for inhibiting elF5a activation in a patient being afflicted with a hyperproliferative or fibrotic disorder.
In accordance with a further more preferred embodiment of the method of the invention, the sample is a tissue sample or a blood sample. In order to be capable of determining the efficacy of a compound for inhibiting elF5a activation in a patient the sample has to be a sample that has been obtained from the body of the patient and has to comprise cells being M2 macrophages or capable of differentiating
into M2 macrophages. Such a sample is preferably a tissue sample or a blood sample. Macrophages are known to be distributed in tissues throughout the body, e.g. in the brain, liver, lung, spleen, intestine, skin, heart, kidney and peritoneum (Epelman et al. (2014), Immunity; 41(1 ):21— 35). As the patient is preferably afflicted with a hyperproliferative disorder, a fibrotic disorder or MAS also the tissue sample is preferably obtained from such a patient. Macrophages are likewise found in the blood, including the peripheral blood. The blood sample may be a whole blood sample, serum or plasma.
In accordance with a further preferred embodiment of the method of the invention, the compound is guanyl-1 ,7-diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I or CNI-1493.
Guanyl-1 ,7-diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I and CNI-1493 are non-limiting examples of compounds interfering with an enzyme being involved in elF5a activation.
Guanyl-1 ,7-diaminoheptane (GC7) (CAS 150333-69-0) is an efficient inhibitor of deoxyhypusine synthase. As a competitive inhibitor it binds to the active site of deoxyhypusine synthase and prevents the first step in the post-translational conversion of a single lysine to hypusine in elF5A. GC-7 is used in the examples of the application.
L-mimosine (CAS 500-44-7) is a plant non-protein amino acid, chemically similar to tyrosine. The synthesis of the amino acid hypusine by deoxyhypusine hydroxylase is blocked by L- mimosine, further noting that hypusine is necessary for the activation of elF-5A.
The drugs ciclopirox (CAS 29342-05-0) and deferiprone (CAS 30652-1 1-0) are inhibitors of deoxyhypusine hydroxylase and hence the posttranslational modification by hypusine being requiring for the activation of elF-5A (Memin et al al. (2014), Cancer Res. 2014 Jan 15;74(2):552-62).
Hydralazine (CAS 86-54-4) inhibits the posttranslational hydroxylation of deoxyhypusine (Paz et al. (1984), Biochem Pharmacol, 33: 779-785).
Agent I (Hoechst 768159, [2-(4-hydroxytoluene-3-yl)-4,5-dihydro-4- carboxythiazole]) is an iron chelator that blocks the function of deoxyhypusine hydroxylase and hence the hypusination of elF5a; see Hanauske-Abel et al. (1994) Biochimica et Biophysica Acta 1221 115-124.
CNI-1493 (or semapimod; CAS 164301-51 -3) is an inhibitor of deoxyhypusine synthase (Specht et al. (2008), Parasitol Res; 102(6): 1 177-84). In a second aspect the present invention relates to a compound inhibiting elF5a activation for use in the treatment or prevention of a disease being mediated by M2 macrophages, wherein the disease is preferably a hyperproliferative, a fibrotic disorder or the macrophage activation syndrome. The present invention likewise relates to a method for treating or preventing a disease being mediated by M2 macrophages, wherein the disease is preferably a hyperproliferative, a fibrotic disorder or the or the macrophage activation syndrome comprising administering a therapeutically effective amount of a compound inhibiting elF5a activation to a subject in need thereof.
In a third aspect the present invention relates to a compound inhibiting elF5a activation for use in the treatment or prevention of a hyperproliferative, a fibrotic disorder or the macrophage activation syndrome by inhibiting cellular differentiation into M2 macrophages. The present invention also relates to a method for treating or preventing a hyperproliferative disorder, a fibrotic disorder or the macrophage activation syndrome by inhibiting cellular differentiation into M2 macrophages comprising administering a therapeutically effective amount of a compound inhibiting elF5a activation to a subject in need thereof, thereby inhibiting the cellular differentiation into M2 macrophages in the subject.
The preferred embodiments, definitions and explanations described herein above in connection with the first aspect of the invention as far as being applicable to the second and third aspect of the invention apply mutatis mutandis to the second and third aspect of the invention.
The subject to be treated in accordance with the second and third aspect of the invention is preferably a mammal, more preferably a primate and most preferably a human.
The compounds inhibiting elF5a activation are preferably admixed with a pharmaceutically acceptable carrier or excipient to form a pharmaceutical composition. By "pharmaceutically acceptable carrier or excipient" is meant a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type (see also Handbook of
Pharmaceutical Excipients 6ed. 2010, Published by the Pharmaceutical Press). The compounds inhibiting elF5a activation or the pharmaceutical composition may be administered, for example, orally, parenterally, such as subcutaneously, intravenously, intramuscularly, intraperitoneally, intrathecally, transdermally, transmucosally, subdurally, locally or topically via iontopheresis, sublingually, by inhalation spray, aerosol or rectally and the like in dosage unit formulations optionally comprising conventional pharmaceutically acceptable carriers or excipients.
The compounds inhibiting elF5a activation may be formulated as vesicles, such as liposomes. Liposomes have attracted great interest because of their specificity and the duration of action they offer from the standpoint of drug delivery. Liposomal delivery systems have been used to effectively deliver nucleic acids, such as siRNA in vivo into cells (Zimmermann et al. (2006) Nature, 441 :11 1-114). Liposomes are unilamellar or multilamellar vesicles which have a membrane formed from a lipophilic material and an aqueous interior. The aqueous portion contains the composition to be delivered. Cationic liposomes possess the advantage of being able to fuse to the cell wall. Non-cationic liposomes, although not able to fuse as efficiently with the cell wall, are phagocytosed by macrophages and other cells in vivo. The compounds inhibiting elF5a activation can be administered to the subject at a suitable dose. The dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. The therapeutically effective amount for a given situation will readily be determined by routine experimentation and is within the skills and judgement of the ordinary clinician or physician. Generally, the regimen as a regular administration of the pharmaceutical composition should be in the range of 1 g to 5 g units per day. However, a more preferred dosage is in the range of 0.01 mg to 100 mg, even more preferably 0.01 mg to 50 mg and most preferably 0.01 mg to 10 mg per day. Furthermore, if for example said compound comprises or is an nucleic acid molecule, such as an siRNA, the total pharmaceutically effective amount of pharmaceutical composition administered will typically be less than about 75 mg per kg of body weight, such as for example less than about 70, 60, 50, 40, 30, 20, 10, 5, 2, 1 , 0.5, 0.1 , 0.05, 0.01 , 0.005, 0.001 , or 0.0005 mg per kg of body weight. More preferably, the amount will be less than 2000 nmol of nucleic acid molecule per kg of body weight, such as for example less than 1500, 750, 300, 150, 75, 15, 7.5, 1.5, 0.75, 0.15, 0.075, 0.015, 0.0075, 0.0015, 0.00075 or 0.00015 nmol per kg of body weight.
The length of treatment needed to observe changes and the interval following treatment for responses to occur vary depending on the desired effect. The particular amounts may be determined by conventional tests which are well known to the person skilled in the art. Suitable tests are, for example, described in Tamhane and Logan (2002), Journal of the American statistical association, 97(457):1-9.
As detailed above, M2 macrophages play an important role in promoting the formation or hyperproliferative or fibrotic disorders as well as the macrophage activation syndrome and therefore these diseases can be treated or prevented by interfering with the formation of M2 macrophages. By the surprising finding of the inventors that the activation of elF5a is important for the alternative activation of M2 cells, it has also been revealed that a compound inhibiting elF5a activation can be used to treat or prevent a disease being mediated by M2 macrophages or a hyperproliferative or fibrotic disorder or the macrophage activation syndrome by inhibiting cellular differentiation into M2 macrophages. In other words, the finding that the formation of M2 macrophages can be inhibited by inhibiting the activation of elF5a also revealed that diseases that are mediated by M2 macrophages can be treated or prevented by inhibiting the activation of elF5a. In accordance with a preferred embodiment of the second and third aspect of the invention, the compound targets ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH) and preferably DHPS or DOHH. As can be taken form Figures 1 and 2, ornithine decarboxylase, spermidine synthase, spermine synthase, DHPS and DOHH are enzymes being involved in the activation of elF5a. Further details on how the compounds of the invention target these enzymes are provided herein above in connection with the corresponding embodiment of the first aspect of the invention. These details apply mutatis mutandis to the second and third aspect of the invention.
In accordance with a further preferred embodiment of the second and third aspect of the invention, the compound is an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, or protein drug, small molecule.
In connection with the second aspect and third of the invention the compound inhibiting elF5a activation may be any one of an antisense molecule, siRNA, shRNA, antibody,
ribozyme, aptamer, protein drug and small molecule as defined herein above in connection with the first aspect of the invention. The details on the compounds of the invention as provided herein above in connection with the corresponding embodiment of the first aspect of the invention apply mutatis mutandis to the second and third aspect of the invention.
In accordance with a more preferred embodiment of the second and third aspect of the invention, the compound is guanyl-1 ,7-diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I or CNI-1493. These compounds compound are known to inhibit elF5a activation. Further details on these inhibitors are provided herein above in connection with the first aspect of the invention.
In accordance with a still further preferred embodiment of the second and third aspect of the invention, the hyperproliferative disorder is a neoplasm, tumor or cancer and is preferably selected from cancer of the breast, lung, prostate, kidney, skin, neural, ovary, uterus, liver, pancreas, epithelial, gastric, intestinal, exocrine, endocrine, lymphatic, hematopoietic system or a head and neck tissue; and/or (ii) the fibrotic disorder is selected from sarcoidosis, renal fibrosis, pulmonary fibrosis, idiopathic pulmonary fibrosis, cystic fibrosis, liver fibrosis, cardiac fibrosis, endomyocardial fibrosis, atrial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, chronic kidney disease, nephrogenic systemic fibrosis, Chron's disease, hypertrophic scarring, keloid, scleroderma, organ transplant-associated fibrosis and ischemia-associated fibrosis.
The above-listed examples of hyperproliferative disorders and fibrotic disorders are non- limiting examples or defined diseases that can be treated in accordance with the second and third aspect of the invention. A neoplasm or tumor is an abnormal benign or malignant new growth of tissue that possesses no physiological function and arises from uncontrolled usually rapid cellular proliferation. The WHO classifies neoplasms into four main groups: benign neoplasms, in situ neoplasms, malignant neoplasms, and neoplasms of uncertain or unknown behavior. A malignant neoplasm is also referred to as cancer.
As regards the embodiments characterized in this specification, in particular in the claims, it is intended that each embodiment mentioned in a dependent claim is combined with each embodiment of each claim (independent or dependent) said dependent claim depends from. For example, in case of an independent claim 1 reciting 3 alternatives A, B and C, a dependent claim 2 reciting 3 alternatives D, E and F and a claim 3 depending from claims 1 and 2 and reciting 3 alternatives G, H and I, it is to be understood that the specification
unambiguously discloses embodiments corresponding to combinations A, D, G; A, D, H; A, D, I; A, E, G; A, E, H; A, E, I; A, F, G; A, F, H; A, F, I; B, D, G; B, D, H; B, D, I; B, E, G; B, E, H; B, E, I; B, F, G; B, F, H; B, F, I; C, D, G; C, D, H; C, D, I; C, E, G; C, E, H; C, E, I; C, F, G; C, F, H; C, F, I, unless specifically mentioned otherwise.
Similarly, and also in those cases where independent and/or dependent claims do not recite alternatives, it is understood that if dependent claims refer back to a plurality of preceding claims, any combination of subject-matter covered thereby is considered to be explicitly disclosed. For example, in case of an independent claim 1 , a dependent claim 2 referring back to claim 1 , and a dependent claim 3 referring back to both claims 2 and 1 , it follows that the combination of the subject-matter of claims 3 and 1 is clearly and unambiguously disclosed as is the combination of the subject-matter of claims 3, 2 and 1. In case a further dependent claim 4 is present which refers to any one of claims 1 to 3, it follows that the combination of the subject-matter of claims 4 and 1 , of claims 4, 2 and 1 , of claims 4, 3 and 1 , as well as of claims 4, 3, 2 and 1 is clearly and unambiguously disclosed.
The Figures show
Figure 1. The polyamine synthesis pathway. Polyamine synthesis begins with the production of putrescine from the amino acid ornithine, catalysed by ornithine decarboxylase. Spermidine and spermine are then synthesised from putrescine from spermidine synthase and spermine synthase, respectively.
Figure 2. elF5aHyp formation. In the first step, deoxyhypusine synthase (DHS) catalyses the transfer of an aminobutyl moiety of spermidine to one specific lysine residue of elF5a to form an intermediate deoxyhypusine residue. This intermediate is subsequently hydroxylated by deoxyhypusine hydroxylase (DOHH) to complete hypusination and elF5a activation.
Figure 3. The deoxyhypusine synthase inhibitor GC7 blocks M2 macrophage differentiation, a) Bone marrow-derived macrophages (BMDMs) on day 7 of culture were polarised into M2 cells with IL-4 in the presence or absence of 10 uM GC7. 18 hours later, M2 phenotype was assessed by flow cytometry analysis of CD301 and RELMa expression, two prototypic markers of M2 cells, b) BMDMs were differentiated into M1 cells with LPS and IFN-y in the presence or absence of 10 uM GC7. 18 hours later, the expression of nitric oxide synthase 2 (NOS2), a hallmark of M1 cells, was assessed by flow cytometry.
Figure 4. GC7 disrupts oxidative phosphorylation in M2 macrophages. BMDCs were polarised toward a M2 phenotype, as described above, in the presence or absence of 10 uM GC7. After 18 hours, oxygen consumption rate (OCR - an indicator of OXPHOS) was measured using a Seahorse Analyser.
Figure 5. elF5a-hypusine levels in macrophages treated with 10uM GC7 for 18 hours.
Figure 6. The polyamine biosynthesis pathway regulates OXPHOS via hypusinated elF5A. (a) The polyamine pathway comprises the cationic metabolites putrescine, spermidine, and spermine that are synthesized downstream of the amino acid ornithine. Spermidine acts as a substrate for the hypusination of elF5A, catalyzed by DHPS and DOHH. DFMO inhibits ODC, whereas DENSPM induces polyamine catabolism. Both GC7 and CPX act as inhibitors of hypusination. (b) Relative OCR of MEFs (NIH3T3) incubated with 2.5 mM DFMO ± 50 μΜ DENSPM (compared to untreated) assessed by Seahorse Extracellular Flux Analyzer (EFA). (c) Intracellular ornithine, putrescine, and spermidine levels detected by LCMS of MEFs treated as in (b). Relative OCR and maximum OCR in MEFs treated with (d) 10 μΜ GC7, (e) or 20 μΜ CPX, for 24 hours, (f) Immunoblot analysis of elF5A and (g) relative OCR of MEFs (NIH3T3) expressing IPTG-inducible £/Y5a-shRNA. (h) DHPS and elF5A-hypusine levels and (i) relative OCR in Dhps ox/nm MEFs expressing a 4-OHT-inducible Cre-ER (controls are Dhpsflox/flox Cre-ER"). (J) S2 cells (D. melanogaster), MDCK (C. familiaris), MCF-7 (H. sapiens) cells treated with 10 μΜ GC7 and 20 μΜ CPX for 24 hours. All data are mean ± SEM (p**<0.005, p***<0.0005, compared to control or untreated), (b-e, j) Represents two experiments, (f-i) representative of three experiments. Figure 7. Relative ECAR of (a) DFMO and DENSPM-treated MEFs. (b) Hypusine inhibition in MEFS by GC7. Relative ECAR of (c) GC7- and (d) CPX-treated MEFs; (e) MEFs transduced with an IPTG-inducible E/'f5a-inducible shRNA; (f) Dhpsitoxinox MEFs expressing a 4-OHT-inducible Cre-ER; (g) S2 cells (D. melanogaster), MDCK (C. familiaris), MCF-7 (H. sapiens) cells ± GC7 or CPX. All data are mean ± SEM (p*<0.05, p**<0.005, p***<0.0005, compared to control), (a) Representative of two experiments, (b, e, f) representative of three experiments, (c, d, g) represents two experiments.
Figure 8. (a) ECAR of E/f5a-shRNA transduced MEFs at baseline, or in response to 2- deoxyglucose (2-DG), Oligo, FCCP, and R+A. (b) Fold increase in cell number of Eif5a- shRNA-expressing MEFs over 5 days of culture. Relative ECAR of (c) GC7- and (d) CPX- treated M(IL-4); (e-g) and M(IL-4) transduced with indicated shRNA. (h) Immunoblot analysis
of specified proteins in M(IL-4) expressing control, Eif5a, Dhps or EG7-OVA cells expressing control or Dohh-shRNA. (i) LCMS relative quantification of TCA cycle-associated metabolites in £/ 5a-shRNA-expressing MEFs. LCMS relative quantification of indicated metabolites in (j) GC7-treated M(IL-4) and (k) £/T5a-shRNA MEFs. All data are mean ± SEM (p*«D.05, p**<0.005, p***<0.0005). (a, j) representative of two experiments, (b) represents one experiment, (c) represents four experiments, (d-g) represents of two experiments, (h) representative of one to two experiments, (i,k) represents one experiment.
Figure 9. Hypusinated elF5A maintains TCA cycle and ETC integrity in macrophages. (a) OCR of £/75a-shRNA transduced MEFs (NIH3T3) at baseline, or in response to 2- deoxyglucose (2-DG), oligomycin (Oligo), FCCP, and rotenone plus antimycin A (R+A). Relative and max OCR of M(IL-4)_(b,c) treated with 10 μΜ GC7 or 20 μΜ CPX or (d-f) retrovirally transduced with the indicated shRNA (hp). (g,h) LCMS quantification of indicated metabolites in M(IL-4) exposed to GC7, relative to control cells, (i) D-13C-glucose and (j) 13C- glutamine GCMS trace analysis of GC7-treated M(IL-4). (k) Proteomic analysis of M(IL-4) treated with 10 μΜ GC7 for 24 hours. Immunoblot assessment of (I) selected TCA cycle enzymes, as depicted in (g), and (m) ETC-associated proteins in GC7-treated M0, M(LPS/IFNy) = M(L/y), and M(IL-4). All data are mean ± SEM (p**<0.005, p***<0.0005). (a, h- j, m) representative of two experiments, (b) represents four experiments, (c-f) represents two experiments, (k) represents one experiment (n=3/group), (I) representative of three experiments. *Duplicate loading control. Due to overlapping sizes, loading controls were analyzed on separate gels. Same amount of protein was run for analyses (I, m).
Figure 10. (a) 13C-palmitate, (b) D-13C-glucose, and (c) 3C-glutamine GCMS trace analysis of specified metabolites in M(IL-4) ± GC7 for 24 hours, (c-e) Isotopologue evaluation of indicated metabolites in 13C-glutamine-labelled M(IL-4) ± GC7, where d,e indicate oxidative and reductive metabolism, respectively. Red X indicates TCA cycle breaks from proteomics data. All data are mean ± SEM (p*<0.05, p**<0.005, p***<0.0005). (a) Represents one experiment, (b-f) representative of two experiments.
Figure 11. Immunoblot analysis of specified proteins in (a, b) M(IL-4) ± 10 μΜ GC7 for 22 hours; (c) M0, M(L/y), M(IL-4) (left panel) and M(IL-4) (right panel) ± GC7; (d) EG7-OVA cells ± DFMO/DENSPM for 48 hours; (e) M(IL-4) ± DFMO for 48 hours; (f) M(IL-4) ± 20 μΜ CPX for 24 hours; (g) M(IL-4) expressing control, Dhps, or Eif5a shRNA. (h) OCR in M(IL-4) treated for 24 hours with GC7 ± dimethylsuccinate. (a-b,f) Representative of three experiments, (c-e,g) representative of two experiments. ABAT (4-aminobutyrate
aminotransferase), ACC (acetyl CoA Carboxylase), ACO (aconitase), ASL (argininosuccinate lyase), DLD (dihydrolipoamide dehydrogenase), FAS (fatty acid synthase), GLDH (glutamate dehydrogenase), G6PDH (glucose-6-phosphate dehydrogenase), HKI (hexokinase I), IDH1 (isocitrate dehydrogenase 1 ) IDH2 (isocitrate dehydrogenase 2), MCM (methylmalonyl-CoA mutase), PDH (pyruvate dehydrogenase), MDH2 (malate dehydrogenase 2), SCS (succinyl- CoA synthetase), SDHA (succinate dehydrogenase A). *Duplicate loading control. Due to overlapping sizes, loading controls were analyzed on separate gels. Same amount of protein was run for analyses (Fig 2I, m). Figure 12. eIF5A regulates macrophage activation and T cell differentiation. CD301 and RELMa expression assayed by flow cytometry in (a) GC7 and (b) CPX-treated M(IL-4). (c) (IL-4) transduced with the indicated shRNA were assessed for CD301 expression, (d) Absolute number of peritoneal macrophages elicited from mice treated with IL-4 complex ± 10mg/kg GC7 (n=4-5/group). (e) Immunoblot analysis of indicated proteins in MO, M(L/y), and M(IL-4) with relative densitometry of elF5AH, values were normalized to GADPH. (f) Relative mRNA expression of indicated genes as revealed by RNA-Seq in OT-I CD8+ T cells activated with anti-CD3/CD28 for 24 and 48 hours (left panel) or adoptively transferred and sorted from spleen of L. monocyfogenes-expressing-ovalbumin (LmOVA) infected congenic mice at the indicated time point (right panel, derived from ImmGen Database), (g) OCR and ECAR of CD8+ T cells activated in vitro with anti-CD3/CD28 ± 10 μΜ GC7. (h) Expression of CD69 on cells in (g). (i) OVA-peptide-activated OT-I CD8+ T cells were differentiated into IL-2 TE or IL- 15 T after 3 days of treatment with IL-2 or IL-15, respectively, starting on day 3 post- activation. OCR was assessed in indicated cells ± GC7 at baseline and in response to PMA/ionomycin restimulation. (j) Activated P14 CD8+ T cells were transduced with Eif5a- shRNA and adoptively transferred into congenic recipient mice infected with LCMV Armstrong and tracked over time in blood. Representative contour plots are gated on CD8+ CD45.1 + gp33-tetramer+ cells, n=10/group. All data are mean ± SEM (p*<0.05, p**<0.005, compared to M(IL-4) condition), (a) Represents three experiments, (b, c, g-i) representative of two experiments, (e) representative of two-three experiments, densitometry represents three experiments (f) represents one experiment.
Figure 13. Expression of markers associated with alternative activation in M(IL-4) (a) ± CPX, (b) ± GC7, (c) expressing control or £/r~5a-shRNA, (d) expressing control or Dhps-s R A, (e) expressing control or Dohft-shRNA. (f, g) Western blots of indicated proteins in M0, M(LJy), M(IL-4) ± GC7 and (g) analysis of NOS2 expression by flow cytometry in M0, M(L/y) ± GC7 (right panel). (h,i) Markers of classical activation in M0 and M(L/y) transduced with the
indicated shRNA by flow cytometry, (j) Activation marker expression on OT-I CD8+ T cells ± GC7, activated in vitro with OVA peptide and IL-2. (k) OVA-peptide-activated OT-I CD8+ T cells were differentiated into IL-2 TE or IL-15 TM after 3 days of treatment with IL-2 or IL-15, respectively, starting on day 3 post-activation. ECAR was assessed in indicated cells ± GC7 at baseline and in response to PMA/ionomycin restimulation. All data are mean ± SEM (p*<0.05, p**<0.005, p***<0.0005, compared to M(IL-4) controls), (a, c-e, j-k) Representative of two experiments, (b) represents one to three experiments, (f) represents one experiment, (g) Representative of one to two experiments, (h) Represents two experiments, (j-k) representative of two experiments. *Duplicate loading control. Due to overlapping sizes, loading controls were analyzed on separate gels. Same amount of protein was run for analyses (Fig 21, m).
Figure 14. (a) Activated and control OT-I CD8+ T cells ± the indicated concentration of GC7 were assessed for proliferation on day 3 of culture, (b) CFSE proliferation analysis of OT-I CD8+ T cells treated with GC7 for the indicated time periods, (a-b) representative of one to three experiments.
Figure 15. Mitochondrial metabolism is controlled by hypusinated elF5A-dependent translation of metabolic machinery, (a) Relative mRNA expression of indicated genes in GC7-treated M(IL-4). (b) HA-tagged elF5A construct was retrovirally transduced into MEFs and bound mRNAs were immunoprecipitated and submitted for microarray analysis against total RNA. (c) KEGG pathway analysis of enriched mRNAs bound to elF5A, numbers above bars indicate percent of genes mapped from submitted list, (d) Target sequences were cloned into the N-terminus of mCherry fused to a degron (to limit its half-life) separated by a GSGSG flexible linker to allow correct and independent folding of the introduced sequences and mCherry. These were sub-cloned into the MIGR1 vector and transduced into MEFs (Suclgl is a subunit of Succinyl CoA Synthetase), (e) Representative confocal images of cloned constructs scale bar = 10 μηι, SV40 NLS and IDH2 MTS are featured with GFP control, (f) Representative histograms of indicated constructs ± GC7. All data are mean ± SEM (p*<0.05, p**<0.005, p***<0.0005). (a) Representative of two to three experiments (n=3/group), (e) representative of two experiments, (f) representative of four experiments.
Figure 16. (a) IDH2 MTS and MCM MTS target sequences fused to mCherry. (b) Representative histograms of indicated constructs ± GC7, representative of six independent experiments.
The examples illustrate the invention.
Example 1 - elF5a and the alternative activation of macrophages (M2 cells)
It was reasoned that elF5a and the polyamine pathway could be important for the alternative activation of macrophages (M2 cells) given that they express arginase, the enzyme that converts the amino acid arginine into ornithine, the precursor to polyamines. Indeed, using N1-Guanyl-1 ,7-diaminoheptane (GC7), a drug that inhibits the function of deoxypusine synthase and therefore blocks elF5aHyp formation, it was found that M2 macrophage differentiation is impaired in the absence of functional elF5a, unlike M1 cells which are unaffected (figure 3). The action of GC7 also had profound effects on macrophage metabolism. M2 cells engage oxidative phosphorylation (OXPHOS) to help drive their differentiation, GC7 treatment significantly decreased OXPHOS in M2 cells (figure 4). M1 metabolism was unaffected in the presence of GC7 (data not shown). Thus, hypusinated elF5a was found to be an important factor in regulating macrophage differentiation and respiration.
Example 2 - elF5a-hypusine levels in untreated or GC7-treated kidney cells
The ability of compounds to inhibit elF5a hypusination can be determined by analyzing the quantity of elF5a-hypusine by western blot in untreated control cells versus treated cells. In figure 5, an example of this is shown - here, kidney cells were incubated with the deoxyhypusine synthase inhibitor GC7 and the quantity of elF5a-hypusine determined by western blot. GC7-treated cells showed a significant reduction in elF5a-hypusine levels.
Example 3 - Exposure of murine embryonic fibroblasts (MEFs) to 2-difluromethylornithine (DFMO) and to diethylnorspermine (DENSPM)
Polyamines are synthesized in metabolically active cells7 (Fig. 6a). To investigate how this pathway contributes to metabolism murine embryonic fibroblasts (MEFs) were exposed to 2- difluromethylornithine (DFMO), an ornithine decarboxylase (ODC) inhibitor8, and to diethylnorspermine (DENSPM), which activates the spermidine catabolising enzyme SSAT9. Blocking polyamine biosynthesis, or inducing catabolism of spermidine, inhibited oxygen consumption rates (OCR, an indicator of OXPHOS), while having a limited effect on extracellular acidification rates (ECAR, an indicator of aerobic glycolysis) (Fig. 6b, Fig. 7a). LCMS analysis of treated cells confirmed a drop in spermidine levels, whereas the upstream metabolite ornithine was unaffected (Fig. 6c). An important cellular function of spermidine is to provide substrate for DHPS, the rate-limiting enzyme during elF5AH formation (Fig 6a). Cells exposed to N1-guanyl-diaminoheptane (GC7), a spermidine analogue that_inhibits
DHPS10 and thus elF5AH (Fig. 7b), or ciclopirox (CPX), an inhibitor of DOHH1 , also dampened OXPHOS (Fig. 6d, e). To examine the loss of respiration genetically, IPTG- inducible £/Y5a-shRNA was transduced into MEFs (Fig. 6f) and a direct correlation between elF5A expression and respiration was observed (Fig. 6g). Tamoxifen-induced deletion of DHPS (Fig. 6h) displayed a similar loss of OCR (Fig. 6i). For reasons that are not clear at this time, these various treatments had differential effects on ECAR (Fig. 7c-g)._Collectively, the results suggest that the polyamine biosynthesis pathway controls OXPHOS via elF5AH (i.e hypusinated elF5A), a process conserved across species (Fig. 6j). Example 4 - Role of via elF5AH in the regulation of respiration
When MEFs were exposed to 2-deoxyglucose (2-DG), which enforces OXPHOS by limiting glycolysis (Fig. 8a), those expressing £//5a-shRNA did not compensate by enhancing OCR when compared to controls (Fig. 9a), revealing that when forced, cells with deficient levels of elF5A are unable to upregulate mitochondrial respiration. To further probe respiratory function controlled by elF5A, ΒΜΜΦ were generated and then these cells were activated with IL-4 [M(IL-4)]. IL-4 driven activation of these cells is dependent on mitochondrial respiration12. Since M(IL-4) and LPS/IFN-y-activated macrophages [M(LPS/IFN-y)] do not proliferate to any appreciable extent in v/'fro13,14, it was possible to assess the role of polyamine biosynthesis on respiration dissociated from its known ability to regulate proliferation (Fig. 8b)15. Inhibiting elF5AH, either pharmacologically or genetically, blocked respiration in M(IL-4) (Fig. 9b-f, Fig. 8h), while again having differential effects on ECAR (Fig. 8c-g). To further understand the function of elF5AH in regulating respiration, metabolites were analyzed by LCMS. GC7-treated M(IL-4) (Fig. 9g, h) and £/75a-shRNA-expressing MEFs (Fig. 8i) displayed decreased metabolites associated with the first half of the TCA cycle, as well as additional alterations in other metabolic pathways (Fig. 8j, k). Next, M(IL-4) were cultured with 13C-labeled glucose, glutamine, or palmitate and carbons were traced from these substrates into metabolites. GC7-treated M(IL-4) incorporated significantly less carbon from 13C-glucose into TCA cycle metabolites compared to control cells (Fig. 9i), indicating that DHPS function, and thus elF5AH, regulates TCA cycle engagement. Incorporation of 13C- glutamine and palmitate carbons into TCA cycle metabolites was also decreased in GC7- treated cells (Fig. 9j, Fig. 10a). However, overall glutamine utilization was increased after GC7-treatment, indicated by greater fractional contribution of 13C-glutamine to glutamate (Fig. 9j). Increased incorporation of 13C-glucose into lactate (Fig. 10b) in GC7-treated M(IL- 4) supports increased glycolysis (Fig. 7c). Accumulation of citrate m+5, and malate,
fumarate, and aspartate m+3 in cells cultured in 13C-glutamine with GC7 indicate these metabolites are generated from reductive, rather than oxidative, glutamine metabolism (Fig. 10c-e), a phenotype consistent with cells with respiratory defects16. Example 5 - Proteomics analysis of GC7-treated M(IL-4)
To ascertain why TCA cycle flux was inhibited, a proteomics analysis of GC7-treated M(IL-4) was performed. Of 153 proteins with significantly altered expression, 63 were mitochondrial, including TCA cycle enzymes and ETC proteins, consistent with dysregulated metabolism (Fig. 9k, Table 2).
Table 2 - Proteins with significantly altered expression Significance Pvalue Log2FC Gene names Gro
1 ,61638562 -2,6822497 Dap3 2
3,23904194 -2,6599585 Did 2
2,54358627 -2,338143 Mpst
2,07361303 -2,3342241 Mut 2
3,44141341 -2,3190365 ΙΙ4Ϊ1 4
2,18938402 -2,2387053 Plau 4
3,55548473 -2,1451321 Pdp2 2
2,42947291 -1 ,9730466 Tacol 2
2,69287908 -1 ,9594231 C1qb 6
2,86588295 -1 ,8919748 Mrpl15 2
2,97469533 -1 ,7968871 Endog 2
2,73933999 -1 ,7271277 Txnip 4
3,71543358 - ,643837 Mrpl53 2
2,83706452 -1 ,6322257 Slirp 2
2,59569418 -1 ,6053034 Fech 2
4,56995079 -1 ,6048381 Gatm 2
2,89614111 -1 ,5880718 Mrpl4 2
3,43220561 -1 ,5437393 Mgl2 8
1 ,35991293 -1 ,53339 Ly86 8
2,29431058 -1 ,5095889 C1qa 6
3,64872233 -1 ,4866397 Nrp2 8
3,60572886 -1 ,4790204 Tbrg4 2
3,07273817 -1 ,4754645 Lrpprc 2
3,6767102 -1 ,4688657 Mrps5 2
3,38499629 -1 ,4618829 Mrps23 2
4,08669921 -1 ,4353498 Acatl 2
2,39097266 -1 ,4323832 Mrpl24 2
1 ,73617642 -1 ,4228071 Plxdc2
,43451734 -1 ,3898404 Aldh6a1 2,31607484 -1,3504715 Gm9755;Tufm 2,46383754 -1,3470567 Bckdha 2,64514881 -1 ,3433749 Cyp51a1 4,58691312 -1,3167547 Suclg2 2 ,63533768 -1,2827237 Tnip3 4,41839073 -1,2631302 Suclgl 2,12159429 -1,2479337 Adck4 2,45507633 -1,2378165 Mrps6 2,62963139 -1,2362143 Spryd4 4,66745116 -1,1789284 Aco2 2,33048461 -1,1746095 Sdf4 10,39543356 -1,1741689 2,11950796 -1,1582934 Mmp12 6 ,2335743 -1,1499882 S100a9 4,24242332 -1,1119525 Fcrls 8,45313354 •1,1111806 Hist1h1a 14 ,06227802 -1,1054045 Hdhd2;ler3ip1;Gm10784 10,46321394 -1,1034241 Rapla 4,80466568 -1,099844 Gfm2 2,07457289 -1,0978165 Mrpl43 2 ,2755759 •1,0907281 Tars2 2,93988764 ■1,0809441 Hfe 8,20465084 ■1 ,0758966 Sill 10,68812696 -1,051501 lsoc2a 2,11087396 1 ,0505759 Coq9 2,67610593 1,0502205 Lipa 12,73767296 ■1,0465832 Iscu 2,35668348 1,0416698 Rbm5 4,12035521 1 ,0349407 Fundc2 2,70756973 1 ,0064894 P4ha1 4,89422251 0,9979471 C1qc 6,00641581 0,9955037 Aldh2 2,12565541 -0,99333 IHrap 8,63896178 0,9920311 Cdk4 4,95283459 -0,970431 Mipep 2,01181725 0,9704189 Tceb3 14,16621371 0,9676393 Atp5e 2,28814293 -0,967289 Nold 14,47918096 0,9612605 Hadhb 2,38858253 0,9548855 Clybl 2,59824647 0,9482288 Icam2 8,41761695 0,9349454 Csflr 8,90422824 0,9244715 Sucla2 2
,22366559 -0,9241314 Itgb3 8,14579813 -0,9196065 Lpl 6,77657933 -0,9118894 Oxctl 2 ,45330453 -0,9023914 Ddx54 14,35774591 -0,9020297 Pdhal 2,51386158 -0,8980109 Aldh1 b1 2 ,7364232 -0,8964577 Sdhb 2 ,7421013 -0,8949674 Xyltl 4,41194281 -0,8789094 Cfp 8 ,54889862 -0,8778667 Hist1 h1 b 14,26757313 -0,8777682 Ncoa5 14,51498086 -0,8712813 Clpp 2,42798443 -0,8712114 Ndufv2 2,53738176 -0,8699748 Histl e 14,80109801 -0,8687051 Top2a 14,07196151 -0,867609 Ndufa9 2 ,67965609 -0,8586871 Sumfl 10,70045778 -0,858202 Rrp1 14,19609011 -0,851291 Pdhb 2,01831974 -0,8498898 Csf2ra 8,01582197 -0,8468628 UqcrlO 2 ,50379676 -0,8465913 Hist1 h1c 14,82670852 -0,8419902 Auh 2,26641122 -0,8403238 Brixl 4,86709888 -0,8314832 Mccc2 2,80206646 -0,8297551 Pltp 4,71715257 -0,8271624 Ndufs7 2,27316052 -0,823417 Ndufb9 2
2,547406 -0,8209324 Usmg5 2,41982039 -0,8171088 Clec7a 8,23319415 -0,8108686 Grn 6,91526189 -0,8087025 Ndufaf2 2,19621169 -0,789978 Cd72 8,61634201 -0,7883053 Meed 2,30568402 -0,7841689 mt-Co3 2,12019742 -0,7806079 Ndufa6 2,36049226 -0,7407722 Lgals3bp 8,87389065 0,74764252 Nars 4,20063436 0,75276947 Fam195b 4,46258339 0,75688171 Gars 4,50742278 0,76665497 Wars 4,10527426 0,77888934 Prpsapl
,27540546 0,78190931 Chuk 4,03498657 0,78310776 Got1 4
2,50675605 0,78502019 Pgp 4
2,88152031 0,79515584 Cars 4
1,90164828 0,79519717 Pycrl 2
2,18255168 0,7986838 Gdi1 4
1,95027297 0,79903412 Trove2 4
2,51830632 0,80703926 Asl 4
1,68500164 0,8124307 Stmnl 4
2,08287112 0,81960487 Eif2b5 14
2,48402834 0,82046763 Sms 4
2,56792453 0,82109006 Naip2 2
2,80228428 0,82948812 Blmh 4
1,64845403 0,82990519 Cryl1 4
1,23293145 0,84697215 MarcksH 4
2,40338474 0,84962018 Ufm1 4
1,32418452 0,8542436 Cnripl 4
1,85587519 0,86110751 Psph 4
2,26021611 0,8707854 Akr1b8 4
2,21152278 0,87606621 Ca13 4
2,13080199 0,8809255 Serpinb6a;Serpinb6 4
2,05953659 0,90220451 Ccdc9
1,34682833 0,91723379 Wbp2 4
2,46712882 0,91964277 Plin2 4
2,27757354 0,92434438 Cebpb 14 2 3,26981017 0,92744827 Aars 4
1,44714079 0,93698883 Nt5c3b 4
1,422935 0,94647662 Carhspl 4
1,67602144 0,9753863 Sashl 4
2,65966073 1,01736641 Sphk2 14
2,47951926 1,06537628 Wdr44 4
1,41719775 1,08094025 Aldh7a1 2
2,58846716 1,10059802 Ppmel 14
2,73078213 1,11097272 Aspscrl 4
1,5827671 1,18353907 Aril 4
2,12550234 1,23897298 Sra1 4
2,98120236 1,28174782 Asns 4
1,64301185 1,28371048 Rpe 4
3,08537371 1,34967677 Naa20 4
4,85099564 1,44808896 Sqstml 4
2,2625328 1,70083173 Myl12b; yl9 4
2,87783113 1,75317446 Alasl 2
Legend Table 2
Significance values columnl is 1
Group column5 Mitochondrial is 2
Group column5 Cytoplasmic is 4
Group column5 Secreted is 6
Group column5 CellSurface is 8
Group column5 ER resident is 10
Group column5 Lysosomal is 12
Group column5 Nuclear is 14
Subgroup column6 M IL4 is 2
To validate our proteomics data, TCA cycle enzymes in GC7-treated M(IL-4), as well as in resting macrophages (MO) and M(LPS/IFN-y) were assessed, by western blot. In each cell type decreased expression of several TCA proteins was found, including succinyl-CoA synthetase (Suclgl ) and succinate dehydrogenase (SDH), supporting a break in the TCA cycle, while citrate synthase (CS) and isocitrate dehydrogenase (IDH) were less affected (Fig. 91, Fig. 11 a). Other enzymes identified by proteomics analysis that feed substrates into the TCA cycle, such as methylmalonyl-CoA mutase (MCM), were also diminished after GC7 treatment (Fig. 11 b), while expression of many enzymes in glycolysis, fatty acid synthesis, and the aspartate-arginosuccinate shunt remained stable (Fig. 11c). Exposing M(IL-4) or lymphoma cells to polyamine synthesis inhibitors also resulted in TCA cycle enzyme deficiencies, as did treatment with CPX or expression of Eif5a and Dhps-s RNA (Fig. 11d- g)-
Example 6 - Assay of ETC complexes
Since SDH also participates in the ETC as complex I I, ETC complexes were assayed and it was found that GC7-treated MO and M(IL-4) had dampened expression of complexes I, II, and IV, while M(LPS/IFN-Y) had decreased expression of these complexes regardless of GC7 treatment (Fig. 9m). Consistent with decreased TCA cycle metabolites in GC7-treated cells (Fig. 9h), a partial rescue of respiration upon exposure to succinate was observed (Fig. 11 h). These data indicate that although SDH/complex II expression is decreased in GC7- treated M(ll_-4), the absence of TCA-derived metabolites contributes to the block in respiration.
Given that M(IL-4) rely on OXPHOS while M(LPS/IFN-y) do not6'12, markers of alternative activation in M(IL-4) upon elF5AH inhibition were assessed. Pharmacologic inhibition of elF5AH blunted RELMa expression (Fig. 12a, Fig. 13a), while GC7 and CPX treatment, or genetic ablation of Eif5a, Dhps or Dohh blunted Arg1 , CD301 , and CD206 to varying degrees (Fig. 12a-c, Fig. 13a-e), indicating inhibition of alternative activation, despite intact IL-4 signaling through STAT6 (Fig. 13f). Proteomics data of GC7-treated M(IL-4) confirmed the down-regulation of many proteins associated with alternative activation, including CD301 (Table 2). GC7 also diminished accumulation in vivo of IL-4 complex (IL-4c)-elicited macrophages in the peritoneal cavity (Fig. 12d). Markers of classical activation, such as nitric oxide synthase 2 (NOS2), were unchanged in M(LPS/IFN-y) (Fig. 13g-i). Proteins in the polyamine-elF5AH axis were enriched in M(IL-4) compared to M(LPS/IFN-y) (Fig. 12e), supporting the notion that this pathway is dynamically regulated in these cells.
Example 7 - Expression of genes associated with the polyamine-elF5AH axis
Expression of genes associated with the polyamine-elF5AH axis was also increased after activation in primary T cells (Fig. 12f). Data from the ImmGen database showed that genes in this pathway are induced in T cells acutely after Listeria monocytogenes infection, decreased in mature effector T cells, and increased in memory T cells (Fig. 12f). These data are in line with the fact that mitochondrial respiration is important in T cells during activation for antigen-driven proliferation17,18 and for memory T cell development and survival19"21, but is not required in fully differentiated effector T cells18. Consistent with the idea that elF5AH controls respiration, GC7-treated T cells did not engage OXPHOS following activation, although ECAR was unaffected (Fig. 12g), and expression of T cell activation markers were unperturbed in the presence of GC7 (Fig. 12h, Fig. 13j). Similarly, in vitro generated memory T cells cultured with GC7 augmented ECAR in response to restimulation, but could not increase OXPHOS (Fig. 12i, Fig. 13k). GC7 also blocked T cell proliferation in a dose- dependent manner when present during naive T cell activation (Fig. 14a), but this effect was mitigated when GC7 was introduced days later (Fig. 14b), consistent with an initial requirement for OXPHOS to induce clonal expansion during T cell activation17,18. To confirm these findings genetically and in vivo, LCMV-specific P14 T cells (gp33+) were transduced with a retrovirus expressing £/T5a-shRNA, which was introduced after activation, and adoptively transferred these cells into LCMV-infected recipients. Donor cells after infection were tracked and it was found that while both control and £/75a-shRNA-transduced cells participated in the primary effector T cell response 8 days after infection, the frequency of £/75a-shRNA-transduced donor cells was decreased in the weeks after infection compared to
control cells, indicating that the cells expressing £/T5a-shRNA were less able to form memory T cells (Fig 3j). These results suggest that elF5AH regulates the phenotype of immune cells that rely on mitochondrial metabolism. Example 8 - Control of OXHPOS by elF5AH
It was next assessed how elF5A controls OXHPOS. Although proteins of many TCA cycle enzymes were decreased, treatment with GC7 did not diminish their transcription, and in fact enhanced transcription of many genes (Fig. 15a), indicating that the TCA cycle defects conferred by GC7 are not transcriptional and suggests that elF5AH regulates TCA cycle enzyme translation. To investigate further MEFs were transduced with HA tagged-elF5A and compared immunoprecipitated bound mRNAs (RNA-IP) to total mRNA (Fig. 15b). As elF5A is ribosomally bound, this approach identifies ribosome-associated transcripts that are elF5AH-dependent. 1023 mRNAs that specifically co-precipitated with elF5A were identified. KEGG analysis revealed these transcripts were enriched for carbon metabolism, specifically for the TCA cycle and amino acid utilization (Fig. 15c, Table 3), suggesting that elF5AH is a critical factor for the posttranscriptional regulation of mitochondrial metabolism.
Table 3 - KEGG analysis of the 1023 mRNAs that specifically co-precipitated with elF5A
ID Description GeneRatio BgRatio p-value genelD
60525/14719/18642/14381/ 8293/1 10208/108037/11409/18563/11674/ 15926/14751/66904/227095/12359/ mmuO 110821/18641/15929/11428/972121 1200 Carbon metabolism 24/350 118/8022 1.69E-10 78920/56451/170718/236539
12039/11669/78038/257633/56357/ 11409/113868/78894/52538/66904/ mmuO Valine, leucine and 227095/110821/97212/12036/1167 0280 isoleucine degradation 15/350 56/8022 9.32E-09 1
11670/319625/12039/12583/60525/ 11669/14719/71780/78038/18642/1 4381/11966/104112/171210/18293/ 110208/108037/26922/227620/563 57/109900/14085/11486/50917/686 31/72157/11409/18799/22436/7042 8/13026/76238/18563/113868/1167 4/15926/12660/109652/12408/1475 1 /52538/216134/20698/22275/1464 5/66904/227095/225326/110821/31 9945/11655/11717/18641/69080/14 854/15929/235386/76952/110119/1 1428/71743/68603/218138/50798/7 mmuO 0266/26897/226518/11677/11898/6 1100 Metabolic pathways 97/350 1309/8022 2.6E-08 6054/11430/192156/22247/234730/
67873/17995/56749/97212/14584/1
9062/78920/14431 /16832/69719/27
0076/623661/56451/171567/20975/
18971/170718/69772/14187/14528/
12036/11671/236539
14719/18642/108037/109900/1856
3/11674/15926/109652/14645/1864 mmuO Biosynthesis of amino 1 /15929/11428/11898/170718/1203
1230 acids 16/350 78/8022 1.78E-07 6/236539
mmuO 104112/18293/18563/15926/15929/
0020 Citrate cycle (TCA cycle) 9/350 32/8022 5.95E-06 11428/78920/56451/170718 mmuO 2-Oxocarboxylic acid 14719/15926/109652/15929/11428/
1210 metabolism 7/350 19/8022 9.07E-06 170718/12036
mmuO Fructose and mannose 18642/11674/18641/69080/110119/
0051 metabolism 9/350 35/8022 1.33E-05 218138/11677/234730/14187 mmuO 72774/56626/11545/18207/16882/1
3410 Base excision repair 9/350 35/8022 1.33E-05 8971/11792/235587/22594 mmuO 11669/11409/113868/52538/12896/
0071 Fatty acid degradation 10/350 49/8022 3.94E-05 11430/97212/12894/270076/11671
Amino sugar and 227620/72157/14751/69080/11011 mmuO nucleotide sugar 9/218138/50798/245847/234730/14
0520 metabolism 10/350 49/8022 3.94E-05 584
mmuO 12039/60525/66904/227095/11082
0640 Propanoate metabolism 8/350 31/8022 3.96E-05 1/97212/16832/56451
11670/319625/60525/11669/18642/ mmuO Glycolysis / 72157/11674/14751/18641 /16832/1
0010 Gluconeogenesis 11/350 66/8022 0,000116 1671
mmuO Biosynthesis of 171210/113868/111175/26897/114
1040 unsaturated fatty acids 7/350 28/8022 0,000152 30/70025/97212
mmuO Glyoxylate and 108037/76238/14645/66904/12359/
0630 dicarboxylate metabolism 7/350 29/8022 0,000192 110821/11428
224824/67528/22436/113868/1592 mmuO 6/12359/111175/68603/16922/1143
4146 Peroxisome 12/350 83/8022 0,000232 0/26874/56794
mmuO 26922/11409/113868/52538/11117
1212 Fatty acid metabolism 9/350 52/8022 0,00036 5/12896/11430/97212/12894 mmuO Pentose phosphate 18642/14381/110208/72157/11674/
0030 pathway 7/350 32/8022 0,000371 14751/18641
It was next questioned how elF5AH might exert selective control over these proteins. elF5AH facilitates translation elongation of difficult to translate proteins or motifs, such as those with proline, glycine, or charged amino acids, which can lead to ribosome stalling4,5. Given that MTS are common to mitochondrial proteins, and are rich in positively charged residues, it was hypothesized that the MTS of specific mitochondrial proteins could make them dependent on elF5AH. Supporting this idea, GO analysis of subcellular localization of mRNAs co-precipitated with elF5A revealed significant enrichment for mitochondrial localization (p=1.4x10"27). To test how elF5A regulates translation of specific mitochondrial proteins, the MTS of several proteins (Suclgl , SDHA, and MCM) identified by our proteomic and RNA-IP
analyses were cloned (Tables 1, 2) to be elF5AH-dependent and were fused to the N- terminus of mCherry in MIGR1 , whereby mCherry expression is dependent on the translation of the preceding sequence (Fig. 15d, Fig. 16a). As two controls, in place of the MTS, SV40 nuclear localization sequence (NLS) was used, which the inventors would predict to be elF5A-independent, or a polyproline stretch described to be elF5A-dependent22 (Fig. 15d). The MTS of IDH2 (Fig. 16a), a protein that was found to be elF5A-independent by proteomics and RNA-IP, was also examined (Tables 1, 2). Using confocal microscopy, it was confirmed that the MTS mCherry constructs localized to mitochondria, whereas SV40 NLS mCherry was in the nucleus (Fig. 15e). IRES-driven GFP expression was equivalent in cells transduced with all constructs, and unaffected by GC7 treatment (Fig. 15f).
In the absence of GC7, Suclgl , SDHA, and MCM MTS mCherry expression was decreased compared to control, NLS, or polyproline mCherry, indicating that the MTS of these proteins are difficult to translate (Fig. 15f, Fig. 16b). In contrast, IDH2 MTS mCherry was more highly expressed (Fig. 16b). When transduced cells were exposed to GC7, the expression of Suclgl , SDHA, and MCM MTS mCherry was further abrogated (Fig. 15f, Fig. 16b). This effect was much less apparent on IDH2 MTS or SV40 NLS mCherry expression. As expected, a modest reduction of polyproline mCherry was also observed (Fig. 15f). Together these data suggest that a subset of MTS are difficult to translate and are therefore dependent on elF5AH, a specialized factor known to facilitate translation of transcripts with specific sequence properties4'5.
Example 9 - Discussion
It is shown herein that the polyamine-elF5AH axis regulates mitochondrial respiration by enabling translation of distinct mitochondrial enzymes. These findings are supported by a recent report that genetic and GC7-driven inhibition of elF5AH silences mitochondria in kidney cells, preventing anoxic cell death and improving kidney transplant outcome10. Our data suggest that MTS of specific proteins confer dependency on elF5AH. However, given that the SV40 NLS, which is rich in positively charged residues, is not subject to elF5AH regulation, it is likely that the presence of positively charged residues alone does not confer specificity. Recent evidence notes that the location of these residues may play a role in regulation by elF5AH, suggesting that MTS secondary structure or some other element may dictate ribosome stalling and hence reliance on elF5AH 4'5. Our data indicate that elF5AH- dependent MTS are difficult to translate regardless of GC7 treatment, supporting that the selectivity of elF5AH regulation is, at least in one aspect, at the level of translational competency. The precise nature of MTS differences, or how additional factors modulate the ultimate effect elF5AH has on protein translation, has yet to be determined. Bacteria lack the
amino acid modification hypusine, and its assembly machinery is incomplete in many archaea23. That the hypusine modification became so ubiquitous in eukaryotes might reflect strong selective forces in early-nucleated cells attempting to overcome the novel problem of routing host proteins to the mitochondria.
Both polyamine synthesis and elF5A expression are upregulated in cancer cells24,25. Though these changes were largely thought to be important for driving proliferation, our data suggest these alterations might also provide cancer cells with the ability to fine-tune OXPHOS, an area of considerable interest in cancer biology. Recent studies have highlighted TCA cycle breaks in inflammatory macrophages26,27. Our data showing that M(LPS/IFN-Y) lose expression of selected ETC complexes suggests the possibility that TLR signaling may modulate the elF5AH-axis to regulate respiration. Previous studies have demonstrated the engagement of polyamine biosynthesis and elF5A in immune cells28"30. Our findings expand on this knowledge by showing this pathway controls fate and function of different immune cell types. A new mechanistic understanding into the role of polyamine biosynthesis in cells via elF5AH is provided herein and it is believed that this pathway can be modulated in immune cells for therapeutic benefit.
Example 10 - Material and Methods
Mice and Immunizations
C57BL/6, C57BL/6 CD45.1 , Arg1 -YFP C57BL/6, ovalbumin (OVA)-specific TCR OT-I transgenic and P14 TCR transgenic mice specific for LCMV were purchased from Jackson Laboratories. All mice were bred and maintained under specific pathogen free conditions under protools approved by the Animal Welfare Committee of the Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany. Mice used in all in vitro and in vivo experiments were 6-10 weeks of age and were age/sex matched. For lymphocytic choriomeningitis virus (LCMV) infections, mice were immunised with 2x105 plaque forming units (PFU) LCMV Armstrong strain by intraperitoneal injection.
Cell Culture
Bone marrow cells were differentiated for 7 days into bone marrow macrophages (ΒΜΜφ) by culturing in complete medium (RPMI 1640 media supplemented with 10% FCS, 2mM L- glutamine, 100 U/mL penicillin/streptomycin) with 20 ng/mL macrophage colony-stimulating factor (M-CSF; PeproTech). M(IL-4) were generated with 20 ng/mL IL-4 overnight from day 7 of culture; M(LPS/IFNy) were generated with 20 ng/mL LPS (Sigma) and 50 ng/mL IFN-γ (R&D Systems) overnight from day 7 of culture. All drug treatments on ΒΜΜφ began from
day 7 of culture. N1-guanyl-1 7-diaminoheptane (GC7; Enzo Life Sciences) and ciclopirox (Sigma) were typically used at 10 μΜ and 20 μΜ, respectively, unless otherwise stated. Difluormethyl ornithine (DFMO) and diethylnorspermine (DENSPM; Tocris) were used 2.5 mM and 50 μΜ, respectively. Dimethylsuccinate (Sigma) was used at 5mM. DHPSFIOXROX Cre-ER MEFs, generated as previously described31, were cultured in complete DMEM (DMEM supplemented with 10% FCS, 2mM L-glutamine, 100 U/mL penicillin/streptomycin) and generated as previously described31, Cre-ER expression was induced with 1 μΜ 4-OHT (Sigma) for the indicated time period. NIH3T3 MEFs (purchased from ATCC) stably transduced with Eif5a-shRNA were grown in complete DMEM and E/"f5a-shRNA expression was induced with 100 μΜ isopropyl^-D-1 thiogalactopyranoside (IPTG, Sigma) for the indicated period of time. Madin-Darby Kidney Canine (MDCK) cells were grown in complete DMEM, as was the human breast adenocarcinoma line MCF-7 but with 0.01mg/ml recombinant human insulin. D. melanogaster Schneider 2 (S2) cells were cultured without C02 at 28°C in complete Schneider's Drosophila medium (Gibco; supplemented with 10% FCS, 50 U/mL penicillin/streptomycin, 25% conditioned complete Schneider's medium). OTI splenocytes were activated with OVA peptide (SINFEKL, New England Peptide), or with anti- CD3 and anti-CD28 antibodies, with IL-2 (100 U/mL) in T cell media (RPMI 1640 media supplemented with 10% FCS, 2mM L-glutamine, 100 U/mL penicillin/streptomycin and 55 μΜ β-mercaptoethanol) for the indicated length of time. To generate in vitro IL-2 TE and IL-15 TM cells, OT-I splenocytes were activated with OVA-peptide and IL-2 (100 U/mL) for 3 days and subsequently cultured in the presence of either IL-2 or IL-15 (10 ng/mL), respectively, for an additional 3 days in TCM.
Metabolic Profiling
Extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) were measured using the Seahorse XFe Bioanalyser (Seahorse Bioscience). 8x104 ΒΜΜφ were added to seahorse 96 well plates on day 7 of culture and analysed in XF media (non-buffered RPMI 1640 containing 25 mM glucose, 2 mM L-glutamine, and 1 mM sodium pyruvate) the following day after cytokine and drug treatment. For T cells, 2x105 were spun down on a poly-D-lysine-coated seahorse 96 well plate. MEFs were plated at 4x104 cells per well of a 96 well seahorse plate in XF. Prior to analysis, cells were incubated for a minimum of 45 minutes at 37°C in the absence of C02. OCR and ECAR were measured under basal conditions, after restimulation with 50 ng/mL phorbal 12-myristate 13-acetate (PMA) and 0.5 μg/mL ionomycin, or after the addition of the following drugs: 1 μΜ oligomycin, 1.5 μΜ fluoro- carbonyl cyanide phenylhydrazone (FCCP), 100 nM rotenone, and 1 μΜ antimycin A (all
Sigma). Measurements were taken using a 96 well Extracellular Flux Analyser (Seahorse Bioscience).
Metabolite Tracing
ΒΜΜφ on day 7 of culture were washed and cultured in complete RPMI 1640 (minus glucose or glutamine), supplemented with either 11 mM 13C-glucose or 4mM 13C-glutamine, for 24 hours. 13C-palmitate (20 μΜ) was added to complete RMPI 1640 overnight. MEFs were washed and cultured in complete DMEM (with 10% dialysed serum, minus glucose or glutamine), supplemented with 25 mM 13C-glucose or 4 mM 13C-glutamine, for 24 hours. For harvest, cells were rinsed with cold 0.9% NaCI and metabolites extracted using 1.2 mL of 80% MeOH kept on dry ice. 10 nM norvaline (internal standard) was added. Following mixing and centrifugation, the supernatant was collected and dried via centrifugal evaporation. Dried metabolite extracts were resuspended in pyridine and derivatized with methoxyamine (sc- 263468 Santa Cruz Bio) for 60 minutes at 37 °C and subsequently with N-(tert- butyldimethylsilyl)-N-methyl-trifluoroacetamid, with 1% tert-butyldimethylchlorosilane (375934 Sigma-Aldrich) for 30 minutes at 80 °C. Isotopomer distributions were measured using a DB5-MS GC column in a 7890 GC system (Agilent Technologies) combined with a 5977 MS system (Agilent Technologies). Correction for natural isotope abundance and calculation of fractional contribution was performed as described elsewhere32.
Metabolite Quantification
Metabolites were quantified by LC-MS using HILIC Chromatography on an Acquity UPLC BEH Amide column 1.7 μιτι, 2.1x100 mm (polyamines) or a Luna NH2 column (all other metabolites) on a 1290 Infinity II UHPLC system (Agilent Technologies) combined with targeted detection in a 6495 MS system (Agilent Technologies). Peak areas were normalized to 13C labelled internal standard (ISOtopic Solutions).
Western blot
For western blot analysis, cells were washed with ice cold PBS and lysed in 1 x Cell Signaling lysis buffer (20 mM Tris-HCI, [pH 7.5], 150 mM NaCI, 1 mM Na2EDTA, 1 mM EGTA, 1% Triton X-100, 2.5 mM sodium pyrophosphate, 1 mM β- glycerophosphate, 1 mM Na3V04, 1 g/mL leupeptin (Cell Signaling Technologies), supplemented with 1 mM PMSF. Samples were frozen and thawed 3 times followed by centrifugation at 20,000 x g for 10 min at 4°C. Cleared protein lysate was denatured with LDS loading buffer for 10 min at 70°C, and loaded on precast 4% to 12% bis-tris protein gels (Life Technologies). Proteins were transferred onto nitrocellulose membranes using the iBLOT 2 system (Life Technologies)
following the manufacturer's protocols. Membranes were blocked with 5% w/v milk and 0.1 % Tween-20 in TBS and incubated with the appropriate antibodies in 5% w/v BSA in TBS with 0.1% Tween-20 overnight at 4°C. All primary antibody incubations were followed by incubation with secondary HRP- conjugated antibody (Pierce) in 5% milk and 0.1 % Tween- 20 in TBS and visualized using SuperSignal West Pico or femto Chemiluminescent Substrate (Pierce) on Biomax MR film (Kodak). Optical density of the signals on film was quantified using grayscale measurements in ImageJ software (NIH) and converted to fold change. All antibodies were from Cell Signaling Technologies except for anti-ABAT, Anti-ACC, anti- Aconitase 1 , anti-ASL, anti-DLD, anti-DOHH, anti-DHPS, anti-MCM, anti-ODC (Abeam), anti- elF5A (BD Bioscience), anti-hypusine (Merck-Millipore). Electron transport chain complexes were probed with the Total OXPHOS Rodent WB Antibody Cocktail (Abeam).
Retroviral Transduction
In the case of T cells, activated P14 splenocytes were transduced with luciferase (empty vector) or £ 5a-expressing retrovirus by centrifugation for 90 minutes in media containing hexadimethrine bromide (8 pg/mL; Sigma) and IL-2 (100 U/mL). GFP was used as a marker for retroviral expression. In ΒΜΜφ, bone marrow cells exposed to M-CSF were transduced with luciferase (empty vector) or Eif5a-,Dhps-, or ΟοΛΛ-expressing retrovirus by centrifugation for 90 minutes in media containing hexadimethrine bromide on day 2 of culture. Transduced cells were subsequently drug or cytokine-treated on day 7 of culture and assayed on day 8, sometimes following sorting on day 6. GFP was used as a marker for transduction in these cells.
Targeted mCherry
A G-block construct (IDT) containing mCherry fused to the degron from ODC 33 (HGFPPEVEEQDDGTLPMSCAQESGMDRH*) (mCherry*39) was constructed to reduce half- life of the mCherry fusion protein. Between the cloning sites and mCherry a Gly-Ser-Gly-Ser- Gly flexible linker was included, to allow correct and independent folding of the introduced sequences and mCherry. The mCherrydeg was cloned into MSCV-I-GFP using Xhol and EcoRI. MTS, NLS, or control sequences were ordered as G-blocks (IDT) or as phosphorylated oligos containg Xhol and BamHI compatible overhangs and cloned into MSCV-mCherryde9-l-GFP using Xhol and BamHI. The targeted sequences were as follows:
MTS-IDH
MAGYLRAVSSLCRASGSARTWAPAALTVPSWPEQPRRHY (SEQ ID NO: 1 ) MTS-Suclgl
MTATWAAAATATMVSSSSGLAAARLLSRTFLLQQNGIRHG (SEQ ID NO: 2) MTS-SDHA
MAGVGAVSRLLRGRRLALTGAWPGTLQKQTCGFHFSVGENKKASAKVSDAISTQYPWD
(SEQ ID NO: 3)
SV40-NLS
MPKKKRKV (SEQ ID NO: 4) PP (MPPPPPP) (SEQ ID NO: 5)
GFP-Control (MSKGEEL) (SEQ ID NO: 6)
MTS-MCM (MLRAKNQLFLLSPHYLKQLNIPSASRWKRLL) (SEQ ID NO: 7) Lentiviral Transductions
The IPTG-inducible MISSION shRNA lentiviral vector pl_KO-puro-IPTG-3xl_acO was purchased either with a shRNA against the 3"-UTR of the murine elF5A mRNA sequence (custom-made from #SHCLND-NM181582-TRCN0000125229; Sigma) or a corresponding non-target shRNA control (#SHC332-1 EA; Sigma). Stable transduction of the lentiviral was performed as previously described34 using HEK293T cells, the packaging plasmids: pMDLg/pRRE (Gag/Pol), pRSV-Rev (Rev) and phCMV-VSV-G (envelope) as well as the ProFection Mammalian Transfection System Calcium Phosphate Kit (Promega). Positive cells were selected using puromycin. Proteomics
Sample Preparation
After cell collection, protein sample preparation was carried out as described in Kulak et al. 35 with minor modifications. In brief, macrophage cells were lysed in urea buffer (8M urea, 10mM TCEP, 40mM CAA, and 100mM Tris pH8.5) and protein concentration estimation was carried out using BradfordRed reagent (Expedeon). 50 pg total protein was digested with endoproteinase lys C at a ratio of 1 :50 (enzyme to protein) for 3 hours at room temperature followed by dilution of the lysate to an urea concentration below 2M and digestion with
trypsin at a ratio of 1 :50 (enzyme to protein) for 13 hours at 37°C. Tryptic peptides were transferred to commercial centrifugal iST devices (PreOmics) and fractionated into 3 different fractions followed by clean-up/desalting and eluted as described in 35. Mass Spectrometry Acquisition
General nanoLC-MS setup was similar as previously described36 with modifications described in the following. QExactive mass spectrometer (Thermo Fisher Scientific, Germany) and Easy nanoLC-1000 were used for all experiments. Chromatographic separation of peptides was carried out on in-house packed fused-silica emitter nanoLC columns (75pm χ 20cm) (Silica PicoTip; New Objective, U.S.A.) packed with 1.9pm reverse- phase ReproSil-Pur C18-AQ beads (Dr. Maisch, Germany). Peptides were separated by a 3h linear gradient of 5-80% buffer B (80% acetonitrile, 0.1 % formic acid) at constant flowrate of 300 nl/min. For MS data acquisition the "fast" method from Kelstrup et al.37 was adopted. Mass spectrometry data analysis
MS raw files were analyzed by MaxQuant software and peak lists were searched against the mouse Uniprot FASTA database (concatenated with a database containing common contaminants) by the Andromeda search engine embedded in MaxQuant 38,39. MS1 -based label free quantification (LFQ) was done using maxLFQ algorithm 40. A minimum of two peptide ratios was requires in order to consider a given protein as valid (protein and peptide ID FDR=0.01 ). Perseus platform41 was used to perform data filtering and statistical testing. In step 1 , contaminant hits, reverse identification hits, and proteins "only identified by site" were removed from the dataset. In step 2, LFQ intensities were log2 transformed. This was followed by categorical annotation to create two samples groups based on their treatment. Step 3 involved removal of missing quantitative data points to minimize the number of missing values in the dataset and this was followed by missing value data imputation using a normal distribution simulating the distribution of low abundant proteins in the dataset. Lastly, Student's T-test was utilized to define differentially expressed proteins employing a two-fold change as a cut-off at a 5% FDR.
Flow cytometry and Confocal Microscopy
Flow cytometric staining was performed as previously described42. All fluorochrome- conjugated monoclonal antibodies were from (eBioscience), except for anti-CD301 (BioRad). Both NOS2 and RELMa protein levels were quantified after fixation and permeabilisation using the transcription buffer staining set (eBioscience) and monoclonal antibodies against NOS2 (Santa Cruz) and RELMa (Peprotech). Cells were stained with Live/Dead viability dye (Thermo) prior to antibody staining. Cells were labelled with CFSE as described43. P14 TCR
transgenic T cells were identified in vivo and ex vivo using the congenic marker CD45.1 and H-2DbGP33-4i MHC Class I tetramer. Cells were collected on LSR II and Fortessa flow cytometers (BD Biosciences) and analysed using FlowJo (TreeStar) software. Cells were sorted using a FACS Aria II. Cells were imaged using a Zeiss spinning disk confocal microscope with an Evolve (EMCCD) camera. Cells were kept in a humidified incubation chamber at 37°C with 5% C02 during image collection. Images were deconvolved and analysed using ImageJ (NIH).
RT-PCR and RNA Sequencing
RNA isolations were done by using the RNeasy kit (Qiagen) and single-strand cDNA was synthesized using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems). All RT-PCR was performed with Taqman primers using an Applied Biosystems 7000 sequence detection system. The expression levels of mRNA were normalized to the expression of a housekeeping gene (β-actin). For RNA-Seq, total RNA samples were extracted using RNeasy isolation kit (Qiagen). Libraries were prepared using the TruSeq stranded mRNA kit (lllumina) and sequenced in a HISeq 3000 (lllumina) by the staff at the Deep-sequencing Facility at the Max-Planck Institute for Immunobiology and Epigenetics. Sequenced libraries were processed with the Galaxy platform and deepTools44,45, using STAR46, for trimming and mapping, featureCounts (Liao et al) to quantify mapped reads, and DESeq247, to determine differentially expressed genes and generate normalised read counts to visualise as heatmaps using Morpheus (Broad Institute). elF5A-RIP-Chip (RNA Immunoprecipitation and Microarray Analysis)
Murine elF5A-1 open reading frame were PCR-amplified, fused with a C-terminal HA-tag and cloned into the pMSCV-puro vector (Clontech, pMSCV-elF-5A1-HA-puro). The following primers were used for cloning:
C-terminal-elF-5A1 -HA_fwd
(CTAGAGATCTGCCACCATGATCAAACGGAATGACTT) (SEQ ID NO: 8), and
C-terminal-elF-5A1-HA_rev
(CTAGGAATTCCTAAGCGTAATCTGGAACATCGTATGGGTATTTTGCCATGGCCTTGATT G) (SEQ ID NO: 9).
Ecotropic retroviruses for the transduction of NIH3T3 cells were obtained by transient calcium-phosphate-mediated transfection of the retroviral vectors into the packaging cell line Phoenix eco. NIH-3T3 cells were transduced by adding the filtered retrovirus-containing supernatant and selected with puromycin. For immunoprecipitation 3.5x106 transduced cells were seeded in triplicates on a 100-mm3 culture dish the day before. Cells were homogenized in pre-cooled lysis buffer containing 100 mM KCI, 5 mM MgCI2, 10 mM
HEPES, 0.5 % Nonidet-P40, 1 mM DTT, 100 U/mL Ribolock (Invitrogen) and 25 μΙ/mL Protease Inhibitor Cocktail (Sigma). Lysates were incubated for 15 minutes, cellular debris was pelleted at 20,000 g for 15 minutes and the supernatants were incubated with 50 pL magnetic anit-HA-MicroBeads (Miltenyi Biotec) for 30 minutes on ice in the dark. Afterwards the lysates were applied to μ Columns in the magnetic field of a pMACS Separator. After rinsing the columns three times with lysis buffer with and without 1 M urea, the RNA-protein- anti-HA-MicroBead complexes were eluted with lysis buffer containing 0.1 % SDS and 0.3 pg/pL proteinase K and incubated for 35 minutes at 50 C. Then the anti-HA-MicroBeads were removed by applying the solution to the μ Columns and eluting the RNA containing fraction with DEPC-treated water. The RNA was cleaned and concentrated with the NucleoSpin RNA XS Kit (Macherey-Nagel). Furthermore, RNA from the same cells (triplicates) were isolated as back ground control. RNA quantity and quality were evaluated by Nanodrop ND1000 Spectrophotometer and Agilent 2100 Bioanalyzer measurement. Procedures for cDNA synthesis, labelling and hybridisation were carried out according to the manufacturer's protocol (Affymetrix 3'-IVT Express Kit) starting with 100 ng of each RNA sample (three samples from immunoprecipitation and three samples of total RNA without immunoprecipitation). The experiments were performed using Affymetrix Mouse Genome 430 2.0 GeneChip. All reactions were performed in triplicates. The signals were processed with a target value of 300 using Affymetrix GeneChip Operating Software 1.4. After immunoprecipitation, significant enriched genes were reported as log2 ratio > 1 (p-value < 0.01 ) compared with the gene expression in total RNA control samples. Statistical analysis for identification of enriched genes were performed by the Functional Genomics Center Zurich using the LIMMA package48 . KEGG pathway annotation were performed using the R package 'clusterProfiler' 49.
Adoptive Transfers
For in vivo T cell experiments, P14 TCR transgenic CD45.1 T cells were activated in vitro with gp33-41 peptide and transduced with E/"f5a-shRNA. A day later 5x105 T cells were transferred into CD45.2 C57BL/6 congenic recipient mice on day one of infection. Blood samples were collected at the indicated time points and analysed by flow cytometry.
Quantification and Statistical Analysis
Statistical analysis was performed using prism 6 software (Graph pad) and results are represented as mean ± SEM, unless otherwise indicated. Comparisons for two groups were calculated using unpaired two-tailed Student's t tests, comparisons of more than two groups were calculated using one-way ANOVA with Bonferroni's multiple comparison tests. Normal
distribution and no difference in variance between groups in individual comparisons were observed.
References
1 . Park, M. H., Cooper, H. L. & Folk, J. E. Identification of Hypusine, an Unusual Amino- Acid, in a Protein From Human-Lymphocytes and of Spermidine as Its Biosynthetic Precursor. Proceedings of the National Academy of Sciences 78, 2869-2873 (1981 ).
2. Wolff, E. C, Lee, Y. B„ Chung, S. I., Folk, J. E. & Park, M. H. Deoxyhypusine
Synthase From Rat Testis - Purification and Characterization. Journal of Biological Chemistry 270, 8660-8666 (1995).
3. Abbruzzese, A., Park, M. H. & Folk, J. E. Deoxyhypusine Hydroxylase From Rat Testis - Partial-Purification and Characterization. Journal of Biological Chemistry 261 , 3085-
3089 (1986).
4. Pelechano, V. & Alepuz, P. elF5A facilitates translation termination globally and
promotes the elongation of many non polyproline-specific tripeptide sequences.
Nucleic Acids Res. 45, 7326-7338 (2017).
5. Schuller, A. P., Wu, C. C.C., Dever, T. E., Buskirk, A. R. & Green, R. elF5A Functions Globally in Translation Elongation and Termination. Molecular Cell 66, 194-+ (2017).
6. Huang, S. C.C. et al. Cell-intrinsic lysosomal lipolysis is essential for alternative
activation of macrophages. Nat Immunol 15, 846-855 (2014).
7. Miller-Fleming, L., Olin-Sandoval, V., Campbell, K. & Raiser, M. Remaining Mysteries of Molecular Biology: The Role of Polyamines in the Cell. J. Mol. Biol. 427, 3389-3406
(2015).
8. Poulin, R., Lu, L., Ackermann, B., Bey, P. & Pegg, A. E. Mechanism of the Irreversible Inactivation of Mouse Ornithine Decarboxylase by Alpha-Difluoromethylornithine - Characterization of Sequences at the Inhibitor and Coenzyme Binding-Sites. Journal of Biological Chemistry 267, 150-158 ( 1992).
9. Gabrielson, E. et al. Induction of spermidine/spermine 1-acetyltransferase in breast cancer tissues treated with the polyamine analogue N1 , N1 1-diethylnorspermine. Cancer Chemother. Pharmacol. 54, 122-126 (2004).
10. Melis, N. et al. Targeting elF5A Hypusination Prevents Anoxic Cell Death through Mitochondrial Silencing and Improves Kidney Transplant Outcome. J. Am. Soc.
Nephrol. ASN.2016010012 (2016). doi:10.1681/ASN.2016010012
1 1. Hoque, M. et al. Inhibition of HIV-1 gene expression by Ciclopirox and Deferiprone, drugs that prevent hypusination of eukaryotic initiation factor 5A. Retrovirology 6, (2009).
12. Vats, D. et al. Oxidative metabolism and PGC-1 beta attenuate macrophage-mediated inflammation. Cell Metab. 4, 13-24 (2006).
13. Liu, L. et al. Proinflammatory signal suppresses proliferation and shifts macrophage metabolism from Myc-dependent to HIF1a-dependent. Proc. Natl. Acad. Sci. U.S.A.
113, 1564-1569 (2016).
14. Arpa, L., Valledor, A. F., Lloberas, J. & Celada, A. IL-4 blocks M-CSF-dependent macrophage proliferation by inducing p21Waf1 in a STAT6-dependent way. Eur. J.
Immunol. 39, 514-526 (2009).
15. Thomas, T. & Thomas, T. J. Polyamines in cell growth and cell death: molecular
mechanisms and therapeutic applications. Cell. Mol. Life Sci. 58, 244-258 (2001 ). 16. Jiang, L. et al. Reductive carboxylation supports redox homeostasis during anchorage- independent growth. Nature 532, 255-+ (2016).
17. Sena, L. A. et al. Mitochondria Are Required for Antigen-Specific T Cell Activation through Reactive Oxygen Species Signaling. Immunity 38, 225-236 (2013).
18. Chang, C.H. et al, Posttranscriptional control of T cell effector function by aerobic glycolysis. Ce// 153, 1239-1251 (2013).
19. van der Windt, G. J. W. ef al. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36, 68-78 (2012).
20. Pearce, E. L. ef al. Enhancing CD8 T-cell memory by modulating fatty acid
metabolism. Nature 460, 103-107 (2009).
21 . Buck, M. D. et al. Mitochondrial Dynamics Controls T Cell Fate through Metabolic Programming. Cell 166, 63-76 (2016).
22. Gutierrez, E. et al. elF5A Promotes Translation of Polyproline Motifs. Molecular Cell
51 , 35-45 (2013).
23. Park, M. H., Nishimura, K., Zanelli, C. F. & Valentini, S. R. Functional significance of elF5A and its hypusine modification in eukaryotes. Amino Acids 38, 491-500 (2010). 24. Mathews, M. B. & Hershey, J. W. B. The translation factor elF5A and human cancer.
Biochim. Biophys. Acta 1849, 836-844 (2015).
25. Casero, R. A. & Marton, L. J. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov 6, 373-390 (2007).
26. Jha, A. K. et al. Network integration of parallel metabolic and transcriptional data
reveals metabolic modules that regulate macrophage polarization. Immunity 42, 419-
430 (2015).
27. Mills, E. L. et al. Succinate Dehydrogenase Supports Metabolic Repurposing of
Mitochondria to Drive Inflammatory Macrophages. Cell 167, 457-470.e13 (2016).
28. Wang, R. ef al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35, 871-882 (201 1 ).
29. Hukelmann, J. L. et al. The cytotoxic T cell proteome and its shaping by the kinase mTOR. Nat Immunol 17, 104-112 (2016).
30. Bevec, D. et al. Induced gene expression of the hypusine-containing protein
eukaryotic initiation factor 5A in activated human T lymphocytes. Proceedings of the National Academy of Sciences 91 , 10829-10833 (1994).
31. Pallmann, N. et al. Biological Relevance and Therapeutic Potential of the Hypusine Modification System. J. Biol. Chem. 290, 18343-18360 (2015).
32. Buescher, J. M. et al. A roadmap for interpreting (13)C metabolite labeling patterns from cells. Curr. Opin. Biotechnol. 34, 189-201 (2015).
33. Kelly, S. M., Vanslyke, J. K. & Musil, L. S. Regulation of ubiquitin-proteasome system mediated degradation by cytosolic stress. Mol. Biol. Cell 18, 4279-4291 (2007).
34. Preukschas, M. ef al. Expression of eukaryotic initiation factor 5A and hypusine
forming enzymes in glioblastoma patient samples: implications for new targeted therapies. PLoS ONE 7, e43468 (2012).
35. Kulak, N. A., Pichler, G., Paron, I., Nagaraj, N. & Mann, M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells.
Nat. Methods 11 , 319-324 (2014).
36. Engeike, R. ef al. The quantitative nuclear matrix proteome as a biochemical snapshot of nuclear organization. J. Proteome Res. 13, 3940-3956 (2014).
37. Kelstrup, C. D., Young, C, Lavallee, R., Nielsen, M. L. & Olsen, J. V. Optimized fast and sensitive acquisition methods for shotgun proteomics on a quadrupole orbitrap mass spectrometer. J. Proteome Res. 11 , 3487-3497 (2012).
38. Cox, J. et al. Andromeda: a peptide search engine integrated into the MaxQuant
environment. J. Proteome Res. 10, 1794-1805 (201 1 ).
39. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p. p. b. -range mass accuracies and proteome-wide protein quantification. Nat.
Biotechnol. 26, 1367-1372 (2008).
40. Cox, J. et al. Accurate proteome-wide label-free quantification by delayed
normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol. Cell Proteomics 13, 2513-2526 (2014).
41. Tyanova, S. ef al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731-740 (2016).
42. Chang, C.-H. et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Ce// 162, 1229-1241 (2015).
43. Kaech, S. M. & Ahmed, R. Memory CD8+ T cell differentiation: initial antigen
encounter triggers a developmental program in naive cells. Nat Immunol 2, 415-422 (2001 ).
44. Afgan, E. ef al. The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2016 update. Nucleic Acids Res. 44, W3-W10 (2016).
45. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160-5 (2016).
46. Dobin, A. ef al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15-21 (2013).
47. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and
dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
48. Smyth, G. K., Michaud, J. & Scott, H. S. Use of within-array replicate spots for
assessing differential expression in microarray experiments. Bioinformatics 21 , 2067- 2075 (2005).
49. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284-287 (2012).
Claims
A method for determining whether a compound has the capability of inhibiting elF5a activation, comprising
(a) contacting a sample comprising cells being capable of differentiating into M2 macrophages with the compound under conditions that allow for the differentiation of cells into M2-type macrophages in the absence of the compound; and
(b) quantifying in the sample the cells having a M2-phenotype,
wherein a reduction of the number of cells having a M2-phenotype as compared to a control sample not contacted with the compound indicates that the compound has the capability of inhibiting elF5a activation.
The method of claim 1 , wherein quantifying the cells having a M2-phenotype comprises measuring the amounts of at least one marker molecule selected from the group consisting of GAT A3, IRF4, SOCS1 , CCL4, CCL13, CCL17, CCL18, MRC1 , STAB1 , F13A1 , TGFB1 , MMP12, TGM2, ALOX15, CD200R, SOCS3, IL-4Ra, CD163, STAB1 , MARCO, TGFBR2, ADORA3, ID3, RGS1 , pSMAD2, TGFBR2, ALOX5AP, CD206 and IL17RB.
3. The method of claim 1 or 2, wherein quantifying the cells having a M2-phenotype comprises flow cytrometry analysis, quantitative PCR, and/or western blot analysis.
4. The method of any one of claims 1 to 3, wherein the compound is an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, protein drug or small molecule.
5. The method of any one of claims 1 to 4, wherein the compound is directed against ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH) and is preferably directed against DHPS or DOHH.
6. The method of any one of claims 1 to 5, wherein the efficacy of a compound for inhibiting elF5a activation in a patient is determined,
wherein the sample of step (a) and the control sample are samples that have been obtained from the patient, and
wherein a reduction of the number of cells having a M2-phenotype in the sample as compared to thecontrol sample not contacted with the compound indicates that the
compound is effective for inhibiting elF5a activation in the patient.
7. The method of claim 6, wherein the patient is afflicted with a hyperproliferative disorder, a fibrotic disorder or the macrophage activation syndrome.
8. The method of claim 6 or 7, wherein the sample is a tissue sample or a blood sample.
9. The method of any one of claims 6 to 8, wherein the compound is guanyl-1 ,7- diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I or CNI-1493.
10. A compound inhibiting elF5a activation for use in the treatment or prevention of a disease being mediated by M2 macrophages, wherein the disease is preferably a hyperproliferative disorder, a fibrotic disorder or the macrophage activation syndrome.
11. A compound inhibiting elF5a activation for use in the treatment or prevention of a hyperproliferative disorder, a fibrotic disorder or the macrophage activation syndrome by inhibiting cellular differentiation into M2 macrophages.
12. The compound for use of claim 10 or 11 , wherein the compound targets ornithine decarboxylase, spermidine synthase, spermine synthase, deoxyhypusine synthase (DHPS) or deoxyhypusine hydroxylase (DOHH) and preferably DHPS or DOHH.
13. The compound for use of any one of claims 10 to 12, wherein the compound is an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, protein drug or small molecule.
14. The compound for use of claim 13, wherein the compound is guanyl-1 ,7- diaminoheptane (GC7), L-mimosine, ciclopirox, deferiprone, hydralazine, agent I or CNI-1493.
15. The compound for use of any one of claims 10 to 14 or the method of any one of claims 7 to 9, wherein
(i) the hyperproliferative disorder is a neoplasm, tumor or cancer and is preferably selected from cancer of the breast, lung, prostate, kidney, skin, neural, ovary, uterus, liver, pancreas, epithelial, gastric, intestinal, exocrine, endocrine, lymphatic, hematopoietic system or a head and neck tissue; and/or
(ii) the fibrotic disorder is selected from sarcoidosis, renal fibrosis, pulmonary fibrosis, idiopathic pulmonary fibrosis, cystic fibrosis, liver fibrosis, cardiac fibrosis, endomyocardial fibrosis, atrial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, chronic kidney disease, nephrogenic systemic fibrosis, Chron's disease, hypertrophic scarring, keloid, scleroderma, organ transplant-associated fibrosis and ischemia-associated fibrosis.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201762459833P | 2017-02-16 | 2017-02-16 | |
US62/459,833 | 2017-02-16 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018149978A1 true WO2018149978A1 (en) | 2018-08-23 |
Family
ID=61249637
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2018/053913 WO2018149978A1 (en) | 2017-02-16 | 2018-02-16 | ALTERNATIVE ACTIVATION OF MACROPHAGES (M2 CELLS) THROUGH THE POLYAMINE-eIF5a-HYPUSINE AXIS |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2018149978A1 (en) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2020227385A1 (en) * | 2019-05-07 | 2020-11-12 | The Regents Of The University Of California | Compounds and methods for treating or preventing fibrosis |
CN113133995A (en) * | 2021-05-09 | 2021-07-20 | 河南牧业经济学院 | Application of inhibitor CNI-1493 in porcine reproductive and respiratory syndrome |
RU2810558C1 (en) * | 2023-05-02 | 2023-12-27 | федеральное государственное автономное образовательное учреждение высшего образования "Казанский (Приволжский) федеральный университет" (ФГАОУ ВО КФУ) | Method of selecting medicinal products for pharmacological induction of mitochondrial dysfunction in macrophages for antitumor therapy |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
US6080560A (en) | 1994-07-25 | 2000-06-27 | Monsanto Company | Method for producing antibodies in plant cells |
US20110177089A1 (en) * | 2008-05-27 | 2011-07-21 | The University Of Tokyo | Apoptosis inducer |
US20150224132A1 (en) * | 2014-02-07 | 2015-08-13 | Effector Therapeutics, Inc. | Compositions and methods for treating fibrotic disease |
US20160054304A1 (en) * | 2013-03-14 | 2016-02-25 | Galapagos Nv | Molecular targets and compounds, and methods to identify the same, useful in the treatment of fibrotic diseases |
-
2018
- 2018-02-16 WO PCT/EP2018/053913 patent/WO2018149978A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
US6080560A (en) | 1994-07-25 | 2000-06-27 | Monsanto Company | Method for producing antibodies in plant cells |
US20110177089A1 (en) * | 2008-05-27 | 2011-07-21 | The University Of Tokyo | Apoptosis inducer |
US20160054304A1 (en) * | 2013-03-14 | 2016-02-25 | Galapagos Nv | Molecular targets and compounds, and methods to identify the same, useful in the treatment of fibrotic diseases |
US20150224132A1 (en) * | 2014-02-07 | 2015-08-13 | Effector Therapeutics, Inc. | Compositions and methods for treating fibrotic disease |
Non-Patent Citations (100)
Title |
---|
"Handbook of Pharmaceutical Excipients", 2010, PHARMACEUTICAL PRESS |
ABBRUZZESE, A.; PARK, M. H.; FOLK, J. E.: "Deoxyhypusine Hydroxylase From Rat Testis - Partial-Purification and Characterization", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 261, 1986, pages 3085 - 3089 |
AFGAN, E. ET AL.: "The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2016 update", NUCLEIC ACIDS RES., vol. 44, 2016, pages W3 - W10 |
ALLAY ET AL., HUM GENE THER., vol. 22, no. 5, May 2011 (2011-05-01), pages 595 - 604 |
ALTSHULER ET AL., BIOCHEMISTRY (MOSC, vol. 75, no. 13, December 2010 (2010-12-01), pages 1584 - 605 |
ARPA, L.; VALLEDOR, A. F.; LLOBERAS, J.; CELADA, A.: "IL-4 blocks M-CSF-dependent macrophage proliferation by inducing p21Waf1 in a STAT6-dependent way", EUR. J. IMMUNOL., vol. 39, 2009, pages 514 - 526 |
AUSUBEL ET AL., HUM GENE THER., vol. 22, no. 4, April 2011 (2011-04-01), pages 489 - 97 |
BALABANOV STEFAN ET AL: "Hypusination of eukaryotic imtiation factor 5A (eIF5A): a novel therapeutic target in BCR-ABL-positive leukemias identified by a proteomics approach", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 109, no. 4, 15 February 2007 (2007-02-15), pages 1701 - 1711, XP009152218, ISSN: 0006-4971, [retrieved on 20061010], DOI: 10.1182/BLOOD-2005-03-037648 * |
BEBBINGTON ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 169 - 175 |
BEVEC, D. ET AL.: "Induced gene expression of the hypusine-containing protein eukaryotic initiation factor 5A in activated human T lymphocytes", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 91, 1994, pages 10829 - 10833 |
BRAGA, FRONT IMMUNOL, vol. 6, 2015, pages 602 |
BUCK, M. D. ET AL.: "Mitochondrial Dynamics Controls T Cell Fate through Metabolic Programming", CELL, vol. 166, 2016, pages 63 - 76, XP029627880, DOI: doi:10.1016/j.cell.2016.05.035 |
BUESCHER, J. M. ET AL.: "A roadmap for interpreting (13)C metabolite labeling patterns from cells", CURR. OPIN. BIOTECHNOL., vol. 34, 2015, pages 189 - 201 |
CASERO, R. A.; MARTON, L. J.: "Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases", NAT REV DRUG DISCOV, vol. 6, 2007, pages 373 - 390, XP007910866, DOI: doi:10.1038/nrd2243 |
CHANG, C.-H. ET AL.: "Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression", CELL, vol. 162, 2015, pages 1229 - 1241 |
CHANG, C.H. ET AL.: "Posttranscriptional control of T cell effector function by aerobic glycolysis", CELL, vol. 153, 2013, pages 1239 - 1251 |
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 150333-69-0 |
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 164301-51-3 |
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 29342-05-0 |
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 30652-11-0 |
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 500-44-7 |
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 86-54-4 |
COLE ET AL.: "Monoclonal Antibodies and Cancer Therapy", 1985, ALAN R. LISS, INC., pages: 77 - 96 |
COX, J. ET AL.: "Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ", MOL. CELL PROTEOMICS, vol. 13, 2014, pages 2513 - 2526 |
COX, J. ET AL.: "Andromeda: a peptide search engine integrated into the MaxQuant environment", J. PROTEOME RES., vol. 10, 2011, pages 1794 - 1805 |
COX, J.; MANN, M.: "MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification", NAT. BIOTECHNOL., vol. 26, 2008, pages 1367 - 1372 |
DE FOUGEROLLES ET AL., CURRENT OPINION IN PHARMACOLOGY, vol. 8, 2008, pages 280 - 285 |
DOBIN, A. ET AL.: "STAR: ultrafast universal RNA-seq aligner", BIOINFORMATICS, vol. 29, 2013, pages 15 - 21 |
ELBASHIR ET AL., NATURE, vol. 411, no. 6836, 24 May 2001 (2001-05-24), pages 494 - 8 |
ELISABETH MÉMIN ET AL: "Blocking eIF5A Modification in Cervical Cancer Cells Alters the Expression of Cancer-Related Genes and Suppresses Cell Proliferation", CANCER RESEARCH, vol. 74, no. 2, 12 November 2013 (2013-11-12), US, pages 552 - 562, XP055478361, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-13-0474 * |
ENGELKE, R. ET AL.: "The quantitative nuclear matrix proteome as a biochemical snapshot of nuclear organization", J. PROTEOME RES., vol. 13, 2014, pages 3940 - 3956 |
EPELMAN ET AL., IMMUNITY, vol. 41, no. 1, 2014, pages 21 - 35 |
GABRIELSON, E. ET AL.: "Induction of spermidine/spermine N1-acetyltransferase in breast cancer tissues treated with the polyamine analogue N1, N11-diethylnorspermine", CANCER CHEMOTHER. PHARMACOL., vol. 54, 2004, pages 122 - 126 |
GHAHROUDI ET AL., FEBS LETTERS, vol. 414, 1997, pages 521 - 526 |
GUTIERREZ E ET AL.: "eIF5A promotes translation of polyproline motifs", MOLECULAR CELL, vol. 51, 2013, pages 35 - 45, XP028675710, DOI: doi:10.1016/j.molcel.2013.04.021 |
GUTIERREZ, E. ET AL.: "elF5A Promotes Translation of Polyproline Motifs", MOLECULAR CELL, vol. 51, 2013, pages 35 - 45, XP028675710, DOI: doi:10.1016/j.molcel.2013.04.021 |
HANAUSKE-ABEL ET AL., BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1221, 1994, pages 115 - 124 |
HARLOW; LANE: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY PRESS |
HARLOW; LANE: "Using Antibodies: A Laboratory Manual", 1999, COLD SPRING HARBOR LABORATORY PRESS |
HOLLIGER; HUDSON, NAT BIOTECHNOL., vol. 23, no. 9, 2005, pages 1126 - 36 |
HOQUE, M. ET AL.: "Inhibition of HIV-1 gene expression by Ciclopirox and Deferiprone, drugs that prevent hypusination of eukaryotic initiation factor 5A", RETROVIROLOGY, vol. 6, 2009, XP021063732, DOI: doi:10.1186/1742-4690-6-90 |
HUANG, S. C.C. ET AL.: "Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages", NAT IMMUNOL, vol. 15, 2014, pages 846 - 855 |
HUKELMANN, J. L. ET AL.: "The cytotoxic T cell proteome and its shaping by the kinase mTOR", NAT IMMUNOL, vol. 17, 2016, pages 104 - 112 |
JASIULIONIS MIRIAM G ET AL: "Inhibition of eukaryotic translation initiation factor 5A (eIF5A) hypusination impairs melanoma growth", CELL BIOCHEMISTRY AND FUNCTION, BUTTERWORTH, GUILDFORD, GB, vol. 25, no. 1, 1 January 2007 (2007-01-01), pages 109 - 114, XP002454030, ISSN: 0263-6484, DOI: 10.1002/CBF.1351 * |
JHA, A. K. ET AL.: "Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization", IMMUNITY, vol. 42, 2015, pages 419 - 430 |
JIANG, L. ET AL.: "Reductive carboxylation supports redox homeostasis during anchorage-independent growth", NATURE, vol. 532, 2016, pages 255 |
KAECH, S. M.; AHMED, R.: "Memory CD8+ T cell differentiation: initial antigen encounter triggers a developmental program in naive cells", NAT IMMUNOL, vol. 2, 2001, pages 415 - 422 |
KELLY, S. M.; VANSLYKE, J. K.; MUSIL, L. S.: "Regulation of ubiquitin-proteasome system mediated degradation by cytosolic stress", MOL. BIOL. CELL, vol. 18, 2007, pages 4279 - 4291 |
KELSTRUP, C. D.; YOUNG, C.; LAVALLEE, R.; NIELSEN, M. L.; OLSEN, J. V.: "Optimized fast and sensitive acquisition methods for shotgun proteomics on a quadrupole orbitrap mass spectrometer", J. PROTEOME RES., vol. 11, 2012, pages 3487 - 3497 |
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495 - 497 |
KOZBOR, IMMUNOLOGY TODAY, vol. 4, 1983, pages 72 |
KULAK, N. A.; PICHLER, G.; PARON, I.; NAGARAJ, N.; MANN, M.: "Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells", NAT. METHODS, vol. 11, 2014, pages 319 - 324 |
LEVIN, GENE THER., vol. 19, no. 11, 2012, pages 1041 - 7 |
LIU, L. ET AL.: "Proinflammatory signal suppresses proliferation and shifts macrophage metabolism from Myc-dependent to HIF1a-dependent", PROC. NATL. ACAD. SCI. U.S.A., vol. 113, 2016, pages 1564 - 1569 |
LOVE, M. I.; HUBER, W.; ANDERS, S.: "Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2", GENOME BIOL., vol. 15, 2014, pages 550, XP021210395, DOI: doi:10.1186/s13059-014-0550-8 |
MATHEWS, M. B.; HERSHEY, J. W. B.: "The translation factor elF5A and human cancer", BIOCHIM. BIOPHYS. ACTA, vol. 1849, 2015, pages 836 - 844, XP055342151, DOI: doi:10.1016/j.bbagrm.2015.05.002 |
MELIS, N. ET AL.: "Targeting elF5A Hypusination Prevents Anoxic Cell Death through Mitochondrial Silencing and Improves Kidney Transplant Outcome", J. AM. SOC. NEPHROL. |
MEMIN ET AL., CANCER RES., vol. 74, no. 2, 15 January 2014 (2014-01-15), pages 552 - 62 |
MILLER-FLEMING, L.; OLIN-SANDOVAL, V.; CAMPBELL, K.; RAISER, M.: "Remaining Mysteries of Molecular Biology: The Role of Polyamines in the Cell", J. MOL. BIOL., vol. 427, 2015, pages 3389 - 3406 |
MILLS, CRIT REV IMMUNOL., vol. 32, no. 6, 2012, pages 463 - 88 |
MILLS, E. L. ET AL.: "Succinate Dehydrogenase Supports Metabolic Repurposing of Mitochondria to Drive Inflammatory Macrophages", CELL, vol. 167, 2016, pages 457 - 470 |
MILSTEIN, C, BIOESSAYS, vol. 21, no. 11, 1999, pages 966 - 73 |
MURPHY, BIOCHEM J., vol. 227, 1991, pages 277 - 279 |
NAKANISHI SHIMA ET AL: "Targeting the polyamine-hypusine circuit for the prevention and treatment of cancer", AMINO ACIDS, SPRINGER VERLAG, AU, vol. 48, no. 10, 29 June 2016 (2016-06-29), pages 2353 - 2362, XP036064386, ISSN: 0939-4451, [retrieved on 20160629], DOI: 10.1007/S00726-016-2275-3 * |
NAKANISHI, S.; CLEVELAND, J.L.: "Targeting the polyamine-hypusine circuit for the prevention and treatment of cancer", AMINO ACIDS, 2016, pages 1 - 10 |
NICOLAS MELIS ET AL: "Targeting eIF5A Hypusination Prevents Anoxic Cell Death through Mitochondrial Silencing and Improves Kidney Transplant Outcome", JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY., vol. 28, no. 3, 9 September 2016 (2016-09-09), US, pages 811 - 822, XP055478682, ISSN: 1046-6673, DOI: 10.1681/ASN.2016010012 * |
NIKKI P. LEE ET AL: "Prognostic significance and therapeutic potential of eukaryotic translation initiation factor 5A (eIF5A) in hepatocellular carcinoma", INTERNATIONAL JOURNAL OF CANCER, vol. 127, 1 January 2010 (2010-01-01), pages 968 - 976, XP055165907, ISSN: 0020-7136, DOI: 10.1002/ijc.25100 * |
OEDEM JR ET AL: "Hypusine Modification of the Ribosome-binding Protein eIF5A, a Target for New Anti-Inflammatory Drugs: Understanding the Action of the Inhibitor GC7 on a Murine Macrophage Cell Line", CURRENT PHARMACEUTICAL DESIGN, vol. 20, no. 2, 31 January 2014 (2014-01-31), NL, pages 284 - 292, XP055464214, ISSN: 1381-6128, DOI: 10.2174/13816128113199990036 * |
OWENS, PROC. NATL. ACAD. SCI. USA, vol. 98, 2001, pages 1471 - 1476 |
PALLMANN, N. ET AL.: "Biological Relevance and Therapeutic Potential of the Hypusine Modification System", J. BIOL. CHEM., vol. 290, 2015, pages 18343 - 18360 |
PARK, M. H.; COOPER, H. L.; FOLK, J. E.: "Identification of Hypusine, an Unusual AminoAcid, in a Protein From Human-Lymphocytes and of Spermidine as Its Biosynthetic Precursor", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 78, 1981, pages 2869 - 2873 |
PARK, M. H.; NISHIMURA, K.; ZANELLI, C. F.; VALENTINI, S. R.: "Functional significance of elF5A and its hypusine modification in eukaryotes", AMINO ACIDS, vol. 38, 2010, pages 491 - 500, XP019783380 |
PATRICK ZIEGLER ET AL: "Evaluation of deoxyhypusine synthase inhibitors targeting BCR-ABL positive leukemias", INVESTIGATIONAL NEW DRUGS ; THE JOURNAL OF NEW ANTICANCER AGENTS, KLUWER ACADEMIC PUBLISHERS, BO, vol. 30, no. 6, 14 March 2012 (2012-03-14), pages 2274 - 2283, XP035131664, ISSN: 1573-0646, DOI: 10.1007/S10637-012-9810-1 * |
PAZ ET AL., BIOCHEM PHARMACOL, vol. 33, 1984, pages 779 - 785 |
PEARCE, E. L. ET AL.: "Enhancing CD8 T-cell memory by modulating fatty acid metabolism", NATURE, vol. 460, 2009, pages 103 - 107 |
PELECHANO, V.; ALEPUZ, P.: "elF5A facilitates translation termination globally and promotes the elongation of many non polyproline-specific tripeptide sequences", NUCLEIC ACIDS RES., vol. 45, 2017, pages 7326 - 7338 |
POULIN, R.; LU, L.; ACKERMANN, B.; BEY, P.; PEGG, A. E.: "Mechanism of the Irreversible Inactivation of Mouse Ornithine Decarboxylase by Alpha-Difluoromethylornithine - Characterization of Sequences at the Inhibitor and Coenzyme Binding-Sites", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 267, 1992, pages 150 - 158 |
PREUKSCHAS, M. ET AL.: "Expression of eukaryotic initiation factor 5A and hypusine forming enzymes in glioblastoma patient samples: implications for new targeted therapies", PLOS ONE, vol. 7, 2012, pages e43468, XP055165911, DOI: doi:10.1371/journal.pone.0043468 |
RAMIREZ, F. ET AL.: "deepTools2: a next generation web server for deep-sequencing data analysis", NUCLEIC ACIDS RES., vol. 44, 2016, pages 160 - 5 |
RE, OCHSNER J., vol. 2, no. 4, October 2000 (2000-10-01), pages 233 - 236 |
SAMBROOK; RUSSEL: "Molecular Cloning: A Laboratory Manual", vol. 3, 2001 |
SCHULLER, A. P.; WU, C. C.C.; DEVER, T. E.; BUSKIRK, A. R.; GREEN, R.: "elF5A Functions Globally in Translation Elongation and Termination", MOLECULAR CELL, vol. 66, 2017, pages 194 |
SENA, L. A. ET AL.: "Mitochondria Are Required for Antigen-Specific T Cell Activation through Reactive Oxygen Species Signaling", IMMUNITY, vol. 38, 2013, pages 225 - 236 |
SMYTH, G. K.; MICHAUD, J.; SCOTT, H. S.: "Use of within-array replicate spots for assessing differential expression in microarray experiments", BIOINFORMATICS, vol. 21, 2005, pages 2067 - 2075 |
SPECHT ET AL., PARASITOL RES, vol. 102, no. 6, 2008, pages 1177 - 84 |
TAMHANE; LOGAN, JOURNAL OF THE AMERICAN STATISTICAL ASSOCIATION, vol. 97, no. 457, 2002, pages 1 - 9 |
THOMAS, T.; THOMAS, T. J.: "Polyamines in cell growth and cell death: molecular mechanisms and therapeutic applications", CELL. MOL. LIFE SCI., vol. 58, 2001, pages 244 - 258 |
TOMLINSON ET AL., NATURE BIOTECHNOLOGY, vol. 22, no. 5, 2004, pages 521 - 522 |
TYANOVA, S. ET AL.: "The Perseus computational platform for comprehensive analysis of (prote)omics data", NAT. METHODS, vol. 13, 2016, pages 731 - 740 |
TYLOER; POSH, BIOMED RESEARCH INTERNATIONAL, vol. 2014, 2014 |
V. BOOM ET AL: "Evidence-based diagnosis and treatment of macrophage activation syndrome in systemic juvenile idiopathic arthritis", PEDIATRIC RHEUMATOLOGY, vol. 13, no. 1, 1 December 2015 (2015-12-01), XP055478725, DOI: 10.1186/s12969-015-0055-3 * |
VAN DER WINDT, G. J. W. ET AL.: "Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development", IMMUNITY, vol. 36, 2012, pages 68 - 78 |
VATS, D. ET AL.: "Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation", CELL METAB., vol. 4, 2006, pages 13 - 24 |
WANG, R. ET AL.: "The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation", IMMUNITY, vol. 35, 2011, pages 871 - 882, XP028348596, DOI: doi:10.1016/j.immuni.2011.09.021 |
WERMUTH; JIMENEZ, CLIN TRANSL MED, vol. 4, 2015, pages 2 |
WOLFF, E. C.; LEE, Y. B.; CHUNG, S. I.; FOLK, J. E.; PARK, M. H.: "Deoxyhypusine Synthase From Rat Testis - Purification and Characterization", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 270, 1995, pages 8660 - 8666, XP002978937, DOI: doi:10.1074/jbc.270.15.8660 |
WOOK-BIN LEE ET AL: "Mincle-mediated translational regulation is required for strong nitric oxide production and inflammation resolution", NATURE COMMUNICATIONS, vol. 7, 18 April 2016 (2016-04-18), GB, pages 11322, XP055464223, ISSN: 2041-1723, DOI: 10.1038/ncomms11322 * |
YU, G.; WANG, L.-G.; HAN, Y.; HE, Q.-Y.: "clusterProfiler: an R package for comparing biological themes among gene clusters", OMICS, vol. 16, 2012, pages 284 - 287 |
ZAHER; UNRAU, RNA, vol. 13, no. 7, 2007, pages 1017 - 1026 |
ZIMMERMANN ET AL., NATURE, vol. 441, 2006, pages 111 - 114 |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2020227385A1 (en) * | 2019-05-07 | 2020-11-12 | The Regents Of The University Of California | Compounds and methods for treating or preventing fibrosis |
CN113133995A (en) * | 2021-05-09 | 2021-07-20 | 河南牧业经济学院 | Application of inhibitor CNI-1493 in porcine reproductive and respiratory syndrome |
RU2810558C1 (en) * | 2023-05-02 | 2023-12-27 | федеральное государственное автономное образовательное учреждение высшего образования "Казанский (Приволжский) федеральный университет" (ФГАОУ ВО КФУ) | Method of selecting medicinal products for pharmacological induction of mitochondrial dysfunction in macrophages for antitumor therapy |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Dreishpoon et al. | FDX1 regulates cellular protein lipoylation through direct binding to LIAS | |
Puleston et al. | Polyamines and eIF5A hypusination modulate mitochondrial respiration and macrophage activation | |
Lou et al. | Excessive polyamine generation in keratinocytes promotes self-RNA sensing by dendritic cells in psoriasis | |
Singh et al. | DDX41 is required for cGAS-STING activation against DNA virus infection | |
Nishikawa et al. | BRCA1-associated protein 1 interferes with BRCA1/BARD1 RING heterodimer activity | |
CA3008289C (en) | Inhibition of cytokine-induced sh2 protein in nk cells | |
Ducker et al. | Two N-myristoyltransferase isozymes play unique roles in protein myristoylation, proliferation, and apoptosis | |
Stonestrom et al. | Functions of BET proteins in erythroid gene expression | |
Li et al. | The histone mark H3K36me3 regulates human DNA mismatch repair through its interaction with MutSα | |
Brechmann et al. | A PP4 holoenzyme balances physiological and oncogenic nuclear factor-kappa B signaling in T lymphocytes | |
Tsai et al. | SMYD3-mediated H2A. Z. 1 methylation promotes cell cycle and cancer proliferation | |
Lachapelle et al. | Proteome-wide identification of WRN-interacting proteins in untreated and nuclease-treated samples | |
Liu et al. | Distinct pathways for NF-κB regulation are associated with aberrant macrophage IL-12 production in lupus-and diabetes-prone mouse strains | |
Chen et al. | Elp3‐mediated codon‐dependent translation promotes mTORC2 activation and regulates macrophage polarization | |
Chang et al. | OTUB2 exerts tumor-suppressive roles via STAT1-mediated CALML3 activation and increased phosphatidylserine synthesis | |
Palombo et al. | The promoter-associated noncoding RNA PNCCCND1_B assembles a protein–RNA complex to regulate cyclin D1 transcription in Ewing sarcoma | |
Wolf et al. | EVI/WLS function is regulated by ubiquitylation and is linked to ER-associated degradation by ERLIN2 | |
Ma et al. | DOCK2 regulates antifungal immunity by regulating RAC GTPase activity | |
Rasool et al. | Loss of LCMT1 and biased protein phosphatase 2A heterotrimerization drive prostate cancer progression and therapy resistance | |
Wang et al. | SATB1 regulates 3D genome architecture in T cells by constraining chromatin interactions surrounding CTCF-binding sites | |
Chapman et al. | Ataxin-3 links NOD2 and TLR2 mediated innate immune sensing and metabolism in myeloid cells | |
Tagnères et al. | SURF2 is a MDM2 antagonist in triggering the nucleolar stress response | |
WO2018149978A1 (en) | ALTERNATIVE ACTIVATION OF MACROPHAGES (M2 CELLS) THROUGH THE POLYAMINE-eIF5a-HYPUSINE AXIS | |
Ali et al. | A methyltransferase-independent role for METTL1 in tRNA aminoacylation and oncogenic transformation | |
Castro et al. | A Potent, Selective, Small-Molecule Inhibitor of DHX9 Abrogates Proliferation of Microsatellite Instable Cancers with Deficient Mismatch Repair |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 18706249 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 18706249 Country of ref document: EP Kind code of ref document: A1 |