MX2008004412A - Conserved membrane activator of calcineurin (cmac), a novel therapeutic protein and target - Google Patents
Conserved membrane activator of calcineurin (cmac), a novel therapeutic protein and targetInfo
- Publication number
- MX2008004412A MX2008004412A MXMX/A/2008/004412A MX2008004412A MX2008004412A MX 2008004412 A MX2008004412 A MX 2008004412A MX 2008004412 A MX2008004412 A MX 2008004412A MX 2008004412 A MX2008004412 A MX 2008004412A
- Authority
- MX
- Mexico
- Prior art keywords
- seq
- cmac
- expression
- agent
- disorder
- Prior art date
Links
- 102000004169 proteins and genes Human genes 0.000 title claims description 162
- 108090000623 proteins and genes Proteins 0.000 title claims description 162
- 102000004631 Calcineurin Human genes 0.000 title claims description 22
- 108010042955 Calcineurin Proteins 0.000 title claims description 22
- 230000001225 therapeutic Effects 0.000 title abstract description 27
- 239000012528 membrane Substances 0.000 title description 10
- 239000012190 activator Substances 0.000 title description 6
- 230000014509 gene expression Effects 0.000 claims abstract description 163
- 230000000694 effects Effects 0.000 claims abstract description 148
- 201000010099 disease Diseases 0.000 claims abstract description 107
- 150000001875 compounds Chemical class 0.000 claims abstract description 93
- 230000000051 modifying Effects 0.000 claims abstract description 70
- 230000004913 activation Effects 0.000 claims abstract description 68
- 210000001744 T-Lymphocytes Anatomy 0.000 claims abstract description 55
- 230000002401 inhibitory effect Effects 0.000 claims abstract description 52
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 32
- 239000003814 drug Substances 0.000 claims abstract description 21
- 210000004027 cells Anatomy 0.000 claims description 249
- 102000004965 antibodies Human genes 0.000 claims description 138
- 108090001123 antibodies Proteins 0.000 claims description 138
- 229920001184 polypeptide Polymers 0.000 claims description 129
- 150000007523 nucleic acids Chemical class 0.000 claims description 88
- 239000003795 chemical substances by application Substances 0.000 claims description 83
- 108020004707 nucleic acids Proteins 0.000 claims description 77
- 229920000160 (ribonucleotides)n+m Polymers 0.000 claims description 64
- 230000027455 binding Effects 0.000 claims description 53
- 230000017105 transposition Effects 0.000 claims description 52
- 229920001850 Nucleic acid sequence Polymers 0.000 claims description 43
- OYPRJOBELJOOCE-UHFFFAOYSA-N calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 claims description 32
- 239000011575 calcium Substances 0.000 claims description 32
- 229910052791 calcium Inorganic materials 0.000 claims description 32
- 229920002033 ribozyme Polymers 0.000 claims description 28
- 229920000272 Oligonucleotide Polymers 0.000 claims description 27
- 230000001419 dependent Effects 0.000 claims description 25
- 238000004519 manufacturing process Methods 0.000 claims description 22
- 238000001415 gene therapy Methods 0.000 claims description 20
- 150000002500 ions Chemical class 0.000 claims description 17
- 230000001603 reducing Effects 0.000 claims description 15
- 108020004459 Small Interfering RNA Proteins 0.000 claims description 14
- 238000006722 reduction reaction Methods 0.000 claims description 14
- 239000002924 silencing RNA Substances 0.000 claims description 14
- 230000004048 modification Effects 0.000 claims description 13
- 238000006011 modification reaction Methods 0.000 claims description 13
- 229920001785 Response element Polymers 0.000 claims description 12
- 230000002708 enhancing Effects 0.000 claims description 12
- 239000004055 small Interfering RNA Substances 0.000 claims description 10
- 229920001891 Small hairpin RNA Polymers 0.000 claims description 9
- 239000003623 enhancer Substances 0.000 claims description 9
- 108010045030 monoclonal antibodies Proteins 0.000 claims description 9
- 102000005614 monoclonal antibodies Human genes 0.000 claims description 9
- 108020000948 Antisense Oligonucleotides Proteins 0.000 claims description 8
- 239000000074 antisense oligonucleotide Substances 0.000 claims description 8
- 201000011510 cancer Diseases 0.000 claims description 8
- 238000009792 diffusion process Methods 0.000 claims description 8
- 230000032258 transport Effects 0.000 claims description 8
- 206010003816 Autoimmune disease Diseases 0.000 claims description 7
- 210000004698 Lymphocytes Anatomy 0.000 claims description 7
- 230000003993 interaction Effects 0.000 claims description 7
- 208000008787 Cardiovascular Disease Diseases 0.000 claims description 6
- 201000009596 autoimmune hypersensitivity disease Diseases 0.000 claims description 6
- 200000000018 inflammatory disease Diseases 0.000 claims description 6
- 230000035693 Fab Effects 0.000 claims description 5
- 206010062016 Immunosuppression Diseases 0.000 claims description 5
- 238000010171 animal model Methods 0.000 claims description 5
- 230000001506 immunosuppresive Effects 0.000 claims description 5
- 230000004044 response Effects 0.000 claims description 5
- 229920001985 Small interfering RNA Polymers 0.000 claims description 4
- 230000000926 neurological Effects 0.000 claims description 4
- 239000002253 acid Substances 0.000 claims description 3
- 239000003937 drug carrier Substances 0.000 claims description 3
- 210000000173 Precursor Cells, T-Lymphoid Anatomy 0.000 claims description 2
- 230000002103 transcriptional Effects 0.000 claims 1
- 230000001575 pathological Effects 0.000 abstract description 23
- 239000000556 agonist Substances 0.000 abstract description 13
- 102100001295 TMEM203 Human genes 0.000 abstract description 5
- 101710009717 TMEM203 Proteins 0.000 abstract description 5
- 230000004952 protein activity Effects 0.000 abstract description 5
- 238000011161 development Methods 0.000 abstract description 4
- 230000018109 developmental process Effects 0.000 abstract description 4
- 239000003596 drug target Substances 0.000 abstract description 3
- 235000018102 proteins Nutrition 0.000 description 150
- 238000000034 method Methods 0.000 description 51
- 241000282414 Homo sapiens Species 0.000 description 49
- 229920003013 deoxyribonucleic acid Polymers 0.000 description 47
- 108020004999 Messenger RNA Proteins 0.000 description 42
- 229920002106 messenger RNA Polymers 0.000 description 42
- 229920002676 Complementary DNA Polymers 0.000 description 34
- 230000035897 transcription Effects 0.000 description 32
- 239000000203 mixture Substances 0.000 description 30
- 102000000588 Interleukin-2 Human genes 0.000 description 28
- 108010002350 Interleukin-2 Proteins 0.000 description 28
- 239000002299 complementary DNA Substances 0.000 description 28
- 210000001519 tissues Anatomy 0.000 description 26
- 241000700605 Viruses Species 0.000 description 24
- 230000003612 virological Effects 0.000 description 24
- 239000002609 media Substances 0.000 description 23
- PHEDXBVPIONUQT-RGYGYFBISA-N 12-O-Tetradecanoylphorbol-13-acetate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(OC(C)=O)C1(C)C PHEDXBVPIONUQT-RGYGYFBISA-N 0.000 description 21
- 102000003569 TRPV6 Human genes 0.000 description 20
- 108060008570 TRPV6 Proteins 0.000 description 20
- 239000000047 product Substances 0.000 description 20
- PMATZTZNYRCHOR-CGLBZJNRSA-N (3S,6S,9S,12R,15S,18S,21S,24S,30S,33S)-30-ethyl-33-[(E,1R,2R)-1-hydroxy-2-methylhex-4-enyl]-1,4,7,10,12,15,19,25,28-nonamethyl-6,9,18,24-tetrakis(2-methylpropyl)-3,21-di(propan-2-yl)-1,4,7,10,13,16,19,22,25,28,31-undecazacyclotritriacontane-2,5,8,11,14,17 Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 19
- 229920001405 Coding region Polymers 0.000 description 19
- 108010036949 Cyclosporine Proteins 0.000 description 19
- 229960001265 ciclosporin Drugs 0.000 description 19
- 102000018358 Immunoglobulins Human genes 0.000 description 18
- 108060003951 Immunoglobulins Proteins 0.000 description 18
- 235000001014 amino acid Nutrition 0.000 description 18
- 239000000523 sample Substances 0.000 description 18
- 230000026683 transduction Effects 0.000 description 18
- 150000001413 amino acids Chemical class 0.000 description 17
- 238000010361 transduction Methods 0.000 description 17
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 16
- 125000003275 alpha amino acid group Chemical group 0.000 description 16
- 239000000427 antigen Substances 0.000 description 16
- 238000000338 in vitro Methods 0.000 description 16
- 239000003112 inhibitor Substances 0.000 description 16
- 230000000875 corresponding Effects 0.000 description 15
- 239000000463 material Substances 0.000 description 15
- 210000000130 stem cell Anatomy 0.000 description 15
- 239000000126 substance Substances 0.000 description 15
- 108091007172 antigens Proteins 0.000 description 14
- 102000038129 antigens Human genes 0.000 description 14
- 108020001507 fusion proteins Proteins 0.000 description 14
- 102000037240 fusion proteins Human genes 0.000 description 14
- 241000894007 species Species 0.000 description 14
- 102000004190 Enzymes Human genes 0.000 description 13
- 108090000790 Enzymes Proteins 0.000 description 13
- 108090000994 Catalytic RNA Proteins 0.000 description 12
- 230000000692 anti-sense Effects 0.000 description 12
- 230000001404 mediated Effects 0.000 description 12
- 230000001105 regulatory Effects 0.000 description 12
- 238000001890 transfection Methods 0.000 description 12
- 241001465754 Metazoa Species 0.000 description 11
- 230000000295 complement Effects 0.000 description 11
- 238000007796 conventional method Methods 0.000 description 11
- 229940079593 drugs Drugs 0.000 description 11
- 229920000023 polynucleotide Polymers 0.000 description 11
- 239000002157 polynucleotide Substances 0.000 description 11
- 102000004196 processed proteins & peptides Human genes 0.000 description 11
- 108090000765 processed proteins & peptides Proteins 0.000 description 11
- 108090000312 Calcium Channels Proteins 0.000 description 10
- 102000003922 Calcium Channels Human genes 0.000 description 10
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 10
- 238000004166 bioassay Methods 0.000 description 10
- 238000006243 chemical reaction Methods 0.000 description 10
- 150000003384 small molecules Chemical class 0.000 description 10
- 241000701161 unidentified adenovirus Species 0.000 description 10
- 210000003958 Hematopoietic Stem Cells Anatomy 0.000 description 9
- 229940072221 IMMUNOGLOBULINS Drugs 0.000 description 9
- 230000002159 abnormal effect Effects 0.000 description 9
- 238000004458 analytical method Methods 0.000 description 9
- 230000001939 inductive effect Effects 0.000 description 9
- 239000003550 marker Substances 0.000 description 9
- 125000003729 nucleotide group Chemical group 0.000 description 9
- 230000002018 overexpression Effects 0.000 description 9
- 208000006673 Asthma Diseases 0.000 description 8
- 241000282412 Homo Species 0.000 description 8
- 206010025135 Lupus erythematosus Diseases 0.000 description 8
- 210000004379 Membranes Anatomy 0.000 description 8
- 210000004940 Nucleus Anatomy 0.000 description 8
- 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 8
- 229950010131 PUROMYCIN Drugs 0.000 description 8
- 108091008153 T cell receptors Proteins 0.000 description 8
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 8
- 230000003115 biocidal Effects 0.000 description 8
- 239000002773 nucleotide Substances 0.000 description 8
- 239000000758 substrate Substances 0.000 description 8
- 229940088598 Enzyme Drugs 0.000 description 7
- 102000008135 Mechanistic Target of Rapamycin Complex 1 Human genes 0.000 description 7
- 108010035196 Mechanistic Target of Rapamycin Complex 1 Proteins 0.000 description 7
- 102000003923 Protein Kinase C Human genes 0.000 description 7
- 108090000315 Protein Kinase C Proteins 0.000 description 7
- 230000003042 antagnostic Effects 0.000 description 7
- 239000005557 antagonist Substances 0.000 description 7
- 230000015572 biosynthetic process Effects 0.000 description 7
- 238000001514 detection method Methods 0.000 description 7
- 238000009396 hybridization Methods 0.000 description 7
- 238000006366 phosphorylation reaction Methods 0.000 description 7
- 102000005962 receptors Human genes 0.000 description 7
- 108020003175 receptors Proteins 0.000 description 7
- 238000003786 synthesis reaction Methods 0.000 description 7
- 238000002560 therapeutic procedure Methods 0.000 description 7
- 229940064005 Antibiotic throat preparations Drugs 0.000 description 6
- 229940083879 Antibiotics FOR TREATMENT OF HEMORRHOIDS AND ANAL FISSURES FOR TOPICAL USE Drugs 0.000 description 6
- 229940042052 Antibiotics for systemic use Drugs 0.000 description 6
- 229940042786 Antitubercular Antibiotics Drugs 0.000 description 6
- 229920002395 Aptamer Polymers 0.000 description 6
- 210000003719 B-Lymphocytes Anatomy 0.000 description 6
- 229940093922 Gynecological Antibiotics Drugs 0.000 description 6
- 239000005089 Luciferase Substances 0.000 description 6
- 241000124008 Mammalia Species 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 210000002966 Serum Anatomy 0.000 description 6
- 210000003491 Skin Anatomy 0.000 description 6
- 229940024982 Topical Antifungal Antibiotics Drugs 0.000 description 6
- 239000003242 anti bacterial agent Substances 0.000 description 6
- 230000020411 cell activation Effects 0.000 description 6
- 230000001413 cellular Effects 0.000 description 6
- 238000010276 construction Methods 0.000 description 6
- 230000001809 detectable Effects 0.000 description 6
- 201000009910 diseases by infectious agent Diseases 0.000 description 6
- 102000034448 gene-regulatory proteins Human genes 0.000 description 6
- 108091006088 gene-regulatory proteins Proteins 0.000 description 6
- 230000001965 increased Effects 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 229940079866 intestinal antibiotics Drugs 0.000 description 6
- 230000003834 intracellular Effects 0.000 description 6
- 239000003446 ligand Substances 0.000 description 6
- 229940005935 ophthalmologic Antibiotics Drugs 0.000 description 6
- 238000004806 packaging method and process Methods 0.000 description 6
- 230000000865 phosphorylative Effects 0.000 description 6
- 238000002360 preparation method Methods 0.000 description 6
- 230000001177 retroviral Effects 0.000 description 6
- -1 single chain Proteins 0.000 description 6
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 230000002194 synthesizing Effects 0.000 description 6
- 102000003995 transcription factors Human genes 0.000 description 6
- 108090000464 transcription factors Proteins 0.000 description 6
- 101710006661 ITGB1 Proteins 0.000 description 5
- 102100001478 ITGB1 Human genes 0.000 description 5
- 210000000936 Intestines Anatomy 0.000 description 5
- 210000004185 Liver Anatomy 0.000 description 5
- 108060001084 Luciferase family Proteins 0.000 description 5
- 241000288906 Primates Species 0.000 description 5
- 230000001154 acute Effects 0.000 description 5
- 238000004113 cell culture Methods 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 108091006028 chimera Proteins 0.000 description 5
- 230000001684 chronic Effects 0.000 description 5
- KCXVZYZYPLLWCC-UHFFFAOYSA-N edta Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 230000028993 immune response Effects 0.000 description 5
- 238000002955 isolation Methods 0.000 description 5
- 230000003389 potentiating Effects 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 230000001629 suppression Effects 0.000 description 5
- 238000005406 washing Methods 0.000 description 5
- 101710027066 ALB Proteins 0.000 description 4
- 108010066057 CABIN1 Proteins 0.000 description 4
- 102100009787 CABIN1 Human genes 0.000 description 4
- 102100018499 CRTC1 Human genes 0.000 description 4
- 101700057734 CRTC1 Proteins 0.000 description 4
- 210000000170 Cell Membrane Anatomy 0.000 description 4
- 108020004461 Double-Stranded RNA Proteins 0.000 description 4
- 241000588724 Escherichia coli Species 0.000 description 4
- 241000238631 Hexapoda Species 0.000 description 4
- 210000004408 Hybridomas Anatomy 0.000 description 4
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 4
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 4
- 241000699660 Mus musculus Species 0.000 description 4
- 241000283973 Oryctolagus cuniculus Species 0.000 description 4
- 101700040790 PH Proteins 0.000 description 4
- 102000007079 Peptide Fragments Human genes 0.000 description 4
- 108010033276 Peptide Fragments Proteins 0.000 description 4
- 108020004511 Recombinant DNA Proteins 0.000 description 4
- 102000005632 Single-Chain Antibodies Human genes 0.000 description 4
- 108010070144 Single-Chain Antibodies Proteins 0.000 description 4
- 229920000978 Start codon Polymers 0.000 description 4
- 238000007792 addition Methods 0.000 description 4
- 239000002671 adjuvant Substances 0.000 description 4
- 230000000240 adjuvant Effects 0.000 description 4
- 125000000539 amino acid group Chemical group 0.000 description 4
- 230000001580 bacterial Effects 0.000 description 4
- 230000031018 biological processes and functions Effects 0.000 description 4
- 239000000969 carrier Substances 0.000 description 4
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 4
- 238000010494 dissociation reaction Methods 0.000 description 4
- 230000005593 dissociations Effects 0.000 description 4
- WSFSSNUMVMOOMR-UHFFFAOYSA-N formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 4
- 230000004927 fusion Effects 0.000 description 4
- 230000003899 glycosylation Effects 0.000 description 4
- 238000006206 glycosylation reaction Methods 0.000 description 4
- 230000001976 improved Effects 0.000 description 4
- 238000002372 labelling Methods 0.000 description 4
- 230000004481 post-translational protein modification Effects 0.000 description 4
- 239000000843 powder Substances 0.000 description 4
- 238000005215 recombination Methods 0.000 description 4
- 230000002829 reduced Effects 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- 230000004083 survival Effects 0.000 description 4
- 238000011144 upstream manufacturing Methods 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 3
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 3
- 241000201370 Autographa californica nucleopolyhedrovirus Species 0.000 description 3
- 102000005636 Cyclic AMP Response Element-Binding Protein Human genes 0.000 description 3
- 108010045171 Cyclic AMP Response Element-Binding Protein Proteins 0.000 description 3
- MTCFGRXMJLQNBG-UWTATZPHSA-N D-serine Chemical compound OC[C@@H](N)C(O)=O MTCFGRXMJLQNBG-UWTATZPHSA-N 0.000 description 3
- 241000252212 Danio rerio Species 0.000 description 3
- 229940022766 EGTA Drugs 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- 102000005720 Glutathione Transferase family Human genes 0.000 description 3
- 108010070675 Glutathione Transferase family Proteins 0.000 description 3
- 230000036499 Half live Effects 0.000 description 3
- 210000002510 Keratinocytes Anatomy 0.000 description 3
- 241000699670 Mus sp. Species 0.000 description 3
- 108010029485 Protein Isoforms Proteins 0.000 description 3
- 102000001708 Protein Isoforms Human genes 0.000 description 3
- 241000700159 Rattus Species 0.000 description 3
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 3
- 206010070834 Sensitisation Diseases 0.000 description 3
- 108010002487 Thomsen-Friedenreich antibodies Proteins 0.000 description 3
- 229920001949 Transfer RNA Polymers 0.000 description 3
- FPKOPBFLPLFWAD-UHFFFAOYSA-N Trinitrotoluene Chemical compound CC1=CC=C([N+]([O-])=O)C([N+]([O-])=O)=C1[N+]([O-])=O FPKOPBFLPLFWAD-UHFFFAOYSA-N 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 229960000070 antineoplastic Monoclonal antibodies Drugs 0.000 description 3
- 201000008937 atopic dermatitis Diseases 0.000 description 3
- 210000002798 bone marrow cell Anatomy 0.000 description 3
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 3
- 229910000389 calcium phosphate Inorganic materials 0.000 description 3
- 238000010367 cloning Methods 0.000 description 3
- 230000003081 coactivator Effects 0.000 description 3
- 239000011248 coating agent Substances 0.000 description 3
- 238000000576 coating method Methods 0.000 description 3
- 230000001086 cytosolic Effects 0.000 description 3
- 230000002950 deficient Effects 0.000 description 3
- 239000002552 dosage form Substances 0.000 description 3
- 238000004520 electroporation Methods 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 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 3
- 230000001747 exhibiting Effects 0.000 description 3
- 239000000284 extract Substances 0.000 description 3
- 125000002485 formyl group Chemical group [H]C(*)=O 0.000 description 3
- 230000002068 genetic Effects 0.000 description 3
- 230000002209 hydrophobic Effects 0.000 description 3
- 238000003384 imaging method Methods 0.000 description 3
- 230000001900 immune effect Effects 0.000 description 3
- 150000002632 lipids Chemical class 0.000 description 3
- 230000004807 localization Effects 0.000 description 3
- 210000004962 mammalian cells Anatomy 0.000 description 3
- 230000003278 mimic Effects 0.000 description 3
- 229960000060 monoclonal antibodies Drugs 0.000 description 3
- 230000036961 partial Effects 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- 108091008117 polyclonal antibodies Proteins 0.000 description 3
- 230000002335 preservative Effects 0.000 description 3
- 239000003755 preservative agent Substances 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 150000003839 salts Chemical class 0.000 description 3
- 230000008313 sensitization Effects 0.000 description 3
- 230000001235 sensitizing Effects 0.000 description 3
- 235000004400 serine Nutrition 0.000 description 3
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 239000003826 tablet Substances 0.000 description 3
- 238000010998 test method Methods 0.000 description 3
- 238000002054 transplantation Methods 0.000 description 3
- 229960005486 vaccines Drugs 0.000 description 3
- HVYWMOMLDIMFJA-DPAQBDIFSA-N (3β)-Cholest-5-en-3-ol Chemical compound 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 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N 1-[(1S,2R,3R,4S,5R,6R)-3-carbamimidamido-6-{[(2R,3R,4R,5S)-3-{[(2S,3S,4S,5R,6S)-4,5-dihydroxy-6-(hydroxymethyl)-3-(methylamino)oxan-2-yl]oxy}-4-formyl-4-hydroxy-5-methyloxolan-2-yl]oxy}-2,4,5-trihydroxycyclohexyl]guanidine 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 2
- 241000432074 Adeno-associated virus Species 0.000 description 2
- 229920000936 Agarose Polymers 0.000 description 2
- 244000105975 Antidesma platyphyllum Species 0.000 description 2
- 229960000190 Bacillus Calmette–Guérin vaccine Drugs 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- 208000009137 Behcet Syndrome Diseases 0.000 description 2
- 201000008335 Behcet's disease Diseases 0.000 description 2
- 210000000601 Blood Cells Anatomy 0.000 description 2
- 210000001185 Bone Marrow Anatomy 0.000 description 2
- 241000251535 Branchiostoma floridae Species 0.000 description 2
- 108010029697 CD40 Ligand Proteins 0.000 description 2
- 102100003729 CD40LG Human genes 0.000 description 2
- 102100005832 CD69 Human genes 0.000 description 2
- 101700080416 CD69 Proteins 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 210000000349 Chromosomes Anatomy 0.000 description 2
- 210000000805 Cytoplasm Anatomy 0.000 description 2
- 210000000172 Cytosol Anatomy 0.000 description 2
- KDXKERNSBIXSRK-RXMQYKEDSA-N D-lysine group Chemical group N[C@H](CCCCN)C(=O)O KDXKERNSBIXSRK-RXMQYKEDSA-N 0.000 description 2
- 206010012438 Dermatitis atopic Diseases 0.000 description 2
- 102000033147 ERVK-25 Human genes 0.000 description 2
- 210000002889 Endothelial Cells Anatomy 0.000 description 2
- 210000000981 Epithelium Anatomy 0.000 description 2
- 210000004700 Fetal Blood Anatomy 0.000 description 2
- 101700014779 GLB1 Proteins 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N HEPES Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 2
- 208000006454 Hepatitis Diseases 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 101700082799 IL2RA Proteins 0.000 description 2
- 101700015336 ISG20 Proteins 0.000 description 2
- 102100002950 ISG20 Human genes 0.000 description 2
- RAXXELZNTBOGNW-UHFFFAOYSA-N Imidazole Chemical compound C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 2
- 210000000987 Immune System Anatomy 0.000 description 2
- 206010061218 Inflammation Diseases 0.000 description 2
- 229920002459 Intron Polymers 0.000 description 2
- 108020004391 Introns Proteins 0.000 description 2
- 102000004310 Ion Channels Human genes 0.000 description 2
- 108090000862 Ion Channels Proteins 0.000 description 2
- 206010023332 Keratitis Diseases 0.000 description 2
- XUJNEKJLAYXESH-REOHCLBHSA-N L-cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 2
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 2
- GUBGYTABKSRVRQ-UUNJERMWSA-N Lactose Natural products O([C@@H]1[C@H](O)[C@H](O)[C@H](O)O[C@@H]1CO)[C@H]1[C@@H](O)[C@@H](O)[C@H](O)[C@H](CO)O1 GUBGYTABKSRVRQ-UUNJERMWSA-N 0.000 description 2
- 239000012097 Lipofectamine 2000 Substances 0.000 description 2
- 210000004072 Lung Anatomy 0.000 description 2
- 102100000165 MS4A1 Human genes 0.000 description 2
- 101710010909 MS4A1 Proteins 0.000 description 2
- 102000018697 Membrane Proteins Human genes 0.000 description 2
- 108010052285 Membrane Proteins Proteins 0.000 description 2
- 210000003205 Muscles Anatomy 0.000 description 2
- 241001467552 Mycobacterium bovis BCG Species 0.000 description 2
- 102000003945 NF-kappa B Human genes 0.000 description 2
- 108010057466 NF-kappa B Proteins 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 206010029164 Nephrotic syndrome Diseases 0.000 description 2
- 239000002033 PVDF binder Substances 0.000 description 2
- LLKYUHGUYSLMPA-UHFFFAOYSA-N Phosphoramidite Chemical compound NP([O-])[O-] LLKYUHGUYSLMPA-UHFFFAOYSA-N 0.000 description 2
- 230000025458 RNA interference Effects 0.000 description 2
- 239000007759 RPMI Media 1640 Substances 0.000 description 2
- PYWVYCXTNDRMGF-UHFFFAOYSA-N Rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M Sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 241000256251 Spodoptera frugiperda Species 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine Kinase Proteins 0.000 description 2
- 108020004566 Transfer RNA Proteins 0.000 description 2
- QORWJWZARLRLPR-UHFFFAOYSA-H Tricalcium 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
- YHBHQYRDAVETGQ-UHFFFAOYSA-N Triton X 100 Chemical compound CC(C)(C)CC(C)(C)C1=CC=C(OCCCCCCCCCCO)C=C1 YHBHQYRDAVETGQ-UHFFFAOYSA-N 0.000 description 2
- 206010046851 Uveitis Diseases 0.000 description 2
- 206010047461 Viral infection Diseases 0.000 description 2
- 208000001756 Virus Disease Diseases 0.000 description 2
- 241000269457 Xenopus tropicalis Species 0.000 description 2
- 230000003044 adaptive Effects 0.000 description 2
- 230000001464 adherent Effects 0.000 description 2
- 239000000443 aerosol Substances 0.000 description 2
- 230000000843 anti-fungal Effects 0.000 description 2
- 108010005774 beta-Galactosidase Proteins 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 239000012472 biological sample Substances 0.000 description 2
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 230000000903 blocking Effects 0.000 description 2
- ODINCKMPIJJUCX-UHFFFAOYSA-N calcium monoxide Chemical compound [Ca]=O ODINCKMPIJJUCX-UHFFFAOYSA-N 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 235000014633 carbohydrates Nutrition 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- 239000003184 complementary RNA Substances 0.000 description 2
- 235000018417 cysteine Nutrition 0.000 description 2
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 2
- 230000003247 decreasing Effects 0.000 description 2
- 230000002616 endonucleolytic Effects 0.000 description 2
- 230000001605 fetal Effects 0.000 description 2
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 2
- 238000003260 fluorescence intensity Methods 0.000 description 2
- 239000007850 fluorescent dye Substances 0.000 description 2
- 238000005755 formation reaction Methods 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 235000009424 haa Nutrition 0.000 description 2
- 231100000283 hepatitis Toxicity 0.000 description 2
- 238000002744 homologous recombination Methods 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 239000000411 inducer Substances 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 230000004941 influx Effects 0.000 description 2
- 230000000977 initiatory Effects 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000000302 ischemic Effects 0.000 description 2
- GUBGYTABKSRVRQ-XLOQQCSPSA-N lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 2
- 239000008101 lactose Substances 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 201000004044 liver cirrhosis Diseases 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 238000002493 microarray Methods 0.000 description 2
- 238000000520 microinjection Methods 0.000 description 2
- 239000011859 microparticle Substances 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 239000005445 natural product Substances 0.000 description 2
- 230000004112 neuroprotection Effects 0.000 description 2
- 239000003921 oil Substances 0.000 description 2
- 235000019198 oils Nutrition 0.000 description 2
- 238000002515 oligonucleotide synthesis Methods 0.000 description 2
- 239000000546 pharmaceutic aid Substances 0.000 description 2
- 239000000825 pharmaceutical preparation Substances 0.000 description 2
- 229920000740 poly(D-lysine) polymer Polymers 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 230000004850 protein–protein interaction Effects 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 230000002285 radioactive Effects 0.000 description 2
- 238000003259 recombinant expression Methods 0.000 description 2
- 230000000241 respiratory Effects 0.000 description 2
- 108091007521 restriction endonucleases Proteins 0.000 description 2
- 230000002987 rna-interference Effects 0.000 description 2
- 238000007423 screening assay Methods 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 230000035939 shock Effects 0.000 description 2
- 231100000486 side effect Toxicity 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 230000011664 signaling Effects 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 150000003431 steroids Chemical class 0.000 description 2
- 238000005556 structure-activity relationship Methods 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- ORHBXUUXSCNDEV-UHFFFAOYSA-N umbelliferone Chemical compound C1=CC(=O)OC2=CC(O)=CC=C21 ORHBXUUXSCNDEV-UHFFFAOYSA-N 0.000 description 2
- 241001515965 unidentified phage Species 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- 230000017613 viral reproduction Effects 0.000 description 2
- 239000002699 waste material Substances 0.000 description 2
- NFGXHKASABOEEW-UHFFFAOYSA-N (+)-methoprene Chemical compound COC(C)(C)CCCC(C)CC=CC(C)=CC(=O)OC(C)C NFGXHKASABOEEW-UHFFFAOYSA-N 0.000 description 1
- NKCXQMYPWXSLIZ-PSRDDEIFSA-N (2S)-2-[[(2S)-1-[(2S)-5-amino-2-[[2-[[(2S)-6-amino-2-[[2-[[(2S)-2-[[(2S)-4-amino-2-[[(2S)-2-[[(2S,3R)-2-[[(2S)-2-[[(2S,3R)-2-amino-3-hydroxybutanoyl]amino]-3-(1H-indol-3-yl)propanoyl]amino]-3-hydroxybutanoyl]amino]propanoyl]amino]-4-oxobutanoyl]amino]-3-m Chemical compound O=C([C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CCCCN)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@@H](N)[C@@H](C)O)[C@@H](C)O)C(C)C)N1CCC[C@H]1C(=O)N[C@@H](CO)C(O)=O NKCXQMYPWXSLIZ-PSRDDEIFSA-N 0.000 description 1
- DDMOUSALMHHKOS-UHFFFAOYSA-N 1,2-dichloro-1,1,2,2-tetrafluoroethane Chemical compound FC(F)(Cl)C(F)(F)Cl DDMOUSALMHHKOS-UHFFFAOYSA-N 0.000 description 1
- YAMUFBLWGFFICM-PTGWMXDISA-N 1-O-oleoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@@H](O)COP([O-])(=O)OCC[N+](C)(C)C YAMUFBLWGFFICM-PTGWMXDISA-N 0.000 description 1
- JLPULHDHAOZNQI-ZTIMHPMXSA-N 1-hexadecanoyl-2-(9Z,12Z-octadecadienoyl)-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/C\C=C/CCCCC JLPULHDHAOZNQI-ZTIMHPMXSA-N 0.000 description 1
- MHKBMNACOMRIAW-UHFFFAOYSA-N 2,3-dinitrophenol Chemical compound OC1=CC=CC([N+]([O-])=O)=C1[N+]([O-])=O MHKBMNACOMRIAW-UHFFFAOYSA-N 0.000 description 1
- RTGDFNSFWBGLEC-SYZQJQIISA-N 2-(morpholin-4-yl)ethyl (4E)-6-(4-hydroxy-6-methoxy-7-methyl-3-oxo-1,3-dihydro-2-benzofuran-5-yl)-4-methylhex-4-enoate Chemical compound COC1=C(C)C=2COC(=O)C=2C(O)=C1C\C=C(/C)CCC(=O)OCCN1CCOCC1 RTGDFNSFWBGLEC-SYZQJQIISA-N 0.000 description 1
- VUKAUDKDFVSVFT-UHFFFAOYSA-N 2-[6-[4,5-bis(2-hydroxypropoxy)-2-(2-hydroxypropoxymethyl)-6-methoxyoxan-3-yl]oxy-4,5-dimethoxy-2-(methoxymethyl)oxan-3-yl]oxy-6-(hydroxymethyl)-5-methoxyoxane-3,4-diol Chemical compound COC1C(OC)C(OC2C(C(O)C(OC)C(CO)O2)O)C(COC)OC1OC1C(COCC(C)O)OC(OC)C(OCC(C)O)C1OCC(C)O VUKAUDKDFVSVFT-UHFFFAOYSA-N 0.000 description 1
- DGVVWUTYPXICAM-UHFFFAOYSA-N 2-mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 1
- PMATZTZNYRCHOR-IMVLJIQESA-N 30-ethyl-33-[(E)-1-hydroxy-2-methylhex-4-enyl]-1,4,7,10,12,15,19,25,28-nonamethyl-6,9,18,24-tetrakis(2-methylpropyl)-3,21-di(propan-2-yl)-1,4,7,10,13,16,19,22,25,28,31-undecazacyclotritriacontane-2,5,8,11,14,17,20,23,26,29,32-undecone Chemical compound CCC1NC(=O)C(C(O)C(C)C\C=C\C)N(C)C(=O)C(C(C)C)N(C)C(=O)C(CC(C)C)N(C)C(=O)C(CC(C)C)N(C)C(=O)C(C)NC(=O)C(C)NC(=O)C(CC(C)C)N(C)C(=O)C(C(C)C)NC(=O)C(CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-IMVLJIQESA-N 0.000 description 1
- XZKIHKMTEMTJQX-UHFFFAOYSA-N 4-Nitrophenyl dihydrogen phosphate Chemical compound OP(O)(=O)OC1=CC=C([N+]([O-])=O)C=C1 XZKIHKMTEMTJQX-UHFFFAOYSA-N 0.000 description 1
- XPDXVDYUQZHFPV-UHFFFAOYSA-N 5-Dimethylaminonaphthyl-5-sulfonyl chloride Chemical compound C1=CC=C2C(N(C)C)=CC=CC2=C1S(Cl)(=O)=O XPDXVDYUQZHFPV-UHFFFAOYSA-N 0.000 description 1
- GZAJOEGTZDUSKS-UHFFFAOYSA-N 6-amino-3',6'-dihydroxyspiro[2-benzofuran-3,9'-xanthene]-1-one Chemical compound C12=CC=C(O)C=C2OC2=CC(O)=CC=C2C21OC(=O)C1=CC(N)=CC=C21 GZAJOEGTZDUSKS-UHFFFAOYSA-N 0.000 description 1
- 101710006746 7.5K Proteins 0.000 description 1
- KHIWWQKSHDUIBK-UHFFFAOYSA-M AC1L4ZKD Chemical compound [O-]I(=O)(=O)=O KHIWWQKSHDUIBK-UHFFFAOYSA-M 0.000 description 1
- 102100009225 ACHE Human genes 0.000 description 1
- 102100007409 APRT Human genes 0.000 description 1
- 101710006647 APRT Proteins 0.000 description 1
- 231100000582 ATP assay Toxicity 0.000 description 1
- 229940022698 Acetylcholinesterase Drugs 0.000 description 1
- 108010022752 Acetylcholinesterase Proteins 0.000 description 1
- 206010000496 Acne Diseases 0.000 description 1
- 206010000565 Acquired immunodeficiency syndrome Diseases 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- 108010024223 Adenine Phosphoribosyltransferase Proteins 0.000 description 1
- 108010000239 Aequorin Proteins 0.000 description 1
- 206010001497 Agitation Diseases 0.000 description 1
- 229960003767 Alanine Drugs 0.000 description 1
- 239000012103 Alexa Fluor 488 Substances 0.000 description 1
- 235000019489 Almond oil Nutrition 0.000 description 1
- 208000004631 Alopecia Areata Diseases 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K Aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 206010001897 Alzheimer's disease Diseases 0.000 description 1
- 241000269328 Amphibia Species 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
- 206010002026 Amyotrophic lateral sclerosis Diseases 0.000 description 1
- 206010002425 Angioedemas Diseases 0.000 description 1
- 108010049777 Ankyrins Proteins 0.000 description 1
- 102000008102 Ankyrins Human genes 0.000 description 1
- 206010002660 Anoxia Diseases 0.000 description 1
- 241000976983 Anoxia Species 0.000 description 1
- 208000008637 Anti-Glomerular Basement Membrane Disease Diseases 0.000 description 1
- 108010055216 Anti-Idiotypic Antibodies Proteins 0.000 description 1
- 108020005098 Anticodon Proteins 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 206010003210 Arteriosclerosis Diseases 0.000 description 1
- 229960001230 Asparagine Drugs 0.000 description 1
- 229960005261 Aspartic Acid Drugs 0.000 description 1
- 208000004300 Atrophic Gastritis Diseases 0.000 description 1
- 208000005783 Autoimmune Thyroiditis Diseases 0.000 description 1
- 206010003827 Autoimmune hepatitis Diseases 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 206010003885 Azotaemia Diseases 0.000 description 1
- 235000014469 Bacillus subtilis Nutrition 0.000 description 1
- 229920000195 Bacterial small RNA Polymers 0.000 description 1
- 210000002469 Basement Membrane Anatomy 0.000 description 1
- 241000349774 Bikinia letestui Species 0.000 description 1
- 210000004369 Blood Anatomy 0.000 description 1
- 210000004204 Blood Vessels Anatomy 0.000 description 1
- 229940098773 Bovine Serum Albumin Drugs 0.000 description 1
- 108091003117 Bovine Serum Albumin Proteins 0.000 description 1
- 206010006451 Bronchitis Diseases 0.000 description 1
- 208000000594 Bullous Pemphigoid Diseases 0.000 description 1
- 102100008431 CCL26 Human genes 0.000 description 1
- 101700040437 CCL26 Proteins 0.000 description 1
- 108010080818 Caenorhabditis elegans Proteins Proteins 0.000 description 1
- JHLNERQLKQQLRZ-UHFFFAOYSA-N Calcium silicate Chemical compound [Ca+2].[Ca+2].[O-][Si]([O-])([O-])[O-] JHLNERQLKQQLRZ-UHFFFAOYSA-N 0.000 description 1
- 229960004424 Carbon Dioxide Drugs 0.000 description 1
- 206010007554 Cardiac failure Diseases 0.000 description 1
- 206010007572 Cardiac hypertrophy Diseases 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- 108010078791 Carrier Proteins Proteins 0.000 description 1
- 108010010919 Casein Kinase II Proteins 0.000 description 1
- 108091005504 Cation channels Proteins 0.000 description 1
- 241000701489 Cauliflower mosaic virus Species 0.000 description 1
- 108010029240 Cell-Tak Proteins 0.000 description 1
- 210000003169 Central Nervous System Anatomy 0.000 description 1
- 241000251522 Cephalochordata Species 0.000 description 1
- 229960005091 Chloramphenicol Drugs 0.000 description 1
- 108010035563 Chloramphenicol O-Acetyltransferase Proteins 0.000 description 1
- 206010008609 Cholangitis sclerosing Diseases 0.000 description 1
- 229940107161 Cholesterol Drugs 0.000 description 1
- 210000004507 Chromosomes, Artificial Anatomy 0.000 description 1
- 210000003917 Chromosomes, Human Anatomy 0.000 description 1
- 108020004705 Codon Proteins 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 208000008006 Collagen Disease Diseases 0.000 description 1
- 108020004394 Complementary RNA Proteins 0.000 description 1
- 206010010741 Conjunctivitis Diseases 0.000 description 1
- 206010010744 Conjunctivitis allergic Diseases 0.000 description 1
- 208000010247 Contact Dermatitis Diseases 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 241000186216 Corynebacterium Species 0.000 description 1
- 206010011401 Crohn's disease Diseases 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- 229960004397 Cyclophosphamide Drugs 0.000 description 1
- 229940119017 Cyclosporine Drugs 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N D-Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 1
- QIVBCDIJIAJPQS-SECBINFHSA-N D-tryptophane Chemical compound C1=CC=C2C(C[C@@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-SECBINFHSA-N 0.000 description 1
- 101710007887 DHFR Proteins 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
- 101710028159 DNTT Proteins 0.000 description 1
- 102100002445 DNTT Human genes 0.000 description 1
- 206010012442 Dermatitis contact Diseases 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N Dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 229960003957 Dexamethasone Drugs 0.000 description 1
- 206010012601 Diabetes mellitus Diseases 0.000 description 1
- 208000007342 Diabetic Nephropathy Diseases 0.000 description 1
- 206010061835 Diabetic nephropathy Diseases 0.000 description 1
- PXBRQCKWGAHEHS-UHFFFAOYSA-N Dichlorodifluoromethane Chemical compound FC(F)(Cl)Cl PXBRQCKWGAHEHS-UHFFFAOYSA-N 0.000 description 1
- 239000004338 Dichlorodifluoromethane Substances 0.000 description 1
- 229940087091 Dichlorotetrafluoroethane Drugs 0.000 description 1
- 108010035533 Drosophila Proteins Proteins 0.000 description 1
- 206010059866 Drug resistance Diseases 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 108010015776 EC 1.1.3.4 Proteins 0.000 description 1
- 108010091358 EC 2.4.2.8 Proteins 0.000 description 1
- 108010092799 EC 2.7.7.49 Proteins 0.000 description 1
- 101710003775 ERVK-10 Proteins 0.000 description 1
- 101710037030 ERVK-11 Proteins 0.000 description 1
- 101710009283 ERVK-18 Proteins 0.000 description 1
- 101710009286 ERVK-19 Proteins 0.000 description 1
- 101710035700 ERVK-25 Proteins 0.000 description 1
- 101710038044 ERVK-6 Proteins 0.000 description 1
- 101710014468 ERVK-7 Proteins 0.000 description 1
- 101710014482 ERVK-8 Proteins 0.000 description 1
- 210000002304 ESC Anatomy 0.000 description 1
- 208000005679 Eczema Diseases 0.000 description 1
- 230000035695 Efflux Effects 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 208000000999 Encephalomyelitis, Autoimmune, Experimental Diseases 0.000 description 1
- 206010060742 Endocrine ophthalmopathy Diseases 0.000 description 1
- 210000001163 Endosomes Anatomy 0.000 description 1
- 241000792859 Enema Species 0.000 description 1
- 229940079360 Enema for Constipation Drugs 0.000 description 1
- 206010014950 Eosinophilia Diseases 0.000 description 1
- 210000003979 Eosinophils Anatomy 0.000 description 1
- 206010014989 Epidermolysis bullosa Diseases 0.000 description 1
- 206010015150 Erythema Diseases 0.000 description 1
- 210000003238 Esophagus Anatomy 0.000 description 1
- 208000004332 Evans Syndrome Diseases 0.000 description 1
- 229920000665 Exon Polymers 0.000 description 1
- 108010074860 Factor Xa Proteins 0.000 description 1
- 108010067306 Fibronectins Proteins 0.000 description 1
- 102000016359 Fibronectins Human genes 0.000 description 1
- 102100011343 GLB1 Human genes 0.000 description 1
- 229940116332 GLUCOSE OXIDASE Drugs 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 206010017860 Gastritis atrophic Diseases 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 210000001654 Germ Layers Anatomy 0.000 description 1
- 206010018364 Glomerulonephritis Diseases 0.000 description 1
- 239000004366 Glucose oxidase Substances 0.000 description 1
- 229960002989 Glutamic Acid Drugs 0.000 description 1
- RWSXRVCMGQZWBV-WDSKDSINSA-N Glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 1
- 229960003180 Glutathione Drugs 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 206010018620 Goodpasture's syndrome Diseases 0.000 description 1
- 206010048748 Graft loss Diseases 0.000 description 1
- 206010018651 Graft versus host disease Diseases 0.000 description 1
- 208000009329 Graft vs Host Disease Diseases 0.000 description 1
- 210000003714 Granulocytes Anatomy 0.000 description 1
- 206010018691 Granuloma Diseases 0.000 description 1
- 108060005986 Granzymes Proteins 0.000 description 1
- 102000001398 Granzymes Human genes 0.000 description 1
- UYTPUPDQBNUYGX-UHFFFAOYSA-N Guanine Chemical class O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 1
- 208000002183 Guillain-Barre Syndrome Diseases 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 101710043924 HERVK_113 Proteins 0.000 description 1
- 208000005721 HIV Infections Diseases 0.000 description 1
- 102100016790 HPRT1 Human genes 0.000 description 1
- 229920001364 Hairpin ribozyme Polymers 0.000 description 1
- 229920001941 Hammerhead ribozyme Polymers 0.000 description 1
- 210000002216 Heart Anatomy 0.000 description 1
- 206010019280 Heart failure Diseases 0.000 description 1
- 206010003820 Hepatic autoimmune disease Diseases 0.000 description 1
- 206010019692 Hepatic necrosis Diseases 0.000 description 1
- 208000002672 Hepatitis B Diseases 0.000 description 1
- 206010019755 Hepatitis chronic active Diseases 0.000 description 1
- 206010019772 Hepatitis fulminant Diseases 0.000 description 1
- 206010019799 Hepatitis viral Diseases 0.000 description 1
- 210000003494 Hepatocytes Anatomy 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 208000005100 Herpetic Keratitis Diseases 0.000 description 1
- 108010093488 His-His-His-His-His-His Proteins 0.000 description 1
- 229940088597 Hormone Drugs 0.000 description 1
- 241000725303 Human immunodeficiency virus Species 0.000 description 1
- 102000015434 Humanized Monoclonal Antibodies Human genes 0.000 description 1
- 108010064750 Humanized Monoclonal Antibodies Proteins 0.000 description 1
- 201000001971 Huntington's disease Diseases 0.000 description 1
- 206010048643 Hypereosinophilic syndrome Diseases 0.000 description 1
- 208000000038 Hypoparathyroidism Diseases 0.000 description 1
- 229960000310 ISOLEUCINE Drugs 0.000 description 1
- 210000001822 Immobilized Cells Anatomy 0.000 description 1
- 108010027440 Immunoconjugates Proteins 0.000 description 1
- 102000018748 Immunoconjugates Human genes 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 231100000608 Immunotoxin Toxicity 0.000 description 1
- 108010004484 Immunotoxins Proteins 0.000 description 1
- 206010021972 Inflammatory bowel disease Diseases 0.000 description 1
- 208000007906 Intestinal Disease Diseases 0.000 description 1
- 241000334154 Isatis tinctoria Species 0.000 description 1
- 210000004153 Islets of Langerhans Anatomy 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
- 210000003734 Kidney Anatomy 0.000 description 1
- 208000001083 Kidney Disease Diseases 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-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
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-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
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- 125000000510 L-tryptophano group Chemical group [H]C1=C([H])C([H])=C2N([H])C([H])=C(C([H])([H])[C@@]([H])(C(O[H])=O)N([H])[*])C2=C1[H] 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
- 229920001320 Leader sequence (mRNA) Polymers 0.000 description 1
- 229940067606 Lecithin Drugs 0.000 description 1
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 1
- 240000007472 Leucaena leucocephala Species 0.000 description 1
- 206010024324 Leukaemias Diseases 0.000 description 1
- 102000019298 Lipocalins Human genes 0.000 description 1
- 108050006654 Lipocalins Proteins 0.000 description 1
- HWYHZTIRURJOHG-UHFFFAOYSA-N Luminol Chemical compound O=C1NNC(=O)C2=C1C(N)=CC=C2 HWYHZTIRURJOHG-UHFFFAOYSA-N 0.000 description 1
- 102000008072 Lymphokines Human genes 0.000 description 1
- 108010074338 Lymphokines Proteins 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 210000002540 Macrophages Anatomy 0.000 description 1
- 235000019759 Maize starch Nutrition 0.000 description 1
- 210000003593 Megakaryocytes Anatomy 0.000 description 1
- 206010027183 Meniere's disease Diseases 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 206010027476 Metastasis Diseases 0.000 description 1
- 229960004452 Methionine Drugs 0.000 description 1
- LXCFILQKKLGQFO-UHFFFAOYSA-N Methylparaben Chemical group COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 210000001616 Monocytes Anatomy 0.000 description 1
- 201000009262 Mooren's ulcer Diseases 0.000 description 1
- 108090000143 Mouse Proteins Proteins 0.000 description 1
- 210000000214 Mouth Anatomy 0.000 description 1
- 206010028116 Mucosal inflammation Diseases 0.000 description 1
- 206010028417 Myasthenia gravis Diseases 0.000 description 1
- 229960000951 Mycophenolic Acid Drugs 0.000 description 1
- 210000004413 Myocytes, Cardiac Anatomy 0.000 description 1
- 102000002673 NFATC Transcription Factors Human genes 0.000 description 1
- 108010018525 NFATC Transcription Factors Proteins 0.000 description 1
- 102100000563 NFATC1 Human genes 0.000 description 1
- 101710044747 NFATC1 Proteins 0.000 description 1
- 102100000559 NFATC2 Human genes 0.000 description 1
- 101710044739 NFATC2 Proteins 0.000 description 1
- 108091007229 NSP3 Papain-like protease domain Proteins 0.000 description 1
- 229920002957 Naked DNA Polymers 0.000 description 1
- 206010051606 Necrotising colitis Diseases 0.000 description 1
- 208000009928 Nephrosis Diseases 0.000 description 1
- 108009000551 Nephrotic syndrome Proteins 0.000 description 1
- 206010053643 Neurodegenerative disease Diseases 0.000 description 1
- 210000000440 Neutrophils Anatomy 0.000 description 1
- 240000008962 Nicotiana tabacum Species 0.000 description 1
- 235000002637 Nicotiana tabacum Nutrition 0.000 description 1
- 210000001331 Nose Anatomy 0.000 description 1
- GEYOCULIXLDCMW-UHFFFAOYSA-N O-Phenylenediamine Chemical compound NC1=CC=CC=C1N GEYOCULIXLDCMW-UHFFFAOYSA-N 0.000 description 1
- 206010067776 Ocular pemphigoid Diseases 0.000 description 1
- 108020005187 Oligonucleotide Probes Proteins 0.000 description 1
- 206010073938 Ophthalmic herpes simplex Diseases 0.000 description 1
- 101710040922 Os08g0547100 Proteins 0.000 description 1
- 206010025310 Other lymphomas Diseases 0.000 description 1
- 101710039569 POLM Proteins 0.000 description 1
- 102100019685 PRKCG Human genes 0.000 description 1
- 101710038826 PRKCG Proteins 0.000 description 1
- 241000282577 Pan troglodytes Species 0.000 description 1
- 210000000496 Pancreas Anatomy 0.000 description 1
- 206010061536 Parkinson's disease Diseases 0.000 description 1
- 241000237988 Patellidae Species 0.000 description 1
- 206010034277 Pemphigoid Diseases 0.000 description 1
- 241000721454 Pemphigus Species 0.000 description 1
- 229940049954 Penicillin Drugs 0.000 description 1
- 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 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 102000035443 Peptidases Human genes 0.000 description 1
- 108091005771 Peptidases Proteins 0.000 description 1
- 108010067902 Peptide Library Proteins 0.000 description 1
- 229940072417 Peroxidase Drugs 0.000 description 1
- 108090000437 Peroxidases Proteins 0.000 description 1
- 102000003992 Peroxidases Human genes 0.000 description 1
- 229960005190 Phenylalanine Drugs 0.000 description 1
- 108091000081 Phosphotransferases Proteins 0.000 description 1
- 108010004729 Phycoerythrin Proteins 0.000 description 1
- 241000235648 Pichia Species 0.000 description 1
- 241000255972 Pieris <butterfly> Species 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 208000005987 Polymyositis Diseases 0.000 description 1
- 208000006994 Precancerous Condition Diseases 0.000 description 1
- 229960002429 Proline Drugs 0.000 description 1
- QELSKZZBTMNZEB-UHFFFAOYSA-N Propylparaben Chemical compound CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 102000001253 Protein Kinases Human genes 0.000 description 1
- 206010037162 Psoriatic arthropathy Diseases 0.000 description 1
- 206010037575 Pustular psoriasis Diseases 0.000 description 1
- 241001026602 Quintana Species 0.000 description 1
- 101710006375 RNASEH1 Proteins 0.000 description 1
- 101700078434 RT67 Proteins 0.000 description 1
- 206010037779 Radiculopathy Diseases 0.000 description 1
- 241000700157 Rattus norvegicus Species 0.000 description 1
- 108010033725 Recombinant Proteins Proteins 0.000 description 1
- 102000007312 Recombinant Proteins Human genes 0.000 description 1
- 241000242739 Renilla Species 0.000 description 1
- 208000002200 Respiratory Hypersensitivity Diseases 0.000 description 1
- 206010039073 Rheumatoid arthritis Diseases 0.000 description 1
- 208000003385 Rhinitis, Allergic, Seasonal Diseases 0.000 description 1
- 102000006382 Ribonucleases Human genes 0.000 description 1
- 108010083644 Ribonucleases Proteins 0.000 description 1
- 229920001914 Ribonucleotide Polymers 0.000 description 1
- 241000724205 Rice stripe tenuivirus Species 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 108060007909 STB3 Proteins 0.000 description 1
- 241000235070 Saccharomyces Species 0.000 description 1
- 206010039705 Scleritis Diseases 0.000 description 1
- 206010039710 Scleroderma Diseases 0.000 description 1
- 208000010157 Sclerosing Cholangitis Diseases 0.000 description 1
- 206010048908 Seasonal allergy Diseases 0.000 description 1
- 208000008742 Seborrheic Dermatitis Diseases 0.000 description 1
- 206010039793 Seborrhoeic dermatitis Diseases 0.000 description 1
- 229920000972 Sense strand Polymers 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 206010040767 Sjogren's syndrome Diseases 0.000 description 1
- 210000004927 Skin cells Anatomy 0.000 description 1
- 229940054269 Sodium Pyruvate Drugs 0.000 description 1
- 229940075582 Sorbic Acid Drugs 0.000 description 1
- 241000251131 Sphyrna Species 0.000 description 1
- 229940032147 Starch Drugs 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 208000007107 Stomach Ulcer Diseases 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 229960005322 Streptomycin Drugs 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 229960002180 Tetracycline Drugs 0.000 description 1
- OFVLGDICTFRJMM-WESIUVDSSA-N Tetracycline Chemical compound C1=CC=C2[C@](O)(C)[C@H]3C[C@H]4[C@H](N(C)C)C(O)=C(C(N)=O)C(=O)[C@@]4(O)C(O)=C3C(=O)C2=C1O OFVLGDICTFRJMM-WESIUVDSSA-N 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- FQENQNTWSFEDLI-UHFFFAOYSA-J Tetrasodium pyrophosphate Chemical compound [Na+].[Na+].[Na+].[Na+].[O-]P([O-])(=O)OP([O-])([O-])=O FQENQNTWSFEDLI-UHFFFAOYSA-J 0.000 description 1
- 229920000401 Three prime untranslated region Polymers 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 108090000190 Thrombin Proteins 0.000 description 1
- 208000007536 Thrombosis Diseases 0.000 description 1
- 229960002175 Thyroglobulin Drugs 0.000 description 1
- 108010034949 Thyroglobulin Proteins 0.000 description 1
- 102000009843 Thyroglobulin Human genes 0.000 description 1
- 241000723873 Tobacco mosaic virus Species 0.000 description 1
- 231100000765 Toxin Toxicity 0.000 description 1
- 210000003437 Trachea Anatomy 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- CYRMSUTZVYGINF-UHFFFAOYSA-N Trichlorofluoromethane Chemical compound FC(Cl)(Cl)Cl CYRMSUTZVYGINF-UHFFFAOYSA-N 0.000 description 1
- 229960004799 Tryptophan Drugs 0.000 description 1
- 206010048302 Tubulointerstitial nephritis Diseases 0.000 description 1
- 208000001072 Type 2 Diabetes Mellitus Diseases 0.000 description 1
- 108009000112 Type II diabetes mellitus Proteins 0.000 description 1
- 108090000848 Ubiquitin Proteins 0.000 description 1
- 102400000757 Ubiquitin Human genes 0.000 description 1
- 206010064996 Ulcerative keratitis Diseases 0.000 description 1
- 210000002700 Urine Anatomy 0.000 description 1
- 206010046736 Urticarias Diseases 0.000 description 1
- 206010047112 Vasculitides Diseases 0.000 description 1
- 206010047115 Vasculitis Diseases 0.000 description 1
- 241000269370 Xenopus <genus> Species 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 230000004721 adaptive immunity Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000001058 adult Effects 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 230000000172 allergic Effects 0.000 description 1
- 230000000735 allogeneic Effects 0.000 description 1
- 239000008168 almond oil Substances 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminum Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 230000003466 anti-cipated Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 230000000890 antigenic Effects 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 229920002847 antisense RNA Polymers 0.000 description 1
- 239000008135 aqueous vehicle Substances 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 201000001320 atherosclerosis Diseases 0.000 description 1
- 230000002238 attenuated Effects 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 229960000626 benzylpenicillin Drugs 0.000 description 1
- 102000005936 beta-Galactosidase Human genes 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 239000000560 biocompatible material Substances 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000003995 blood forming stem cell Anatomy 0.000 description 1
- YYRMJZQKEFZXMX-UHFFFAOYSA-L calcium bis(dihydrogenphosphate) Chemical compound [Ca+2].OP(O)([O-])=O.OP(O)([O-])=O YYRMJZQKEFZXMX-UHFFFAOYSA-L 0.000 description 1
- 239000000292 calcium oxide Substances 0.000 description 1
- 230000028956 calcium-mediated signaling Effects 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- CURLTUGMZLYLDI-UHFFFAOYSA-N carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000004956 cell adhesive effect Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 230000004700 cellular uptake Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 235000010980 cellulose Nutrition 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 239000003593 chromogenic compound Substances 0.000 description 1
- 201000010002 cicatricial pemphigoid Diseases 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 230000023298 conjugation with cellular fusion Effects 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 230000001808 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 230000001461 cytolytic Effects 0.000 description 1
- 230000004059 degradation Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 239000003405 delayed action preparation Substances 0.000 description 1
- 238000006209 dephosphorylation reaction Methods 0.000 description 1
- 231100000080 dermatitis contact Toxicity 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 229940042935 dichlorodifluoromethane Drugs 0.000 description 1
- 235000019404 dichlorodifluoromethane Nutrition 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- BFMYDTVEBKDAKJ-UHFFFAOYSA-L disodium;(2',7'-dibromo-3',6'-dioxido-3-oxospiro[2-benzofuran-1,9'-xanthene]-4'-yl)mercury;hydrate Chemical compound O.[Na+].[Na+].O1C(=O)C2=CC=CC=C2C21C1=CC(Br)=C([O-])C([Hg])=C1OC1=C2C=C(Br)C([O-])=C1 BFMYDTVEBKDAKJ-UHFFFAOYSA-L 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 230000002222 downregulating Effects 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 238000009510 drug design Methods 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 238000005538 encapsulation Methods 0.000 description 1
- 239000007920 enema Substances 0.000 description 1
- 230000002255 enzymatic Effects 0.000 description 1
- 230000002327 eosinophilic Effects 0.000 description 1
- 210000002919 epithelial cells Anatomy 0.000 description 1
- 231100000321 erythema Toxicity 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 239000003925 fat Substances 0.000 description 1
- 235000019197 fats Nutrition 0.000 description 1
- 150000002194 fatty esters Chemical class 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 230000037320 fibronectin Effects 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 238000001215 fluorescent labelling Methods 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 230000000799 fusogenic Effects 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- 239000003862 glucocorticoid Substances 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 235000019420 glucose oxidase Nutrition 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 235000004554 glutamine Nutrition 0.000 description 1
- SXRSQZLOMIGNAQ-UHFFFAOYSA-N glutaraldehyde Chemical compound O=CCCCC=O SXRSQZLOMIGNAQ-UHFFFAOYSA-N 0.000 description 1
- 125000005456 glyceride group Chemical group 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 230000003394 haemopoietic Effects 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 201000005787 hematologic cancer Diseases 0.000 description 1
- 108060003552 hemocyanin family Proteins 0.000 description 1
- 230000002949 hemolytic Effects 0.000 description 1
- 125000003372 histidine group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C([H])=N1 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 1
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- 201000003838 idiopathic interstitial pneumonia Diseases 0.000 description 1
- 238000005286 illumination Methods 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000010324 immunological assay Methods 0.000 description 1
- 230000002637 immunotoxin Effects 0.000 description 1
- 239000002596 immunotoxin Substances 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000004073 interleukin-2 production Effects 0.000 description 1
- 201000006334 interstitial nephritis Diseases 0.000 description 1
- 238000010255 intramuscular injection Methods 0.000 description 1
- 239000007927 intramuscular injection Substances 0.000 description 1
- 201000010659 intrinsic asthma Diseases 0.000 description 1
- 238000009114 investigational therapy Methods 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- 201000010666 keratoconjunctivitis Diseases 0.000 description 1
- 201000002287 keratoconus Diseases 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 201000010260 leiomyoma Diseases 0.000 description 1
- 231100000636 lethal dose Toxicity 0.000 description 1
- 229960003136 leucine Drugs 0.000 description 1
- 150000002617 leukotrienes Chemical class 0.000 description 1
- 201000011486 lichen planus Diseases 0.000 description 1
- 230000000670 limiting Effects 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 231100000149 liver necrosis Toxicity 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 230000002132 lysosomal Effects 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 229920001239 microRNA Polymers 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 239000003094 microcapsule Substances 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 235000010755 mineral Nutrition 0.000 description 1
- 230000011278 mitosis Effects 0.000 description 1
- 239000003607 modifier Substances 0.000 description 1
- 238000001823 molecular biology technique Methods 0.000 description 1
- 235000019691 monocalcium phosphate Nutrition 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 210000000663 muscle cells Anatomy 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 229960004866 mycophenolate mofetil Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 229930014626 natural products Natural products 0.000 description 1
- 239000006199 nebulizer Substances 0.000 description 1
- 230000017074 necrotic cell death Effects 0.000 description 1
- 230000001613 neoplastic Effects 0.000 description 1
- 201000008383 nephritis Diseases 0.000 description 1
- 231100001027 nephrosis Toxicity 0.000 description 1
- 230000001537 neural Effects 0.000 description 1
- 230000000626 neurodegenerative Effects 0.000 description 1
- 230000001264 neutralization Effects 0.000 description 1
- 239000002687 nonaqueous vehicle Substances 0.000 description 1
- 231100000956 nontoxicity Toxicity 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 238000007899 nucleic acid hybridization Methods 0.000 description 1
- 230000000414 obstructive Effects 0.000 description 1
- 239000002751 oligonucleotide probe Substances 0.000 description 1
- 210000000056 organs Anatomy 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 239000012071 phase Substances 0.000 description 1
- 239000002644 phorbol ester Substances 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 239000002985 plastic film Substances 0.000 description 1
- 229920001983 poloxamer Polymers 0.000 description 1
- 230000001402 polyadenylating Effects 0.000 description 1
- 229920000447 polyanionic polymer Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 229920001592 potato starch Polymers 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- OZAIFHULBGXAKX-UHFFFAOYSA-N precursor Substances N#CC(C)(C)N=NC(C)(C)C#N OZAIFHULBGXAKX-UHFFFAOYSA-N 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000001737 promoting Effects 0.000 description 1
- 230000000644 propagated Effects 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- 239000004405 propyl p-hydroxybenzoate Substances 0.000 description 1
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 201000004681 psoriasis Diseases 0.000 description 1
- 201000001263 psoriatic arthritis Diseases 0.000 description 1
- 238000000163 radioactive labelling Methods 0.000 description 1
- 230000036647 reaction Effects 0.000 description 1
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 1
- 230000027425 release of sequestered calcium ion into cytosol Effects 0.000 description 1
- 230000000268 renotropic Effects 0.000 description 1
- 238000002271 resection Methods 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 201000003068 rheumatic fever Diseases 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 101700086982 rnh Proteins 0.000 description 1
- 201000000306 sarcoidosis Diseases 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 239000006152 selective media Substances 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 229940048086 sodium pyrophosphate Drugs 0.000 description 1
- 229940079832 sodium starch glycolate Drugs 0.000 description 1
- 239000008109 sodium starch glycolate Substances 0.000 description 1
- 229920003109 sodium starch glycolate Polymers 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- WSWCOQWTEOXDQX-UHFFFAOYSA-N sorbic acid Chemical compound CC=CC=CC(O)=O WSWCOQWTEOXDQX-UHFFFAOYSA-N 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 235000010356 sorbitol Nutrition 0.000 description 1
- 230000002269 spontaneous Effects 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000003892 spreading Methods 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 230000000087 stabilizing Effects 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 230000004936 stimulating Effects 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000002511 suppository base Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 230000005700 syncytium formation by plasma membrane fusion Effects 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 235000019818 tetrasodium diphosphate Nutrition 0.000 description 1
- 239000001577 tetrasodium phosphonato phosphate Substances 0.000 description 1
- 235000008521 threonine Nutrition 0.000 description 1
- 229960004072 thrombin Drugs 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 150000004992 toluidines Chemical class 0.000 description 1
- 230000000699 topical Effects 0.000 description 1
- 230000002588 toxic Effects 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 108020003112 toxins Proteins 0.000 description 1
- 230000005026 transcription initiation Effects 0.000 description 1
- 238000005820 transferase reaction Methods 0.000 description 1
- 230000001131 transforming Effects 0.000 description 1
- 230000001052 transient Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 108091005683 transmembrane proteins Proteins 0.000 description 1
- 102000035402 transmembrane proteins Human genes 0.000 description 1
- 229940029284 trichlorofluoromethane Drugs 0.000 description 1
- 238000007039 two-step reaction Methods 0.000 description 1
- 235000002374 tyrosine Nutrition 0.000 description 1
- 201000006704 ulcerative colitis Diseases 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 229960004295 valine Drugs 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 230000003966 vascular damage Effects 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 201000005539 vernal conjunctivitis Diseases 0.000 description 1
- 201000001862 viral hepatitis Diseases 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 239000011534 wash buffer Substances 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Abstract
The invention discloses the first known function and biological activity of the hypothetical protein MGC14327, now designated cMAC, which is herein identified as an important controller of T-cell activation. It is contemplated herein that cMAC is a suitable drug target for the development of new therapeutics to treat cMAC-associated disorders. The invention relates to methods to treat said pathological conditions and to pharmaceutical compositions therefore. The pharmaceutical compositions comprise modulators with inhibitory or agonist effect on cMAC protein activity and/or cMAC gene expression. The invention also relates to methods to identify compounds with therapeutic usefulness to treat said pathological conditions, comprising identifying compounds that can inhibit or agonize cMAC protein activity and/or cMAC gene expression.
Description
ACTIVATOR OF MEMBRANE OF CALCINEURIN TO PRESERVED IMAC), A NOVEL THERAPEUTIC PROTEIN AND OBJECTIVE
BACKGROUND OF THE INVENTION There is much evidence to support the notion that T lymphocytes are required for the advancement of systemic autoimmune disease. The simple reduction in the number of T cells reduces the host immune response and improves survival in animal models of autoimmune disease. For example, in a murine model of spontaneous lupus, the reduction of T cells with anti-T cell antibodies reduced circulating T cells, decreased autoantibody levels, reduced renal complications and improved survival of animals (Wofsy D, Ledbetter JA, Hendler PL, Seaman WE (1985) Treatment of murine lupus with monoclonal anti-T cell antibody, (Treatment of murine lupus with anti-T cell monoclonal antibody), J Immunol Feb. 134 (2): 852 -7). Antibodies that block T-cell co-stimulatory receptors are also effective in prolonging animal survival (Finck BK, Linsley PS, Wofsy D. (1994) Treatment of murine lupus with CTLA4lg (Treatment of murine lupus with CTLIA4lg) Science. Aug 26; 285 (5176): 1225-7). Drugs that reduce the proliferation or activation of T cells are effective in treating lupus in humans (cyclophosphamide, mycophenolate mofetil and cyclosporin A). More evidence of support than T cells
involved in the autoimmune disease in humans comes from the observation that lupus patients with HIV infections may experience remission due to the reduction of CD4 + T cells (Byrd VM, Sergent JS. (1996) Suppression of systemic lupus erythematosus by the human immunodeficiency virus J Rheumatol Jul; 23 (7): 1295-6.) T lymphocytes also play an important role in acute rejection of grafts. The acute rejection of grafts is classically preceded by the migration of T cells into the graft, the clonal expansion of activated T cells, the proliferation of cytotoxic T cells and the subsequent destruction of tissue and graft loss. The ability of athymic mice to indefinitely accept grafts from other species demonstrates the particular importance that T cells represent in the rejection of grafts. Likewise, drugs that block T cell responses or eliminate all T cells are effective in preventing rejection of grafts in humans (Sykes M, Uchincloss H, Sachs DH (2003) "Transplantation immunology", in Fundamental Immunology, ed WE Paul, Lippincott Williams &Wilkins, Philadelphia, p 1499.) Activation of natural T cells requires stimulation of the T cell receptor (TCR) and a co-stimulator receptor (CD28). The TCR / CD28 binding activates a complex cascade of signaling that causes an increase in intracellular calcium through the release of calcium stores
intracellular and the subsequent influx of calcium through the current calcium channel activated by calcium release (CRAC). The increase in intracellular calcium results in the activation of calcineurin which dephosphorylates NFAT. NFAT proteins are a family of transcription factors that, once dephosphorylated, are transposed in the nucleus where they carry the transcription of one of the most important lymphokines in cellular rejection, IL-2 (Hutchinson I, (2002) "Transplantation and rejection" in Immunology, I Roitt, J Brostoff and D Male, Mosby New York P. 389.). IL-2 stimulates the proliferation of cytotoxic T cells, which release cytolytic, performin and granzyme molecules, which mediate the destruction of the graft. The immunosuppressive drug cyclosporin A inhibits the enzyme phosphatase calcineurin that prevents transposition of NFAT in the nucleus and thus prevents the transcription of IL-2. The TORC proteins are CREB coactivators which, like the NFAT, are transposed in the nucleus in response to the mobilization of the intracellular calcium stores. It is believed that TORCs facilitate gene expression mediated by CRE. The proteins that regulate NFAT and / or TORC may be convenient targets for therapeutic intervention. Applicants report that cMAC is a potent regulator of T cell activation and regulates the nuclear transposition of NFAT and TORC. BRIEF DESCRIPTION OF THE INVENTION The present description refers to the discovery that
a protein with a previously unknown function, called in the present cMAC ("Calcineurin Membrane Conserved"), is a potent regulator of T cell activation, and participates in the calcium-mediated nuclear transposition of NFAT and TORC1. As such, cMAC is a therapeutic protein and an important therapeutic target for the treatment of diseases associated with cMAC (defined herein), using small molecules, antibodies, nucleic acids and other therapeutic agents that modulate the activity or expression of cMAC. In one aspect the invention relates to the mature or original cMAC polypeptide. In accordance with the foregoing, the invention relates to the polypeptide isolated from SEQ ID NO: 2, or a fragment thereof, or a substantially similar protein sequence having a sequence identity of at least 50 percent with SEQ ID NO: 2, or a functional equivalent thereof, and exhibiting a biological activity selected from ion transport, ion diffusion, activation of the calcineurin pathway, calcium-dependent activation of a T cell, nuclear transposition of TORC, nuclear transposition of NFAT or gene expression activity driven by CRE (cAMP Response Element) of the original SEQ ID NO: 2. In other aspects, the invention comprises an isolated nucleic acid molecule encoding cMAC, a vector comprising the nucleic acid molecule, preferably a vector
of expression comprising the nucleic acid molecule operably linked to a promoter, a host cell comprising the vector molecule, including mammalian and bacterial host cells, and a method of using a nucleic acid molecule encoding cMAC to effect the production of cMAC, which comprises culturing a host cell comprising the vector molecule. Another aspect of the invention provides an antibody or antibody fragment that is capable of binding the cMAC polypeptide of the invention. Yet another aspect of the invention relates to an RNA agent capable of down-regulating the expression of cMAC, preferably that RNA agent comprises at least one nucleic acid selected from Table 5 or Table 6. Other aspects of the invention relates to the use of an antibody or RNA agent according to the invention for the manufacture of a medicament for the treatment of a disorder associated with cMAC (as defined herein) and with a method for treating a disorder in a subject comprising administering to the subject an effective amount of an agent that modulates the amount or activity of cMAC. In accordance with the foregoing, the invention comprises the use of an antibody, an antibody fragment or a polypeptide comprising a cMAC specific binding region in the treatment of a disorder in a subject, especially wherein the disorder is an associated disorder with cMAC. Alternatively, the invention comprises the use of an iRNA agent or
SiRNA specific for cMAC in the treatment of a disorder in a subject, especially wherein the disorder is a disorder associated with cMAC. In several aspects, this method comprises administering an agent that inhibits the expression of cMAC, wherein the disorder is a disorder associated with cMAC (as defined herein), or wherein the agent is an antibody, an antibody fragment or a polypeptide that contains a specific binding region of cMAC. Optionally the agent is administered as a pharmaceutical composition. In another aspect, this method comprises administering to the subject an effective amount of an agent that inhibits the expression of cMAC. This agent includes an inhibitory nucleic acid capable of specifically inhibiting the expression of cMAC. Different modalities include that the inhibitory nucleic acid is selected from the group consisting of an antisense oligonucleotide, an RNA agent, and a ribozyme, DsRNA, siRNA, and shRNA. Optionally the agent is administered as a pharmaceutical composition. In another aspect, the invention comprises a method for treating a disorder in a subject, comprising administering to the subject an effective amount of an agent that enhances the activity of cMAC. This method includes a method comprising administering to the subject an effective amount of an agent that increases the expression of cMAC, for example where the agent is a gene therapy vector comprising a nucleic acid encoding cMAC or a fragment of the
same, or where the agent is a transcription enhancer of the cMAC gene. In terms of the compositions, the invention comprises an antibody or antibody fragment that specifically binds to cMAC (SEQ ID NO 2), and any polypeptide comprising a specific binding region with cMAC. Such an antibody includes an antibody fragment that is a Fab or an F (ab ') 2 fragment, or wherein the antibody is a monoclonal antibody. The invention includes a pharmaceutical composition comprising an effective amount of an agent that inhibits the expression of cMAC or inhibits a cMAC activity, and a pharmaceutically acceptable carrier. That agent may be an antisense oligonucleotide, an RNA agent, an antibody fragment, an antibody fragment that specifically binds to cMAC, or a polypeptide comprising a specific binding region for cMAC. In a preferred embodiment, the pharmaceutical composition comprises an antibody or an antibody fragment that specifically binds to cMAC. (SEQ ID NO.2), or any polypeptide comprising a cMAC-specific binding region, which binds to a cMAC epitope selected from SEQ ID NOs.6, 7, 8, 9, 10. The invention also comprises a method for treating a disorder in a subject, comprising administering to the subject an effective amount of a pharmaceutical composition of an agent that inhibits the activity of cMAC, especially wherein the disorder is a disorder associated with cMAC, and wherein the agent is a
antibody or a fragment thereof that specifically binds to cMAC (SEQ ID NO.2) or a polypeptide comprising a specific binding region with cMAC. The agent optionally binds to a cMAC epitope selected from SEQ ID No. 6, 7, 8, 9, 10. The invention further comprises screening test methods to identify a compound useful for the treatment of a disorder associated with cMAC. comprising: (a) contacting a test compound with cMAC; and (b) detecting a change in a biological activity of cMAC compared to cMAC that is not in contact with the test compound, wherein detecting a change identifies the test compound as being useful for the treatment of that disorder. Similarly, the invention comprises filtering test methods for identifying a compound useful for the treatment of a cMAC-associated disorder comprising: (a) contacting a test compound with cMAC under permissible sample conditions for biological activity from cMAC; (b) determine the level of a biological activity of cMAC; (c) comparing that level with that of a control sample that lacks the test compound; and (d) selecting a test compound that causes the level to change to later prove it as a potential agent for the treatment of that disorder. The invention comprises a method for testing whether a compound modulates a biological activity of cMAC comprising: (a) contacting a test compound with cMAC; and (b) detect a change of one
biological activity of cMAC compared to cMAC that had no contact with the test compound, wherein detecting a change identifies that test compound as a modulator of the biological activity of cMAC. Similarly, the invention comprises a method for identifying modulators useful for treating a disorder, comprising testing the ability of a candidate modulator to inhibit the activity of a cMAC protein; and a method for identifying modulators useful for treating a disorder comprising testing the ability of a candidate modulator to inhibit the expression of a cMAC protein. In these methods the change or modulation may be a reduction or an increase in biological activity. In addition, the biological activity can be selected from the group of ion transport, ion diffusion, interaction of cMAC with proteins or modification of cMAC, calcium dependent activation of a T cell, nuclear transposition of TORC, and gene expression guided by the response element (CRE) of cAMP. In accordance with the invention, disorders related to cMAC, or associated with cMAC include, but are not limited to, autoimmune disease, immunosuppression, inflammatory disease, cancer, cardiovascular disease and neurological disease. Description of the Figures Figure 1. cMAC is a predicted integral membrane protein. The prediction of the transmembrane domain of cMAC is ordered in series using the TMHMM algorithm. Two small
Predicted extracellular domains are located in amino acids
36-49 and 101-110. Figure 2. cMAC is a highly conserved protein. The ClustalW alignment of vertebrate proteins with similarity to cMAC. The potential transmembrane helices predicted by the TMHMM algorithm are indicated by lines. Figure 3. cMAC mRNA levels measured by profiling Affymetrix expression. Figure 4. Over-expression of cMAC induces the translocation of TORC in HEK293 cells. Bittenger and collaborators identified TRPV6 and PKA as hits in the TORC-eGFP transposition filter (Bittenger et al., Curr Biol. 2004 Dec.
14; 14 (23): 2156-61). CMAC was also identified although it was not previously described. Figure 5. The cMAC mediated transposition of TORC1-eGFP is blocked with the calcineurin inhibitor CsA. In panel A HeLa cells: TORC1-eGFP were transduced with the stop codon virus (vector), or human TRPV6, or the human CMAC virus (50 microliters). Panel B The cells were treated as in A except the cells were treated with 5 uM of cyclosporin A (CsA) for 1 hour before fixation. Figure 6. cMAC induces NFAT-dependent transcription. The HEK293 cells were co-transfected with a NFAT-luciferase reporter plasmid, the transfection control, and the following constructs: Empty vector (CMV), TRPV6 and cMAC and
treated with either DMSO, 5μ? of CsA, 10 μ? of PMA or 10μ? of PMA and 5μ? of CsA. Figure 7. cMAC induces the transposition of NFAT1 in Jurkat cells. Panel A. Over-expression of cMAC mediated by Lentiviral, control vector (translation stop sequence), and TRPV6 with and without cyclosporin A; B. The same treatment as in A except the cells were sensitized with PMA 6 hours before fixation. Figure 8. cMAC induces the transposition of NFAT2 in Jurkat cells. A. Over-expression of cMAC mediated by viral (pLLB1 -GW-Kan), control vector (translation stop sequence), TRPV6 with and without cyclosporin; B The same treatment as in A except cells sensitized with PMA 6 hours before fixation. Figure 9. The over-expression of murine cMAC and the human homologue activates Jurkat T cells. Jurkat cells were transduced with the viral expression vector (QL-GW-final-Kan) containing the empty negative control vector (translation stop sequence). The calcium channel TRPV6, and NM_177244 (murine cMAC) and NM_053045 (human cMAC). The IL-2 protein (ELISA) and the expression of the ICOS surface marker were measured 72 hours after transduction (48 hours after transduction the cells were sensitized with PMA and the anti-TCR antibody). Figure 10. Multiple viral ssDNA sequences directing the cMAC block to TCR / CD28 activation of Jurkat T cells. The cells were transduced with viral constructions
(pLKO.1), were selected with puromycin and activated with TCR / CD28 6 days after transduction. The levels of the IL-2 protein were measured and normalized by the number of viable cells present in each well. The construction of ssDNA pGL3-Luc served as the negative control ssDNA for viral transductions. Figure 11. Human cMAC: NM_053045: hypothetical Homo sapiens protein MGC14327 (MGC14327), mRNA (gi | 16596685 | ref | NM_053045.1 | [16596685]) (SEQ ID NO: 1) and human hypothetical protein LOC94107 | [ Homo sapiens; > gi | 16596686 | ref | NP_444273.11] (SEQ ID NO: 2). Figure 12. Genomic promoter sequence of human cMAC (NM_053045.1_5 '_- 3000 + 100NT_024000.16886093882993) (SEQ ID NO: 3). Figure 13. Isolated nucleic acid sequence for 5'UTR of cMAC (SEQ ID NO: 4). and isolated nucleic acid sequence for the 3'UTR of cMAC (SEQ ID NO: 5). Figure 14. murine cMAC NM_177344: Mus musculus RIKEN cDNA C730025P13 gene (C730025P13Rik), mRNA (gi | 31340922 | ref | NM_177344.2 |) (SEQ ID NO: 11) and
> gi | 18490941 | gb | BC22606.11 Mus musculus RIKEN cDNA C730025P13 gene, mRNA (cDNA clone MGC: 31129 IMAGE: 4165766) complete cds (SEQ ID N012) and mouse cMAC amino acid sequence translated from NM_177344.2 (SEQ ID NO: 13 ).
Figure 15. Other orthologs of human cMAC from other species: Mus musculus (SEQ ID NO: 14); Rattus norvegicus (SEQ ID NO: 15); Canis familiaris (SEQ ID NO: 16); Pan troglodytes (SEQ ID NO: 17); Xenopus tropicalis (SEQ ID NO: 18); Danio rerio (SEQ ID NO: 19); Gallus gallus (SEQ ID NO: 20); Branchiostoma floridae (SEQ ID NO: 21). DETAILED DESCRIPTION OF THE INVENTION Definitions It is contemplated that the invention described herein is not limited to the particular methodology, protocols and reagents described, since these may be several. It should also be understood that the terminology used herein is for the sole purpose of describing particular embodiments, and is not intended to limit the scope of the present invention in any way. Although some methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, preferred methods, materials and devices are described below. All publications mentioned herein are incorporated by reference for the purpose of describing and disclosing the materials and methodologies reported in the publication that could be used in connection with the invention. In the practice of the present invention, many conventional techniques in molecular biology are used. These techniques are well known and are explained, for example, in Current Protocols in Molecular
Biology, Volumes I, II, and III, 1997 (F. M. Ausubel ed.); Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y .; DNA Cloning: A Practical Approach, Volumes I and II, 1985 (D. N. Glover ed.); Oligonucleotide Synthesis, 1984 (M. L. Gait ed.); Nucleic Acid Hybridization, 1985, (Hames and Higgins); Transcription and Translation, 1984 (Hames and Higgins eds.); Animal Cell Culture, 1986 (R. I. Freshney ed.); Immobilized Cells and Enzymes, 1986 (IRL Press); Perbal, 1984, A Practical Guide to Molecular Cloning; the series, Methods in Enzymology (Academic Press, Inc.); Gene Transfer Vectors for Mammalian Cells, 1987 (J. H. Miller and M. P. Calos eds., Cold Spring Harbor Laboratory); and Methods in Enzymology Vol. 154 and Vol. 155 (Wu and Grossman, and Wu, eds., respectively). As used herein and in the appended claims, the singular forms "a", "an" and "the" include plural reference, unless the context is otherwise stated. Thus, for example, the reference to "antibody" is a reference for one or more antibodies and the like, known to those skilled in the art. The "cMAC" or "Calcineurin Membrane Preservative" is the protein object of the invention and is described in detail below. The various names assigned to this protein and gene can change according to scientific use. Accordingly, the claims of this patent and the content of this specification are intended to refer to the gene and protein objects of the invention, and to their various fragments and forms without regard to the specific name
assigned. Thus, "cMAC" is defined herein as any polypeptide sequence that possesses at least one biological property (as defined below) of a naturally occurring polypeptide comprising the polypeptide sequence of SEQ ID NO: 2 or any another of the orthologs thereof shown in Figures 14 and 15. A "disorder associated with cMAC" or "a cMAC-related disorder" means a disorder that can be treated by modulating the activity of cMAC. These disorders include, but are not limited to, autoimmune disease, immune suppression, inflammatory disease, cancer, cardiovascular disease and neurological disease. Examples of autoimmune diseases include disorders and / or conditions including sarcoidosis, fibroid lung, idiopathic interstitial pneumonia, obstructive airway disease, including conditions such as asthma, intrinsic asthma, extrinsic asthma, powder asthma, particularly chronic or inveterate asthma (eg, airway hyperresponsiveness and late asthma), bronchitis, including bronchial asthma, childhood asthma, allergic rheumatoid arthritis, systemic lupus erythematosus, nephrotic lupus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, diabetes mellitus type I, and complications associated with it, beginning of adult type II diabetes mellitus, uveitis, nephrotic syndrome, steroid-dependent and steroid-resistant nephrosis, pustulosis palmo -plantar, allergic encephalomyelitis, glomerulonephritis, psoriasis, psoriatic arthritis, atopic eczema, (atopic dermatitis), contact dermatitis and other
eczematous dermatitis, seborrheic dermatitis, lichen planus, pemphigus, pemphigoid ampulla, epidermolysis bullosa, urticaria, angioedema, vasculitides, erythema, cutaneous eosinophilia, acne, alopecia areata, eosinophilic faciitis, atherosclerosis, conjunctivitis, keratoconjunctivitis, keratitis, vernal conjunctivitis, associated uveitis with Behcet's disease, herpetic keratitis, conical cornea, corneal epithelial dystrophy, keratoleucoma, ocular pemphigoid, Mooren's ulcer, scleritis, Graves' ophthalmopathy, severe intraocular inflammation, mucosal inflammation or blood vessels such as leukotriene-mediated diseases B4, gastric ulcers, vascular damage caused by ischemic diseases and thrombosis, ischemic intestinal disease, inflammatory bowel disease (eg, Crohn's disease, and ulcerative colitis), necrotising enterocolitis, kidney diseases including interstitial nephritis, Goodpasture syndrome, uraemic syndrome hemolytic, and diabetic nephropathy, selected nervous diseases between multiple myositis, Guillain-Barre syndrome, Meniere's disease and radiculopathy, collagen disease including scleroderma, Wegener's granuloma, and Sjogren's syndrome, chronic autoimmune liver diseases including autoimmune hepatitis, cirrhosis primary biliary, and sclerosing cholangitis), partial liver resection, acute liver necrosis (eg, necrosis caused by toxins, viral hepatitis, shock or anoxia), viral hepatitis B, non-A / non-B hepatitis and cirrhosis, hepatitis fulminant, pustular psoriasis, Behcet's disease, chronic active hepatitis, Evans syndrome, pollinosis, idiopathic hypoparathyroidism, disease
of Addison, autoimmune atrophic gastritis, lupoid hepatitis, tubulointerstitial nephritis, membranous nephritis, amyotrophic lateral sclerosis or rheumatic fever. Immuno suppression is desirable for the treatment of acute or chronic graft rejection, such as allo-or xenograft rejection of acute or chronic graft of cells, tissues or solid organ of eg pancreatic islets, stem cells, bone marrow, skin , muscle, corneal tissue, neuronal tissue, heart, lung, heart-lung combined, kidney, liver, intestines, pancreas, trachea or esophagus. Treatment for graft-versus-host disease is also included. Chronic rejection is also called vessel graft disease or graft vasculopathies. The treatment of immunosuppression, in the sense of treating immunocompromised subjects, can also be achieved by means of the modulation of cMAC, as it can result from the activation of the T cells, which are not sufficiently active in a subject to solve the disease or the condition. Diseases caused by the deficient immune response include but are not limited to AIDS, SLE, and the like. Inflammatory diseases that can be treated by the modulation of cMAC include those inflammatory diseases, disorders and / or conditions that are thought to respond to treatment with CsA. Cancer includes but is not limited to neoplasia and abnormal cell growth associated with pre-cancerous conditions or
cancerous Those skilled in the art are familiar with the numerous forms of cancer, neoplasia and abnormal cell growth, in particular lymphoma, leukemia, and other hematologic cancers. Cardiovascular disease includes but is not limited to cardiovascular diseases, disorders and conditions such as cardiac hypertrophy and heart failure. Diseases and / or neurological conditions include diseases, disorders, and / or conditions that include but are not limited to Alzheimer's disease, Parkinson's disease, and Huntington's disease, and include neuroprotection that can be achieved with methods and compositions of the invention. modulation of cMAC. While it is suggested to use cMAC antagonists or inhibitors in any or all diseases, the disorders and / or conditions noted above, the cMAC agonists are particularly involved and are desirable for the treatment of cancer, the diseases caused by the deficient immune response and for neuroprotection. "The cMAC-associated disorder" is sometimes referred to herein as a "pathological condition" that is associated with an abnormal expression of cMAC, an abnormal cMAC activity, or an abnormal activation of T cells. As used herein "disorder" includes a disease, disorder, or condition either existing or identified in prognosis, which includes symptoms or side effects of diseases, disorders or conditions and the pharmaceutical products used to treat them.
The ability of a substance to "modulate" a cMAC protein (eg, a "cMAC modulator") includes, but is not limited to, the ability of a substance to inhibit or increase one or more biological activities of the cMAC protein and / or inhibit or increase your expression. These modulators include both agonists and antagonists of cMAC activity. Such modulation may also involve the ability of other proteins to interact with cMAC, for example related regulatory proteins or proteins that bind to cMAC. The "biological activity" when used with either "cMAC isolated" or "cMAC" means that it has an activity selected from ion transport activity, ion diffusion activity, a calcium-dependent activation of a T cell activity, nuclear transposition of TORC, nuclear transposition of NFAT or gene expression activity driven by CRE (cAMP Response Element) when compared to mature, original or endogenous cMAC activity of for example, SEQ ID NO: 2. The term "agonist", as used herein, refers to a molecule (i.e., modulator) that can directly or indirectly modulate a polypeptide (e.g., a cMAC polypeptide), and which increases the biological activity of said polypeptide . Agonists can include proteins, nucleic acids, carbohydrates, organic molecules, small organic molecules (with or without inorganic fractions) or other molecules). A modulator that increases the transcription of the gene, the biological activity or the biochemical function of a protein is something that increases the transcription or stimulates the
biochemical properties or activity of that protein, as the case may be. The terms "antagonist" or "inhibitor" as used herein, refer to a molecule (i.e., modulator) that directly or indirectly modulates a polypeptide (e.g., cMAC polypeptide) which blocks or inhibits expression and / or the biological activity of that polypeptide. Antagonists and inhibitors may include proteins, nucleic acids, carbohydrates, or other molecules. A modulator that inhibits the expression or biochemical function of a protein is something that reduces the expression of the gene or the biological activity of that protein, respectively. The "nucleic acid sequence", as used herein, refers to an oligonucleotide, a nucleotide or a polynucleotide, and fragments or portions thereof, the polymeric components of which may be DNA, RNA, modified nucleotides, mimetic nucleotides or combinations thereof; and may be of genomic or synthetic origin, and may be single or double chain, and represent the sense or antisense chain. The term "antisense" as used herein, refers to nucleotide sequences that are complementary to a specific DNA or RNA sequence. The term "antisense strand" is used in reference to a nucleic acid strand that is complementary to the "sense" strand. Antisense molecules can be produced by any method, including synthesis by ligating the gene (s) of interest in an inverse orientation to a viral promoter that allows the synthesis of
the complementary chain. The designation "negative" is sometimes used in reference to the antisense strand, and "positive" is sometimes used in reference to the sense strand. The term "RNA agent" as used herein, refers to compounds and compositions that can act through an RNA (or iRNA) interference mechanism (see, as a general reference, He and Hannon, 2004). ) Nat. Genet 5: 522-532). IRNA agents such as short RNA interference ("siRNA"), double-stranded RNA ("dsRNA"), hairpin RNA ("shRNA", sometimes also called 'synthetic RNA') are commonly used, others are Developing. When introduced into a cell or synthesized within a cell the iRNAs are incorporated into a macromolecular complex that uses chains of the iRNA agent to target and dissociate the RNA strands containing the complementary (or substantially complementary) sequence. As contemplated in this, antisense oligonucleotides, triple helix DNA, RNA aptamers, iRNA agents such as siRNA, dsRNA, and shRNA, ribozymes and single-stranded RNA are designed to inhibit the expression of cMAC in such a way that it is designed the nucleotide sequence of the inhibitory molecule to cause the inhibition of endogenous cMAC protein synthesis. For example, based on the description herein, knowledge of the nucleotide sequence of cMAC can be used to design an siRNA molecule that inhibits the expression of cMAC without undue experimentation. Similarly, ribozymes can
synthesized to recognize specific nucleotide sequences of a protein of interest and dissociate it (Cech. J. Amer. Med Assn. 260: 3030 (1988)). Techniques for designing these molecules for use in the directed inhibition of gene expression are known to those skilled in the art. The term "sample" or "biological sample", as used herein, is used in its broadest sense. A biological sample of a subject may comprise blood, urine, or some other biological material with which the activity or gene expression of the cMAC proteins can be tested. As used herein, the term "antibody" is interchangeable with "immunoglobulin" and refers to immunoglobulins of a general form that are found in vertebrate species including mammals such as humans, primates, rodents, rabbits and many other species in which immunoglobulins have been identified. In particular those immunoglobulins include the heavy chain antibodies, which are found in camelids, which lack light chains and as a result have variable domains that reflect the absence of a VL partner. "Antibody" means molecules that correspond to complete immunoglobulins, as well as fragments thereof, such as Fa, F (ab ') 2. and Fv, which are capable of binding the epitope determinant. The term "antibody fragment" refers more specifically to these fragments and immunoglobulin-like polypeptides that do not comprise a complete immunoglobulin.
The term "humanized antibody" as used herein, refers to antibody molecules in which the amino acids have been replaced in the non-antigen binding regions in order to more closely resemble the human antibody, although still they retain the original link capacity. Depending on the context, this phrase may also include 'primatized' antibodies, wherein an antibody first obtained from a non-primate organism has been modified to closely resemble a primate immunoglobulin. A "polypeptide comprising a cMAC-specific binding region" means a polypeptide that incorporates one or more binding regions that specifically bind to the cMAC protein of SEQ ID NO: 2. A classical type of antigen-specific binding region is a complementary region ("CDR") found in an immunoglobulin. The CDRs are short amino acid sequences that bind to the antigen in question and provide the basis for the binding selectivity of the polypeptide in which it resides. CDRs are typically identified from immunoglobulins but can be generated by other means. The CDRs were originally defined in immunoglobulins using common definitions such as the definition of Kabat, the definition of Chothia (based on the location of the structural regions of curl); the definition of Ab (a compromise between the two used by the Oxford Molecular AbM antibody model software); and the definition of contact, which is possibly the most useful for people who want to make
mutagenesis to modify the affinity of an antibody, since these are residues that take part in the interactions with the antigen. The specific binding regions with antigen also include short sequences of amino acids that bind to an antigen, even if those sequences are not derived from the CDRs. PCT International Publication Number WO 2004/044011, which is incorporated herein by reference, provides an example of how those specific antigen binding regions (those which are not CDRs) can be identified and developed. Other methods are known to those skilled in the art. Once a specific binding region for cMAC is developed, it can be used in a wide variety of known or future frameworks or scaffolds, including any immunoglobulin isotype or fragment thereof., and other scaffold or framework polypeptides which are not immunoglobulins (discussed elsewhere herein) to provide antigen binding specificity. All of these polypeptides are considered herein as "polypeptides comprising a specific binding region for cMAC". A peptide mimetic is a synthetically derived peptide or a non-peptide agent created based on an understanding of the critical residues of a polypeptide subject which can mimic the function of a normal polypeptide. The mimetic peptides can interrupt the binding of a polypeptide to its receptor or other proteins and thus interfere with the normal function of a polypeptide. For example, a cMAC mimic may interfere with the normal function of cMAC.
A "therapeutically effective amount" is the amount of the drug sufficient to treat, prevent or ameliorate pathological conditions related to the function, activity or expression of cMAC. "Treat" includes preventing or improving, as the context may imply, and includes such treatment where the intent is therapeutic, prophylactic or directed to alleviate only the symptoms. The "related regulatory proteins" and "related regulatory polypeptides" as used herein refer to polypeptides involved in the regulation of cMAC proteins, which can be identified by one skilled in the art using conventional methods such as those described in I presented. The abnormal activation of T cells may include excessive activation, for example, in the states in which the mRNA encoding the cMAC protein is up-regulated or the cMAC protein has increased activity or amounts in a cell through, either increases in the absolute amount or in the specific activity; abnormal activation may also include states in which there is a down-regulation of cMAC gene expression, protein level or protein activity or there is an abnormally low activation of T cells. The "subject", when used in relationship to receiving treatment, refers to any human or non-human organism. In its broadest sense, the term "substantially similar" or "equivalent", when used herein with respect to a nucleotide sequence, means a nucleotide sequence.
corresponding to a reference cMAC nucleotide sequence, wherein the corresponding sequence encodes a polypeptide having substantially the same structure and function as the polypeptide encoded by the reference nucleotide sequence, for example, wherein only changes occur in the amino acids that they do not affect the function of the polypeptide. Desirably the substantially similar nucleotide sequence encodes the polypeptide encoded by the reference nucleotide sequence. The percent identity between the substantially similar nucleotide sequence and the reference nucleotide sequence is desirably at least 80 percent, more desirably at least 85 percent, and preferably at least 90 percent, more preferably at at least 95 percent, 96 percent, 97 percent, or 98 percent, still more preferably at least 99 percent. A sequence of nucleotides "substantially similar" to the reference nucleotide sequence hybrid to the reference nucleotide sequence in 7 percent sodium dodecyl sulfate (SDS), 0.5 M NaP04, 1 mM EDTA at 50 ° C washing in 2 X SSC, 0.1 percent SDS at 50 ° C, more desirably at 7 percent sodium dodecyl sulfate (SDS), 0.5 M NaP04, 1 mM EDTA at 50 ° C with washing in 1X SSC, 0.1 percent of SDS at 50 ° C, most desirably still at 7 percent sodium dodecyl sulfate (SDS), 0.5 M NaP04, 1 mM EDTA at 50 ° C with 0.5X SSC wash, 0.1 percent SDS at 50 ° C, preferably in 7 percent sodium dodecyl sulfate (SDS), 0.5 M NaP04, 1 mM EDTA at 50 ° C with
washed in 0.1X SSC, 0.1 percent SDS at 50 ° C, more preferably in 7 percent sodium dodecyl sulfate (SDS), 0.5 M NaP0, 1 mM EDTA at 50 ° C with washing in 0.1X SSC, 0.1 percent SDS at 65 ° C, which still codes for a geneically equivalent gene product. Generally, hybridization conditions can be extremely stringent or less stringent. In some cases when the nucleic acid molecules are deoxyoligonucleotides ("oligos"), extremely stringent conditions refer, for example, to washing in 6X SSC / 0.05 percent sodium pyrophosphate at 37 ° C (for 14 base oligos) ), 48 ° C (for oligos of 17 bases), 55 ° C (for oligos of 20 bases), and 60 ° C (for oligos of 23 bases). The appropriate ranges of those stringent conditions for the nucleic acids of the variant compositions are described in Krause and Aaronson (1991)., Methods in Enzymology, 200: 546-556 in addition to Maniatis et al., Cited above. When used with respect to a polypeptide sequence, "substantially similar" means a protein sequence corresponding to a cMAC polypeptide described herein, such a protein sequence has substantially the same structure and function as the cMAC polypeptide, including isoforms, homologs, orthologs and modified sequences that contain an amino acid sequence identity across the protein length of at least 50 percent, 55 percent, 60 percent, 65 percent, 70 percent, 75 percent, 80 percent, 85 percent, 90 percent, 95 percent
one hundred or 98 percent. "Functionally equivalent", as used herein, may refer to a protein or polypeptide capable of exhibiting substantially similar activity in vivo or in vitro as the differentially expressed endogenous gene products encoded by the differentially expressed gene sequences described above. . "Functionally equivalent" may also refer to proteins or polypeptides capable of interacting with other cellular or extracellular molecules in a manner substantially similar to the way in which the corresponding portion of the endogenous gene product differentially expressed would do so. For example, a "functionally equivalent" peptide would be capable, in an immunological assay, of decreasing the binding of an antibody to the corresponding peptide (ie, the peptide whose amino acid sequence was modified to achieve the "functionally equivalent" peptide) of the endogenous protein or the endogenous protein itself, wherein the antibody was raised against the corresponding peptide of the endogenous protein. An equimolar concentration of the functionally equivalent peptide will decrease the aforementioned binding of the corresponding peptide by at least 5 percent, preferably between about 5 percent and 10 percent, more preferably between about 10 percent and 25 percent, further preferably between about 25 percent and 50 percent, and most preferably between about 40 percent and 50 percent. A "fragment" is a portion of a cMAC sequence of
full length that occurs naturally mature, native or endogenous that has one or more amino acid residues removed. The removed amino acid residue (s) can occur anywhere in the polypeptide, including either the N-terminus, the C-terminus or internally. That fragment will have at least one biological property in common with cMAC. The cMAC fragments will typically have a consecutive sequence of at least 10, 15, 20, 25, 30, 40, 50 or 60 amino acid residues that are identical to the isolated cMAC sequences of a mammal including the cMAC of SEQ ID NO: 2. "High mRNA transcription" refers to a larger amount of the transcribed RNA messenger of the natural endogenous human gene encoding a cMAC polypeptide of the present invention in an appropriate tissue or a cell of an individual suffering from a pathological condition related to the abnormal activation of cMAC gene expression or abnormal activation of T cells compared to control levels, in particular at least about twice, preferably at least about five times, more preferably at least about ten times, and even more preferably 100 times the amount of mRNA found in the corresponding tissues in subjects who do not suffer from this condition. Such high levels of mRNA can eventually lead to increased protein levels translated from that mRNA in an individual suffering from that condition when compared to a healthy individual. A "host cell", as used herein, refers to
a prokaryotic or eukaryotic cell containing heterologous DNA that has been introduced into the cell by any means, for example, electroporation, calcium phosphate precipitation, microinjection, transformation, viral infection, and the like. "Heterologist" as used herein means "of different natural origin" or represents a non-natural state. For example, if a host cell is transformed with a DNA or gene derived from another organism, particularly from another species, that gene is heterologous with respect to the host cell and also with respect to the descendants of the host cell carrying that gene. Similarly, heterologous refers to a nucleotide sequence derived from and inserted into the same type of natural cell, original, but which is present in a non-natural state, for example a different copy number or under the control of different regulatory elements. A "vector" molecule is a nucleic acid molecule into which the heterologous nucleic acid can be inserted and which can be introduced into an appropriate host cell. The vectors preferably have one or more origins of replication, and one or more sites where the recombinant DNA can be inserted. Vectors often have convenient means by which cells can be selected with vectors from those that do not, for example, they encode drug resistance genes. Common vectors include plasmids, viral genomes, and (mainly in yeast and bacteria) "artificial chromosomes". The term "isolated" means that the material was removed from its
original environment (for example, the natural environment if it is occurring naturally). For example, a naturally occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of its coexistence materials in the natural system, is isolated, although it is subsequently reintroduced into the natural system These polynucleotides can be part of a vector and / or those polynucleotides or polypeptides could be part of a composition, and still be isolated in that vector or composition if that is not part of their natural environment. As used herein, the term "transcription control sequence" refers to DNA sequences, such as primer sequences, enhancer sequences, and promoter sequences, which induce, repress or otherwise. they control the transcription of the nucleic acid sequences that encode proteins to which they are operatively linked. As used herein, "human transcription control sequences" are any of those transcription control sequences normally found associated with a human gene encoding the cMAC protein of the present invention as found on the respective human chromosome . As used herein, the term "non-human transcription control sequence" is any of the transcriptional control sequences not found in the human genome. As used herein, a "chemical derivative" of a
polypeptide of the invention, is a polypeptide of the invention that contains additional chemical fractions that are not normally part of the molecule. These fractions can improve the solubility, absorption, biological half-life, etc. of the molecule. Alternatively, the fractions can lessen the toxicity of the molecule, eliminate or attenuate any undesirable side effects of the molecule, and so on. Fractions capable of mediating these effects are described, for example, in Remington's Pharmaceutical Sciences, 16th ed., Mack Publishing Co., Easton, Pa. (1980). Introduction The present invention is based on the surprising discovery that the cMAC ("Calcineurin-Conserved Membrane Activator") protein previously referred to in the sequence database as "hypothetical Homo sapiens protein MGC14327 (MGC14327), mRNA. (GenBank) Access Number NM_053045) "and henceforth of unknown function, is a potent regulator of the nuclear transposition of NFAT and TORC1, functions via the trajectory of Calcineurin, and has an important role in the activation of T cells. 1, Figure 11). Table 1
As used herein "cMAC" means, depending on the context, a cMAC protein, or a nucleic acid comprising a sequence encoding the cMAC protein (e.g., the cMAC gene), or fragments or fusions thereof. The cMAC protein described herein includes the cMAC polypeptide identified herein, any and all forms of these polypeptides including, but not limited to, partial forms, homologs, isoforms, precursor forms, full length polypeptides, fusion proteins containing the protein sequence, proteins that are substantially similar or equivalent to cMAC, or fragments of any of the foregoing, from humans, primates, mammals, vertebrates, invertebrates or any other species. The invention also covers nucleic acids that are related to the transcription, function and stability of cMAC mRNA (Table 2, Figures 12 and 13):
Table 2
The fragments of interest of cMAC include, but are not limited to, those fragments that contain amino acids of particular importance for the normal function of cMAC. Based on the predicted transmembrane structure of cMAC as illustrated in Table 3 and Figure 1, the polypeptide can be subdivided into the following domains: Table 3
The designation of the inside and outside of the membrane needs to be confirmed experimentally, however, according to the analysis of the TMHMM prediction for the human cMAC NM_053045, NP_444273.1, the regions (e.g., epitopes) of particular interest for therapeutic antibodies include the extra-membrane amino acid sequences 1-12, 36-49, 73-78, 102-110, 131-136. Structure and conservation of cMAC The human cMAC cDNA encodes a hydrophobic protein of 136 amino acids. The analysis of the amino acid sequence
Primary with algorithms that predict transmembrane helices indicates that cMAC is an integral membrane protein with four transmembrane domains and cytoplasmic domains at the short N and C terminals. Figure 1. Two potential sites for post-translational modification of the protein were found, using the MotifsGCG programs. A potential phosphorylation site of protein kinase C (PKC) and a phosphorylation site of Casein kinase II were identified in Serine 4. The proposed PKC phosphorylation site in Serine 4 is predicted and conserved in all vertebrate cMAC genes identified. Potential additional PKC sites also exist at residue 73 (SVR) and 97 (SLK). The predominant PKC isoform present in T cells is PKC9, and it is known to be important in mediating the activation of T cells. During the activation of T cells, PKCG localizes a large number of lipids to the plasma membrane (Khoshnan et al. J. Immunol. 165 (12): 6933-40 (2000)), which are insoluble insoluble cholesterol-containing membrane domains containing many components (sometimes transiently on stimulation) that contribute to signal transduction. Interestingly, the calcium channel proposed in B cells, CD20, is also a 4TM protein critical to the activation of B cells. CD20 is known to be constitutively associated with a large number of lipids. It remains to be determined if cMAC is a substrate for PKC but we are trying to find a motif in the cMAC amino acid sequence that can serve as a regulatory function through which PKC is
activates following TCR / CD28 stimulation. An isoprenylation site was also identified in cysteine 134 of the predicted cMAC proteins of human, rat, mouse, zebrafish and Xenopus. cMAC is a highly conserved protein. CMAC orthologs of other species described here are presented in Figures 2 and 15. The human cMAC protein is 97 percent identical to the predicted mouse protein, and 82 percent and 78 percent identical to the proteins of Xenopus tropicalis and Danio rerio, respectively. Interestingly, there is also a gene encoded by the ancient vertebral species Amphioxus floridae that is 54 percent identical to human cMAC. In total, 63 of the 136 amino acids are conserved through each vertebrate cMAC and 99/136 amino acids are identical or represent conservative changes. Interestingly, there are also similar invertebrate proteins with the Drosophila and C elegans proteins of significantly lower levels of homology (39 percent and 27 percent identical, respectively) compared to the vertebrate orthologs. It is not clear if the genes of the insects are also orthologs of cMAC. The vertebrate and invertebrate proteins described here were mutually qualified as the best guesses suggesting that they are probably orthologs and / or derivatives of a single ancestral gene. It is interesting to note that the cMAC protein sequence is the most highly conserved in organisms that contain a modern adaptive immune system. In particular, cMAC is highly conserved in animals that contain T cells (e.g., > 80 per
percent identity in mammals, fish and amphibians), is more divergent in Amphioxus (54 percent identical), which has many orthologs and gene homologs destined to be recruited for adaptive immunity but has not yet developed lymphocytes, and is highly divergent in invertebrates that lack some correlation with lymphocytes. Thus, it is tempting to speculate that cMAC may have evolved over the development of the adaptive immune system of vertebrates and is a central player in the signaling of capacitive calcium that is required in the activation of T cells and perhaps in the activation of B cells. In each species, a single ortholog of cMAC is present and conserved. The cMAC is predicted to encode a 4 TM domain protein (illustrated in Figure 1). It is speculated that cMAC represents a novel gene family of signal transducers mediated by calcium. The cMAC homologs and orthologs include those described herein (e.g., Table 4 and Figures 14 and 15), and those that would be apparent to one skilled in the art, and are considered to be included within the scope of the invention. For example, screening assays for small molecules as contemplated in this invention may use a human cMAC homolog or a cMAC ortholog of different species such as another primate, mammal, vertebrate or invertebrate. It is also contemplated that cMAC proteins include those isolated from naturally occurring sources of any species such as
genomic DNA libraries as well as genetically engineered host cells that comprise expression systems, or produced by chemical synthesis, using, for example, automatic peptide synthesizers or a combination of those methods. The means for isolating and preparing such polypeptides are well known in the art. Table 4
The present invention also includes any fragments of proteins or nucleic acids encoding protein fragments found in SEQ ID NOs: 1-21. Additional homologs can be identified and isolated
easily, without undue experimentation, by molecular biology techniques well known in the field. In addition, there may be genes and other genetic sites within the genome that encode proteins that have extensive homology to one or more domains of those gene products. These genes can also be identified via similar techniques. Function and role of cMAC, and cell type localization Without wishing to be bound by any particular theory, it is possible that human cMAC is a transmembrane protein as illustrated in Figure 1. Bioinformatic analyzes indicate several domains that are likely to adopt an intramembrane conformation. and extramembrana. This prediction highlights the potential use of antibodies to inhibit or activate cMAC in a therapeutic setting. A biological function or activity of the cMAC polypeptide is clearly established in the functional activation of T cells, as demonstrated in the Examples herein. Other biological activities of cMAC can be further elucidated as the studies progress. Some of the most specific biological activities of cMAC, such as conclusions can be drawn based on the examples, include nuclear transposition of TORC, nuclear transposition of NFAT, and expression of the gene driven by CRE (cAMP Response Element). The cMAC could have different biochemical activities including as an example an ion channel, for example a calcium channel (voltage gate or ligand gate), and can have activity in the activation
calcium-dependent T cells. Specific biochemical activities also include interactions with cell membranes and cell membrane components, as an objective for myristilization, glycosylation, phosphorylation, dephosphorylation and other post-translational modifications. Based on the description herein, those skilled in the art will be able to identify these and other biological activities of the cMAC. The invention describes a method for modulating (eg inhibiting or increasing) one or more of these cMAC activities. These methods may be for therapeutic application with modulators identified from cMAC, or for research and discovery use such as for selection trials or other research tools. CMAC mRNA has been identified in a variety of human cell types. Figure 3 identifies the highest level of cMAC in lymphocytes, specifically T cells. Most other tissues also show a level of cMAC indicating that cMAC may be commonly used to modulate diseases related to calcium signaling. Expression of cMAC. In order to obtain an overview of cells expressing cMAC, mRNA levels were examined in different tissues and cell types as indicated by Affymetrix expression profiling. As shown in Figure 3, cMAC was widely expressed. However, the highest levels of expression seen were in the populations of
T and B cells. The average expression level in these lymphocyte preparations was 3 to 10 times higher than in the median expression seen through all tissues examined. Although the expression of mRNA and cMAC protein should be examined by other methods, these results suggest that cMAC could be enriched in lymphocyte populations. The predominant presence in T cells indicates that anti-cMAC antibodies and other cMAC binding compounds will predominantly be directed to T cells; those antibodies or compounds have many significant therapeutic and research uses, as described more fully elsewhere in this specification. Thus, the present invention provides isolated polypeptides comprising or consisting of an amino acid sequence as set forth in SEQ ID NO. 2 or a fragment thereof, or a substantially similar protein sequence having a sequence identity of at least 50 percent with SEQ ID NO: 2, or a functional equivalent thereof exhibiting an activity selected from an ion transport , ion diffusion, calcium-dependent activation of T cells, nuclear transposition of TORC, nuclear transposition of NFAT or gene expression activity driven by CRE (cAMP Response Element) of the original SEQ ID NO: 2. Accordingly, the present invention further provides the polypeptides of SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19,
SEQ ID NO: 20 and SEQ ID NO: 21. In addition, these polypeptides can be, for example, a fusion protein including all or a part of SEQ ID NO.2. The invention also includes isolated nucleic acid or nucleotide molecules, preferably DNA molecules, in particular SEQ ID NO. 1, SEQ ID NO: 11 or SEQ ID NO: 12 that encode the cMAC protein. The invention also discloses an isolated nucleic acid molecule, preferably a DNA molecule, of the present invention that encodes a polypeptide comprising the amino acid sequence presented in SEQ ID NO: 2 or SEQ ID NO: 13 to 21. The invention it also encompasses: (a) vectors comprising a nucleotide sequence of a cMAC protein, in particular SEQ ID NO. 1, SEQ ID NO: 11 or SEQ ID NO: 12 or a fragment thereof and / or its complements (ie, antisense); (b) vector molecules, preferably vector molecules comprising transcriptional control sequences, in particular expression vectors, comprising encoding sequences of any of the cMAC proteins described herein operatively associated with a regulatory element that directs the expression of the coding sequences; and (c) genetically engineered host cells containing a vector molecule as mentioned herein or at least one fragment of any of the above nucleotide sequences operatively associated with a regulatory element that directs the expression of the
coding in the host cell. As used herein, regulatory elements include, but are not limited to, inducible and non-inducible promoters, enhancers, operators and other elements known to those skilled in the art that drive and regulate expression. Preferably, the host cells can be vertebrate host cells, preferably mammalian host cells, such as human cells or rodent cells, such as CHO or BHK cells. Equally preferred, the host cells can be bacterial host cells, in particular E. coli cells. Therefore, the invention covers a vector molecule comprising the nucleic acid sequence of cMAC (SEQ ID NO: 1), and a host cell comprising this vector molecule. Particularly preferred is a host cell, in particular of the type described above, which can be propagated in vitro and which is capable after growth in culture of producing a cMAC polypeptide, in particular a polypeptide comprising or consisting of an amino acid sequence presented in SEQ ID NO: 2, wherein the cell comprises at least one transcription control sequence that is not a transcriptional control sequence of the natural endogenous human gene encoding the polypeptide, wherein the one or more sequences of transcription control control the transcription of a DNA encoding these polypeptides. This vector or host cell can be used in a method for
producing a cMAC polypeptide of SEQ ID NO. 2, which comprises culturing a host cell that has incorporated therein an expression vector comprising the cMAC vector under conditions sufficient for expression of the polypeptide in the host cell. The invention also includes fragments of any of the nucleic acid sequences described herein. Fragments of the nucleic acid sequences of SEQ ID NO. 1 and SEQ ID NO. 3 as a hybridization probe for a cDNA library to isolate the full length gene and to isolate other genes having a high sequence similarity to a cMAC gene of similar biological activity. Probes of this type of preference have at least about 20 bases and may contain, for example, from about 30 to about 50 bases, from about 50 to about 100 bases, from about 100 to about 200 bases, or more than 200 bases. The probe can also be used to identify a cDNA clone corresponding to an entire length transcript and one or more genomic clones containing a complete cMAC gene including regulatory and promoter regions., exons, and introns. An example of a selection comprises isolating the coding region of a cMAC gene using the known DNA sequence to synthesize an oligonucleotide probe. Labeled oligonucleotides having a sequence complementary to that of the gene of the present invention are used to analyze a human cDNA library, DNA or
Genomic mRNA to determine which members of the hybrid library the probe. The isolated nucleotide sequence of the present invention that encodes a cMAC polypeptide can be labeled and used to analyze a cDNA library constructed from the mRNA obtained from the organism of interest. Hybridization conditions will be of less stringency when the cDNA library is derived from an organism different from the type of organism from which the tagged sequence is derived. Alternatively, the tagged fragment can be used to analyze a genomic library derived from the organism of interest, again, using appropriately stringent conditions. These conditions of low stringency will be well known to those skilled in the art and will vary predictably depending on the specific organisms from which the library and tagged sequences are derived. For guidance regarding these conditions see, for example, Sambrook et al., Cited above. PCR technology can be used to isolate complete or partial cDNA sequences that are substantially similar to cMAC. For example, RNA can be isolated, following normal procedures, from a suitable cell or tissue source. A reverse transcription reaction can be performed on the RNA using an oligonucleotide primer specific for the most 5 'end of the amplified fragment for priming the first chain synthesis. The resulting RNA / DNA hybrid can be
then "putting a tail" of guanines using a standard terminal transferase reaction, the hybrid can be digested with RNase H and the synthesis of the second chain can then be primed with a poly-C primer. In this way, the cDNA sequences upstream of the amplified fragment can easily be isolated. For a review of the cloning strategies that may be used, see, for example, Sambrook et al., 1989, supra. In cases where the gene identified is the normal, or original type, this gene can be used to isolate mutant alleles of the gene. This isolation is preferable in processes and disorders where it is known or suspected to have a known genetic basis. Mutant alleles can be isolated from individuals known or suspected to have a genotype that contributes to disease symptoms related to inflammation or immune response. Mutant alleles and mutant allele products can then be used in the diagnostic test systems described below. A cDNA of the mutant gene can be isolated, for example, using PCR, a technique that is well known to those skilled in the art. In this case, the first strand of cDNA can be synthesized by hybridizing an oligo-dT oligonucleotide to mRNA isolated from tissue that is known or suspected to be expressed in an individual that presumably carries the mutant allele, and spreading the new strand with reverse transcriptase . The second strand of the cDNA is
then synthesized using an oligonucleotide that hybridizes specifically to the 5 'end of the normal gene. Using these two primers, the product is then amplified via PCR, cloned into a convenient vector, and subjected to DNA sequence analysis through methods well known to those skilled in the art. By comparing the DNA sequence of the mutant gene with that of the normal gene, the mutation (s) responsible for the loss or alteration of the function of the mutant gene product can be checked. Alternatively, a genomic or cDNA library can be constructed and selected using DNA or RNA, respectively, from a tissue that is known or suspected to express the gene of interest in an individual that is suspected or known to carry the mutant allele. The normal gene, or any convenient fragment thereof, can then be labeled and used as a probe to identify the corresponding mutant allele in the library. The clone containing this gene can then be purified by methods routinely practiced in the art, and undergo sequence analysis as described above. The present invention includes proteins that represent functionally equivalent cMAC gene products. This equivalent gene product may contain deletions, additions or substitutions of amino acid residues within the amino acid sequence encoded by the sequences of the gene described above, thereby producing a functionally equivalent gene product. Substitutions can be made
amino acids based on the similarity of polarity, charge, solubility, hydrophobicity, hydrophilicity, and / or the antipathetic nature of the residues involved. For example, non-polar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; the neutral polar amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and the negatively charged amino acids (acids) include aspartic acid and glutamic acid. The data described herein indicate particular polypeptide fragments are useful for certain activities of the cMAC protein. Thus, these cMAC peptide fragments as well as the fragments of the nucleic acids encoding the active portion of the polypeptides described herein, and the vectors comprising these fragments, are also within the scope of the present invention. As used herein, a "fragment" of the nucleic acid encoding the active portion of the cMAC polypeptides refers to a nucleotide sequence having fewer nucleotides than the nucleotide sequence encoding the entire amino acid sequence of a cMAC polypeptide and which encodes a peptide having an activity of a cMAC protein (ie, a peptide having at least one biological activity of a cMAC protein) as defined herein. Generally, the nucleic acid encoding the peptide
having an activity of a cMAC protein will be selected from the bases that encode the mature protein. However, in some cases, it may be desirable to select all or part of a peptide from the forward portion of the nucleic acid sequence of a cMAC protein. These nucleic acids can also contain binding sequences, modified restriction endonuclease sites and other sequences useful for cloning, expression or molecular purification or recombinant peptides having at least one biological activity of a cMAC protein. The cMAC peptide fragments as well as the nucleic acids encoding a peptide fragment having an activity of a cMAC protein can be obtained according to conventional methods. In addition, antibodies directed to these peptide fragments can be made as described hereinafter. Modifications to these peptide fragments (e.g., amino acid substitutions) that can increase the immunogenicity of the peptide can also be employed. Similarly, using methods familiar to a person skilled in the art, these peptides of cMAC proteins can be modified to contain forward or signal sequences or conjugate to a linker or other sequence to facilitate molecular manipulations. The polypeptides of the present invention can be produced by recombinant DNA technology using techniques well known in the art. Therefore, a method is provided for
producing a polypeptide of the present invention, this method comprises culturing a host cell that has incorporated therein an expression vector containing an exogenously derived polynucleotide encoding a polypeptide comprising an amino acid sequence as presented in SEQ ID NOs: 2 , 13-21, preferably SEQ ID NO. 2, under conditions sufficient for expression of the polypeptide in the host cell, whereby production of the expressed polypeptide is caused. Optionally, this method further comprises recovering the polypeptide produced by the cell. In a preferred embodiment of this method, the exogenously derived polynucleotide encodes a polypeptide consisting of an amino acid sequence presented in SEQ ID NO: 2. Preferably, this exogenously derived polynucleotide comprises the nucleotide sequence as presented in any of SEQ ID NOS. NOs: 1. So, methods for preparing the polypeptides and peptides of the invention are described herein by expressing the nucleic acid encoding the respective polypeptide sequences. Methods that are well known to those skilled in the art can be used to construct expression vectors containing appropriate protein coding sequences and transcription / translation control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination / genetic recombination. See, for example, the techniques described in Sambrook et al., 1989, supra, and
Ausubel et al., 1989, supra. Alternatively, RNA capable of encoding protein sequences from differentially expressed genes can be chemically synthesized using, for example, synthesizers. See, for example, the techniques described in "Oligonucleotide Synthesis", 1984, Gait, M.J. ed., IRL Press, Oxford, which is incorporated by reference herein in its entirety. A variety of host expression vector systems can be used to express the coding sequences of genes of the invention. These host expression systems represent vehicles by which the coding sequences of interest can be produced and subsequently purified, but also represent the cells which, when transformed or transfected with the appropriate nucleotide coding sequences, can exhibit the protein of the invention on the site. These include, but are not limited to, microorganisms such as bacteria (e.g., E. coli, B. subtilis) transformed with DNA expression vectors of a recombinant bacteriophage, plasmid DNA or cosmid DNA containing the cMAC protein encoding sequences; yeast (eg, Saccharomyces, Pichia) transformed with the recombinant yeast expression vectors containing cMAC protein coding sequences; insect cell systems infected or transfected with recombinant virus expression vectors (eg, baculovirus) containing coding sequences of the
cMAC protein; plant cell systems infected with recombinant virus expression vectors (eg, cauliflower mosaic virus, CaMV, tobacco virus, TMV) or transformed with recombinant vectors, including plasmids, (eg, Ti plasmid) containing cMAC protein coding sequences; or mammalian cell systems (eg, COS, CHO, BHK, 293, 3T3) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (eg, mothalothionein promoter) or from other mammalian viruses (for example, the last adenovirus promoter, the 7.5K promoter of the vaccine virus, or the CMV promoter). The expression of the cMAC protein of the present invention can also be performed by a cell of a gene encoding cMAC that is original of that cell. Methods for that expression are detailed, for example, in U.S. Patent Nos. 5,641,670; 5,733,761; 5,968,502; and 5,994,127, all of which are incorporated by reference herein in their entirety. Cells that have been induced to express cMAC by the methods of any of the patents of the United States of America Numbers 5,641,670; 5,733,761; 5,968,502; and 5,994,127 can be implanted into a desired tissue in a live animal in order to increase the local concentration of cMAC in the tissue. These methods have therapeutic implications, for example, for neurodegenerative states in which it occurs
the loss of CREB function and those agonists and / or exogenous cMAC protein may be useful to treat these states. In bacterial systems, several expression vectors can be advantageously selected depending on the intended use of the protein being expressed. For example, when a large amount of that protein is to be produced, for the generation of antibodies or for selecting peptide libraries, for example, vectors that direct the expression of high levels of fusion protein products may be desirable. they are easily purified. In this respect, fusion proteins comprising hexahistidine tags (Sisk et al., 1994: J. Virol 68: 766-775) can be used as provided by several vendors (eg Qiagen, Valencia, CA). These vectors include, but are not limited to, the E. coli expression vector pUR278 (Ruther et al., 1983, EMBO J. 2: 1791), in which the sequence encoding the protein can be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye &Inouye, 1985, Nucleic Acids Res. 13: 3101-3109; Van Heeke &Schuster, 1989, J. Biol. Chem. 264: 5503-5509); and similar. The pGEX vectors can also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, these fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of glutathione
free. The pGEX vectors are designed to include thrombin or factor Xa protease dissociation sites so that the cloned target gene protein can be released from the GST fraction. The promoter regions of any desired gene can be selected using vectors containing a reporter transcription unit that lacks a promoter region, such as a chloramphenicol acetyl transferase ("CAT"), or the luciferase transcription unit, downstream of the site or restriction sites to introduce a candidate promoter fragment; that is, a fragment that may contain a promoter. For example, the introduction into the vector of a promoter-containing fragment at the restriction site upstream of the CAT gene generates the production of CAT activity, which can be detected by standard assays for CAT. Suitable vectors for this purpose are well known and readily available. Two of these vectors are pKK232-8 and pCM7. Thus, promoters for the expression of polynucleotides of the present invention include not only well-known and readily available promoters, but also promoters that can be readily obtained by the prior art, using a reporter gene. Among the known bacterial promoters suitable for the expression of polynucleotides and polypeptides according to the present invention are the lacZ and lacZ promoters of E. coli, the T3 and 17 promoters, the T5 tac promoter, the lambda PR, the
PL promoters and the trp promoter. Suitable eukaryotic promoters for this purpose include the early CMV promoter, the HSV thymidine kinase promoter, the SV40 early and late promoters, the retroviral LTR promoters, such as those of the Rous sarcoma virus ("RSV"), and the promoters of metallothionein, such as the mouse metallothionein-l promoter.
In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV) is one of several insect systems that can be used as a vector to express foreign genes. The virus grows in the cells of Spodoptera frugiperda. The coding sequence can be cloned individually in the non-essential regions (for example the polyhedrin gene) of the virus and placed under the control of an AcNPV promoter (for example the polyhedrin promoter). Successful insertion of the coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat encoded by the polyhedrin gene). These recombinant viruses are then used to infect Spodoptera frugiperda cells in which the inserted gene is expressed. (For example, see Smith et al., 1983, J. Virol 46: 584, Smith, United States Patent No. 4,215,051). In mammalian host cells, various expression systems can be used. In cases when an adenovirus is used as an expression vector, the coding sequence of interest
it can be linked to an adenovirus transcription / translation control complex, for example, the sequence of the last promoter and tripartite leader. This chimeric gene can then be inserted into the adenovirus genome by in vitro or in vivo recombination. Insertion into a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing the desired protein in infected hosts. (For example, See Logan &Shenk, 1984, Proc. Nati, Acad. Sci. USA 81: 3655-3659). Specific initiation signals may also be required for efficient translation of the coding sequences of the inserted gene. These include the ATG start codon and the adjacent sequences. In cases when a whole gene, which includes its own initiation codon and adjacent sequences, inserted into the appropriate expression vector, additional translation control signals may not be required. However, in cases where only a portion of the gene coding sequence is inserted, exogenous translation control signals must be provided, which include, perhaps, the ATG initiation condom. In addition, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These control signals of the exogenous translation and the initiation codons can have a variety of origins, both natural and synthetic. The efficiency of the expression can be improved by the inclusion of elements that improve the
appropriate transcription, terminators of the transcription, etc., (see Bittner et al., 1987, Methods in Enzymol, 153: 516-544). Other common systems are based on SV40 viruses, retroviruses or adeno-associated viruses. The selection of vectors and associated promoters for expression in a host cell is a well-known procedure and the requisite techniques for the construction of the expression vector, the introduction of the vector into the host and the expression in the host per se are skills of routine in the technique. Generally, recombinant expression vectors will include replication origins, a promoter derived from a gene highly expressed to direct the transcription of a downstream structural sequence, and a selectable marker to allow isolation of cells containing the vector after exposure to the vector. Additionally, a strain of host cells can be chosen that modulates the expression of the inserted sequences, or modifies and processes the product of the gene in the specific manner desired. These modifications (eg, glycosylation) and processing (eg, dissociation) of the protein products may be important for the function of the protein. Different host cells have specific characteristics and mechanisms for the processing and post-translational modification of proteins. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the expressed foreign protein. To this end, you can use the
eukaryotic host cells that possess the cellular machinery for the processing of primary transcription, glycosylation, and phosphorylation of the gene product. These mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, WI38, and the like. The present invention also includes peptides and fragments of cMAC recombinant peptides having an activity of a cMAC protein. The term "recombinant peptide" refers to a protein of the present invention that is produced by recombinant techniques, wherein the DNA that generally encodes the active cMAC fragment is inserted into a convenient expression vector which in turn is used to transform a host cell to produce the heterologous protein. The recombinant proteins of the present invention can also include cMAC chimeric or fusion proteins and different polypeptides which can be made according to techniques known to those skilled in the art (see, for example, Current Protocols in Molecular Biology; Eds Ausubel and collaborators, John Wiley &Sons; 1992; PNAS 85: 4879 (1988)). For long-term, high-yield protein production, stable expression is preferred. For example, cell lines that stably express the differentially expressed gene protein can be genetically engineered. Instead of using expression vectors which contain viral origins of replication, the host cells can be transformed with
DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. After the introduction of foreign DNA, designed cells can be allowed to grow for 1-2 days in an enriched medium, and then changed to selective medium. The selectable marker in the recombinant plasmid confers resistance to selection and allows the cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method can be used advantageously to design cell lines that express the differentially expressed gene protein. These genetically engineered cell lines may be useful in particular for selecting and evaluating compounds that affect the endogenous activity of the expressed protein. Various screening systems, including but not limited to thymidine kinase genes, can be used (Wigler, et al., 1977, Cell 11: 223), hypoxanthine-guanine phosphoribosyl transferase (Szybalska &; Szybalski, 1962, Proc. Nati Acad. Sci. USA 48: 2026), and adenine phosphoribosyl transferase (Lowy, et al., 1980, Cell 22: 817) of the herpes simplex virus, can be used in tk ", hgprt 'or aprt" cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the dhfr genes, which confers resistance to methotrexate (Wigler, et al., 1980, Nati. Acad. Sci. USA
77: 3567; O'Hare, et al., 1981, Proc. Nati Acad. Sci. USA 78: 1527); gpt, which confers resistance to mycophenolic acid (Mulligan &Berg, 1981, Proc Nati Acad Sci USA 78: 2072); neo, which confers resistance to the aminoglucoside G-418 (Colberre-Garapin, et al., 1981, J. Mol. Biol. 150: 1); and hygro, which confers resistance to hygromycin (Santerre, et al., 1984, Gene 30: 147). An alternative fusion protein system allows easy purification of non-denatured fusion proteins expressed in human cell lines (Janknecht, et al., 1991, Proc Nati Acad Sci USA 88: 8972-8976). In this system, the gene of interest is subcloned into a vaccine recombination plasmid so that the open reading frame of the gene is fused in translation to an amino-terminal tag consisting of six histidine residues. Extracts from the cells infected with the recombinant vaccine virus are loaded onto Ni2 + nitriloacetic acid-agarose columns and the histidine-tagged proteins are eluted selectively with imidazole-containing regulators. When used as a component in test systems such as those described below, a protein of the present invention can be labeled, either directly or indirectly, to facilitate the detection of a complex formed between the protein and a test substance. Any of a variety of convenient labeling systems may be used that include but are not
limit to radioisotopes such as 125l; enzyme labeling systems that generate a detectable calorimetric signal or light when exposed to a substrate; and fluorescent labels. When using recombinant DNA technology to produce a protein of the present invention for these test systems, it may be advantageous to design fusion proteins that can facilitate labeling, immobilization, detection and / or isolation. Indirect labeling includes the use of a protein, such as a labeled antibody, which specifically binds to a polypeptide of the present invention. These antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab fragments and fragments produced by a Fab expression library. The use of cMAC as a drug target, in screening assays, and for the identification of cMAC modulators The present invention describes for the first time that cMAC is a useful drug target for therapeutic agents for the treatment of pathological conditions related to activation abnormality of T cells. The description states that modulators (e.g., agonists or inhibitors) of the activity and / or expression of cMAC can have many significant therapeutic uses. Pathological conditions that can be treated with cMAC modulators include, but are not limited to, disorders associated with cMAC (as defined
previously) . Selection In yet another aspect, the present invention relates to a method for identifying modulators useful for treating the pathological conditions discussed above comprising testing the ability of a candidate modulator to inhibit or improve the activity of cMAC and / or inhibit or improve expression. of cMAC in vi tro, ex vivo or in vivo. Based on the present disclosure, conventional selection tests (eg, in vitro, ex vivo and in vivo) can be used to identify modulators that inhibit or enhance the activity of the cMAC protein and / or inhibit or improve the expression of cMAC. Many formats for these tests are available and are known to those skilled in the art. In general terms, these tests are based on radio-labeling, fluorescence, luminescence, substrate accumulation and a wide variety of other basic formats. The tests can be designed to employ the target protein in a multiple cell, whole cell, partially purified, purified cell extract format. Generally, tests can be designed as high performance or low performance. The activity of the target protein can be measured directly or indirectly. The cMAC activity that could be measured in a test includes any activity such as a function or biological activity of the cMAC polypeptide set forth herein
description, which includes the functional activation of T cells. Other biological activities of cMAC that can be improved or inhibited in a screening assay include the nuclear transposition of TORC, the nuclear transposition of NFAT or the increased expression of NFAT-dependent genes transcribed markers or reporters, and gene expression driven by CRE (cAMP Response Element), markers of T cell activation such as ICOS, CD69, CD40L and CD25. cMAC can also function as an ion channel, for example a calcium channel (voltage gate or ligand gate); and may have activity in the calcium-dependent activation of a T cell. In this way, the cMAC activity could be monitored by the effects on the influx or efflux in the cell culture. At a more specific biochemical level, the biological activities that could also be tested include interactions with membranes and components of cell membranes, as an objective for myristilization, glycosylation, phosphorylation, de-phosphorylation and other post-translational modifications. Based on the description herein, those skilled in the art will be able to identify these and other biological activities of cMAC, any of which could result in a convenient screening test. These tests typically employ controls, such as negative and / or positive controls that establish the background activity of cMAC. Potential agents, such as small molecules, antibodies or antibody fragments, and the like,
they are tested sequentially in the test to identify the agents that generate a measurable effect on the activity of interest compared to the controls. Those skilled in the art are familiar with the testing of agents, particularly large agent libraries, in selection formats which may be high performance or low performance. The invention therefore comprises: A method for identifying a compound useful for the treatment of a disorder associated with cMAC comprising contacting a test compound with cMAC; and detecting a change in a biological activity of cMAC as compared to the cMAC not in contact with the test compound, wherein upon detecting a change, the test compound is identified as being useful for the treatment of the disorder. A method for identifying a compound useful for the treatment of a cMAC-associated disorder comprising contacting a test compound with cMAC under sample conditions permissive for a biological activity of cMAC that determines the level of a biological activity of cMAC in vitro or live; compare the level with that of a control sample lacking the test compound; and, selecting a test compound that causes the level to change to further prove as a potential agent for the treatment of the disorder; and a method for testing whether a compound modulates a biological activity of cMAC comprising: contacting in vitro or in vivo a
test compound with cMAC; and detecting a change in a biological activity of cMAC compared to cMAC not contacted with the test compound, wherein the detection of a change identifies the test compound as a modulator of the biological activity of cMAC. In one embodiment, the method identifies inhibitors of the biological activity of cMAC. The biological activity can be selected from ion transport, ion diffusion, protein-cMAC interaction or modification of cMAC, calcium-dependent activation of a T cell, nuclear transposition of TORC or NFAT or another calcium-dependent molecule, activation of the calcineurin trajectory, and gene expression driven by CRE (cAMP Response Element) or NFAT. The invention further includes identifying and confirming whether a compound is useful for the treatment of a disorder associated with cMAC which comprises administering a compound identified in an in vitro selection assay with an animal model of this disorder associated with cMAC and observing a desired response in the animal. As contemplated in this, the present invention includes a method for using the gene and the cMAC gene product described herein to discover agonists and antagonists that induce or repress, respectively, the activity of TORC, the activation of NFAT (nuclear factor of activated T cells). , and / or activation of T cells, and will result in several therapeutic effects.
In other modalities, the invention relates to a method for identifying a useful compound for the treatment of a cMAC-related disorder comprising (a) contacting a test compound with cMAC; and (b) detecting a change in a biological activity of cMAC compared to cMAC not contacted with the test compound, wherein detecting a change identifies the test compound as useful for the treatment of the disorder. The invention includes a method for identifying a useful compound for the treatment of a cMAC-related disorder comprising (a) contacting a test combo with cMAC under sample conditions permissive for the biological activity of cMAC.; (b) Determine the level of a biological activity of cMAC; (c) comparing the level with that of a control sample lacking the test compound; and, (d) selecting a test compound that causes the level to change to further prove it as a potential agent for the treatment of the disorder. Alternatively, the invention relates to a method for testing whether a compound modulates a biological activity of cMAC that comprises (a) contacting a test com- pound with cMAC; and (b) detecting a change in a biological activity of cMAC compared to cMAC not contacted with the test compound, wherein detecting a change identifies the test compound as a modulator of the biological activity of cMAC. As it is related anywhere in the present, the change
to be identified may be a reduction of a biological activity, such as a reduction in ion transport, ion diffusion, protein-cMAC interaction or modification of cMAC, calcium-dependent activation of a T cell, activation of a cell T, or markers of T cell activation including but not limited to (ICOS, CD69, CD25, CD40L), nuclear transposition of NFAT or TORC, gene expression driven by CRE (cAMP Response Element) and expression of gene driven by NFAT or TORC. The invention further comprises the use of any compound identified by a screening assay method herein in the treatment of a disorder associated with cMAC. The invention includes a method for identifying modulators useful for treating a disorder comprising testing the ability of a candidate modulator to improve or inhibit the expression of a cMAC protein. Many formats for these tests are known to those skilled in the art. Inhibitors of cMAC expression can be identified by testing candidate modulators to determine their ability to inhibit transcription, processing, export to the cytosol, stability, translation of cMAC mRNA (or any of the numerous substeps involved in these processes ). Finally, inhibitors of expression are identified by a reduction in the amount of functionally active cMAC protein compared with controls. The
Expression enhancers can improve any of the steps that lead to expression and ultimately result in an increase in the amount of functionally active cMAC protein. Typical expression tests include promoter activity tests. In a standard promoter test, a vector comprising all or part of the cMAC promoter sequence of SEQ ID NO.3 operably linked to a reporter gene sequence (encoding a reporter protein) such as CAT (Chloramphenicol acetyl) is constructed. transferase) or luciferase. The promoter sequence of SEQ ID NO. 3 which does not include the sequences corresponding to the cDNA sequence of SEQ ID NO. 1. The vector is transfected into a cell or cell extract. The candidate modulators are tested to determine if they increase promoter activity by measuring the activity of the reporter protein compared to the controls. Those skilled in the art are aware of and have access to numerous other promoter activity testing formats. The expression of cMAC gene (e.g. mRNA levels) can also be determined using methods known to a person skilled in the art., which include, for example, conventional Northern analysis or commercially available micro-arrays. In addition, the effect of a test compound on cMAC levels and / or regulatory protein levels related to an ELISA antibody-based test or fluorescent labeling reaction test can be detected. These
Techniques are readily available for high performance selection and are known to a person skilled in the art. The promoter fragment can also be easily inserted into any less promoter reporter gene vector designed for expression in human cells (e.g., the improved fluorescent vector minus Clontech promoter pECFP-1, pEGFP-1, or pEYFP, Clontech, Palo Alto , CA). The selection would then consist of culturing the cells for an appropriate length of time with a different compound added to each well and then testing for reporter gene activity. In another embodiment, a test for modulators of cMAC expression comprises first selecting cell lines to find those expressing the cMAC protein of interest. Recombinant cell lines that contain and express an exogenous cMAC gene can also be tested. These cell lines could be grown, for example, in trays of 96, 384 or 1536 wells. A comparison of the effects of some known modifiers of gene expression, for example dexamethasone, phorbol ester, heat shock in primary tissue cultures and cell lines will allow the selection of the most appropriate cell line to use. The selection would consist merely in cultivating the cells for a fixed duration of time with a different compound added to each well and then testing the activity of cMAC.
The data collected from these studies can be used to identify modulators with therapeutic utility for the treatment of pathological conditions mentioned above; for example, other inhibitory substances could be tested in conventional in vitro or in vivo models of these pathological conditions and / or in clinical trials with humans with these pathological conditions according to conventional methods to assess the ability of these compounds to treat the conditions pathological live. The present invention, by making available critical information regarding the active portions of the cMAC polypeptides, allows the development of modulators of the cMAC function eg antibodies, antibody fragments, agonists or small molecule antagonists, em pl by a rational drug design known to those skilled in the art. U se of cMAC modulators in the treatment of disorders associated with cMAC In another aspect, the invention relates to a method for treating disorders associated with cMAC which comprises administering to a subject in need thereof, a pharmaceutical composition comprising an effective amount of a modulated r of c ^ AC. It is contemplated that the modulators identified and discovered through the cMAC screening tests described herein could be agents such as small molecules, which include small organic molecules (with or without characteristics).
similar to drugs), and that include natural products. The modulators of cMAC include agonists of the biological activity of cMAC or of the expression of cMAC; The modulators also include inhibitors of the biological activity of cMAC and / or inhibitors of cMAC expression. Other details are provided elsewhere in the present. Use of cMAC modulators to modulate biological processes The invention describes methods for inhibiting biological processes. In one embodiment, the invention relates to a method for inhibiting the biological activity of cMAC in a cell. This inhibition can be achieved by contacting a cell with a cMAC inhibitor, such as an anti-cMAC antibody, antibody fragment, or polypeptide comprising a specific binding region for cMAC or with a nucleic acid that reduces the expression of cMAC. The biological activity that is inhibited is selected from the group consisting of calcium-dependent activation of a T cell, nuclear transposition of TORC, gene expression driven by CRE (cAMP Response Element), and expression of genes / proteins inducible by NFAT such as IL-2. Alternatively, the biological activity may be a method of selectively inhibiting lymphocyte activity in a multi-cellular organism comprising contacting this organism with an anti-cMAC antibody, antibody fragment, or polypeptide comprising a specific binding region for cMAC or with a
nucleic acid that reduces the expression of cMAC. 'Selectively' as used herein means that it tends to select the identified tissue or cell type in preference to other tissues or cell types. With respect to methods of improving biological processes, the invention also relates to a method for improving T cell activation comprising contacting a T cell or a T cell precursor cell with a purified cMAC polypeptide, a therapy vector of gene comprising the cMAC gene, or an enhancer of cMAC gene expression. Other details on the use of cMAC modulators to modulate biological processes are provided elsewhere in the present. Antibodies to cMAC Convenient antibodies to cMAC proteins can be produced according to conventional methods. For example, methods for the production of antibodies capable of specifically recognizing one or more differentially expressed gene epitopes are described herein. These antibodies may include, but are not limited to, polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F (ab ') 2 fragments, fragments produced by an Fab expression library, anti-idiotypic antibodies (anti-ld), and fragments that bind epitopes of any of the foregoing, all as
described in the definition of 'antibody', supra. For the production of antibodies to the cMAC polypeptides mentioned herein, different host animals can be immunized by injection with the polypeptides, or a portion thereof. These host animals may include, but are not limited to, rabbits, mice and rats. Various adjuvants may be used to increase the immune response, depending on the host species, including, but not limited to, Freund's adjuvant (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, orifice limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (Bacillus Calmette-Guerin) and Corynebacterium parvum. Antibodies binding the cMAC polypeptides described herein can be prepared using full-length cMAC polypeptides or fragments containing small peptides of interest as the immunizing antigen. The polypeptides or peptides used to immunize an animal can be derived from the translation of RNA or chemically synthesized, and can be conjugated to a carrier protein, if desired. Commonly used carriers that chemically attach to the peptides include bovine serum albumin and thyroglobulin. The coupled peptide is then used to immunize an animal (e.g., a mouse, a rat or a rabbit).
Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the serum of animals immunized with an antigen, such as the product of the target gene, or an antigenic functional derivative thereof. For the production of polyclonal antibodies, host animals, such as those described above, can be immunized by injection with the polypeptides, or a portion thereof, supplemented with adjuvants as also described above. Monoclonal antibodies, which are homogeneous populations of antibodies to a particular antigen, can be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the Kohler and Milstein hybridoma technique, (1975, Nature 256: 495-497; and U.S. Patent Number 4,376,110), the human B-cell hybridoma technique (Kosbor. et al., 1983, Immunology Today 4:72, Colé et al., 1983, Proc. Nati, Acad. Sci. USA 80: 2026-2030), and the EBV hybridoma technique (Colé et al., 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). These antibodies may be of any class of immunoglobulin including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The hybridoma that produces the mAb of this invention can be cultured in vitro or in vivo. The production of high titers of mAbs in vivo makes this the
currently preferred production method. Additionally, techniques developed for the production of "chimeric antibodies" can be used (Morrison et al., 1984, Proc. Nati, Acad. Sci., 81: 6851-6855, Neuberger et al., 1984, Nature, 312: 604- 608; Takeda et al., 1985, Nature, 314: 452-454) by separating the genes from an antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity. A chimeric antibody is a molecule in which different portions are derived from different species of animals, such as those having a variable or hypervariable region derived from murine mAb and a constant region of human immunoglobulin. Alternatively, the techniques described for the production of single chain antibodies can be adapted (U.S. Patent Number 4,946,778; Bird, 1988, Science 242: 423-426; Huston et al., 1988, Proc. Nati. Acad. Sci. USA 85: 5879-5883; and Ward et al., 1989, Nature 334: 544-546) to produce single chain antibodies from differentially expressed genes. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Preferably, techniques useful for the production of "humanized antibodies" to produce antibodies can be adapted
for the polypeptides, fragments, derivatives, and functional equivalents described herein. These techniques are described in United States of America Patents Nos. 5,932, 448; 5,693,762; 5,693,761; 5,585,089; 5,530,101; 5,910,771; 5,569,825; 5,625,126; 5,633,425; 5,789,650; 5,545,580; 5,661,016; and 5,770,429, the descriptions of all of which are hereby incorporated by reference in their entirety. Antibody fragments that recognize cMAC-specific epitopes can be generated by known techniques. For example, these fragments include, but are not limited to: the F (ab ') 2 fragments that can be produced by the pepsin digestion of the antibody molecule and the Fab fragments that can be generated by reducing the disulphide bridges of the fragments F (ab ') 2. Alternatively, Fab expression libraries can be constructed (Huse et al., 1989, Science, 246: 1275-1281) to allow rapid and easy identification of the monoclonal Fab fragments with the desired specificity. A wide variety of antibody / immunoglobulin structures or scaffolds may be employed as long as the resulting polypeptide includes one or more binding regions that are specific for the cMAC protein. These structures or scaffolds include the 5 major idiotypes of human immunoglobulins, or fragments thereof (as described elsewhere herein), and include immunoglobulins from other animal species, which preferably have humanized aspects. The
simple heavy chain antibodies such as those identified in camelids are of particular interest in this regard. The structures, scaffolds and novel fragments continue to be discovered and developed by those skilled in the art. Alternatively, known or future non-immunoglobulin structures and scaffolds may be employed, as long as they comprise a specific binding region for the cMAC protein of SEQ ID NO: 2. These compounds are referred to herein as "polypeptides that they comprise a specific binding region for cMAC. " Non-immunoglobulin structures or scaffolds include Adnectins (fibronectin) (Compound T erapeutics, Inc., Walt am, MA), ankyrin (Molecular Partners AG, Zurich, Switzerland), domain antibodies (Domantis, Ltd (Cambridge, MA) and Ablynx nv (Zwijnaarde, Belgium)), lipocalin (Anticalin) (Pieris Proteolab AG, Freising, Germany), small modular immuno-pharmaceutical products (Trubion Pharmaceuticals Inc., Seattle, WA), mammalian (Avidia, Inc. (Mountain View, CA)), Protein A (Affibody AG, Sweden) and afilin (gamma-crystalline or ubiquitin) (Scil Proteins GmbH, Halle, Germany). In accordance with the present invention, the anti-cMAC antibody or fragment thereof, or the polypeptide comprising a cMAC-specific binding region, independent of the structure or scaffold employed, can be ligated, either covalently or non-covalently, to an additional fraction. The additional fraction can be a polypeptide, an inert polymer such
as PEG, small molecule, radiosotope, metal, ion, nucleic acid or another type of biologically relevant molecule. This construct, which may be known as an immunoconjugate, immunotoxin, or the like, is also included in the meaning of antibody, antibody fragment or polypeptide comprising a specific binding region for cMAC, as used herein. The invention further relates to the use of an antibody, a cMAC-specific antibody fragment or a polypeptide comprising a cMAC-specific binding region in the treatment of a disorder in a subject as described herein. The invention also relates to an antibody or antibody fragment that specifically binds to cMAC (SEQ ID NO: 2) or a polypeptide comprising a cMAC-specific binding region, which includes an antibody fragment (e.g. F (ab ') 2) or a monoclonal antibody. The invention also covers a pharmaceutical composition of this antibody, antibody fragment or a polypeptide comprising a binding region that specifically binds to cMAC. The detection of cMAC by the antibodies described herein can be achieved using ELISA, standard FACS, and standard imaging techniques used in vitro or in vivo. Detection can be facilitated by coupling (it is
say, physically bind) cMAC to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, (3-galactosidase, or acetylcholinesterase, examples of suitable prosthetic group complexes include streptavidin / biotin and avidin / biotin, examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinyl amine fluorescein, dansyl chloride or phycoerythrin, an example of a luminescent material includes luminol, examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125 l, 13 l 35 S or 3H In particular, for its easy detection, the sandwich test is preferred, of which there are variants, all of which are intended to encompass the present invention.For example, in a typical forward test, unlabeled anti-cMAC antibody is immobilized on a solid substrate and the sample that is going to pro bar is contacted with the binding molecule. After a convenient incubation period, for a period of time sufficient to allow the formation of an antibody-antigen binary complex, a second antibody is added and incubated, labeled with a reporter molecule capable of inducing a detectable signal,
allowing sufficient time for the formation of a ternary complex of antibody-antigen-labeled antibody. Any unreacted material is discarded, and the presence of antigen is determined by observation of a signal, or it can be quantified by comparing with a control sample containing known amounts of antigen. Variations on the forward test include the simultaneous test, in which both the sample and the antibody are added simultaneously to the bound antibody, or a reverse test, in which the labeled antibody and the sample to be tested are first combined , they are incubated and added to the unlabeled surface bound antibody. These techniques are well known to those skilled in the art, and the possibility of minor variations will be readily apparent. As used herein, "the sandwich test" is intended to cover all variations on the basic technique of two sites. For the immunological tests of the present invention, the only limiting factor is that the antibody labeled is an antibody that is specific for cMAC polypeptides or related regulatory proteins, or fragments thereof. The most commonly used reporter molecules are either enzymes, molecules containing fluorophore or radioniclide. In the case of an enzyme immunological test, an enzyme is conjugated with the second antibody, usually by means of glutaraldehyde or periodate. However, as will be easily recognized, there is a wide variety of different ligation techniques, which are
well known by experts. Commonly used enzymes include horseradish peroxidase, glucose oxidase, beta-galactosidase and alkaline phosphatase, among others. The substrates to be used with the specific enzymes are generally chosen for the production, after hydrolysis by the corresponding enzyme, of a detectable color change. For example, p-nitrophenyl phosphate is convenient for use with alkaline phosphatase conjugates; for peroxidase conjugates, 1,2-phenylenediamine or toluidine is commonly used. It is also possible to employ fluorogenic substrates, which produce a fluorescent product instead of the chromogenic substrates noted above. A solution containing the appropriate substrate is then added to the tertiary complex. The substrate reacts with the enzyme linked to the second antibody, giving a qualitative visual signal, which can also be quantified, usually spectrophotometrically, to give an evaluation of the amount of polypeptide or polypeptide fragment of interest that is present in the serum sample. Alternatively, fluorescent compounds, such as fluorescein and rhodamine, can be chemically coupled to the antibodies without altering their binding capacity. When activated by illumination with light of a particular wavelength, the antibody labeled with fluorochrome absorbs the energy of the light, inducing a state of excitability in the molecule, followed by the emission of light at a longer characteristic wavelength. The
emission appears as a characteristic color visually detectable with a light microscope. The immunofluorescence and EIA techniques are very well established in the field and are particularly preferred for the present method. However, other reporter molecules can also be employed, such as radioisotopes, chemiluminescent or bioluminescent molecules. It will be readily apparent to the expert how to vary the procedure to adapt it to the required use. Therefore, the invention includes pharmaceutical compositions comprising antibodies that are highly selective for human cMAC polypeptides or portions of human cMAC, and methods for using these antibodies. After administration to a subject, these antibodies can inhibit or decrease the activity of cMAC, or in some cases they can increase the activity of cMAC, by interacting directly with the protein. Inhibitors can block active sites or block access of substrates to active sites. CMAC antibodies can also be used to inhibit cMAC activity by preventing protein-protein interactions that may be involved in the regulation of cMAC proteins and necessary for protein activity. Antibodies with inhibitory activity such as those described herein can be produced and identified according to standard tests known to those skilled in the art. CMAC antibodies can also be used for diagnosis. For example, these antibodies could be used
according to conventional methods at quantitative levels of a cMAC protein in a subject; increased levels could, for example, indicate undesirable T-cell activation, excessive activation of CRE-dependent gene expression (e.g., activation of genes that have CRE in their promoter regions) and could possibly indicate the degree of excessive activation and the corresponding severity of the related pathological condition. In this way, different levels of cMAC could be indicative of different clinical forms or severity of pathological conditions such as the disorders associated with cMAC. This information would be useful to identify subsets of patients suffering from a pathological condition that may not respond to treatment with cMAC modulators. Gene Therapy In another embodiment, nucleic acids comprising a sequence encoding a cMAC protein or a functional derivative thereof are administered for therapeutic purposes, by means of gene therapy. Gene therapy refers to therapy performed by administering a nucleic acid to a subject. In this embodiment of the invention, the nucleic acid produces its encoded protein which mediates a therapeutic effect by promoting the normal activation of T cells. Any of the methods for gene therapy available in the art can be used in accordance with the present invention. Example methods are described below.
In a preferred aspect, the therapy comprises a cMAC nucleic acid that is part of an expression vector that expresses a cMAC protein or fragment or chimeric protein thereof in a convenient host. In particular, this nucleic acid has a promoter operably linked to the coding region of cMAC, the promoter is inducible or constitutive, and, optionally, tissue-specific. In another particular embodiment, a nucleic acid molecule is used in which the coding sequences of cMAC and any other desired sequence are flanked by regions that promote homologous recombination at a desired site in the genome, thereby providing intrachromosomal expression of a cMAC nucleic acid (Koller and Smithies, 1989, Proc. Nati, Acad. Sci. USA 86: 8932-8935, Zijlstra et al., 1989, Nature 342: 435-438). The administration of the nucleic acid in a patient can be direct, in which case the patient is exposed directly to the nucleic acid or nucleic acid carrier vector, or indirectly, in which case, the cells are first transformed with nucleic acid in vitro, then They are transplanted to the patient. These two approaches are known, respectively, as gene therapy in vivo or ex vivo.
In a specific embodiment, the nucleic acid is administered directly in vivo, where it is expressed to produce the encoded product. This can be carried out by any of numerous methods known in the art, for example, by building it as part of an acid expression vector
nucleic acid and administering it to become intracellular, for example, by infection using a defective or attenuated viral vector or other viral vector (see, e.g., U.S. Patent No. 4,980,286 and others mentioned below), or by direct injection of naked DNA, or by the use of microparticle bombardment (eg, a gene gun, Biolistic, Dupont), or a coating with lipids or receptors on the cell surface or transfection agents, encapsulation in liposomes, microparticles , or microcapsules, or administering it in binding with a peptide that is known to enter the nucleus, administering it in binding to a ligand subjected to receptor-mediated endocytosis (see, for example, US Patents Numbers 5,166,320; 5,728,399; 5,874,297 and 6,030,954, which are incorporated by reference herein in their entirety) (which may be used for to read cell types that specifically express the receptors), and so on. In another embodiment, a nucleic acid-ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disorganize the endosomes, allowing the nucleic acid to prevent lysosomal degradation. In yet another embodiment, the nucleic acid can be chosen in vivo for specific cellular uptake and expression, by means of choosing a specific receptor (see, for example, PCT International Publication Numbers WO 92/06180, WO 92/22635; WO92 / 20316; W093 / 14188; and WO 93/20221). By way of
Alternatively, the nucleic acid can be introduced intracellularly and incorporated into the DNA of the host cell for expression, by homologous recombination (see, for example, U.S. Patent Nos. 5,413,923, 5,416,260, and 5,574,205; and Zijlstra et al., 1989, Nature 342: 435-438). In a specific embodiment, a viral vector containing a cMAC nucleic acid is used. For example, a retroviral vector can be used (see, for example, U.S. Patent Nos. 5,219,740, 5,604,090, and 5,834,182). These retroviral vectors have been modified to eliminate retroviral sequences that are not necessary to package the viral genome and integrate it into the DNA of the host cell. The cMAC nucleic acid to be used in gene therapy is cloned into a vector, which facilitates the administration of the gene in a patient. Adenoviruses are other viral vectors that can be used in gene therapy. Adenoviruses are especially attractive vehicles for administering genes to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia where they cause mild disease. Other targets for adenovirus-based delivery systems are the liver, central nervous system, endothelial cells, and muscles. Adenoviruses have the advantage that they are capable of infecting cells that are not dividing. The methods to carry out
adenovirus-based gene therapy are described, for example, in US Pat. Nos. 5,824,544; 5,868,040; 5,871,722; 5,880,102; 5,882,877; 5,885,808; 5,932,210; 5,981,225; 5,994,106; 5,994,132; 5,994,134; 6,001,557; and 6,033.8843, all of which are hereby incorporated by reference in their entirety. Adeno-associated viruses (AAV) have also been proposed for use in gene therapy. Methods for producing and using AAV are described, for example, in U.S. Patent Nos. 5,173,414; 5,252,479; 5,552,311; 5,658,785; 5,763,416; 5,773,289; 5,843,742; 5,869,040; 5,942,496; and 5,948,675, all of which are incorporated by reference herein in their entirety. Another approach to gene therapy involves transferring a gene to cells in tissue culture by methods such as electroporation, lipofection, calcium phosphate-mediated transfection, or viral infection. Usually, the transfer method includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate the cells that have absorbed and express the transferred gene. Those cells are then administered to the patient. In this embodiment, the nucleic acid is introduced into a cell prior to the in vivo administration of the resulting recombinant cell. This introduction can be carried out by any method known in the art, which includes but is not limited to
transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequence, cell fusion, chromosome-mediated gene transfer, micro-cell-mediated gene transfer, spheroplast fusion, and so on. Numerous techniques are known in the field for the introduction of foreign genes into cells and can be used according to the present invention, provided that the necessary physiological and development functions of the recipient cells are not interrupted. The technique should provide stable transfer of the nucleic acid to the cell, so that the nucleic acid can be expressed by the cell and preferably heritable and expressible by the progeny of the cell. The resulting recombinant cells can be administered to a patient by various methods known in the art. In a preferred embodiment, epithelial cells are injected, for example, subcutaneously. In another embodiment, recombinant skin cells can be applied as a skin graft on the patient. Preferably, recombinant blood cells (e.g., hematopoietic stem or progenitor cells) are administered intravenously. The amount of cells considered for use depends on the desired effect, the condition of the patient, etc., and can be determined by a person skilled in the art. Cells into which a nucleic acid can be introduced for gene therapy purposes encompass any type of available, desired cell, and include but are not limited to cells
epithelial, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem, for example, such as those obtained from bone marrow, umbilical cord blood, peripheral blood, fetal liver, and the like. In a preferred embodiment, the cell used for gene therapy is autologous to the patient. In an embodiment in which recombinant cells are used in gene therapy, a cMAC nucleic acid is introduced into the cells in a manner that is expressible by the cells or their progeny, and then the recombinant cells are administered in vivo for therapeutic effect . In a specific embodiment, stem or progenitor cells are used. Any stem-and / or progenitor cell that can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention. These stem cells include but are not limited to hematopoietic stem cells (HSC)stem cells from epithelial tissues such as the skin and lining of the intestines, embryonic cardiac muscle cells, liver stem cells (see, for example, WO 94/08598), and stem cells (Stemple and Anderson, 1992, Cell 71: 973-985). Epithelial stem cells (ESCs) or keratinocytes can be obtained from tissues such as the skin and the coating
of the intestines by known procedures (Rheinwald, 1980, Meth Cell Bio 21A: 229). In stratified epithelial tissue like skin renewal occurs by mitosis in the stem cells within the germ layer, the layer closest to the basal lamina. Stem cells within the lining of the intestines provide rapid turnover of tissue. ESC or keratinocytes obtained from the skin or from the lining of the intestines of a patient or donor can be cultured in tissue cultures (Pittelkow and Scott, 1986, Mayo Clinic Proc. 61: 771). If the ESCs are provided by a donor, a method for suppressing the host reaction against the graft (eg, irradiation, drug administration or antibody to promote moderate immunosuppression) can also be used. With respect to hematopoietic stem cells (HSC), any technique that provides for the isolation, propagation, and in vitro maintenance of HSC can be used in this embodiment of the invention. Techniques by which this can be done include (a) the isolation and establishment of HSC cultures from bone marrow cells isolated from the future host or a donor, or (b) the use of HSC cultures previously established in the long term, which may be allogeneic or xenogeneic. Preferably, non-autologous HSCs are used together with a method of suppressing immune reactions to transplantation of the future host / patient. In a particular embodiment of the present invention, human bone marrow cells can be obtained from
from the posterior iliac crest by needle aspiration (see, for example, Kodo et al., 1984, J. Clin Invest 73, 1377-1384). In a preferred embodiment of the present invention, the HSC can be made in highly enriched or substantially pure form. This enrichment can be carried out before, during, or after long-term cultivation and can be done by any technique known in the field. Long-term cultures of bone marrow cells can be established and maintained using, for example, modified Dexter cell culture techniques (Dexter et al., 1977, J. Cell Physiol. 91: 335) or Witlock culture techniques. -Witte (Witlock and Witte, 1982, Proc. Nati, Acad. Sci. USA 79: 3608-3612). In a specific embodiment, the nucleic acid to be introduced for gene therapy purposes comprises an inducible promoter operably linked to the coding region, such that the expression of the nucleic acid is controllable by controlling the absence or presence of the appropriate inducer. of the transcription. The pharmaceutical compositions of the present invention may also comprise substances that inhibit the expression of cMAC proteins at the level of the nucleic acid. These molecules include ribozymes, antisense oligonucleotides, triple helix DNA, RNA aptamer, siRNA, and double or single stranded RNA, directed to an appropriate nucleotide sequence of cMAC nucleic acid. These inhibitory molecules can be created using
conventional techniques by one skilled in the art without undue burden or experimentation. For example, modifications (e.g., inhibition) of gene expression can be obtained by designing antisense molecules, DNA or RNA, for the control regions of a gene encoding a cMAC peptide mentioned herein, i.e., for promoters, enhancers, and introns. For example, oligonucleotides derived from the transcription initiation site can be used, for example, between positions -10 and + 10 from the start site. In any case, all regions of the gene can be used to design an antisense molecule in order to create those that give the strongest hybridization to the mRNA and these convenient oligonucleotides can be produced and identified by test procedures known to those skilled in the art. technique. In a similar way, inhibition of expression gene expression can be achieved using a "triple helix" base pairing methodology. Triple-helix pairing is useful because it causes the inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. Recent therapeutic advances using triple DNA have been described in the literature (Gee, J. E. et al. (1994) In: Huber, B.E. and B. I. Carr, Molecular and Immunologic Approaches, Futura Publishing Co., Mt. Kisco, N.Y.). These molecules can also be designed to block the translation of the mRNA, preventing
the transcription link to the ribozymes. Ribozymes, enzymatic RNA molecules, can also be used to inhibit the expression of the gene by catalyzing the specific dissociation of RNA. The mechanism of ribozyme action involves the sequence-specific hybridization of the ribozyme molecule to choose the complementary RNA followed by the endonucleolytic dissociation. Examples that include ribozyme molecules with "hammerhead" or "hairpin" motifs can be used to specifically and efficiently catalyze the endonucleolytic cleavage of the gene sequence, for example, mRNA for cMAC. The specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule to find sites that include the following sequences: GUA, GUU and GUC. As soon as they are identified, short RNA frequencies of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the solution site can be evaluated to find secondary structural features that can render the oligonucleotide inoperable. The suitability of the candidate targets can also be evaluated by testing the accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays. The ribozyme methods include exposing a cell to ribozymes or introducing the expression into a cell of these molecules
of small RNA ribozyme (Grassi and Marini, 1996, Annals of Medicine 28: 499-510; Gibson, 1996, Cancer and Metastasis Reviews 15: 287-299). The intracellular expression of the hammerhead and hairpin ribozymes chosen for the mRNA corresponding to at least one of the genes mentioned herein can be used to inhibit the protein encoded by the gene. The ribozymes can be delivered directly to the cells, in the form of RNA oligonucleotides that incorporate ribozyme sequences, or they can be introduced into the cell as an expression vector encoding the desired ribozimal RNA. Ribozymes can routinely be expressed in vivo in numbers sufficient to be catalytically effective in dissociating the mRNA, and thereby modifying the abundance of the mRNA in the cell (Cotten et al., 1989 EMBO J. 8: 3861-3866). In particular, a DNA sequence encoding ribozymes, designed in accordance with conventional, well-known rules, can be ligated and synthesized, for example, by standard phosphoramidite chemistry, at a restriction enzyme site in the anticodon stem and the a gene encoding a tRNA, which can then be transformed and expressed in a cell of interest by routine methods in the art. Preferably, an iible promoter (eg, a glucocorticoid or a tetracycline response element) is also introduced into this construct so that the expression of the ribozyme can be selectively controlled. For saturation use, a highly constitutively active promoter can be used. The
cDNA genes (ie, the genes encoding tRNA) are useful in this application due to their small size, high transcription rate and ubiquitous expression in different tissue classes. Therefore, ribozymes can be routinely designed to dissociate virtually any mRNA sequence, and a cell can routinely be transformed with DNA encoding these ribozyme sequences so that a controllable and catalytically effective amount of the ribozyme is expressed. In accordance with the foregoing, the abundance of any RNA species can be virtually modified or disturbed. The ribozyme sequences can be modified in essentially the same manner as described for the antisense nucleotides, for example, the ribozyme sequence can comprise a modified base moiety. RNA aptamers can also be introduced or expressed in a cell to modify the abundance or activity of the RNA. RNA aptamers are RNA ligands for proteins, such as for Tat and Rev RNA (Good et al., 1997, Gene Therapy 4: 45-54) that can specifically inhibit their transduction. Gene-specific inhibition of gene expression can also be achieved using RNA interference strategies
("iNAR"). RNA is a relatively new discovery that is based on double-stranded RNA. A description of that technology can be found in the International Publication Number WO
99/32619 which is incorporated by reference herein in its entirety. IRNA technology has been shown to be useful in inhibiting gene expression (see, eg, Cuiten, BR Nat. Immunol., 2002 Jul; 3 (7): 597-9). An iRNA agent as used herein refers to compounds and compositions that can act through an RNA mechanism (see, as a general reference, He and Hannon, (2004) Nat. Genet 5: 522-532) . Commonly used iRNA agents such as short interfering RNA ("siRNA"), double-stranded RNA ("dsRNA"), short-hair RNA ("shRNA", also sometimes called 'synthetic RNA'), others are Developing. When introduced into a cell or synthesized within a cell the iRNA agents are incorporated into a macromolecular complex using the iRNA agent to select and dissociate RNA strands containing the complementary (or substantially complementary) sequence. IRNA agents can be chemically modified. A variety of chemical modifications known to those skilled in the art are presented in PCT International Publication Number WO 03/070918, incorporated herein by reference. Other modifications and combinations of modifications that do not eliminate the iRNA activity of the compound are also contemplated herein. Suitable iRNA agents for use in the invention include the dsRNA strands resulting from the hybridization of the
single chain sense and antisense chains indicated in Table 5, and Table 6 (see Examples); (See Examples, note that the sequences must be synthesized as RNA (not DNA), and optionally chemically modified). RNA agents can be prepared based on Table 5 or Table 6, or as otherwise designed by one skilled in the art, from 17 to 30 mer of double-chain compounds can be shortened, with or without draperies. 3 'of 1-6 nts, with or without chemical modifications or extreme modifications, and with or without exact complementarity to the target sequence, in which case they are referred to herein as short interfering RNA compounds ("siRNA"). The preferred siRNA compounds, as calculated using the Biopred algorithm (Huesken et al. (2005) Nat.Biotech, 23 (8): 995-1001) are: Table 5:
The invention further relates to the use of an iRNA agent, such as a siRNA specific for cMAC, in the treatment of a disorder in a subject. The antisense molecules, the triple helix ARD, the RNA aptamers and ribozymes of the present invention can be prepared by any method known in the art by the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligonucleotides such as chemical synthesis of solid phase phosphoramidite. Alternatively, RNA molecules can be generated by in vitro and in vivo transcription of DNA sequences encoding the genes of the polypeptides mentioned herein. These DNA sequences can be incorporated into a wide variety of vectors with convenient RNA polymerase promoters such as T7 or SP6. Alternatively, cDNA constructs that synthesize antisense RNA constitutively or inducibly in cell lines, cells or tissues can be introduced. Peptide mimetics It could be predicted that the peptide mimetics of cMAC proteins act as modulators of cMAC. Thus, one embodiment of this invention are peptides derived or designed from cMAC which block the cMAC function. Suitable peptide mimetics for cMAC proteins can be made according to conventional methods based on an understanding of the regions in the polypeptides required for
the protein activity of cMAC. In summary, a short amino acid sequence is identified in a protein by studies of conventional structural functions such as suppression or mutation analysis of the original type protein. As soon as the critical regions are identified, it is anticipated that if they correspond to a highly conserved portion of the protein that this region will be responsible for a critical function (such as a protein-protein interaction). A small synthetic mimic that is designed to look like the critical region could be predicted to compete with the intact protein and thus interfere with its function. The synthetic amino acid sequence could be composed of amino acids that coincide with this region in whole or in part. These amino acids could be replaced with other chemical structures that resemble the original amino acids but impart pharmacologically better properties such as higher inhibitory activity, stability, half-life or better biological availability. Small Molecules It is contemplated that the modulators identified and discovered through the cMAC screening tests described herein could be agents such as small molecules, which include small organic molecules (with or without drug-like characteristics), and which include products natural These small molecule modulators of cMAC include agonists of the biological activity of cMAC or expression of cMAC; as well
they may include inhibitors of the biological activity of cMAC or inhibitors of cMAC expression. Those skilled in the art are familiar with library analyzes of natural compounds, semi-synthetic compounds or libraries of combinatorial compounds, in formats of low yield, medium yield and high yield, and ultra high performance. The compounds found to modulate the activity of cMAC, compared to the control compounds, are identified and grouped by their structure. The chemical structures are then modified and tested for another activity in the test. The structure-activity relationship (SAR) of the compound for the objective is evaluated. The compounds with high potency and high selectivity for the target are developed. These compounds are rigorously tested in a battery of other tests before being tested on animals and humans to determine the therapeutic effect. All these steps are well known to those skilled in the art. Other uses of cMAC investigations It is also noted that the present invention is useful for further investigation. For example, the cDNA encoding the cMAC proteins and / or the cMAC proteins themselves can be used to identify other proteins, for example, kinases, proteases, or transcription factors, which are modified or indirectly activated by a protein cascade. cMAC. The proteins identified in this way can be used and identified, for example, for the selection of drugs to treat pathological conditions
mentioned in this. To identify these genes that are downstream of cMAC proteins, it is contemplated, for example, that conventional methods can be used to treat animals in disease state models with a specific cMAC inhibitor, sacrifice animals, remove relevant tissues and isolate the total RNA of these cells and employing standard microarray assay technologies to identify message levels that are altered in relation to a control animal (animal to which no drug has been administered). Additionally, conventional in vitro or in vivo tests can be used to identify possible genes that lead to overexpression of cMAC proteins. These related regulatory proteins encoded by genes identified in this way can be used to select drugs that could be potent therapies for the treatment of the pathological conditions mentioned herein. For example, a conventional reporter gene test could be used in which the promoter region of a cMAC protein is placed upstream of a reporter gene, the construct is transfected into a convenient cell (eg, from ATCC, Manassas, VA) and using conventional techniques, the cells are tested for an upstream gene that causes activation of the cMAC promoter by detecting the expression of the reporter gene. It is contemplated herein, that the function and / or expression of a gene for a related regulatory protein can be inhibited.
or protein that modifies cMAC as a way to treat the pathological conditions described herein by, for example, designing antibodies for these proteins or peptide mimetics and / or designing inhibitory antisense oligonucleotides, triple helix DNA, ribozymes, siRNA, double-stranded RNA or simple and RNA aptamers directed to the genes for these proteins according to conventional methods. Also contemplated are pharmaceutical compositions comprising these inhibitory substances for the treatment of pathological conditions. Pharmaceutical Compositions and their Administration One embodiment of the invention relates to the administration of a pharmaceutical composition, together with a pharmaceutically acceptable carrier, excipient or diluent, for the treatment of any of the pathological conditions described herein. These pharmaceutical compositions can comprise any of the cMAC modulators described herein, including the cMAC protein, or fragments thereof, antibodies to the cMAC polypeptides, nucleic acids (e.g., gene therapy vectors, antisense agents, ribozyme or iRNA) ), cMAC peptide mimetics, small molecule modulators, and any other cMAC modulator (eg, agonists, antagonists, or inhibitors of cMAC expression and / or function). The compositions can be administered alone or in combination with at least one other agent, such as a stabilizing compound that can be administered in any carrier
biocompatible, sterile pharmaceutical, which includes, but is not limited to,? saline solution, regulated saline solution, dextrose, and water. The compositions can be administered to a patient, alone or in combination with other agents, drugs or hormones. The present invention also comprises a method for treating a disorder in a subject which comprises administering to the subject an effective amount of an agent, or a pharmaceutical composition of an agent, which inhibits or enhances the activity or expression of cMAC. Pharmaceutical compositions comprising cMAC modulators thereof can be administered when they are considered medically beneficial by one skilled in the art, for example, in conditions where the agonists of cMAC function have a therapeutic effect such as in neurodegenerative disorders such as Alzheimer, Parkinson and Huntington. These pharmaceutical compositions for use in the present invention can be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients. The pharmaceutical compositions described herein as useful for preventing, treating or alleviating pathological conditions described herein will be administered to a patient at therapeutically effective doses. A therapeutically effective dose refers to the amount of compound sufficient to result in the prevention, treatment or amelioration of these conditions.
The compounds and their physiologically acceptable salts can be formulated for administration by inhalation or insufflation either through the mouth or nose) or topical, oral, buccal, parenteral or rectal administration. For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared with conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (for example, lactose, microcrystalline cellulose or acid calcium phosphate); lubricants (for example, magnesium stearate, talc or calcium oxide); disintegrants (e.g., potato starch, sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulfate). The tablets can be coated by methods well known in the art. Liquid preparations for oral administration may take the form, for example, of solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. These liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (eg, sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (for example, lecithin or acacia); non-aqueous vehicles (eg, almond oil, fatty esters, ethyl alcohol
or fractionated vegetable oils); and preservatives (for example, methyl or propyl-p-hydroxybenzoate or sorbic acid). The preparations may also contain regulatory salts, flavors, colorants, and sweetening agents as appropriate. Preparations for oral administration can conveniently be formulated to give controlled release of the active compound. For oral administration the compositions can take the form of tablets or troches formulated in a conventional manner. For administration by inhalation, the compounds for use in accordance with the present invention are conveniently administered in the form of an aerosol spray presentation, from pressurized packings or nebulizer, with the use of a suitable propellant, for example, dichlorodifluoromethane , trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of pressurized aerosol the metering unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, for example, gelatin for use in the inhaler or insufflator can be formulated containing a powder mixture of the compound and a convenient powder base such as lactose or starch. The compounds can be formulated for parenteral administration by injection, for example, by bolus injection or continuous infusion. Formulations for injection may be presented in a single dosage form, for example, in ampoules
or in multiple dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions, or emulsions in greasy or aqueous vehicles and may contain formulating agents such as suspending, stabilizing and / or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, eg, sterile, pyrogen-free water, before use. The compounds can also be formulated in rectal compositions such as suppositories or retention enemas, for example containing conventional suppository bases such as cocoa butter or other glycerides. In addition to the formulations described above, the compounds can be formulated as a depot preparation. These long acting formulations can be administered by implant (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as poorly soluble derivatives, for example, as a poorly soluble salt . The compositions, if desired, may be presented in a package or in a dosing device that may contain one or more unit dosage forms containing the ingredient
active. The package may comprise aluminum sheet or plastic sheet, such as a blister pack. The package or the dosing device can be accompanied with instructions for its administration. Suitable pharmaceutical compositions for use in the invention, include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. The determination of an effective dose is very well within the ability of those skilled in the art. For any compound, the effective therapeutic dose can be estimated initially in cell culture tests, for example, neoplastic cells, or in model animals, usually mice, rabbits, dogs or pigs. The animal model can also be used to determine the appropriate concentration range and route of administration. A dose can be formulated in animal models to achieve a range of circulating plasma concentration that includes IC50 (ie, the concentration of a test compound that achieves maximum inhibition of symptoms). This information can then be used to determine useful doses and routes of administration in humans. A therapeutically effective dose refers to the amount of the active ingredient useful to prevent, treat or ameliorate a particular pathological condition of interest. The therapeutic efficacy and toxicity can be determined by standard pharmaceutical procedures in cell or animal cultures
experimental, for example, ED50 (the therapeutically effective dose in 50 percent of the population) and LD50 (the lethal dose for 50 percent of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, and can be expressed as the ratio, LD50 / ED50. Pharmaceutical compositions that exhibit large therapeutic indices are preferred. The data obtained in cell cultures and animal studies are used to formulate a dosage range for human use. The dosage contained in these compositions is preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage varies within this range depending on the dosage form employed, the sensitivity of the patient, and the route of administration. The exact dose will be determined by the doctor, in view of the factors related to the subject that requires the treatment. The dosage and administration are adjusted to provide sufficient levels of the active fraction or to maintain the desired effect. The factors that must be taken into account are the seriousness of the disease state, the general health of the subject, the age, weight and gender of the subject, diet, time and frequency of administration, the combination or combinations of drugs, reaction sensitivities and tolerance / response to therapy. The long-acting pharmaceutical compositions can be administered every 3 to 4 days, every week, or once every two weeks depending on
the half-life and the elimination rate of the particular formulation. Normal dosage amounts may vary from 0.1 to 100,000 micrograms, up to a total dose of approximately 1 gram, depending on the route of administration. The guidance on particular dosages and methods and methods of administration is provided in the literature and is generally available to treating physicians. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, the administration of polynucleotides or polypeptides will be specific for cells, conditions, particular locations, and so on. Suitable pharmaceutical formulations for the oral administration of proteins are described, for example, in U.S. Patent Nos. 5,008,114; 5,505,962; 5,641,515; 5,681,811; 5,700,486; 5,766,633; 5,792,451; 5,853,748; 5,972,387; 5,976,569; and 6,051, 561. It is contemplated herein that monitoring cMAC levels or activity and / or detection of cMAC expression (mRNA levels) can be used as part of the clinical testing procedure, for example, to determine the efficacy of a regimen of given treatment. For example, patients to whom drugs have been administered would be evaluated and the physician would be able to identify those patients in whom the levels, activity and / or expression levels of cMAC are higher than desired (e.g. , the levels higher or lower than the
levels in control patients who do not experience a related disease state or in patients in whom the disease state has been sufficiently alleviated by clinical intervention). Based on these data the doctor could adjust the dosage, the administration regime or type of prescribed medicine. Factors to be considered in optimizing a therapy for a patient include the particular condition being treated, the particular mammal being treated, the clinical condition of the individual patient, the site of delivery of the active compound, the particular type of the active compound, the method of administration, the schedule of administration, and other factors known to medical practitioners. The therapeutically effective amount for an active compound to be administered will be subject to these considerations and is the minimum amount necessary for treatment according to the pathological condition. The following examples illustrate the present invention and are not intended to limit it. Examples General methods Selection of transposition of TORC-1 The selection was made as described (Bittinger et al Curr Biol. 2004 Dec 14; 14 (23): 2156-61.) Briefly, a fluorescent fusion (Tord-eGFP) was constructed and the construct cotransfected with 7680 individual cDNA clones (predominantly from the MGC clone collection) in cells
HeLa. The nuclear transposition of the Torc fusion protein was assessed using an automated fluorescence microscope platform. Quantification of the TORC-eGFP transposition: A stable expression HeLa cell line expressing Torc1-GFP was prepared and the cells were seeded (6000 cells per well in a 96-well tray), and transduced with lentiviral constructs (pLLB1-GW- Kan) containing the empty vector (translation stop sequence), TRPV6 or human cMAC. 48 hours after transduction, the cells were treated with or without cyclosporine (5 μ?) For one hour before fixing them, forming their image and quantifying using a Cell II fix II (the fixation procedure is seen below) . The difference between nuclear cytoplasmic fluorescence intensity and cytoplasmic fluorescence intensity was determined from 500 cell images per well. Moloney retrovirus expression particle packaging: 24 hours before transfection, 1.47X106 GP2 packaging cells were seeded. -293 on PDL plates (poly-d-lysine in 6-well plates, Becton Dickinson) 10 percent serum without antibiotics. 2.5 micrograms of QL-GW-final-kan construction expression vector DNA and 2.5 micrograms of plasmid pvpackVSV-G (Stratagene) were combined in a final volume of 250 microliters Optimem (Life Technologies). 12 microliters of Lipofectamine 2000 reagent were mixed in a final volume of
250 microliters of Optimem and incubated for 5 minutes at room temperature. The diluted DNA was combined with the diluted lipofectamine (20 minutes at room temperature). The complex was added to the GP2 cells (in 2 milliliters of medium without antibiotics) and incubated overnight. The next day the medium was removed and refilled with new medium containing antibiotics. 48 hours after transfection the medium containing the virus was collected and stored at 4 ° C; the cells were refilled with new medium. 72 hours after the transfection, the final virus collection was made and grouped with the previous collection sample (48 hours). The supernatant of the virus was filtered through a 0.45 μ filter. PVDF to remove any non-adherent cells and cellular waste. Lentiviral expression particle packing: 24 hours prior to transfection of the packaging construct, 1.47X106 293T packaging cells PDL plates (poly-d-lysine in 6-well plates, Becton Dickinson) were seeded in 10 percent of serum without antibiotics. 2 micrograms of lentiviral expression construct (pLLB1-GW-Kan) and 1 microgram of plasmid pLP-VSVG, 1 microgram of pLP1, 1 microgram of pLP2 (Invitrogen) suspended in 250 microliters of Optimem (Life Technologies). 12 microliters of Lipofectamine 2000 reagent was mixed in a final volume of 250 microliters of Optimem and incubated for 5 minutes at room temperature. The diluted DNA was combined with the diluted liipofectamine (20 minutes at room temperature). The complex was added to the cells
293 T (in 2 milliliters of medium without antibiotics) and incubated overnight. The next day the medium was removed and refilled with new medium containing antibiotics. 48 hours post transfection the medium containing the virus was collected and stored at 4 ° C; the cells were refilled with new medium. 72 hours after the transfection, the final virus collection was made and grouped with the previous collection sample (48 hours). The excess virus was filtered through a 0.45 μ filter. PVDF to remove any non-adherent cells and cellular waste. The packaging of lentiviral ssDNA constructs: The same general procedure described for the lentiviral expression constructs described with the following exceptions: 2.6X104 293T cells were seeded in 96-well plates 24 hours before transfection. 100 nanograms of the construction of ssDNA (pLKO.1) and 10 nanograms of plasmid pLP-vsvg, 50 nanograms of pLP1, 50 nanograms of pLP2 (Invitrogen) were suspended in 30 microliters of Optimem (Life Technologies) and combined with 0.6 microliters of fugene6 (Roche). The complex was allowed to form for 30 minutes before the addition to the packaging cells. Description of viral constructs: Vector pLL-B1-GW Kan was derived from vector pLL3.7 from the MIT laboratory (Luc VanParijs laboratory). A gate cassette was replaced for the eGFP marker and the U6 (shRNA promoter) was removed. The kanamycin resistance cassette was replaced by the ampicillin cassette.
The final vector QL_GW-final-Kan was derived from the pQCXIX vector of Biosciences. The CMV promoter and the IRES sequence were removed and a gate cassette was inserted into the vector. The 3'LTR was replaced with a 3'LLTR of the original type which drives the expression of the inserted gene. The lentiviral vector of ssDHA pLKO.1 was not modified and was obtained from the Broad Institute (the RNA consortium) in Cambridge MA. Activation of the NFAT transcript HEK293 cells were transfected with 20 nanograms of the indicated plasmids in combination with 10 nanograms of Renilla, 20 nanograms of pCMV-SPORT6, and 50 nanograms of NFAT-Luc (Stratagene Inc). Transfections were carried out in 96-well format using approximately 20,000 cells per well. The cells were exposed to either DMSO, 5 uM of CsA, 10 uM of PMA, or 10 uM of PMA and 5 uM of CsA for 16 h. Reporter activities were determined 72 hours after transfection with the Dual-Glo luciferase reagent following the manufacturer's instructions (Promega). Fixation of HeLa cells: The cells were fixed with 3.7 percent formaldehyde, 0.5 percent Triton X 100 in PBS, 20 minutes at room temperature. The cells were washed twice with 0.5 percent Triton X 100 in PBS. Fixation and staining of Jurkat cells, NFAT transposition assay: After treatment, Jurkat cells were attached to 96-well trays using the Becton cell adhesive
Dickinson Cell-Tak. The suspended cells were centrifuged (800 X G) for 5 minutes on previously covered trays (following the manufacturer's instructions). The bound cells were permeabilized in 3.7 percent formaldehyde, 0.5 percent Triton X100, and washed twice with washing regulators (0.5 percent Triton X100 in PBS) and blocked with 2 percent BSA, 0.1 percent of Triton X100 in PBS. The cells were incubated in primary antibodies NFAT-1 (Cellomics K01-0011-1 diluted 1: 100) and NFAT-2 (Affinity Bioreagents MA3-024 diluted 1: 250) in blocking buffer for 1 hour at room temperature. Cells were washed twice with wash buffer and incubated with conjugated secondary goat anti mouse IgG for Alexa Fluor 488 GaM (Cellomics reagents K01-0011-1 antibody diluted 1: 2000, and Hoechst dye 1: 2000) in regulator of blocking. The cells were washed once with PBS and their image was formed. Gateway transfer cDNA sequences to viral vectors: The cDNA gene sequences used in this study were obtained from the Gateway transfer of clones obtained from the collection of MGC cDNA clones which were used either directly or transferred to vectors viral QL-GW-Kan / pLLB1 -GW-Kan. This was carried out using a single-tube reaction and a two-step reaction process. The BP reaction was performed by combining 100 nanograms of pCMV-Sport6 cDNA plasmids with 100 nanograms of the intermediate plasmid pDONR207 (Invitrogen). The reaction was started by adding 1.5 microliters of BP 5X
Clonase regulator (Invitrogen) and 1.5 microliters of BP Clonase (Invitrogen) in a total volume of 8 microliters, at room temperature overnight. The LR reaction was carried out by combining 4 microliters of BP reaction with 100 nanograms of target vector (QL-GW-Kan or pLLB1-GW-Kan) with 0.4 microliter NaCL 0.75M, 5x 1 microliter LR regulator (Invitrogen) and 1.8 microliters of LR Clonase in a final volume of 12 microliters. They were incubated at room temperature overnight and transformed into STB3 cells (2 microliters in 20 microliters of competent cells). Transduction of HeLa cells. Cells were seeded 24 hours before transduction in 96-well trays treated with tissue culture in a clear bottom at 6000 cells / well (100 microliters per well) in DMEM / FBS (10 percent heat-inactivated serum, Invitrogen) and antibiotic / antifungal (1 percent, Invitrogen). The medium was replaced with a transduction medium at a final concentration of 8 micrograms / milliliter of polybranch (Sigma) and 10 mM of the HEPES regulator (Invitrogen). Fifty microliters of retroviral supernatant was added to each well and the trays were centrifuged and centrifuged at 800 X g for 90 minutes. Transduction and sensitization of Jurkat cells: Jurkat cells were maintained in RPMI 1640 (GIBCO 21870-076) 10 percent FC1 clone Fetal 1 (Hyclone), 1 percent Penicillin / Streptomycin, 1 percent Glutamaxl, 0.1 percent beta mercaptoethanol. Before transduction, the cells
changed to means of transduction which contained: RPMI 1640 (top) fortified with 2.25 grams of glucose 1 percent antibiotic / antifungal, 1 percent 1 M Hepes (Gibco), 1 percent 100mM sodium pyruvate, 10 milliliters 7.5 percent of sodium bicarbonate 10 percent of fetal clone serum 1. Cells were seeded 5X104 in transduction medium combined with virus (volume in legend) and 4 micrograms / milliliter polybranum in final concentration and centrifuged at approximately 800 xg for 3 days. hours. For the activation of ICOS and IL-2 expression experiments the cells were transduced with 50 microliters of retroviral supernatant (QL-GW-Final-Kan) and 48 hours post transduction of the medium was fortified with the PMA (phorbol 12-myristate 13- acetate) 10 nanograms / milliliter and 24 hours after the addition the cells or medium were removed for ICOS or IL-2 measurements. For NFAT transposition experiments the cells were transduced with 50 microliters of supernatant (pLL-B1-GW-Kan) and 48 hours after transduction they were sensitized with PMA 10 nanograms / milliliter for 6 hours before fixing and staining. Analysis of the ICOS surface marker and IL-2 protein expression: 1.5 microliters of ICOS-PE (BD-Parmingen 557802) were combined with approximately 1 × 10 6 Jurkat cells and incubated on ice for 30 minutes. Cells were centrifuged at approximately 500 X g for 5 minutes and washed 2X in PBS and medium channel fluorescence was determined using flow cytometry (BD FACSCaliber). IL-2 levels were measured using the kit
QuantiGlo IL-2 Elisa (R &D Systems). Activation of Jurkat cells for ssDNA inhibition studies: To assess the efficacy of ssDNA, Jurkat cells (15,000) were transduced with 10 microliters of LKO virus as described and 24 hours after transduction the medium was fortified with puromycin (see more ahead). Six days after transduction, half of the cells were activated with antibodies directed to TCR and the CD28 receptors were bound to the 96-well tray surface and incubated overnight and the medium was collected for the determination of IL-2. . The remaining cells were used to determine the fraction of viable cells in each well using the Cell Titer-Glo assay (see below). Activation trays were prepared by coating goat anti-mouse IgB, Fcy fragment-specific antibody (Jackson ImmunoResearch Laboratories) 55 microliters per well at a final concentration of 10 micrograms / milliliter in PBS. The plates were incubated 3 hours at room temperature. The excess IgG was removed and the trays were stained. The trays were blocked with 300 microliters of 2 percent BSA / PBS (BSA, Fraction V lyophilized, Roche) and incubated for 2 hours at room temperature. The trays were washed 3 times with PBS and anti-TCR stimulating antibodies 0.01 micrograms / milliliter (BD Biosciences 347770 clone WT31) and anti-CD28 0.3 micrograms / milliliter (BD Pharmingen 555725) in 2 percent BSA / PBS final volume 50 microliters /water well. The trays were incubated overnight and
washed 3 times with PBS before adding the cells for activation. Puromycin selection of Jurkat cells with transduced ssDNA and normalization for cell survival: The LKO viral vector used in this study contains a puromycin selection marker. Jurkat cells infected with ssDNA constructs (LKO.1) were placed under puromycin selection 24 hours after infection by the addition of puromycin (2 micrograms / milliliter) and were maintained for the duration of the experiment (6 days after infection) , 3 days for the quantification of mRNA). To count the differences in cell number after selection with puromycin (possibly due to variations in viral titration), we adopted a cellular ATP assay that is proportional to the cell number (Luminescent Test Kit Cell Titer-Glo, Promega). The test was performed following the manufacturer's instructions. A standard curve was generated for each cell type to determine the linearity for the assay. The concentrations of IL-2 were normalized to the number of cells by dividing the concentration of IL-2 calculated by the rLU values for the Cell Titer-Glo assay, which is equivalent to the expression of IL-2 / cell number. The levels of mRNA expression were determined 3 days after infection.
Determination of dismantled mRNA for cMAC: Jurkat mRNA expression levels were determined 3 days after infection (2 days after selection with
puromycin). Preparation of ssDNA constructs and ligation in vector LKO.1: DNA oligonucleotides were synthesized with adapters for 5 'Agel and 3' EcoR1 with the loop sequence TTCAAGAGA. The oligonucleotides were annealed and ligated directly into the predigested LKO vector. See Table 6 for the Oligo sequences.
Table 6 Sequence targeting ssDNA and linked oligos vector LKO.1
Example 1 Discovery that cMAC induces the nuclear transposition of TORC1. NFAT, NFkB and AP-1 are probably the three most important transcription factors in the activation of T cells (Quintana Eur J Physiol (2005) 450: 1-12). The three promoter elements are represented in the IL-2 promoter and it has been determined that all three are calcium dependent (NFkB and AP-1 indirectly). The activation of T cells requires the transposition of NFAT in the nucleus. This is mediated by the unmasking of the nuclear localization sequence in NFAT by the enzyme phosphatase calcineurin. Calcineurin is activated through the mobilization of calcium and is blocked by the immunosuppressive drug ciclosporin A. TORC-1 is a coactivator of cAMP-dependent transcription. TORC-1 is similar to NFAT in that Torc-1 transposes in the nucleus after activation after calcium mobilization. It is thought that TRPV6 is a calcium channel operated in storage and, although controversial, it has been suggested that it has similarity with calcium channel activated by calcium release (CRAC) which is responsible for the induction of genes regulated by the Activation of calcium (Feske Nat Immunol.2 (4): 316-24 (2001)). Imaging has been selected to identify genes involved in the transposition of the TORC1 CREB coactivator dependent on cAMP. This selection identified several
genes as inductors of the nuclear transposition of TORC Figure 4 and Table 7 (Bittinger et al., Current Biology 14 (23): 2156-61 (2004)), however, the identity of a previously uncharacterized gene to which we now refer as cMAC (Calcineurin Conserved Membrane Activator) is not described in that publication. The published sequence of murine cMAC (Access Number NM_177344) and the human cMAC orthologue (Access Number NM_053045) are located in GenBank. The clone cMAC found in the analysis was an MGC clone that was noted as similar to NM_177344. However, NM_177344 encodes a protein with an alternative 3 'end that is not present in human cDNAs or predicted orthologs of cMAC. The cDNAs active in the primary selection as well as the human ortholog from murine cMAC were recovered, retransformed, the sequence confirmed, and inserted into viral vectors and introduced into HeLa cells stably expressing TORC1-eGFP, and the relative amounts were calculated. of TORC1-eGFP in the cytosol and nucleus using a microscope platform in example 2 below. The Torc-eGFP transposition was blocked by the calcineurin inhibitor cyclosporin A which also illustrates that the cDNAs undoubtedly induced the transposition of TORC as opposed to affecting the morphology of the cell or other phenotypes that can be misinterpreted by the automated microscope platform as a transposition.
Table 7 Inducers Assumptions of TORC transposition
Abbreviated Annotation Designation
MGC tura
BC022606, Mus musculus RIKEN cDNA C730025P13 gene, cMAC
-P2 mRNA (cDNA clone MGC: 31129 IMAGE: 4165766), complete cds. NM_177344
BC034814 Potential cation channel transient TRPV6 receptor Homo sapiens, subfamily V, member 6 (TRPV6), mRNA. NM_018646
Example 2 cMAC acts by means of calcium and calcineurin To determine if the translocation of TORC by cMAC was through calcium and calcineurin, cMAC was over-expressed in HeLa cells which stably expressed the Tord-eGFP fusion construct by infecting the cells with lentiviral particles containing the TRPV6 calcium channel and the human cMAC sequence. The cells were treated with the calcineurin inhibitor Cyclosporin A (CsA) one hour before imaging. As shown in Figure 5: Treatment with CsA resulted in an inversion of the transposition of TORC1, resulting in
that the majority of TORC1-eGFP was returned to the cytoplasm. These data suggest that the cMAC transposition of TORC-1 was dependent on the activation of calcineurin. In a separate set of experiments, an extracellular calcium requirement using EGTA was also tested. The presence of EGTA in the blocked medium of cMAC induced the nuclear transposition of TORC1. The effect of EGTA could, however, be reversed by the addition of excess calcium to the medium (these data are not shown). Thus, cMAC induces the translocation of TORC1 through the calcium-dependent activation of calcineurin. The effect of overexpression of cMAC on the calcineurin-dependent NFAT transcription factor was also examined. cMAC or TRPV6 activator previously described by or
NFAT was co-transfected with a luciferase reporter powered by NFAT. In the presence of PMA, the overexpression cMAC (and TRPV6) induced a significant increase in the expression driven by NFAT (Figure 6) and this activation was partially blocked by CsA. These data are consistent with the activation of calcineurin. It should be noted that activation by cMAC was not as strong as by the TRPV6 calcium channel. EXAMPLE 3 Effect of cMAC on the nuclear transposition of NFAT and the activation of T cells The effect of cMAC on the nuclear transposition of the transcription factor NFAT and its dependence on the
calcineurin. Jurkat cells were transduced with an empty viral vector (translation stop sequence), the calcium channel TRPV6
0 human cMAC. The cells were examined for localization of endogenous NFAT-1 (Figure 7) and NFAT-2 protein (Figure 8). Although NFAT-1 and NFAT-2 were detected in the cytoplasm of the cells infected by the control virus, both isoforms of NFAT were located mainly in the nucleus in a large proportion of transduced cells of cMAC. The transposition was dependent on the sensitization of the cells with PMA. PMA alone showed no effect on the transposition of NFAT, however, PMA in combination with cMAC or TRPV6 markedly increased nuclear transposition. The calcineurin inhibitor, cyclosporin A, blocked the PMA sensitized cMAC (and TRPV6) induced the transposition of both NFAT-1 and NFAT-2. The mobilization of calcium and the subsequent transposition of NFAT is a critical component in the path of signal transduction in the activation of T cells and NFAT is required for the production of IL-2. These data also support the role of NFAT and calcineurin in the activation of T cells and specifically show that overexpression of cMAC in a T cell line induces transposition of endogenous NFAT-1 and NFAT-2 dependent calcineurin. We assessed the role of TRPV6 and cMAC in an analysis of T cell activation. Jurkat T cells were transduced with TRPV6 and cMAC (Figure 9) and their ability to induce the
T-cell activation markers after priming with anti-TCR antibody and PMA (similar results were obtained with PMA alone). Both TRPV6 and cMAC were potent T-cell activators as assessed by the induction of IL-2 and the expression of the ICOS surface marker, whereas PMA alone or PMA plus anti-TCR did not upregulate IL-2 expression levels 2 or ICOS. Conversely, the expression of the cDNAs showed negligible upregulation of IL-2 and ICOS without PMA sensitization, which is consistent with the findings observed with the transposition of NFAT. In this test, the cDNAs encoding cMAC cDNAs, both human and mouse, were annotated, as well as the RefSeq sequences Numbers NM_0539045 and NM_177344, respectively. Both cMAC cDNA species were equally active to induce IL-2 secretion and ICOS expression. Thus, overexpression of cMAC also induces the activation markers of T cells. Example 4 Use of shRNA to demonstrate that cMAC is critical for the activation of Jurkat T cells To assess whether cMAC was essential for the activation of T cells, we designed several constructs of ssDNA directed to cMAC. In these experiments Jurkat T cells were transduced with viral constructs directed to cMAC or to another unrelated gene. 6 days after transduction, the cells were activated with
TCR / CD28 antibodies and the level of cMAC and IL-2 mRNA secreted in the medium was measured. All but two constructs of ssDNA targeted to cMAC demonstrated significant reductions in IL-2 production. Figure 10. In separate experiments, other negative controls targeting the CD29 protein and a random murine gene demonstrated a similar inhibition profile to the pGL3 negative control. To determine the specificity of the ssDNA construct, the reduction of cMAC mRNA was measured for each construct (Table 8) and compared to the inhibition of IL-2 observed. The ssDNA constructs pGL3-Luc and CD29 served as negative controls. The construction of ssDNA CD29 potently reduced the CD29 mRNA (and the CD29 protein, the data are not shown) but had no effect on the cMAC message. Only one construction (BL8) demonstrated satisfactory dismantling of the message (70%) without any reduction of IL-2, a second construct (BL6) demonstrated marginal dismantling of mRNA (36%) without any reduction of IL-2. The remaining constructs showed significant decreases in IL-2 with the reduction of mRNA although in some cases mRNA reductions were marginal. In those cases it may be that the ssDNA is causing decreases in the expression of the cMAC protein through microRNA effects. Thus, it seems that with the exception of 2 constructs (BL6 and BL8) the phenotypically active IL-2 constructs correlate with reduced levels of mRNA from
cMAC. Table 8 Dismantling of cDNA mRNA-mediated mRNA virus correlates with inhibition of IL-2
Claims (72)
1. A polypeptide isolated from SEQ ID NO: 2, or a fragment thereof, or a substantially similar protein sequence having a sequence identity of at least 50 percent with SEQ ID NO: 2, or a functional equivalent of the the same, and that exhibits a biological activity selected among the transport of ions, the diffusion of ions, the activation of the calcineurin trajectory, the calcium-dependent activation of a T cell, the nuclear transposition of TORC, the nuclear transposition of NFAT or the gene expression activity driven by CRE (cAMP Response Element) of the original SEQ ID NO: 2.
2. The polypeptide of claim 1 having a sequence selected from the group consisting of SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20 and SEQ ID NO: 21.
3. An antibody or antibody fragment that is capable of binding to the polypeptide of claim 1 or 2.
4. An antibody or antibody fragment that binds specifically to cMAC (SEQ ID No. 2), or a polypeptide comprising a specific binding region for cMAC.
5. An antibody fragment according to claim 3 or 4, which is a Fab or F (ab ') 2 fragment.
6. An antibody according to any of claims 3 to 5, which is a monoclonal antibody.
7. An isolated nucleic acid molecule encoding the polypeptide of claim 1 or 2.
8. The nucleic acid molecule of claim 7, comprising SEQ ID NO: 1, SEQ ID NO: 11 or SEO ID NO: 12.
9. The nucleic acid molecule of claim 7 or 8, further comprising a promoter operably linked to the nucleic acid molecule.
10. An isolated nucleic acid sequence selected from SEQ ID NOs 3, 4 and 5.
11. A vector molecule comprising the nucleic acid molecule of any of claims 7 to 10.
12. A vector molecule of claim 11, comprising the nucleic acid sequence of cMAC (SEQ ID NO: 1).
13. A vector comprising the cMAC promoter (SEQ ID NO: 3) operably linked to a reporter protein nucleic acid sequence.
14. A host cell comprising the vector molecule of any of claims 11 to 13.
15. A method for producing the polypeptide of claim 1 or 2, which comprises culturing the host cell having an expression vector incorporated therein. comprising the vector of claim 11 or 12 under conditions sufficient for expression of the polypeptide in the host cell.
16. A method for producing a cMAC polypeptide of SEQ ID NO. 2, which comprises cultivating the host cell that has embedded therein an expression vector comprising the vector of claim 11 or 12 under conditions sufficient for expression of the polypeptide in the host cell.
17. A method for treating a disorder in a subject, comprising administering to the subject an effective amount of an agent that inhibits the activity of cMAC.
18. A method according to claim 17, wherein the disorder is a disorder associated with cMAC.
19. A method according to claim 17 or 18, wherein the agent is antibody, an antibody fragment or a polypeptide that contains a specific binding region for cMAC.
20. An antibody, an antibody fragment or a polypeptide of any of claims 3 to 6, comprising a specific binding region for cMAC as a medicament.
21. The use of an antibody, an antibody fragment or a polypeptide comprising a specific binding region for cMAC-in the treatment of a disorder in a subject.
22. The use of claim 21, wherein the disorder is a disorder associated with cMAC.
23. The use of an antibody of any of claims 3 to 6 for the manufacture of a medicament for the treatment of a disorder associated with cMAC.
24. A method for treating a disorder in a subject comprising administering to the subject an effective amount of an agent which inhibits the expression of cMAC.
25. A method according to claim 24, wherein the disorder is a disorder associated with cMAC.
26. A method according to claim 24 or 25, wherein the agent is an inhibitory nucleic acid capable of specifically inhibiting the expression of cMAC.
27. A method according to claim 26, wherein the inhibitory nucleic acid is selected from the group consisting of an antisense oligonucleotide, an RNA agent, and a ribozyme.
28. A method according to claim 27, wherein the RNA agent is selected from the group consisting of dsRNA, siRNA, and shRNA.
29. The method according to claim 28, wherein the iRNA agent comprises at least one nucleic acid selected from the group consisting of SEQ ID NO: 22 to SEQ ID NO: 101, and SEQ ID NO: 106, SEQ ID NO: 107, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 118, SEQ ID NO: 119, SEQ ID NO: 121, SEQ ID NO: 122, SEQ ID NO: 124, SEQ ID NO: 125, SEQ ID NO: 127, SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 131, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 136, and SEQ ID NO: 137.
30. An iRNA agent comprising at least one nucleic acid selected from the group consisting of SEQ ID NO: 22 a SEQ ID NO: 101, and SEQ ID NO: 106, SEQ ID NO: 107, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 118, SEQ ID NO: 119, SEQ ID NO: 121, SEQ ID NO: 122, SEQ ID NO: 124, SEQ ID NO: 125, SEQ ID NO: 127, SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 131, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 136, and SEQ ID NO: 137.
31. An agent! RNA specific for cMAC selected from the group consisting of dsRNA, siRNA, and shRNA as a medicament, wherein the RNA agent comprises at least one nucleic acid selected from the group consisting of SEQ ID NO: 22 to SEQ ID NO: 101, and SEQ ID NO: 106, SEQ ID NO: 107, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 115, SEQ ID NO: 116 , SEQ ID NO: 118, SEQ ID NO: 119, SEQ ID NO: 121, SEQ ID NO: 122, SEQ ID NO: 124, SEQ ID NO: 125, SEQ ID NO: 127, SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 131, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 136, and SEQ ID NO: 137.
32. The use of a specific RNA or siRNA agent for cMAC in the treatment of a disorder in a subject.
33. The use of claim 32, wherein the disorder is a disorder associated with cMAC.
34. The use of the RNA agent of claim 31 for the manufacture of a medicament for the treatment of a disorder associated with cMAC.
35. A method for treating a disorder in a subject, comprising administering to the subject an effective amount of an agent that improves the activity of cMAC.
36. A method for treating a disorder in a subject comprising administering to the subject an effective amount of an agent that increases the expression of cMAC.
37. A method according to claim 36, wherein the agent is a transcriptional enhancer of cMAC.
38. A method according to claim 36, wherein the agent is a gene therapy vector comprising a nucleic acid encoding cMAC or a fragment thereof.
39. The method of claim 38, wherein the agent is a vector of claim 11 or 12.
40. A method according to any of claims 36 to 39 wherein the disorder is a disorder associated with cMAC.
41. A pharmaceutical composition comprising an effective amount of an agent that inhibits the expression of cMAC or inhibits a cMAC activity, and a pharmaceutically acceptable carrier.
42. A pharmaceutical composition according to claim 41 wherein the agent is an antisense oligonucleotide or an iRNA agent.
43. The pharmaceutical composition according to claim 42, wherein the iRNA agent comprises at least one nucleic acid selected from the group consisting of SEQ ID NO: 22 to SEQ ID NO: 101, and SEQ ID NO: 106, SEQ ID NO: 107, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 118, SEQ ID NO: 119, SEQ ID NO : 121, SEQ ID NO: 122, SEQ ID NO: 124, SEQ ID NO: 125, SEQ ID NO: 127, SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 131, SEQ ID NO: 133 , SEQ ID NO: 134, SEQ ID NO: 136, and SEQ ID NO: 137.
44. A pharmaceutical composition according to claim 41, wherein the agent is an antibody, an antibody fragment that specifically binds to cMAC. , or a polypeptide comprising a specific binding region for cMAC.
45. A pharmaceutical composition of claim 44, wherein the antibody is the antibody of any of claims 3 to 6.
46. A pharmaceutical composition according to claim 44 or 45 wherein the agent binds to a cMAC epitope. selected from SEQ ID NO.6, 7, 8, 9, 10.
47. A method for treating a disorder in a subject comprising administering to the subject an effective amount of a pharmaceutical composition of an agent that inhibits the activity of cMAC.
48. A method according to claim 47 wherein or the disorder is a disorder associated with cMAC.
49. A method according to claim 47 or 48, wherein the agent is an antibody or fragment thereof, which is specifically binds to cMAC (SEQ ID NO: 2) or a polypeptide comprising a specific binding region for cMAC.
50. A method of claim 49, wherein the antibody is the antibody of any of claims 3 to 6.
51. A method according to claim 49, wherein the agent binds to a cMAC epitope selected from SEQ. ID NOs.6, 7, 8, 9 and 10.
52. A method for treating a disorder in a subject, comprising administering to the subject an effective amount of a pharmaceutical composition of an agent that inhibits the expression of cMAC.
53. A method according to claim 52, wherein the agent is an inhibitory nucleic acid capable of specifically inhibiting the expression of cMAC.
54. A method according to claim 53, wherein the inhibitory nucleic acid is selected from the group consisting of an antisense oligonucleotide, an iRNA agent, and a ribozyme.
55. A method according to claim 54, wherein the iRNA agent is selected from the group consisting of dsRNA, SiRNA, and siRNA.
56. A method according to claim 55, wherein the iRNA agent comprises at least one nucleic acid selected from the group consisting of SEQ ID NO: 22 to SEQ ID NO: 101, and SEQ ID NO: 106, SEQ ID NO: 107, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 115, SEO ID NO: 116, SEQ ID NO: 118, SEQ ID NO: 119, SEQ ID NO: 121, SEQ ID NO: 122, SEQ ID NO: 124, SEQ ID NO: 125, SEQ ID NO: 127, SEQ ID NO: 128, SEQ ID NO: 130, SEQ ID NO: 131, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 136, and SEQ ID NO: 137.
57. A method for identifying a compound useful for the treatment of a disorder associated with cMAC comprising: (a) contacting a test compound with cMAC; and (b) detecting a change in a biological activity of cMAC compared to cMAC that is not in contact with the test compound, wherein upon detecting a change, the test compound is identified as being useful for the treatment of the disorder.
58. A method for identifying a compound useful for the treatment of a cMAC-associated disorder comprising: (a) contacting a test compound with cMAC under sample conditions that allow the biological activity of cMAC; (b) determine the level of a biological activity of cMAC; (c) comparing the level with that of a control sample lacking the test compound; and, (d) selecting a test compound that causes the level for the change to also prove it as a potential agent for the treatment of the disorder.
59. A method according to claim 57 or 58, in where the change is a reduction of that biological activity.
60. A method according to claims 57 to 59, wherein the biological activity is selected from ion transport, ion diffusion, protein-cMAC interaction or modification of cMAC, calcium-dependent activation of a T cell, nuclear transposition of TORC, and expression of gene driven by CRE (Response element of cAMP).
61. A method for testing whether a compound modulates a biological activity of cMAC, comprising: (a) contacting a test compound with cMAC; and (b) detecting a change in a biological activity of cMAC compared to cMAC not contacted with the test compound, wherein upon detecting a change that compound is identified as a modulator of the biological activity of cMAC.
62. A method for identifying modulators useful for treating a disorder, comprising testing the ability of a candidate modulator to inhibit the activity of a cMAC protein.
63. A method for identifying modulators useful for treating a disorder, comprising testing the ability of a candidate modulator to inhibit the expression of a cMAC protein.
64. A compound identified by a method according to any of claims 57 to 63.
65. A method for identifying a compound useful for the treatment of a disorder associated with cMAC, comprising administering a compound identified by a method according to claim 65, to an animal model of the condition associated with cMAC.
66. The method according to any of claims 18, 25, 40, 57 to 60, or 65, or the use of claim 22, 23, 33 or 34, wherein the disorder associated with cMAC is selected from the group which consists of autoimmune disease, immunosuppression, inflammatory disease, cancer, cardiovascular disease and neurological disease.
67. A method for inhibiting the biological activity of cMAC in a cell, comprising contacting a cell with an anti-cMAC antibody or fragment thereof, with a polypeptide comprising a specific binding region for cMAC or with a nucleic acid that reduces the expression of cMAC.
68. A method according to claim 67 wherein the biological activity is selected from the group consisting of calcium-dependent activation of a T cell, nuclear transposition of TORC, nuclear transposition of NFAT and gene expression driven by CRE (Element of Response cAMP).
69. A method for selectively inhibiting lymphocyte activity in a multi-cellular organism, comprising contacting the organism with an anti-cMAC antibody or fragment thereof, with a polypeptide comprising a specific region for cMAC or with an acid nucleic acid that reduces the expression of cMAC.
70. A method to improve the activation of T cells that comprises contacting a T cell or a T cell precursor cell with a purified cMAC polypeptide, a gene therapy vector comprising the cMAC gene, or an expression enhancer of the cMAC gene.
71. The method of claim 70 wherein the cMAC polypeptide is the polypeptide of claim 1 or 2.
72. The method of claim 70 wherein the gene therapy vector comprising the cMAC gene is the vector of the claim 11 or 12
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US60/723,181 | 2005-10-03 |
Publications (1)
Publication Number | Publication Date |
---|---|
MX2008004412A true MX2008004412A (en) | 2008-10-03 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11492404B2 (en) | Inhibition of cytokine-induced SH2 protein in NK cells | |
Lemaire et al. | SF2 and SRp55 regulation of CD45 exon 4 skipping during T cell activation | |
EP2830662A1 (en) | Methods for treating hair loss disorders | |
JP3451092B2 (en) | Monoclonal antibody specifically reacting with Fas ligand and method for producing the same | |
KR20130107203A (en) | Detection and treatment of fibrosis | |
JP2004520027A (en) | Histone deacetylase-related genes and proteins | |
WO2006093337A1 (en) | Preventive/therapeutic agent for cancer | |
US20090136506A1 (en) | Conserved Membrane Activator of Calcineurin (CMAC), a Novel Therapeutic Protein and Target | |
MX2008004412A (en) | Conserved membrane activator of calcineurin (cmac), a novel therapeutic protein and target | |
JP7325075B2 (en) | For screening at least one agent selected from the group consisting of agents for increasing or decreasing the number of frizzled 3-expressing cells, agents for preventing or treating diabetes, agents for preventing or treating insulinoma, and agents for promoting or suppressing insulin secretion Screening Agent, Screening Kit, and Screening Method | |
US8153764B2 (en) | Biomarker specific to brain/nerve or specific to neuronal differentiation | |
WO2004069869A1 (en) | Apoptosis-inducing gene and utilization of the same | |
US20060099676A1 (en) | Mammalian genes involved in rapamycin resistance and tumorgenesis: rapr6 genes | |
CA2535976A1 (en) | A novel gene and protein associated with angiogenesis and endothelial apoptosis | |
WO2003094841A2 (en) | Mammalian genes involed in rapamycin resistance and tumorgenesis: annexin xiii genes | |
US20060240021A1 (en) | Mammalian genes involved in rapamycin resistance and tumorgenesis: rapr7 genes | |
JP2004517619A (en) | Interleukin-1 related genes and proteins | |
WO2013070563A1 (en) | Treatment of autoimmune and inflammatory disorders by inhibition of blimp-1 | |
KR102569644B1 (en) | Composition for preventing and treating immune diseases comprising gdf15 | |
US8309687B2 (en) | Biomarker specific for cancer | |
US6787646B1 (en) | Tumor suppressor molecules and methods of use | |
WO2001010883A1 (en) | Cell death gene and uses thereof | |
WO2003083117A2 (en) | Vascularization controlling gene | |
WO2004047853A2 (en) | Methods for the treatment of chronic pain and compositions therefor | |
JP2006166703A (en) | Cartilage differentiation regulating gene |