WO2016141108A1 - Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier - Google Patents
Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier Download PDFInfo
- Publication number
- WO2016141108A1 WO2016141108A1 PCT/US2016/020530 US2016020530W WO2016141108A1 WO 2016141108 A1 WO2016141108 A1 WO 2016141108A1 US 2016020530 W US2016020530 W US 2016020530W WO 2016141108 A1 WO2016141108 A1 WO 2016141108A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- gene
- bacterium
- promoter
- disease
- genetically engineered
- Prior art date
Links
- 241000894006 Bacteria Species 0.000 title claims abstract description 442
- 206010061218 Inflammation Diseases 0.000 title claims description 164
- 230000004054 inflammatory process Effects 0.000 title claims description 73
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 title claims description 56
- 201000010099 disease Diseases 0.000 title claims description 42
- 230000004682 mucosal barrier function Effects 0.000 title abstract description 8
- 230000002829 reductive effect Effects 0.000 title description 8
- 230000008901 benefit Effects 0.000 title description 7
- 208000023275 Autoimmune disease Diseases 0.000 claims abstract description 27
- 238000000034 method Methods 0.000 claims abstract description 24
- 108090000623 proteins and genes Proteins 0.000 claims description 715
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Natural products CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 claims description 166
- 230000014509 gene expression Effects 0.000 claims description 166
- FERIUCNNQQJTOY-UHFFFAOYSA-M Butyrate Chemical compound CCCC([O-])=O FERIUCNNQQJTOY-UHFFFAOYSA-M 0.000 claims description 163
- 230000004888 barrier function Effects 0.000 claims description 147
- 239000003623 enhancer Substances 0.000 claims description 108
- 239000001301 oxygen Substances 0.000 claims description 107
- 229910052760 oxygen Inorganic materials 0.000 claims description 107
- XBDQKXXYIPTUBI-UHFFFAOYSA-M Propionate Chemical compound CCC([O-])=O XBDQKXXYIPTUBI-UHFFFAOYSA-M 0.000 claims description 93
- 239000003642 reactive oxygen metabolite Substances 0.000 claims description 93
- 239000007845 reactive nitrogen species Substances 0.000 claims description 85
- 241000588724 Escherichia coli Species 0.000 claims description 64
- 239000013612 plasmid Substances 0.000 claims description 63
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 claims description 39
- 210000000349 chromosome Anatomy 0.000 claims description 37
- 208000022559 Inflammatory bowel disease Diseases 0.000 claims description 35
- TWSALRJGPBVBQU-PKQQPRCHSA-N glucagon-like peptide 2 Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(O)=O)C(O)=O)[C@@H](C)CC)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@@H](NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCSC)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC=1NC=NC=1)[C@@H](C)O)[C@@H](C)CC)C1=CC=CC=C1 TWSALRJGPBVBQU-PKQQPRCHSA-N 0.000 claims description 35
- 102400000326 Glucagon-like peptide 2 Human genes 0.000 claims description 34
- 101800000221 Glucagon-like peptide 2 Proteins 0.000 claims description 34
- -1 NAPEs Chemical compound 0.000 claims description 34
- 230000007613 environmental effect Effects 0.000 claims description 30
- 102000004190 Enzymes Human genes 0.000 claims description 24
- 108090000790 Enzymes Proteins 0.000 claims description 24
- 230000006696 biosynthetic metabolic pathway Effects 0.000 claims description 23
- 108010066979 Interleukin-27 Proteins 0.000 claims description 18
- 230000001010 compromised effect Effects 0.000 claims description 13
- 208000035475 disorder Diseases 0.000 claims description 13
- 230000000770 proinflammatory effect Effects 0.000 claims description 13
- 241000193403 Clostridium Species 0.000 claims description 12
- 230000001363 autoimmune Effects 0.000 claims description 12
- 239000004055 small Interfering RNA Substances 0.000 claims description 12
- 230000000529 probiotic effect Effects 0.000 claims description 11
- 239000006041 probiotic Substances 0.000 claims description 10
- 235000018291 probiotics Nutrition 0.000 claims description 10
- 208000031981 Thrombocytopenic Idiopathic Purpura Diseases 0.000 claims description 9
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 9
- 201000003710 autoimmune thrombocytopenic purpura Diseases 0.000 claims description 9
- 206010012735 Diarrhoea Diseases 0.000 claims description 8
- 208000007465 Giant cell arteritis Diseases 0.000 claims description 8
- 206010043207 temporal arteritis Diseases 0.000 claims description 8
- 241000606125 Bacteroides Species 0.000 claims description 7
- 206010009900 Colitis ulcerative Diseases 0.000 claims description 7
- 108020004459 Small interfering RNA Proteins 0.000 claims description 7
- 201000006704 Ulcerative Colitis Diseases 0.000 claims description 7
- 208000009299 Benign Mucous Membrane Pemphigoid Diseases 0.000 claims description 6
- 241000186000 Bifidobacterium Species 0.000 claims description 6
- 208000011231 Crohn disease Diseases 0.000 claims description 6
- 108010015972 Elafin Proteins 0.000 claims description 6
- 241000186660 Lactobacillus Species 0.000 claims description 6
- 108091027967 Small hairpin RNA Proteins 0.000 claims description 6
- 208000002552 acute disseminated encephalomyelitis Diseases 0.000 claims description 6
- MDCUNMLZLNGCQA-HWOAGHQOSA-N elafin Chemical compound N([C@H](C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N1CCC[C@H]1C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@@H](CCCCN)C(=O)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H]1C(=O)N2CCC[C@H]2C(=O)N[C@H](C(=O)N[C@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H]2CSSC[C@H]3C(=O)NCC(=O)N[C@@H](CCSC)C(=O)N[C@@H](C)C(=O)N[C@@H](CSSC[C@H]4C(=O)N5CCC[C@H]5C(=O)NCC(=O)N[C@H](C(N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H]5N(CCC5)C(=O)[C@H]5N(CCC5)C(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCSC)NC(=O)[C@H](C)NC2=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(O)=O)C(=O)N4)C(=O)N[C@@H](CSSC1)C(=O)N[C@@H](CCC(O)=O)C(=O)NCC(=O)N[C@@H](CO)C(=O)N3)=O)[C@@H](C)CC)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C(C)C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCC(N)=O)C(O)=O)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)C(C)C)C(C)C)C(=O)[C@@H]1CCCN1C(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](C)N MDCUNMLZLNGCQA-HWOAGHQOSA-N 0.000 claims description 6
- 229940039696 lactobacillus Drugs 0.000 claims description 6
- 239000000203 mixture Substances 0.000 claims description 6
- 208000008795 neuromyelitis optica Diseases 0.000 claims description 6
- 239000000126 substance Substances 0.000 claims description 6
- 206010072579 Granulomatosis with polyangiitis Diseases 0.000 claims description 5
- 208000003456 Juvenile Arthritis Diseases 0.000 claims description 5
- 206010059176 Juvenile idiopathic arthritis Diseases 0.000 claims description 5
- 206010025135 lupus erythematosus Diseases 0.000 claims description 5
- 150000004666 short chain fatty acids Chemical class 0.000 claims description 5
- 208000008190 Agammaglobulinemia Diseases 0.000 claims description 4
- 208000003343 Antiphospholipid Syndrome Diseases 0.000 claims description 4
- 208000031212 Autoimmune polyendocrinopathy Diseases 0.000 claims description 4
- 208000030939 Chronic inflammatory demyelinating polyneuropathy Diseases 0.000 claims description 4
- 208000015943 Coeliac disease Diseases 0.000 claims description 4
- 208000021866 Dressler syndrome Diseases 0.000 claims description 4
- 208000035186 Hemolytic Autoimmune Anemia Diseases 0.000 claims description 4
- 206010020983 Hypogammaglobulinaemia Diseases 0.000 claims description 4
- 201000009794 Idiopathic Pulmonary Fibrosis Diseases 0.000 claims description 4
- 206010021245 Idiopathic thrombocytopenic purpura Diseases 0.000 claims description 4
- 208000012309 Linear IgA disease Diseases 0.000 claims description 4
- 208000003250 Mixed connective tissue disease Diseases 0.000 claims description 4
- 208000000733 Paroxysmal Hemoglobinuria Diseases 0.000 claims description 4
- 206010034277 Pemphigoid Diseases 0.000 claims description 4
- 102100036050 Phosphatidylinositol N-acetylglucosaminyltransferase subunit A Human genes 0.000 claims description 4
- RJKFOVLPORLFTN-LEKSSAKUSA-N Progesterone Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)C)[C@@]1(C)CC2 RJKFOVLPORLFTN-LEKSSAKUSA-N 0.000 claims description 4
- 206010042276 Subacute endocarditis Diseases 0.000 claims description 4
- 206010043561 Thrombocytopenic purpura Diseases 0.000 claims description 4
- 102000007641 Trefoil Factors Human genes 0.000 claims description 4
- 108010007389 Trefoil Factors Proteins 0.000 claims description 4
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 claims description 4
- 208000025851 Undifferentiated connective tissue disease Diseases 0.000 claims description 4
- 208000017379 Undifferentiated connective tissue syndrome Diseases 0.000 claims description 4
- 206010046851 Uveitis Diseases 0.000 claims description 4
- 208000027625 autoimmune inner ear disease Diseases 0.000 claims description 4
- 201000005795 chronic inflammatory demyelinating polyneuritis Diseases 0.000 claims description 4
- 201000001981 dermatomyositis Diseases 0.000 claims description 4
- 201000003045 paroxysmal nocturnal hemoglobinuria Diseases 0.000 claims description 4
- 208000002574 reactive arthritis Diseases 0.000 claims description 4
- 206010039073 rheumatoid arthritis Diseases 0.000 claims description 4
- 208000008467 subacute bacterial endocarditis Diseases 0.000 claims description 4
- 206010002556 Ankylosing Spondylitis Diseases 0.000 claims description 3
- 108020005544 Antisense RNA Proteins 0.000 claims description 3
- 208000009137 Behcet syndrome Diseases 0.000 claims description 3
- 201000004624 Dermatitis Diseases 0.000 claims description 3
- 241000588722 Escherichia Species 0.000 claims description 3
- 102000003814 Interleukin-10 Human genes 0.000 claims description 3
- 108090000174 Interleukin-10 Proteins 0.000 claims description 3
- 241000194036 Lactococcus Species 0.000 claims description 3
- 108090001030 Lipoproteins Proteins 0.000 claims description 3
- 102000004895 Lipoproteins Human genes 0.000 claims description 3
- 201000004681 Psoriasis Diseases 0.000 claims description 3
- 201000001263 Psoriatic Arthritis Diseases 0.000 claims description 3
- 208000036824 Psoriatic arthropathy Diseases 0.000 claims description 3
- 208000006673 asthma Diseases 0.000 claims description 3
- 230000001684 chronic effect Effects 0.000 claims description 3
- 239000003184 complementary RNA Substances 0.000 claims description 3
- 230000003111 delayed effect Effects 0.000 claims description 3
- 208000011580 syndromic disease Diseases 0.000 claims description 3
- 231100000331 toxic Toxicity 0.000 claims description 3
- 230000002588 toxic effect Effects 0.000 claims description 3
- 208000032194 Acute haemorrhagic leukoencephalitis Diseases 0.000 claims description 2
- 208000026872 Addison Disease Diseases 0.000 claims description 2
- 208000032671 Allergic granulomatous angiitis Diseases 0.000 claims description 2
- 206010001935 American trypanosomiasis Diseases 0.000 claims description 2
- 208000028185 Angioedema Diseases 0.000 claims description 2
- 206010003267 Arthritis reactive Diseases 0.000 claims description 2
- 206010003827 Autoimmune hepatitis Diseases 0.000 claims description 2
- 206010071577 Autoimmune hyperlipidaemia Diseases 0.000 claims description 2
- 206010064539 Autoimmune myocarditis Diseases 0.000 claims description 2
- 206010069002 Autoimmune pancreatitis Diseases 0.000 claims description 2
- 208000022106 Autoimmune polyendocrinopathy type 2 Diseases 0.000 claims description 2
- 206010003840 Autonomic nervous system imbalance Diseases 0.000 claims description 2
- 208000023328 Basedow disease Diseases 0.000 claims description 2
- 208000008439 Biliary Liver Cirrhosis Diseases 0.000 claims description 2
- 208000033222 Biliary cirrhosis primary Diseases 0.000 claims description 2
- 208000031229 Cardiomyopathies Diseases 0.000 claims description 2
- 208000005024 Castleman disease Diseases 0.000 claims description 2
- 208000024699 Chagas disease Diseases 0.000 claims description 2
- 206010008609 Cholangitis sclerosing Diseases 0.000 claims description 2
- 208000006344 Churg-Strauss Syndrome Diseases 0.000 claims description 2
- 208000010007 Cogan syndrome Diseases 0.000 claims description 2
- 208000011038 Cold agglutinin disease Diseases 0.000 claims description 2
- 206010009868 Cold type haemolytic anaemia Diseases 0.000 claims description 2
- 208000013586 Complex regional pain syndrome type 1 Diseases 0.000 claims description 2
- 206010011258 Coxsackie myocarditis Diseases 0.000 claims description 2
- 208000019707 Cryoglobulinemic vasculitis Diseases 0.000 claims description 2
- 206010012468 Dermatitis herpetiformis Diseases 0.000 claims description 2
- 206010048768 Dermatosis Diseases 0.000 claims description 2
- 201000009273 Endometriosis Diseases 0.000 claims description 2
- 206010014954 Eosinophilic fasciitis Diseases 0.000 claims description 2
- 208000018428 Eosinophilic granulomatosis with polyangiitis Diseases 0.000 claims description 2
- 206010064212 Eosinophilic oesophagitis Diseases 0.000 claims description 2
- 206010015226 Erythema nodosum Diseases 0.000 claims description 2
- 208000004332 Evans syndrome Diseases 0.000 claims description 2
- 206010018364 Glomerulonephritis Diseases 0.000 claims description 2
- 208000024869 Goodpasture syndrome Diseases 0.000 claims description 2
- 208000015023 Graves' disease Diseases 0.000 claims description 2
- 208000035895 Guillain-Barré syndrome Diseases 0.000 claims description 2
- 208000030836 Hashimoto thyroiditis Diseases 0.000 claims description 2
- 206010019263 Heart block congenital Diseases 0.000 claims description 2
- 201000004331 Henoch-Schoenlein purpura Diseases 0.000 claims description 2
- 206010019617 Henoch-Schonlein purpura Diseases 0.000 claims description 2
- 206010019939 Herpes gestationis Diseases 0.000 claims description 2
- 208000031814 IgA Vasculitis Diseases 0.000 claims description 2
- 208000010159 IgA glomerulonephritis Diseases 0.000 claims description 2
- 206010021263 IgA nephropathy Diseases 0.000 claims description 2
- 208000014919 IgG4-related retroperitoneal fibrosis Diseases 0.000 claims description 2
- 206010061598 Immunodeficiency Diseases 0.000 claims description 2
- 208000029462 Immunodeficiency disease Diseases 0.000 claims description 2
- 102100037850 Interferon gamma Human genes 0.000 claims description 2
- 108010074328 Interferon-gamma Proteins 0.000 claims description 2
- 102000013691 Interleukin-17 Human genes 0.000 claims description 2
- 108050003558 Interleukin-17 Proteins 0.000 claims description 2
- 206010022557 Intermediate uveitis Diseases 0.000 claims description 2
- 208000005615 Interstitial Cystitis Diseases 0.000 claims description 2
- 201000010743 Lambert-Eaton myasthenic syndrome Diseases 0.000 claims description 2
- 208000032514 Leukocytoclastic vasculitis Diseases 0.000 claims description 2
- 206010024434 Lichen sclerosus Diseases 0.000 claims description 2
- 208000027530 Meniere disease Diseases 0.000 claims description 2
- 206010049567 Miller Fisher syndrome Diseases 0.000 claims description 2
- 208000024599 Mooren ulcer Diseases 0.000 claims description 2
- 208000012192 Mucous membrane pemphigoid Diseases 0.000 claims description 2
- 208000000112 Myalgia Diseases 0.000 claims description 2
- 201000002481 Myositis Diseases 0.000 claims description 2
- 206010071579 Neuronal neuropathy Diseases 0.000 claims description 2
- 208000003435 Optic Neuritis Diseases 0.000 claims description 2
- 206010053869 POEMS syndrome Diseases 0.000 claims description 2
- 206010048705 Paraneoplastic cerebellar degeneration Diseases 0.000 claims description 2
- 208000004788 Pars Planitis Diseases 0.000 claims description 2
- 208000008223 Pemphigoid Gestationis Diseases 0.000 claims description 2
- 241000721454 Pemphigus Species 0.000 claims description 2
- 208000031845 Pernicious anaemia Diseases 0.000 claims description 2
- 208000000766 Pityriasis Lichenoides Diseases 0.000 claims description 2
- 206010048895 Pityriasis lichenoides et varioliformis acuta Diseases 0.000 claims description 2
- 206010065159 Polychondritis Diseases 0.000 claims description 2
- 208000031732 Post-Lyme Disease Syndrome Diseases 0.000 claims description 2
- 208000004347 Postpericardiotomy Syndrome Diseases 0.000 claims description 2
- 208000012654 Primary biliary cholangitis Diseases 0.000 claims description 2
- 208000037534 Progressive hemifacial atrophy Diseases 0.000 claims description 2
- 208000003670 Pure Red-Cell Aplasia Diseases 0.000 claims description 2
- 208000012322 Raynaud phenomenon Diseases 0.000 claims description 2
- 201000001947 Reflex Sympathetic Dystrophy Diseases 0.000 claims description 2
- 208000033464 Reiter syndrome Diseases 0.000 claims description 2
- 208000005793 Restless legs syndrome Diseases 0.000 claims description 2
- 206010038979 Retroperitoneal fibrosis Diseases 0.000 claims description 2
- 208000025747 Rheumatic disease Diseases 0.000 claims description 2
- 206010039705 Scleritis Diseases 0.000 claims description 2
- 206010039710 Scleroderma Diseases 0.000 claims description 2
- 208000021386 Sjogren Syndrome Diseases 0.000 claims description 2
- 206010072148 Stiff-Person syndrome Diseases 0.000 claims description 2
- 241000194017 Streptococcus Species 0.000 claims description 2
- 206010042742 Sympathetic ophthalmia Diseases 0.000 claims description 2
- 208000001106 Takayasu Arteritis Diseases 0.000 claims description 2
- 206010071574 Testicular autoimmunity Diseases 0.000 claims description 2
- 206010051526 Tolosa-Hunt syndrome Diseases 0.000 claims description 2
- 241000223109 Trypanosoma cruzi Species 0.000 claims description 2
- 208000026928 Turner syndrome Diseases 0.000 claims description 2
- 108700036309 Type I Plasminogen Deficiency Proteins 0.000 claims description 2
- 206010064996 Ulcerative keratitis Diseases 0.000 claims description 2
- 208000024780 Urticaria Diseases 0.000 claims description 2
- 206010047115 Vasculitis Diseases 0.000 claims description 2
- 206010047642 Vitiligo Diseases 0.000 claims description 2
- 208000004631 alopecia areata Diseases 0.000 claims description 2
- 206010002022 amyloidosis Diseases 0.000 claims description 2
- 208000007502 anemia Diseases 0.000 claims description 2
- 201000000448 autoimmune hemolytic anemia Diseases 0.000 claims description 2
- 208000006424 autoimmune oophoritis Diseases 0.000 claims description 2
- 201000009780 autoimmune polyendocrine syndrome type 2 Diseases 0.000 claims description 2
- 206010071578 autoimmune retinopathy Diseases 0.000 claims description 2
- 208000010928 autoimmune thyroid disease Diseases 0.000 claims description 2
- 230000003376 axonal effect Effects 0.000 claims description 2
- 208000000594 bullous pemphigoid Diseases 0.000 claims description 2
- 208000025302 chronic primary adrenal insufficiency Diseases 0.000 claims description 2
- 201000010002 cicatricial pemphigoid Diseases 0.000 claims description 2
- 201000004395 congenital heart block Diseases 0.000 claims description 2
- 201000003278 cryoglobulinemia Diseases 0.000 claims description 2
- 230000003210 demyelinating effect Effects 0.000 claims description 2
- 239000003937 drug carrier Substances 0.000 claims description 2
- 208000019479 dysautonomia Diseases 0.000 claims description 2
- 206010014599 encephalitis Diseases 0.000 claims description 2
- 201000000708 eosinophilic esophagitis Diseases 0.000 claims description 2
- 208000002980 facial hemiatrophy Diseases 0.000 claims description 2
- ZZUFCTLCJUWOSV-UHFFFAOYSA-N furosemide Chemical compound C1=C(Cl)C(S(=O)(=O)N)=CC(C(O)=O)=C1NCC1=CC=CO1 ZZUFCTLCJUWOSV-UHFFFAOYSA-N 0.000 claims description 2
- 208000018090 giant cell myocarditis Diseases 0.000 claims description 2
- 208000007475 hemolytic anemia Diseases 0.000 claims description 2
- 201000006362 hypersensitivity vasculitis Diseases 0.000 claims description 2
- 230000007813 immunodeficiency Effects 0.000 claims description 2
- 208000015446 immunoglobulin a vasculitis Diseases 0.000 claims description 2
- 230000004957 immunoregulator effect Effects 0.000 claims description 2
- 201000008319 inclusion body myositis Diseases 0.000 claims description 2
- 208000036971 interstitial lung disease 2 Diseases 0.000 claims description 2
- 201000002215 juvenile rheumatoid arthritis Diseases 0.000 claims description 2
- 201000011486 lichen planus Diseases 0.000 claims description 2
- 206010071570 ligneous conjunctivitis Diseases 0.000 claims description 2
- 206010063344 microscopic polyangiitis Diseases 0.000 claims description 2
- 201000006417 multiple sclerosis Diseases 0.000 claims description 2
- 206010028417 myasthenia gravis Diseases 0.000 claims description 2
- 201000003631 narcolepsy Diseases 0.000 claims description 2
- 201000008383 nephritis Diseases 0.000 claims description 2
- 201000001119 neuropathy Diseases 0.000 claims description 2
- 230000007823 neuropathy Effects 0.000 claims description 2
- 208000004235 neutropenia Diseases 0.000 claims description 2
- 208000015200 ocular cicatricial pemphigoid Diseases 0.000 claims description 2
- 201000005580 palindromic rheumatism Diseases 0.000 claims description 2
- 208000033808 peripheral neuropathy Diseases 0.000 claims description 2
- 201000006292 polyarteritis nodosa Diseases 0.000 claims description 2
- 208000005987 polymyositis Diseases 0.000 claims description 2
- 208000018290 primary dysautonomia Diseases 0.000 claims description 2
- 201000000742 primary sclerosing cholangitis Diseases 0.000 claims description 2
- 239000000186 progesterone Substances 0.000 claims description 2
- 229960003387 progesterone Drugs 0.000 claims description 2
- 208000005069 pulmonary fibrosis Diseases 0.000 claims description 2
- 208000009954 pyoderma gangrenosum Diseases 0.000 claims description 2
- 230000000306 recurrent effect Effects 0.000 claims description 2
- 208000009169 relapsing polychondritis Diseases 0.000 claims description 2
- 230000000552 rheumatic effect Effects 0.000 claims description 2
- 201000003068 rheumatic fever Diseases 0.000 claims description 2
- 201000000306 sarcoidosis Diseases 0.000 claims description 2
- 208000010157 sclerosing cholangitis Diseases 0.000 claims description 2
- 208000017520 skin disease Diseases 0.000 claims description 2
- 201000000596 systemic lupus erythematosus Diseases 0.000 claims description 2
- 208000009174 transverse myelitis Diseases 0.000 claims description 2
- 230000002568 urticarial effect Effects 0.000 claims description 2
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 claims 2
- GMKMEZVLHJARHF-UHFFFAOYSA-N (2R,6R)-form-2.6-Diaminoheptanedioic acid Natural products OC(=O)C(N)CCCC(N)C(O)=O GMKMEZVLHJARHF-UHFFFAOYSA-N 0.000 claims 1
- 208000032116 Autoimmune Experimental Encephalomyelitis Diseases 0.000 claims 1
- 102000002149 Elafin Human genes 0.000 claims 1
- 108090001005 Interleukin-6 Proteins 0.000 claims 1
- 108090001007 Interleukin-8 Proteins 0.000 claims 1
- 208000002286 Susac Syndrome Diseases 0.000 claims 1
- 102100040247 Tumor necrosis factor Human genes 0.000 claims 1
- 239000012467 final product Substances 0.000 claims 1
- GMKMEZVLHJARHF-SYDPRGILSA-N meso-2,6-diaminopimelic acid Chemical compound [O-]C(=O)[C@@H]([NH3+])CCC[C@@H]([NH3+])C([O-])=O GMKMEZVLHJARHF-SYDPRGILSA-N 0.000 claims 1
- 239000002924 silencing RNA Substances 0.000 claims 1
- 229940113082 thymine Drugs 0.000 claims 1
- 231100000167 toxic agent Toxicity 0.000 claims 1
- 239000003440 toxic substance Substances 0.000 claims 1
- 230000002401 inhibitory effect Effects 0.000 abstract description 8
- 239000008194 pharmaceutical composition Substances 0.000 abstract description 6
- 230000037456 inflammatory mechanism Effects 0.000 abstract description 2
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 312
- 230000001105 regulatory effect Effects 0.000 description 275
- 108091023040 Transcription factor Proteins 0.000 description 209
- 102000040945 Transcription factor Human genes 0.000 description 209
- 210000001035 gastrointestinal tract Anatomy 0.000 description 194
- 108091028043 Nucleic acid sequence Proteins 0.000 description 110
- 230000015572 biosynthetic process Effects 0.000 description 89
- 150000007523 nucleic acids Chemical group 0.000 description 74
- 230000001939 inductive effect Effects 0.000 description 72
- 108010046335 Ferredoxin-NADP Reductase Proteins 0.000 description 70
- 230000027455 binding Effects 0.000 description 70
- 238000004519 manufacturing process Methods 0.000 description 69
- 230000001628 butyrogenic effect Effects 0.000 description 66
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 63
- 230000001225 therapeutic effect Effects 0.000 description 61
- 230000005945 translocation Effects 0.000 description 57
- 210000004027 cell Anatomy 0.000 description 47
- 230000028327 secretion Effects 0.000 description 46
- JTTIOYHBNXDJOD-UHFFFAOYSA-N 2,4,6-triaminopyrimidine Chemical compound NC1=CC(N)=NC(N)=N1 JTTIOYHBNXDJOD-UHFFFAOYSA-N 0.000 description 36
- 101000724418 Homo sapiens Neutral amino acid transporter B(0) Proteins 0.000 description 36
- 102100028267 Neutral amino acid transporter B(0) Human genes 0.000 description 36
- 102100040870 Glycine amidinotransferase, mitochondrial Human genes 0.000 description 34
- 101000893303 Homo sapiens Glycine amidinotransferase, mitochondrial Proteins 0.000 description 34
- 230000001419 dependent effect Effects 0.000 description 33
- YGPSJZOEDVAXAB-UHFFFAOYSA-N kynurenine Chemical compound OC(=O)C(N)CC(=O)C1=CC=CC=C1N YGPSJZOEDVAXAB-UHFFFAOYSA-N 0.000 description 32
- 239000003795 chemical substances by application Substances 0.000 description 31
- 241000894007 species Species 0.000 description 30
- 108700020534 tetracycline resistance-encoding transposon repressor Proteins 0.000 description 30
- 230000001580 bacterial effect Effects 0.000 description 28
- 241000193163 Clostridioides difficile Species 0.000 description 27
- 239000012634 fragment Substances 0.000 description 27
- 230000001965 increasing effect Effects 0.000 description 25
- 102000019197 Superoxide Dismutase Human genes 0.000 description 24
- 108010012715 Superoxide dismutase Proteins 0.000 description 24
- HCZHHEIFKROPDY-UHFFFAOYSA-N kynurenic acid Chemical compound C1=CC=C2NC(C(=O)O)=CC(=O)C2=C1 HCZHHEIFKROPDY-UHFFFAOYSA-N 0.000 description 24
- 108700012359 toxins Proteins 0.000 description 24
- 230000001147 anti-toxic effect Effects 0.000 description 23
- 229940088598 enzyme Drugs 0.000 description 23
- 230000000694 effects Effects 0.000 description 22
- 102000004169 proteins and genes Human genes 0.000 description 22
- 239000003053 toxin Substances 0.000 description 22
- 231100000765 toxin Toxicity 0.000 description 22
- 229940023064 escherichia coli Drugs 0.000 description 21
- 108090000765 processed proteins & peptides Proteins 0.000 description 21
- 101100028324 Streptomyces rimosus subsp. rimosus (strain ATCC 10970 / DSM 40260 / JCM 4667 / NRRL 2234) oxyS gene Proteins 0.000 description 19
- 102100030703 Interleukin-22 Human genes 0.000 description 18
- 108010074109 interleukin-22 Proteins 0.000 description 18
- 235000018102 proteins Nutrition 0.000 description 18
- 238000013518 transcription Methods 0.000 description 18
- 230000035897 transcription Effects 0.000 description 18
- 108010002350 Interleukin-2 Proteins 0.000 description 17
- 102000000588 Interleukin-2 Human genes 0.000 description 17
- 102100036678 Interleukin-27 subunit alpha Human genes 0.000 description 17
- PYMYPHUHKUWMLA-WDCZJNDASA-N arabinose Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)C=O PYMYPHUHKUWMLA-WDCZJNDASA-N 0.000 description 17
- PYMYPHUHKUWMLA-UHFFFAOYSA-N arabinose Natural products OCC(O)C(O)C(O)C=O PYMYPHUHKUWMLA-UHFFFAOYSA-N 0.000 description 17
- SRBFZHDQGSBBOR-UHFFFAOYSA-N beta-D-Pyranose-Lyxose Natural products OC1COC(O)C(O)C1O SRBFZHDQGSBBOR-UHFFFAOYSA-N 0.000 description 17
- 230000037361 pathway Effects 0.000 description 17
- 101150026728 tesB gene Proteins 0.000 description 17
- 230000003213 activating effect Effects 0.000 description 15
- 230000006870 function Effects 0.000 description 15
- 230000007246 mechanism Effects 0.000 description 15
- 210000003289 regulatory T cell Anatomy 0.000 description 15
- 108010039224 Amidophosphoribosyltransferase Proteins 0.000 description 14
- 101100162202 Aspergillus parasiticus (strain ATCC 56775 / NRRL 5862 / SRRC 143 / SU-1) aflF gene Proteins 0.000 description 14
- 101150076456 norB gene Proteins 0.000 description 14
- 230000008520 organization Effects 0.000 description 14
- 102000004127 Cytokines Human genes 0.000 description 13
- 108090000695 Cytokines Proteins 0.000 description 13
- 230000004913 activation Effects 0.000 description 13
- 210000004379 membrane Anatomy 0.000 description 13
- 239000012528 membrane Substances 0.000 description 13
- 230000003110 anti-inflammatory effect Effects 0.000 description 12
- 230000002708 enhancing effect Effects 0.000 description 12
- 230000012010 growth Effects 0.000 description 12
- 230000002757 inflammatory effect Effects 0.000 description 12
- 230000000670 limiting effect Effects 0.000 description 12
- 108010048816 AraC Transcription Factor Proteins 0.000 description 11
- 244000052616 bacterial pathogen Species 0.000 description 11
- 230000000447 dimerizing effect Effects 0.000 description 11
- 238000001727 in vivo Methods 0.000 description 11
- QJJXYPPXXYFBGM-SHYZHZOCSA-N tacrolimus Natural products CO[C@H]1C[C@H](CC[C@@H]1O)C=C(C)[C@H]2OC(=O)[C@H]3CCCCN3C(=O)C(=O)[C@@]4(O)O[C@@H]([C@H](C[C@H]4C)OC)[C@@H](C[C@H](C)CC(=C[C@@H](CC=C)C(=O)C[C@H](O)[C@H]2C)C)OC QJJXYPPXXYFBGM-SHYZHZOCSA-N 0.000 description 11
- 102000018120 Recombinases Human genes 0.000 description 10
- 108010091086 Recombinases Proteins 0.000 description 10
- 239000004098 Tetracycline Substances 0.000 description 10
- 241000589892 Treponema denticola Species 0.000 description 10
- 238000000338 in vitro Methods 0.000 description 10
- 238000003780 insertion Methods 0.000 description 10
- 230000037431 insertion Effects 0.000 description 10
- 229960002180 tetracycline Drugs 0.000 description 10
- 229930101283 tetracycline Natural products 0.000 description 10
- 235000019364 tetracycline Nutrition 0.000 description 10
- 150000003522 tetracyclines Chemical class 0.000 description 10
- 102100031051 Cysteine and glycine-rich protein 1 Human genes 0.000 description 9
- 102100023795 Elafin Human genes 0.000 description 9
- 108700039887 Essential Genes Proteins 0.000 description 9
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 9
- 230000008859 change Effects 0.000 description 9
- 210000000805 cytoplasm Anatomy 0.000 description 9
- 239000008103 glucose Substances 0.000 description 9
- 239000000758 substrate Substances 0.000 description 9
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerol Natural products OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 8
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 8
- 229940024606 amino acid Drugs 0.000 description 8
- 229920003045 dextran sodium sulfate Polymers 0.000 description 8
- 239000000411 inducer Substances 0.000 description 8
- 239000002609 medium Substances 0.000 description 8
- FVFVNNKYKYZTJU-UHFFFAOYSA-N 6-chloro-1,3,5-triazine-2,4-diamine Chemical compound NC1=NC(N)=NC(Cl)=N1 FVFVNNKYKYZTJU-UHFFFAOYSA-N 0.000 description 7
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 7
- 108010069584 Type III Secretion Systems Proteins 0.000 description 7
- 108010073429 Type V Secretion Systems Proteins 0.000 description 7
- 230000009471 action Effects 0.000 description 7
- 210000003578 bacterial chromosome Anatomy 0.000 description 7
- 230000003247 decreasing effect Effects 0.000 description 7
- 108020004201 indoleamine 2,3-dioxygenase Proteins 0.000 description 7
- 102000006639 indoleamine 2,3-dioxygenase Human genes 0.000 description 7
- 230000006698 induction Effects 0.000 description 7
- 230000000754 repressing effect Effects 0.000 description 7
- 230000004083 survival effect Effects 0.000 description 7
- 208000024891 symptom Diseases 0.000 description 7
- 238000011144 upstream manufacturing Methods 0.000 description 7
- VWEWCZSUWOEEFM-WDSKDSINSA-N Ala-Gly-Ala-Gly Chemical compound C[C@H](N)C(=O)NCC(=O)N[C@@H](C)C(=O)NCC(O)=O VWEWCZSUWOEEFM-WDSKDSINSA-N 0.000 description 6
- 108010078791 Carrier Proteins Proteins 0.000 description 6
- 108020004705 Codon Proteins 0.000 description 6
- 108700039691 Genetic Promoter Regions Proteins 0.000 description 6
- 239000000427 antigen Substances 0.000 description 6
- 238000013459 approach Methods 0.000 description 6
- 206010009887 colitis Diseases 0.000 description 6
- 210000001723 extracellular space Anatomy 0.000 description 6
- 230000000968 intestinal effect Effects 0.000 description 6
- 102000004196 processed proteins & peptides Human genes 0.000 description 6
- 239000000047 product Substances 0.000 description 6
- 230000003248 secreting effect Effects 0.000 description 6
- 101150061166 tetR gene Proteins 0.000 description 6
- 101150072448 thrB gene Proteins 0.000 description 6
- 101150000850 thrC gene Proteins 0.000 description 6
- 238000011282 treatment Methods 0.000 description 6
- 102000003390 tumor necrosis factor Human genes 0.000 description 6
- 101100235003 Acetobacterium woodii (strain ATCC 29683 / DSM 1030 / JCM 2381 / KCTC 1655 / WB1) lctC gene Proteins 0.000 description 5
- 241000193830 Bacillus <bacterium> Species 0.000 description 5
- 241000193171 Clostridium butyricum Species 0.000 description 5
- 108020004414 DNA Proteins 0.000 description 5
- 101150110799 ETFA gene Proteins 0.000 description 5
- 101100394050 Escherichia coli (strain K12) gyrB gene Proteins 0.000 description 5
- 241000192125 Firmicutes Species 0.000 description 5
- 102000015696 Interleukins Human genes 0.000 description 5
- 108010063738 Interleukins Proteins 0.000 description 5
- 241000124008 Mammalia Species 0.000 description 5
- 229910002651 NO3 Inorganic materials 0.000 description 5
- NHNBFGGVMKEFGY-UHFFFAOYSA-N Nitrate Chemical compound [O-][N+]([O-])=O NHNBFGGVMKEFGY-UHFFFAOYSA-N 0.000 description 5
- 102000035554 Proglucagon Human genes 0.000 description 5
- 108010076504 Protein Sorting Signals Proteins 0.000 description 5
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 5
- 241000607142 Salmonella Species 0.000 description 5
- 108010008681 Type II Secretion Systems Proteins 0.000 description 5
- 101150077561 aceF gene Proteins 0.000 description 5
- 101150004068 acrB gene Proteins 0.000 description 5
- 101150068316 acrC gene Proteins 0.000 description 5
- 101150044616 araC gene Proteins 0.000 description 5
- 230000033228 biological regulation Effects 0.000 description 5
- FLKYBGKDCCEQQM-WYUVZMMLSA-M cefazolin sodium Chemical compound [Na+].S1C(C)=NN=C1SCC1=C(C([O-])=O)N2C(=O)[C@@H](NC(=O)CN3N=NN=C3)[C@H]2SC1 FLKYBGKDCCEQQM-WYUVZMMLSA-M 0.000 description 5
- 150000001875 compounds Chemical class 0.000 description 5
- 230000009368 gene silencing by RNA Effects 0.000 description 5
- 101150066555 lacZ gene Proteins 0.000 description 5
- 238000010172 mouse model Methods 0.000 description 5
- 101150029849 nsrR gene Proteins 0.000 description 5
- 230000008506 pathogenesis Effects 0.000 description 5
- 210000001322 periplasm Anatomy 0.000 description 5
- 229920001184 polypeptide Polymers 0.000 description 5
- 230000008685 targeting Effects 0.000 description 5
- 108091006106 transcriptional activators Proteins 0.000 description 5
- 101150096946 ydfZ gene Proteins 0.000 description 5
- 102100025230 2-amino-3-ketobutyrate coenzyme A ligase, mitochondrial Human genes 0.000 description 4
- 108010087522 Aeromonas hydrophilia lipase-acyltransferase Proteins 0.000 description 4
- 241001136167 Anaerotignum propionicum Species 0.000 description 4
- 101100480489 Arabidopsis thaliana TAAC gene Proteins 0.000 description 4
- 101100404144 Bacillus subtilis (strain 168) nasD gene Proteins 0.000 description 4
- 241000186016 Bifidobacterium bifidum Species 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 101001048718 Homo sapiens Elafin Proteins 0.000 description 4
- 240000001046 Lactobacillus acidophilus Species 0.000 description 4
- 235000013956 Lactobacillus acidophilus Nutrition 0.000 description 4
- 240000006024 Lactobacillus plantarum Species 0.000 description 4
- 235000013965 Lactobacillus plantarum Nutrition 0.000 description 4
- YJPIGAIKUZMOQA-UHFFFAOYSA-N Melatonin Natural products COC1=CC=C2N(C(C)=O)C=C(CCN)C2=C1 YJPIGAIKUZMOQA-UHFFFAOYSA-N 0.000 description 4
- 241000588652 Neisseria gonorrhoeae Species 0.000 description 4
- 102000004316 Oxidoreductases Human genes 0.000 description 4
- 108090000854 Oxidoreductases Proteins 0.000 description 4
- 102000035195 Peptidases Human genes 0.000 description 4
- 108091005804 Peptidases Proteins 0.000 description 4
- 108010058003 Proglucagon Proteins 0.000 description 4
- 210000001744 T-lymphocyte Anatomy 0.000 description 4
- 108010064721 Type I Secretion Systems Proteins 0.000 description 4
- 101150015189 aceE gene Proteins 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N acetic acid Substances CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 4
- 230000005784 autoimmunity Effects 0.000 description 4
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 4
- 229940002008 bifidobacterium bifidum Drugs 0.000 description 4
- 230000024245 cell differentiation Effects 0.000 description 4
- 238000003776 cleavage reaction Methods 0.000 description 4
- 238000010276 construction Methods 0.000 description 4
- 230000006378 damage Effects 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 230000004069 differentiation Effects 0.000 description 4
- 230000004890 epithelial barrier function Effects 0.000 description 4
- 238000004817 gas chromatography Methods 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- 239000005090 green fluorescent protein Substances 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 230000010354 integration Effects 0.000 description 4
- 102000004114 interleukin 20 Human genes 0.000 description 4
- 108090000681 interleukin 20 Proteins 0.000 description 4
- 101150013110 katG gene Proteins 0.000 description 4
- 229940039695 lactobacillus acidophilus Drugs 0.000 description 4
- 229940072205 lactobacillus plantarum Drugs 0.000 description 4
- 230000003902 lesion Effects 0.000 description 4
- 210000004185 liver Anatomy 0.000 description 4
- 238000004949 mass spectrometry Methods 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- DRLFMBDRBRZALE-UHFFFAOYSA-N melatonin Chemical compound COC1=CC=C2NC=C(CCNC(C)=O)C2=C1 DRLFMBDRBRZALE-UHFFFAOYSA-N 0.000 description 4
- 229960003987 melatonin Drugs 0.000 description 4
- 108020004999 messenger RNA Proteins 0.000 description 4
- 230000000813 microbial effect Effects 0.000 description 4
- 101150044129 nirB gene Proteins 0.000 description 4
- 230000003647 oxidation Effects 0.000 description 4
- 238000007254 oxidation reaction Methods 0.000 description 4
- IWDCLRJOBJJRNH-UHFFFAOYSA-N p-cresol Chemical compound CC1=CC=C(O)C=C1 IWDCLRJOBJJRNH-UHFFFAOYSA-N 0.000 description 4
- 230000029058 respiratory gaseous exchange Effects 0.000 description 4
- 210000003705 ribosome Anatomy 0.000 description 4
- 230000007017 scission Effects 0.000 description 4
- QZAYGJVTTNCVMB-UHFFFAOYSA-N serotonin Chemical compound C1=C(O)C=C2C(CCN)=CNC2=C1 QZAYGJVTTNCVMB-UHFFFAOYSA-N 0.000 description 4
- 210000000813 small intestine Anatomy 0.000 description 4
- 238000006467 substitution reaction Methods 0.000 description 4
- 230000002103 transcriptional effect Effects 0.000 description 4
- 230000032258 transport Effects 0.000 description 4
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 3
- FJKROLUGYXJWQN-UHFFFAOYSA-N 4-hydroxybenzoic acid Chemical compound OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 3
- 101000651036 Arabidopsis thaliana Galactolipid galactosyltransferase SFR2, chloroplastic Proteins 0.000 description 3
- 101100427060 Bacillus spizizenii (strain ATCC 23059 / NRRL B-14472 / W23) thyA1 gene Proteins 0.000 description 3
- 244000063299 Bacillus subtilis Species 0.000 description 3
- 235000014469 Bacillus subtilis Nutrition 0.000 description 3
- 101100002068 Bacillus subtilis (strain 168) araR gene Proteins 0.000 description 3
- 101100462127 Bacillus subtilis (strain 168) ohrA gene Proteins 0.000 description 3
- 241000606124 Bacteroides fragilis Species 0.000 description 3
- 241000606123 Bacteroides thetaiotaomicron Species 0.000 description 3
- 241000901050 Bifidobacterium animalis subsp. lactis Species 0.000 description 3
- 241000186015 Bifidobacterium longum subsp. infantis Species 0.000 description 3
- 108091006146 Channels Proteins 0.000 description 3
- 229920002307 Dextran Polymers 0.000 description 3
- 108700035208 EC 7.-.-.- Proteins 0.000 description 3
- 241000194031 Enterococcus faecium Species 0.000 description 3
- 101100153154 Escherichia phage T5 thy gene Proteins 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 241000186394 Eubacterium Species 0.000 description 3
- 101710198884 GATA-type zinc finger protein 1 Proteins 0.000 description 3
- DTHNMHAUYICORS-KTKZVXAJSA-N Glucagon-like peptide 1 Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](CCCNC(N)=N)C(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC=1N=CNC=1)[C@@H](C)O)[C@@H](C)O)C(C)C)C1=CC=CC=C1 DTHNMHAUYICORS-KTKZVXAJSA-N 0.000 description 3
- 102100024977 Glutamine-tRNA ligase Human genes 0.000 description 3
- 101150108784 INSB gene Proteins 0.000 description 3
- 244000199885 Lactobacillus bulgaricus Species 0.000 description 3
- 235000013960 Lactobacillus bulgaricus Nutrition 0.000 description 3
- 241001468157 Lactobacillus johnsonii Species 0.000 description 3
- 241000186605 Lactobacillus paracasei Species 0.000 description 3
- 241000186604 Lactobacillus reuteri Species 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 241000699670 Mus sp. Species 0.000 description 3
- 206010028980 Neoplasm Diseases 0.000 description 3
- 101710116435 Outer membrane protein Proteins 0.000 description 3
- 102100040918 Pro-glucagon Human genes 0.000 description 3
- LCTONWCANYUPML-UHFFFAOYSA-M Pyruvate Chemical compound CC(=O)C([O-])=O LCTONWCANYUPML-UHFFFAOYSA-M 0.000 description 3
- 238000011529 RT qPCR Methods 0.000 description 3
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 3
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 3
- 101100313751 Rickettsia conorii (strain ATCC VR-613 / Malish 7) thyX gene Proteins 0.000 description 3
- 241000235070 Saccharomyces Species 0.000 description 3
- 244000057717 Streptococcus lactis Species 0.000 description 3
- 235000014897 Streptococcus lactis Nutrition 0.000 description 3
- OUUQCZGPVNCOIJ-UHFFFAOYSA-M Superoxide Chemical compound [O-][O] OUUQCZGPVNCOIJ-UHFFFAOYSA-M 0.000 description 3
- 108010000134 Vascular Cell Adhesion Molecule-1 Proteins 0.000 description 3
- 102100023543 Vascular cell adhesion protein 1 Human genes 0.000 description 3
- 230000001154 acute effect Effects 0.000 description 3
- 101150024831 ahpC gene Proteins 0.000 description 3
- 150000001413 amino acids Chemical class 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 238000003491 array Methods 0.000 description 3
- 238000003149 assay kit Methods 0.000 description 3
- 210000004082 barrier epithelial cell Anatomy 0.000 description 3
- 229940004120 bifidobacterium infantis Drugs 0.000 description 3
- 229940009289 bifidobacterium lactis Drugs 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 239000000090 biomarker Substances 0.000 description 3
- 230000001851 biosynthetic effect Effects 0.000 description 3
- CRFNGMNYKDXRTN-CITAKDKDSA-N butyryl-CoA Chemical compound O[C@@H]1[C@H](OP(O)(O)=O)[C@@H](COP(O)(=O)OP(O)(=O)OCC(C)(C)[C@@H](O)C(=O)NCCC(=O)NCCSC(=O)CCC)O[C@H]1N1C2=NC=NC(N)=C2N=C1 CRFNGMNYKDXRTN-CITAKDKDSA-N 0.000 description 3
- 210000004899 c-terminal region Anatomy 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 230000030833 cell death Effects 0.000 description 3
- 210000004922 colonic epithelial cell Anatomy 0.000 description 3
- 230000001276 controlling effect Effects 0.000 description 3
- 230000000593 degrading effect Effects 0.000 description 3
- 229960002086 dextran Drugs 0.000 description 3
- LZDSILRDTDCIQT-UHFFFAOYSA-N dinitrogen trioxide Chemical compound [O-][N+](=O)N=O LZDSILRDTDCIQT-UHFFFAOYSA-N 0.000 description 3
- 238000001839 endoscopy Methods 0.000 description 3
- 210000003495 flagella Anatomy 0.000 description 3
- 108010051239 glutaminyl-tRNA synthetase Proteins 0.000 description 3
- 210000000987 immune system Anatomy 0.000 description 3
- 238000003018 immunoassay Methods 0.000 description 3
- 102000003898 interleukin-24 Human genes 0.000 description 3
- 108090000237 interleukin-24 Proteins 0.000 description 3
- 108010067653 lactate dehydratase Proteins 0.000 description 3
- 229940004208 lactobacillus bulgaricus Drugs 0.000 description 3
- 229940001882 lactobacillus reuteri Drugs 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 230000004060 metabolic process Effects 0.000 description 3
- 210000000822 natural killer cell Anatomy 0.000 description 3
- 210000000440 neutrophil Anatomy 0.000 description 3
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 3
- BOPGDPNILDQYTO-NNYOXOHSSA-N nicotinamide-adenine dinucleotide Chemical compound C1=CCC(C(=O)N)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C3=NC=NC(N)=C3N=C2)O)O1 BOPGDPNILDQYTO-NNYOXOHSSA-N 0.000 description 3
- 102000039446 nucleic acids Human genes 0.000 description 3
- 108020004707 nucleic acids Proteins 0.000 description 3
- 230000003287 optical effect Effects 0.000 description 3
- 230000001590 oxidative effect Effects 0.000 description 3
- 101150114893 oxyR gene Proteins 0.000 description 3
- 230000007170 pathology Effects 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- CMFNMSMUKZHDEY-UHFFFAOYSA-N peroxynitrous acid Chemical compound OON=O CMFNMSMUKZHDEY-UHFFFAOYSA-N 0.000 description 3
- 238000003566 phosphorylation assay Methods 0.000 description 3
- 238000003752 polymerase chain reaction Methods 0.000 description 3
- 230000002265 prevention Effects 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 230000009467 reduction Effects 0.000 description 3
- 229960002181 saccharomyces boulardii Drugs 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 101150033650 soxS gene Proteins 0.000 description 3
- 125000003003 spiro group Chemical group 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000009885 systemic effect Effects 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 101150072314 thyA gene Proteins 0.000 description 3
- 210000001578 tight junction Anatomy 0.000 description 3
- 210000001519 tissue Anatomy 0.000 description 3
- 108091008023 transcriptional regulators Proteins 0.000 description 3
- 108091006107 transcriptional repressors Proteins 0.000 description 3
- 230000014616 translation Effects 0.000 description 3
- 230000001960 triggered effect Effects 0.000 description 3
- 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
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 2
- XQXPVVBIMDBYFF-UHFFFAOYSA-N 4-hydroxyphenylacetic acid Chemical compound OC(=O)CC1=CC=C(O)C=C1 XQXPVVBIMDBYFF-UHFFFAOYSA-N 0.000 description 2
- 102000005416 ATP-Binding Cassette Transporters Human genes 0.000 description 2
- 108010006533 ATP-Binding Cassette Transporters Proteins 0.000 description 2
- RZVAJINKPMORJF-UHFFFAOYSA-N Acetaminophen Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 2
- 241001430271 Acetitomaculum Species 0.000 description 2
- 241001112780 Acetoanaerobium Species 0.000 description 2
- 241001135190 Acetohalobium Species 0.000 description 2
- 241000204396 Acetonema Species 0.000 description 2
- NIXOWILDQLNWCW-UHFFFAOYSA-M Acrylate Chemical compound [O-]C(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-M 0.000 description 2
- 101100059196 Alkalihalobacillus pseudofirmus (strain ATCC BAA-2126 / JCM 17055 / OF4) katE gene Proteins 0.000 description 2
- 239000004475 Arginine Substances 0.000 description 2
- 241000193749 Bacillus coagulans Species 0.000 description 2
- 241001608472 Bifidobacterium longum Species 0.000 description 2
- 241000605902 Butyrivibrio Species 0.000 description 2
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 102000014914 Carrier Proteins Human genes 0.000 description 2
- 102000019034 Chemokines Human genes 0.000 description 2
- 108010012236 Chemokines Proteins 0.000 description 2
- 241000588923 Citrobacter Species 0.000 description 2
- 208000035473 Communicable disease Diseases 0.000 description 2
- 108091035707 Consensus sequence Proteins 0.000 description 2
- 241000186226 Corynebacterium glutamicum Species 0.000 description 2
- IVOMOUWHDPKRLL-KQYNXXCUSA-N Cyclic adenosine monophosphate Chemical compound C([C@H]1O2)OP(O)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=CN=C2N)=C2N=C1 IVOMOUWHDPKRLL-KQYNXXCUSA-N 0.000 description 2
- 102100029133 DNA damage-induced apoptosis suppressor protein Human genes 0.000 description 2
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 2
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 2
- 101100465553 Dictyostelium discoideum psmB6 gene Proteins 0.000 description 2
- 108010028127 Dihydrolipoamide Dehydrogenase Proteins 0.000 description 2
- 102000028526 Dihydrolipoamide Dehydrogenase Human genes 0.000 description 2
- MYMOFIZGZYHOMD-UHFFFAOYSA-N Dioxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 2
- 241000792859 Enema Species 0.000 description 2
- 241000588914 Enterobacter Species 0.000 description 2
- 241000194033 Enterococcus Species 0.000 description 2
- 241001608234 Faecalibacterium Species 0.000 description 2
- 108091026922 FnrS RNA Proteins 0.000 description 2
- 241000605909 Fusobacterium Species 0.000 description 2
- 101000918646 Homo sapiens DNA damage-induced apoptosis suppressor protein Proteins 0.000 description 2
- 101000853000 Homo sapiens Interleukin-26 Proteins 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- SIKJAQJRHWYJAI-UHFFFAOYSA-N Indole Chemical compound C1=CC=C2NC=CC2=C1 SIKJAQJRHWYJAI-UHFFFAOYSA-N 0.000 description 2
- 102100036679 Interleukin-26 Human genes 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- 241001026509 Kata Species 0.000 description 2
- 241000588748 Klebsiella Species 0.000 description 2
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 2
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 2
- 244000199866 Lactobacillus casei Species 0.000 description 2
- 235000013958 Lactobacillus casei Nutrition 0.000 description 2
- 241000218588 Lactobacillus rhamnosus Species 0.000 description 2
- 102100025169 Max-binding protein MNT Human genes 0.000 description 2
- 241000604448 Megasphaera elsdenii Species 0.000 description 2
- 102000018897 Membrane Fusion Proteins Human genes 0.000 description 2
- 108010027796 Membrane Fusion Proteins Proteins 0.000 description 2
- 108700011259 MicroRNAs Proteins 0.000 description 2
- 241000736262 Microbiota Species 0.000 description 2
- 241000178985 Moorella Species 0.000 description 2
- 101100509674 Mycolicibacterium smegmatis (strain ATCC 700084 / mc(2)155) katG3 gene Proteins 0.000 description 2
- 150000001200 N-acyl ethanolamides Chemical class 0.000 description 2
- GQPLMRYTRLFLPF-UHFFFAOYSA-N Nitrous Oxide Chemical compound [O-][N+]#N GQPLMRYTRLFLPF-UHFFFAOYSA-N 0.000 description 2
- 241000178986 Oxobacter Species 0.000 description 2
- 101150032327 PI3 gene Proteins 0.000 description 2
- 241001607889 Peptoclostridium Species 0.000 description 2
- 239000004365 Protease Substances 0.000 description 2
- 102100029812 Protein S100-A12 Human genes 0.000 description 2
- 101710110949 Protein S100-A12 Proteins 0.000 description 2
- 241000202386 Pseudobutyrivibrio Species 0.000 description 2
- 101100169519 Pyrococcus abyssi (strain GE5 / Orsay) dapAL gene Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- AUNGANRZJHBGPY-SCRDCRAPSA-N Riboflavin Chemical compound OC[C@@H](O)[C@@H](O)[C@@H](O)CN1C=2C=C(C)C(C)=CC=2N=C2C1=NC(=O)NC2=O AUNGANRZJHBGPY-SCRDCRAPSA-N 0.000 description 2
- 241000192031 Ruminococcus Species 0.000 description 2
- 241000607768 Shigella Species 0.000 description 2
- 206010049416 Short-bowel syndrome Diseases 0.000 description 2
- 241000204388 Sporomusa Species 0.000 description 2
- 241000191940 Staphylococcus Species 0.000 description 2
- 241001234687 Thermacetogenium Species 0.000 description 2
- 241000589886 Treponema Species 0.000 description 2
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 2
- 108010046504 Type IV Secretion Systems Proteins 0.000 description 2
- 108010046179 Type VI Secretion Systems Proteins 0.000 description 2
- IVOMOUWHDPKRLL-UHFFFAOYSA-N UNPD107823 Natural products O1C2COP(O)(=O)OC2C(O)C1N1C(N=CN=C2N)=C2N=C1 IVOMOUWHDPKRLL-UHFFFAOYSA-N 0.000 description 2
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 2
- 241000607734 Yersinia <bacteria> Species 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 108010041620 acryloyl-CoA reductase Proteins 0.000 description 2
- 238000013019 agitation Methods 0.000 description 2
- 230000001668 ameliorated effect Effects 0.000 description 2
- 230000009604 anaerobic growth Effects 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 108700003859 araC Genes Proteins 0.000 description 2
- YZXBAPSDXZZRGB-DOFZRALJSA-N arachidonic acid Chemical compound CCCCC\C=C/C\C=C/C\C=C/C\C=C/CCCC(O)=O YZXBAPSDXZZRGB-DOFZRALJSA-N 0.000 description 2
- 229940054340 bacillus coagulans Drugs 0.000 description 2
- 229940009291 bifidobacterium longum Drugs 0.000 description 2
- 108091008324 binding proteins Proteins 0.000 description 2
- 238000001574 biopsy Methods 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 239000006227 byproduct Substances 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 229910052799 carbon Inorganic materials 0.000 description 2
- 229910002092 carbon dioxide Inorganic materials 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 235000010980 cellulose Nutrition 0.000 description 2
- 229920002678 cellulose Polymers 0.000 description 2
- 239000001913 cellulose Substances 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol 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
- 230000002759 chromosomal effect Effects 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- CVSVTCORWBXHQV-UHFFFAOYSA-N creatine Chemical compound NC(=[NH2+])N(C)CC([O-])=O CVSVTCORWBXHQV-UHFFFAOYSA-N 0.000 description 2
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 2
- 229940095074 cyclic amp Drugs 0.000 description 2
- 101150011371 dapA gene Proteins 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 230000002939 deleterious effect Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 235000014113 dietary fatty acids Nutrition 0.000 description 2
- XEYBRNLFEZDVAW-ARSRFYASSA-N dinoprostone Chemical compound CCCCC[C@H](O)\C=C\[C@H]1[C@H](O)CC(=O)[C@@H]1C\C=C/CCCC(O)=O XEYBRNLFEZDVAW-ARSRFYASSA-N 0.000 description 2
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 239000002621 endocannabinoid Substances 0.000 description 2
- 239000007920 enema Substances 0.000 description 2
- 229940079360 enema for constipation Drugs 0.000 description 2
- 210000002919 epithelial cell Anatomy 0.000 description 2
- 210000003238 esophagus Anatomy 0.000 description 2
- 229930195729 fatty acid Natural products 0.000 description 2
- 239000000194 fatty acid Substances 0.000 description 2
- 150000004665 fatty acids Chemical class 0.000 description 2
- 230000002349 favourable effect Effects 0.000 description 2
- 230000002550 fecal effect Effects 0.000 description 2
- 239000007789 gas Substances 0.000 description 2
- 235000021472 generally recognized as safe Nutrition 0.000 description 2
- 210000004907 gland Anatomy 0.000 description 2
- 229930195712 glutamate Natural products 0.000 description 2
- 208000035474 group of disease Diseases 0.000 description 2
- 244000005709 gut microbiome Species 0.000 description 2
- 230000010243 gut motility Effects 0.000 description 2
- 239000003228 hemolysin Substances 0.000 description 2
- 101150063051 hom gene Proteins 0.000 description 2
- HBMCQTHGYMTCOF-UHFFFAOYSA-N hydroquinone monoacetate Natural products CC(=O)OC1=CC=C(O)C=C1 HBMCQTHGYMTCOF-UHFFFAOYSA-N 0.000 description 2
- QWPPOHNGKGFGJK-UHFFFAOYSA-N hypochlorous acid Chemical compound ClO QWPPOHNGKGFGJK-UHFFFAOYSA-N 0.000 description 2
- 101150095957 ilvA gene Proteins 0.000 description 2
- 230000008676 import Effects 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- BXFFHSIDQOFMLE-UHFFFAOYSA-N indoxyl sulfate Chemical compound C1=CC=C2C(OS(=O)(=O)O)=CNC2=C1 BXFFHSIDQOFMLE-UHFFFAOYSA-N 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 229940076144 interleukin-10 Drugs 0.000 description 2
- 230000003870 intestinal permeability Effects 0.000 description 2
- 210000000936 intestine Anatomy 0.000 description 2
- 150000002500 ions Chemical class 0.000 description 2
- 101150115163 katA gene Proteins 0.000 description 2
- 101150108683 katG2 gene Proteins 0.000 description 2
- 229940017800 lactobacillus casei Drugs 0.000 description 2
- 210000002429 large intestine Anatomy 0.000 description 2
- 210000003750 lower gastrointestinal tract Anatomy 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 239000002207 metabolite Substances 0.000 description 2
- 239000002679 microRNA Substances 0.000 description 2
- 244000005700 microbiome Species 0.000 description 2
- 230000003472 neutralizing effect Effects 0.000 description 2
- 101150035558 nuoB gene Proteins 0.000 description 2
- 235000015097 nutrients Nutrition 0.000 description 2
- 230000009437 off-target effect Effects 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 150000001451 organic peroxides Chemical class 0.000 description 2
- 239000007800 oxidant agent Substances 0.000 description 2
- 230000007918 pathogenicity Effects 0.000 description 2
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 2
- 150000002978 peroxides Chemical class 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 239000011148 porous material Substances 0.000 description 2
- 230000001737 promoting effect Effects 0.000 description 2
- 230000000644 propagated effect Effects 0.000 description 2
- BHMBVRSPMRCCGG-OUTUXVNYSA-N prostaglandin D2 Chemical compound CCCCC[C@H](O)\C=C\[C@@H]1[C@@H](C\C=C/CCCC(O)=O)[C@@H](O)CC1=O BHMBVRSPMRCCGG-OUTUXVNYSA-N 0.000 description 2
- 235000019833 protease Nutrition 0.000 description 2
- 235000019419 proteases Nutrition 0.000 description 2
- 230000001681 protective effect Effects 0.000 description 2
- KIDHWZJUCRJVML-UHFFFAOYSA-N putrescine Chemical compound NCCCCN KIDHWZJUCRJVML-UHFFFAOYSA-N 0.000 description 2
- 150000003254 radicals Chemical class 0.000 description 2
- 238000009790 rate-determining step (RDS) Methods 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 229940076279 serotonin Drugs 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 230000011664 signaling Effects 0.000 description 2
- ATHGHQPFGPMSJY-UHFFFAOYSA-N spermidine Chemical compound NCCCCNCCCN ATHGHQPFGPMSJY-UHFFFAOYSA-N 0.000 description 2
- PFNFFQXMRSDOHW-UHFFFAOYSA-N spermine Chemical compound NCCCNCCCCNCCCN PFNFFQXMRSDOHW-UHFFFAOYSA-N 0.000 description 2
- 230000006641 stabilisation Effects 0.000 description 2
- 238000011105 stabilization Methods 0.000 description 2
- 239000007858 starting material Substances 0.000 description 2
- 210000002784 stomach Anatomy 0.000 description 2
- 230000009469 supplementation Effects 0.000 description 2
- 101150014006 thrA gene Proteins 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 210000002438 upper gastrointestinal tract Anatomy 0.000 description 2
- 230000002485 urinary effect Effects 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 239000011782 vitamin Substances 0.000 description 2
- 235000013343 vitamin Nutrition 0.000 description 2
- 229940088594 vitamin Drugs 0.000 description 2
- 229930003231 vitamin Natural products 0.000 description 2
- 208000016261 weight loss Diseases 0.000 description 2
- 230000004580 weight loss Effects 0.000 description 2
- YAPRWCFMWHUXRS-UHFFFAOYSA-N (2-hydroxyphenyl) propanoate Chemical group CCC(=O)OC1=CC=CC=C1O YAPRWCFMWHUXRS-UHFFFAOYSA-N 0.000 description 1
- KIKDKMOYZRBMLH-MLXZFANPSA-N (5e,9e,14e)-11-[(2r)-2-[[(2s)-2-amino-4-carboxybutanoyl]amino]-3-(carboxymethylamino)-3-oxopropyl]sulfanyl-8,12-dihydroxyicosa-5,9,14-trienoic acid Chemical compound OC(=O)CC[C@H](N)C(=O)N[C@H](C(=O)NCC(O)=O)CSC(C(O)C/C=C/CCCCC)\C=C\C(O)C\C=C\CCCC(O)=O KIKDKMOYZRBMLH-MLXZFANPSA-N 0.000 description 1
- ZSCHDSYZOUKNFL-DOFZRALJSA-N (8z,11z,14z,17z)-1,2,3-trihydroxytricosa-8,11,14,17-tetraen-4-one Chemical compound CCCCC\C=C/C\C=C/C\C=C/C\C=C/CCCC(=O)C(O)C(O)CO ZSCHDSYZOUKNFL-DOFZRALJSA-N 0.000 description 1
- JVTAAEKCZFNVCJ-UWTATZPHSA-M (R)-lactate Chemical compound C[C@@H](O)C([O-])=O JVTAAEKCZFNVCJ-UWTATZPHSA-M 0.000 description 1
- JHFAEUICJHBVHB-UHFFFAOYSA-N 1h-indol-2-ol Chemical compound C1=CC=C2NC(O)=CC2=C1 JHFAEUICJHBVHB-UHFFFAOYSA-N 0.000 description 1
- PPEOLFJCQDBTKT-UHFFFAOYSA-N 2-[[2-(1h-indol-2-yl)acetyl]amino]acetic acid Chemical compound C1=CC=C2NC(CC(=O)NCC(=O)O)=CC2=C1 PPEOLFJCQDBTKT-UHFFFAOYSA-N 0.000 description 1
- QVWAEZJXDYOKEH-UHFFFAOYSA-N 3-(3-hydroxyphenyl)propanoic acid Chemical compound OC(=O)CCC1=CC=CC(O)=C1 QVWAEZJXDYOKEH-UHFFFAOYSA-N 0.000 description 1
- 108010055682 3-hydroxybutyryl-CoA dehydrogenase Proteins 0.000 description 1
- QHKABHOOEWYVLI-UHFFFAOYSA-N 3-methyl-2-oxobutanoic acid Chemical compound CC(C)C(=O)C(O)=O QHKABHOOEWYVLI-UHFFFAOYSA-N 0.000 description 1
- MGWGWNFMUOTEHG-UHFFFAOYSA-N 4-(3,5-dimethylphenyl)-1,3-thiazol-2-amine Chemical compound CC1=CC(C)=CC(C=2N=C(N)SC=2)=C1 MGWGWNFMUOTEHG-UHFFFAOYSA-N 0.000 description 1
- 229940090248 4-hydroxybenzoic acid Drugs 0.000 description 1
- 208000004998 Abdominal Pain Diseases 0.000 description 1
- 108010006229 Acetyl-CoA C-acetyltransferase Proteins 0.000 description 1
- 102000005345 Acetyl-CoA C-acetyltransferase Human genes 0.000 description 1
- 101710120269 Acyl-CoA thioester hydrolase YbgC Proteins 0.000 description 1
- 101710092462 Alpha-hemolysin Proteins 0.000 description 1
- 241001233887 Ania Species 0.000 description 1
- 101710109637 Antigen 43 Proteins 0.000 description 1
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 1
- 206010003805 Autism Diseases 0.000 description 1
- 208000020706 Autistic disease Diseases 0.000 description 1
- 206010071576 Autoimmune aplastic anaemia Diseases 0.000 description 1
- 241000193738 Bacillus anthracis Species 0.000 description 1
- 101100402524 Bacillus subtilis (strain 168) mrgA gene Proteins 0.000 description 1
- 101100197953 Bacillus subtilis (strain 168) rlbA gene Proteins 0.000 description 1
- 108091032955 Bacterial small RNA Proteins 0.000 description 1
- 241001453380 Burkholderia Species 0.000 description 1
- 108700024126 Butyrate kinases Proteins 0.000 description 1
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 1
- 101150045267 CEA gene Proteins 0.000 description 1
- 241000589876 Campylobacter Species 0.000 description 1
- 241000589875 Campylobacter jejuni Species 0.000 description 1
- 241000606161 Chlamydia Species 0.000 description 1
- 206010008631 Cholera Diseases 0.000 description 1
- 241001112696 Clostridia Species 0.000 description 1
- 241001112695 Clostridiales Species 0.000 description 1
- 241000193470 Clostridium sporogenes Species 0.000 description 1
- 241000193452 Clostridium tyrobutyricum Species 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 206010056979 Colitis microscopic Diseases 0.000 description 1
- 101710154303 Cyclic AMP receptor protein Proteins 0.000 description 1
- AUNGANRZJHBGPY-UHFFFAOYSA-N D-Lyxoflavin Natural products OCC(O)C(O)C(O)CN1C=2C=C(C)C(C)=CC=2N=C2C1=NC(=O)NC2=O AUNGANRZJHBGPY-UHFFFAOYSA-N 0.000 description 1
- 238000007702 DNA assembly Methods 0.000 description 1
- 102000052510 DNA-Binding Proteins Human genes 0.000 description 1
- 101710096438 DNA-binding protein Proteins 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- 206010012741 Diarrhoea haemorrhagic Diseases 0.000 description 1
- 102000016680 Dioxygenases Human genes 0.000 description 1
- 108010028143 Dioxygenases Proteins 0.000 description 1
- 102000003779 Dipeptidyl-peptidases and tripeptidyl-peptidases Human genes 0.000 description 1
- 108090000194 Dipeptidyl-peptidases and tripeptidyl-peptidases Proteins 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 101710105586 Electron transfer flavoprotein subunit alpha Proteins 0.000 description 1
- 102100030695 Electron transfer flavoprotein subunit alpha, mitochondrial Human genes 0.000 description 1
- 102100027262 Electron transfer flavoprotein subunit beta Human genes 0.000 description 1
- 101710186952 Electron transfer flavoprotein subunit beta Proteins 0.000 description 1
- 108010079426 Electron-Transferring Flavoproteins Proteins 0.000 description 1
- 102000012737 Electron-Transferring Flavoproteins Human genes 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 108010023922 Enoyl-CoA hydratase Proteins 0.000 description 1
- 102000011426 Enoyl-CoA hydratase Human genes 0.000 description 1
- 208000004232 Enteritis Diseases 0.000 description 1
- 241000701832 Enterobacteria phage T3 Species 0.000 description 1
- 241000588921 Enterobacteriaceae Species 0.000 description 1
- 101000658547 Escherichia coli (strain K12) Type I restriction enzyme EcoKI endonuclease subunit Proteins 0.000 description 1
- 101100390720 Escherichia coli (strain K12) fhuF gene Proteins 0.000 description 1
- 101100126128 Escherichia coli (strain K12) insE6 gene Proteins 0.000 description 1
- 241001013691 Escherichia coli BW25113 Species 0.000 description 1
- 241001302654 Escherichia coli Nissle 1917 Species 0.000 description 1
- 101000658543 Escherichia coli Type I restriction enzyme EcoAI endonuclease subunit Proteins 0.000 description 1
- 101000658546 Escherichia coli Type I restriction enzyme EcoEI endonuclease subunit Proteins 0.000 description 1
- 101000658530 Escherichia coli Type I restriction enzyme EcoR124II endonuclease subunit Proteins 0.000 description 1
- 101000658540 Escherichia coli Type I restriction enzyme EcoprrI endonuclease subunit Proteins 0.000 description 1
- 101100383218 Escherichia coli colE7 gene Proteins 0.000 description 1
- 108010014531 FMN Reductase Proteins 0.000 description 1
- 241000605980 Faecalibacterium prausnitzii Species 0.000 description 1
- VTLYFUHAOXGGBS-UHFFFAOYSA-N Fe3+ Chemical compound [Fe+3] VTLYFUHAOXGGBS-UHFFFAOYSA-N 0.000 description 1
- 102000003983 Flavoproteins Human genes 0.000 description 1
- 108010057573 Flavoproteins Proteins 0.000 description 1
- BDAGIHXWWSANSR-UHFFFAOYSA-M Formate Chemical compound [O-]C=O BDAGIHXWWSANSR-UHFFFAOYSA-M 0.000 description 1
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 1
- 102000018899 Glutamate Receptors Human genes 0.000 description 1
- 108010027915 Glutamate Receptors Proteins 0.000 description 1
- 108010070675 Glutathione transferase Proteins 0.000 description 1
- 102000005720 Glutathione transferase Human genes 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- 101000658545 Haemophilus influenzae (strain ATCC 51907 / DSM 11121 / KW20 / Rd) Type I restriction enyme HindI endonuclease subunit Proteins 0.000 description 1
- 102100039894 Hemoglobin subunit delta Human genes 0.000 description 1
- 108010006464 Hemolysin Proteins Proteins 0.000 description 1
- 108010072039 Histidine kinase Proteins 0.000 description 1
- 101500028771 Homo sapiens Glucagon-like peptide 2 Proteins 0.000 description 1
- 101001002470 Homo sapiens Interferon lambda-1 Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101001033233 Homo sapiens Interleukin-10 Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101001039966 Homo sapiens Pro-glucagon Proteins 0.000 description 1
- 101000617830 Homo sapiens Sterol O-acyltransferase 1 Proteins 0.000 description 1
- 108010064711 Homoserine dehydrogenase Proteins 0.000 description 1
- 206010060378 Hyperinsulinaemia Diseases 0.000 description 1
- 101150085950 IL10 gene Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 206010062016 Immunosuppression Diseases 0.000 description 1
- 102100020990 Interferon lambda-1 Human genes 0.000 description 1
- 102100020989 Interferon lambda-2 Human genes 0.000 description 1
- 101710099622 Interferon lambda-2 Proteins 0.000 description 1
- 102100020992 Interferon lambda-3 Human genes 0.000 description 1
- 101710099621 Interferon lambda-3 Proteins 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 102100030236 Interleukin-10 receptor subunit alpha Human genes 0.000 description 1
- 101710146672 Interleukin-10 receptor subunit alpha Proteins 0.000 description 1
- 102100020788 Interleukin-10 receptor subunit beta Human genes 0.000 description 1
- 101710199214 Interleukin-10 receptor subunit beta Proteins 0.000 description 1
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 102000006541 Ionotropic Glutamate Receptors Human genes 0.000 description 1
- 108010008812 Ionotropic Glutamate Receptors Proteins 0.000 description 1
- 206010022971 Iron Deficiencies Diseases 0.000 description 1
- 108010035210 Iron-Binding Proteins Proteins 0.000 description 1
- 102000008133 Iron-Binding Proteins Human genes 0.000 description 1
- 230000004163 JAK-STAT signaling pathway Effects 0.000 description 1
- 108010068073 Kynurenine-oxoglutarate transaminase Proteins 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-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
- 102100023162 L-serine dehydratase/L-threonine deaminase Human genes 0.000 description 1
- 101710193388 L-serine dehydratase/L-threonine deaminase Proteins 0.000 description 1
- 101710125839 L-threonine dehydratase Proteins 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
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 1
- 241000589248 Legionella Species 0.000 description 1
- 208000007764 Legionnaires' Disease Diseases 0.000 description 1
- 102000001109 Leukocyte L1 Antigen Complex Human genes 0.000 description 1
- 108010069316 Leukocyte L1 Antigen Complex Proteins 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 101000658548 Methanocaldococcus jannaschii (strain ATCC 43067 / DSM 2661 / JAL-1 / JCM 10045 / NBRC 100440) Putative type I restriction enzyme MjaIXP endonuclease subunit Proteins 0.000 description 1
- 101000658542 Methanocaldococcus jannaschii (strain ATCC 43067 / DSM 2661 / JAL-1 / JCM 10045 / NBRC 100440) Putative type I restriction enzyme MjaVIIIP endonuclease subunit Proteins 0.000 description 1
- 101000658529 Methanocaldococcus jannaschii (strain ATCC 43067 / DSM 2661 / JAL-1 / JCM 10045 / NBRC 100440) Putative type I restriction enzyme MjaVIIP endonuclease subunit Proteins 0.000 description 1
- 101710163328 Methionine synthase Proteins 0.000 description 1
- 102100031551 Methionine synthase Human genes 0.000 description 1
- 102000008109 Mixed Function Oxygenases Human genes 0.000 description 1
- 108010074633 Mixed Function Oxygenases Proteins 0.000 description 1
- MSFSPUZXLOGKHJ-UHFFFAOYSA-N Muraminsaeure Natural products OC(=O)C(C)OC1C(N)C(O)OC(CO)C1O MSFSPUZXLOGKHJ-UHFFFAOYSA-N 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 101001023834 Mus musculus Neutrophil gelatinase-associated lipocalin Proteins 0.000 description 1
- 102000003896 Myeloperoxidases Human genes 0.000 description 1
- 108090000235 Myeloperoxidases Proteins 0.000 description 1
- HOKKHZGPKSLGJE-GSVOUGTGSA-N N-Methyl-D-aspartic acid Chemical compound CN[C@@H](C(O)=O)CC(O)=O HOKKHZGPKSLGJE-GSVOUGTGSA-N 0.000 description 1
- DZTHIGRZJZPRDV-LBPRGKRZSA-N N-acetyl-L-tryptophan Chemical compound C1=CC=C2C(C[C@H](NC(=O)C)C(O)=O)=CNC2=C1 DZTHIGRZJZPRDV-LBPRGKRZSA-N 0.000 description 1
- 108090000913 Nitrate Reductases Proteins 0.000 description 1
- SUHOOTKUPISOBE-UHFFFAOYSA-N O-phosphoethanolamine Chemical class NCCOP(O)(O)=O SUHOOTKUPISOBE-UHFFFAOYSA-N 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- CBENFWSGALASAD-UHFFFAOYSA-N Ozone Chemical compound [O-][O+]=O CBENFWSGALASAD-UHFFFAOYSA-N 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 101100026007 Paulinella chromatophora ndhK gene Proteins 0.000 description 1
- 108010013639 Peptidoglycan Proteins 0.000 description 1
- 108700024327 Phosphate butyryltransferases Proteins 0.000 description 1
- 206010035664 Pneumonia Diseases 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 208000002389 Pouchitis Diseases 0.000 description 1
- 241000605861 Prevotella Species 0.000 description 1
- 241000605860 Prevotella ruminicola Species 0.000 description 1
- 101710113072 Probable methionine synthase Proteins 0.000 description 1
- 206010036774 Proctitis Diseases 0.000 description 1
- 206010036783 Proctitis ulcerative Diseases 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 241000589516 Pseudomonas Species 0.000 description 1
- 241000589517 Pseudomonas aeruginosa Species 0.000 description 1
- 108700014910 Pseudomonas aeruginosa DNR Proteins 0.000 description 1
- 241000589540 Pseudomonas fluorescens Species 0.000 description 1
- 241000589755 Pseudomonas mendocina Species 0.000 description 1
- 241000589776 Pseudomonas putida Species 0.000 description 1
- 241000589615 Pseudomonas syringae Species 0.000 description 1
- 101710193192 Putative transcriptional regulator Proteins 0.000 description 1
- 239000005700 Putrescine Substances 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 108091027981 Response element Proteins 0.000 description 1
- 101100091507 Rhizobium etli (strain CFN 42 / ATCC 51251) rosR gene Proteins 0.000 description 1
- 241000605947 Roseburia Species 0.000 description 1
- 108010091732 SEC Translocation Channels Proteins 0.000 description 1
- 102000018673 SEC Translocation Channels Human genes 0.000 description 1
- 108010017324 STAT3 Transcription Factor Proteins 0.000 description 1
- 241001468001 Salmonella virus SP6 Species 0.000 description 1
- 101710170075 Serine dehydratase-like Proteins 0.000 description 1
- 102100024040 Signal transducer and activator of transcription 3 Human genes 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 239000005708 Sodium hypochlorite Substances 0.000 description 1
- 241000191967 Staphylococcus aureus Species 0.000 description 1
- 101001042773 Staphylococcus aureus (strain COL) Type I restriction enzyme SauCOLORF180P endonuclease subunit Proteins 0.000 description 1
- 101000838760 Staphylococcus aureus (strain MRSA252) Type I restriction enzyme SauMRSORF196P endonuclease subunit Proteins 0.000 description 1
- 101000838761 Staphylococcus aureus (strain MSSA476) Type I restriction enzyme SauMSSORF170P endonuclease subunit Proteins 0.000 description 1
- 101000838758 Staphylococcus aureus (strain MW2) Type I restriction enzyme SauMW2ORF169P endonuclease subunit Proteins 0.000 description 1
- 101001042566 Staphylococcus aureus (strain Mu50 / ATCC 700699) Type I restriction enzyme SauMu50ORF195P endonuclease subunit Proteins 0.000 description 1
- 101000838763 Staphylococcus aureus (strain N315) Type I restriction enzyme SauN315I endonuclease subunit Proteins 0.000 description 1
- 101000838759 Staphylococcus epidermidis (strain ATCC 35984 / RP62A) Type I restriction enzyme SepRPIP endonuclease subunit Proteins 0.000 description 1
- 101000838756 Staphylococcus saprophyticus subsp. saprophyticus (strain ATCC 15305 / DSM 20229 / NCIMB 8711 / NCTC 7292 / S-41) Type I restriction enzyme SsaAORF53P endonuclease subunit Proteins 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 102100021993 Sterol O-acyltransferase 1 Human genes 0.000 description 1
- 241000194026 Streptococcus gordonii Species 0.000 description 1
- 101000697584 Streptomyces lavendulae Streptothricin acetyltransferase Proteins 0.000 description 1
- 241001136694 Subdoligranulum Species 0.000 description 1
- 108010012901 Succinate Dehydrogenase Proteins 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- 101100329526 Synechocystis sp. (strain PCC 6803 / Kazusa) csd gene Proteins 0.000 description 1
- 201000008754 Tenosynovial giant cell tumor Diseases 0.000 description 1
- JZRWCGZRTZMZEH-UHFFFAOYSA-N Thiamine Natural products CC1=C(CCO)SC=[N+]1CC1=CN=C(C)N=C1N JZRWCGZRTZMZEH-UHFFFAOYSA-N 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 102100033451 Thyroid hormone receptor beta Human genes 0.000 description 1
- 102000009618 Transforming Growth Factors Human genes 0.000 description 1
- 108010009583 Transforming Growth Factors Proteins 0.000 description 1
- UATJOMSPNYCXIX-UHFFFAOYSA-N Trinitrobenzene Chemical compound [O-][N+](=O)C1=CC([N+]([O-])=O)=CC([N+]([O-])=O)=C1 UATJOMSPNYCXIX-UHFFFAOYSA-N 0.000 description 1
- 208000025865 Ulcer Diseases 0.000 description 1
- 229930003779 Vitamin B12 Natural products 0.000 description 1
- 229930003448 Vitamin K Natural products 0.000 description 1
- 241001522212 [Clostridium] populeti Species 0.000 description 1
- 241001656794 [Clostridium] saccharolyticum Species 0.000 description 1
- 230000003187 abdominal effect Effects 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- IPBVNPXQWQGGJP-UHFFFAOYSA-N acetic acid phenyl ester Natural products CC(=O)OC1=CC=CC=C1 IPBVNPXQWQGGJP-UHFFFAOYSA-N 0.000 description 1
- 230000000789 acetogenic effect Effects 0.000 description 1
- 108020002494 acetyltransferase Proteins 0.000 description 1
- 102000005421 acetyltransferase Human genes 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 108091006088 activator proteins Proteins 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 108010069175 acyl-CoA transferase Proteins 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 238000011374 additional therapy Methods 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000009603 aerobic growth Effects 0.000 description 1
- 230000004103 aerobic respiration Effects 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 210000002255 anal canal Anatomy 0.000 description 1
- 208000022531 anorexia Diseases 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000000845 anti-microbial effect Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 210000000612 antigen-presenting cell Anatomy 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 229940054051 antipsychotic indole derivative Drugs 0.000 description 1
- 210000000436 anus Anatomy 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 230000004596 appetite loss Effects 0.000 description 1
- 229940114079 arachidonic acid Drugs 0.000 description 1
- 235000021342 arachidonic acid Nutrition 0.000 description 1
- LGEQQWMQCRIYKG-UHFFFAOYSA-N arachidonic acid ethanolamide Natural products CCCCCC=CCC=CCC=CCC=CCCCC(=O)NCCO LGEQQWMQCRIYKG-UHFFFAOYSA-N 0.000 description 1
- 101150010487 are gene Proteins 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 229940065181 bacillus anthracis Drugs 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 210000000013 bile duct Anatomy 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 229960000074 biopharmaceutical Drugs 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 208000027503 bloody stool Diseases 0.000 description 1
- NKWPZUCBCARRDP-UHFFFAOYSA-L calcium bicarbonate Chemical compound [Ca+2].OC([O-])=O.OC([O-])=O NKWPZUCBCARRDP-UHFFFAOYSA-L 0.000 description 1
- 229910000020 calcium bicarbonate Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 239000004202 carbamide Substances 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 230000009015 carbon catabolite repression of transcription Effects 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 210000004534 cecum Anatomy 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000010001 cellular homeostasis Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 239000003638 chemical reducing agent Substances 0.000 description 1
- 108091006116 chimeric peptides Proteins 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- AGVAZMGAQJOSFJ-WZHZPDAFSA-M cobalt(2+);[(2r,3s,4r,5s)-5-(5,6-dimethylbenzimidazol-1-yl)-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] [(2r)-1-[3-[(1r,2r,3r,4z,7s,9z,12s,13s,14z,17s,18s,19r)-2,13,18-tris(2-amino-2-oxoethyl)-7,12,17-tris(3-amino-3-oxopropyl)-3,5,8,8,13,15,18,19-octamethyl-2 Chemical compound [Co+2].N#[C-].[N-]([C@@H]1[C@H](CC(N)=O)[C@@]2(C)CCC(=O)NC[C@@H](C)OP(O)(=O)O[C@H]3[C@H]([C@H](O[C@@H]3CO)N3C4=CC(C)=C(C)C=C4N=C3)O)\C2=C(C)/C([C@H](C\2(C)C)CCC(N)=O)=N/C/2=C\C([C@H]([C@@]/2(CC(N)=O)C)CCC(N)=O)=N\C\2=C(C)/C2=N[C@]1(C)[C@@](C)(CC(N)=O)[C@@H]2CCC(N)=O AGVAZMGAQJOSFJ-WZHZPDAFSA-M 0.000 description 1
- 208000008609 collagenous colitis Diseases 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 230000000112 colonic effect Effects 0.000 description 1
- 230000001447 compensatory effect Effects 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 101150000386 cpfC gene Proteins 0.000 description 1
- 229960003624 creatine Drugs 0.000 description 1
- 239000006046 creatine Substances 0.000 description 1
- 229940109239 creatinine Drugs 0.000 description 1
- 230000009260 cross reactivity Effects 0.000 description 1
- KFWWCMJSYSSPSK-PAXLJYGASA-N crotonoyl-CoA Chemical compound O[C@@H]1[C@H](OP(O)(O)=O)[C@@H](COP(O)(=O)OP(O)(=O)OCC(C)(C)[C@@H](O)C(=O)NCCC(=O)NCCSC(=O)/C=C/C)O[C@H]1N1C2=NC=NC(N)=C2N=C1 KFWWCMJSYSSPSK-PAXLJYGASA-N 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 231100000599 cytotoxic agent Toxicity 0.000 description 1
- 239000002619 cytotoxin Substances 0.000 description 1
- 230000007423 decrease Effects 0.000 description 1
- 206010061428 decreased appetite Diseases 0.000 description 1
- 230000007123 defense Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 238000001784 detoxification Methods 0.000 description 1
- 235000013325 dietary fiber Nutrition 0.000 description 1
- 208000035647 diffuse type tenosynovial giant cell tumor Diseases 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 229910001882 dioxygen Inorganic materials 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 201000008243 diversion colitis Diseases 0.000 description 1
- 239000003651 drinking water Substances 0.000 description 1
- 235000020188 drinking water Nutrition 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 101150026863 dsbG gene Proteins 0.000 description 1
- 210000001198 duodenum Anatomy 0.000 description 1
- 208000001848 dysentery Diseases 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 201000002491 encephalomyelitis Diseases 0.000 description 1
- 210000003890 endocrine cell Anatomy 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 210000005216 enteric neuron Anatomy 0.000 description 1
- 239000000147 enterotoxin Substances 0.000 description 1
- 231100000655 enterotoxin Toxicity 0.000 description 1
- 230000001973 epigenetic effect Effects 0.000 description 1
- 210000005081 epithelial layer Anatomy 0.000 description 1
- 210000000981 epithelium Anatomy 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- 230000002964 excitative effect Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 206010016256 fatigue Diseases 0.000 description 1
- 238000000855 fermentation Methods 0.000 description 1
- 230000004151 fermentation Effects 0.000 description 1
- 229910001447 ferric ion Inorganic materials 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
- 230000004907 flux Effects 0.000 description 1
- 229940014144 folate Drugs 0.000 description 1
- OVBPIULPVIDEAO-LBPRGKRZSA-N folic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 1
- 235000019152 folic acid Nutrition 0.000 description 1
- 239000011724 folic acid Substances 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 238000002825 functional assay Methods 0.000 description 1
- 210000000232 gallbladder Anatomy 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 238000003304 gavage Methods 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000012224 gene deletion Methods 0.000 description 1
- 238000012239 gene modification Methods 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 230000005017 genetic modification Effects 0.000 description 1
- 235000013617 genetically modified food Nutrition 0.000 description 1
- 231100000024 genotoxic Toxicity 0.000 description 1
- 230000001738 genotoxic effect Effects 0.000 description 1
- 230000014101 glucose homeostasis Effects 0.000 description 1
- 229930182480 glucuronide Natural products 0.000 description 1
- 101150077538 gor gene Proteins 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 101150002138 grxA gene Proteins 0.000 description 1
- 101150107068 gsiB gene Proteins 0.000 description 1
- 230000007149 gut brain axis pathway Effects 0.000 description 1
- 150000003278 haem Chemical class 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000035876 healing Effects 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 230000007407 health benefit Effects 0.000 description 1
- 101150042363 hemH gene Proteins 0.000 description 1
- 208000035861 hematochezia Diseases 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 108010071598 homoserine kinase Proteins 0.000 description 1
- 230000005745 host immune response Effects 0.000 description 1
- 101150099805 htpG gene Proteins 0.000 description 1
- 102000052620 human IL10 Human genes 0.000 description 1
- 229940048921 humira Drugs 0.000 description 1
- 150000002432 hydroperoxides Chemical class 0.000 description 1
- OUUQCZGPVNCOIJ-UHFFFAOYSA-N hydroperoxyl Chemical compound O[O] OUUQCZGPVNCOIJ-UHFFFAOYSA-N 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-M hydroxide Chemical compound [OH-] XLYOFNOQVPJJNP-UHFFFAOYSA-M 0.000 description 1
- TUJKJAMUKRIRHC-UHFFFAOYSA-N hydroxyl Chemical compound [OH] TUJKJAMUKRIRHC-UHFFFAOYSA-N 0.000 description 1
- 230000003451 hyperinsulinaemic effect Effects 0.000 description 1
- 201000008980 hyperinsulinism Diseases 0.000 description 1
- WQYVRQLZKVEZGA-UHFFFAOYSA-N hypochlorite Chemical compound Cl[O-] WQYVRQLZKVEZGA-UHFFFAOYSA-N 0.000 description 1
- 208000009326 ileitis Diseases 0.000 description 1
- 210000003405 ileum Anatomy 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 239000003018 immunosuppressive agent Substances 0.000 description 1
- 229940124589 immunosuppressive drug Drugs 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 208000027138 indeterminate colitis Diseases 0.000 description 1
- PZOUSPYUWWUPPK-UHFFFAOYSA-N indole Natural products CC1=CC=CC2=C1C=CN2 PZOUSPYUWWUPPK-UHFFFAOYSA-N 0.000 description 1
- SEOVTRFCIGRIMH-UHFFFAOYSA-N indole-3-acetic acid Chemical compound C1=CC=C2C(CC(=O)O)=CNC2=C1 SEOVTRFCIGRIMH-UHFFFAOYSA-N 0.000 description 1
- RKJUIXBNRJVNHR-UHFFFAOYSA-N indolenine Natural products C1=CC=C2CC=NC2=C1 RKJUIXBNRJVNHR-UHFFFAOYSA-N 0.000 description 1
- 150000002475 indoles Chemical class 0.000 description 1
- 230000004968 inflammatory condition Effects 0.000 description 1
- 208000027866 inflammatory disease Diseases 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 239000002054 inoculum Substances 0.000 description 1
- 229910001872 inorganic gas Inorganic materials 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 229940047124 interferons Drugs 0.000 description 1
- 230000031261 interleukin-10 production Effects 0.000 description 1
- 210000005027 intestinal barrier Anatomy 0.000 description 1
- 230000007358 intestinal barrier function Effects 0.000 description 1
- 210000002490 intestinal epithelial cell Anatomy 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- BKWBIMSGEOYWCJ-UHFFFAOYSA-L iron;iron(2+);sulfanide Chemical compound [SH-].[SH-].[Fe].[Fe+2] BKWBIMSGEOYWCJ-UHFFFAOYSA-L 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 238000006317 isomerization reaction Methods 0.000 description 1
- 210000001630 jejunum Anatomy 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- VNYSSYRCGWBHLG-AMOLWHMGSA-N leukotriene B4 Chemical compound CCCCC\C=C/C[C@@H](O)\C=C\C=C\C=C/[C@@H](O)CCCC(O)=O VNYSSYRCGWBHLG-AMOLWHMGSA-N 0.000 description 1
- 150000002617 leukotrienes Chemical class 0.000 description 1
- 229920005610 lignin Polymers 0.000 description 1
- 108010052322 limitin Proteins 0.000 description 1
- 230000003859 lipid peroxidation Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 238000012153 long-term therapy Methods 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 208000004341 lymphocytic colitis Diseases 0.000 description 1
- 210000003563 lymphoid tissue Anatomy 0.000 description 1
- 101150076563 malP gene Proteins 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- NUJOXMJBOLGQSY-UHFFFAOYSA-N manganese dioxide Chemical compound O=[Mn]=O NUJOXMJBOLGQSY-UHFFFAOYSA-N 0.000 description 1
- 238000013178 mathematical model Methods 0.000 description 1
- 230000007102 metabolic function Effects 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 239000013264 metal-organic assembly Substances 0.000 description 1
- 101150055934 mgrA gene Proteins 0.000 description 1
- 244000000010 microbial pathogen Species 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 208000018962 mouth sore Diseases 0.000 description 1
- 230000003843 mucus production Effects 0.000 description 1
- 210000003249 myenteric plexus Anatomy 0.000 description 1
- 230000012666 negative regulation of transcription by glucose Effects 0.000 description 1
- 210000005036 nerve Anatomy 0.000 description 1
- 230000001537 neural effect Effects 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 239000002858 neurotransmitter agent Substances 0.000 description 1
- JCXJVPUVTGWSNB-UHFFFAOYSA-N nitrogen dioxide Inorganic materials O=[N]=O JCXJVPUVTGWSNB-UHFFFAOYSA-N 0.000 description 1
- 230000008599 nitrosative stress Effects 0.000 description 1
- 239000001272 nitrous oxide Substances 0.000 description 1
- 229960001730 nitrous oxide Drugs 0.000 description 1
- 102000042567 non-coding RNA Human genes 0.000 description 1
- 101150002988 norR gene Proteins 0.000 description 1
- 101150103941 nuoBCD gene Proteins 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 206010073131 oligoastrocytoma Diseases 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 101150008884 osmY gene Proteins 0.000 description 1
- 230000004792 oxidative damage Effects 0.000 description 1
- 229940094443 oxytocics prostaglandins Drugs 0.000 description 1
- WGNAKZGUSRVWRH-UHFFFAOYSA-N p-cresol sulfate Chemical compound CC1=CC=C(OS(O)(=O)=O)C=C1 WGNAKZGUSRVWRH-UHFFFAOYSA-N 0.000 description 1
- 208000014965 pancolitis Diseases 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- 230000009984 peri-natal effect Effects 0.000 description 1
- 210000001428 peripheral nervous system Anatomy 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 229940049953 phenylacetate Drugs 0.000 description 1
- WLJVXDMOQOGPHL-UHFFFAOYSA-N phenylacetic acid Chemical compound OC(=O)CC1=CC=CC=C1 WLJVXDMOQOGPHL-UHFFFAOYSA-N 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 150000008105 phosphatidylcholines Chemical class 0.000 description 1
- SHUZOJHMOBOZST-UHFFFAOYSA-N phylloquinone Natural products CC(C)CCCCC(C)CCC(C)CCCC(=CCC1=C(C)C(=O)c2ccccc2C1=O)C SHUZOJHMOBOZST-UHFFFAOYSA-N 0.000 description 1
- 230000035479 physiological effects, processes and functions Effects 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 229920000768 polyamine Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 229940068977 polysorbate 20 Drugs 0.000 description 1
- 230000032361 posttranscriptional gene silencing Effects 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 229960004618 prednisone Drugs 0.000 description 1
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 238000011165 process development Methods 0.000 description 1
- 206010036784 proctocolitis Diseases 0.000 description 1
- 208000017048 proctosigmoiditis Diseases 0.000 description 1
- KAQKFAOMNZTLHT-OZUDYXHBSA-N prostaglandin I2 Chemical compound O1\C(=C/CCCC(O)=O)C[C@@H]2[C@@H](/C=C/[C@@H](O)CCCCC)[C@H](O)C[C@@H]21 KAQKFAOMNZTLHT-OZUDYXHBSA-N 0.000 description 1
- 150000003180 prostaglandins Chemical class 0.000 description 1
- 230000009993 protective function Effects 0.000 description 1
- 230000004850 protein–protein interaction Effects 0.000 description 1
- 230000008141 pubertal development Effects 0.000 description 1
- 125000002181 pyridoxine group Chemical group 0.000 description 1
- 238000012372 quality testing Methods 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000011506 response to oxidative stress Effects 0.000 description 1
- 230000003938 response to stress Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 235000019192 riboflavin Nutrition 0.000 description 1
- 229960002477 riboflavin Drugs 0.000 description 1
- 239000002151 riboflavin Substances 0.000 description 1
- 230000009962 secretion pathway Effects 0.000 description 1
- 230000009758 senescence Effects 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 230000001953 sensory effect Effects 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 235000021391 short chain fatty acids Nutrition 0.000 description 1
- 239000002356 single layer Substances 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- SUKJFIGYRHOWBL-UHFFFAOYSA-N sodium hypochlorite Chemical compound [Na+].Cl[O-] SUKJFIGYRHOWBL-UHFFFAOYSA-N 0.000 description 1
- 101150100021 soxR gene Proteins 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 229940063673 spermidine Drugs 0.000 description 1
- 229940063675 spermine Drugs 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 229940032147 starch Drugs 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 230000035882 stress Effects 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-L succinate(2-) Chemical compound [O-]C(=O)CCC([O-])=O KDYFGRWQOYBRFD-UHFFFAOYSA-L 0.000 description 1
- 101150118796 sufS gene Proteins 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000001502 supplementing effect Effects 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- CILIXQOJUNDIDU-ASQIGDHWSA-N teduglutide Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(O)=O)C(O)=O)[C@@H](C)CC)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@@H](NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCSC)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CC=1NC=NC=1)[C@@H](C)O)[C@@H](C)CC)C1=CC=CC=C1 CILIXQOJUNDIDU-ASQIGDHWSA-N 0.000 description 1
- 108010073046 teduglutide Proteins 0.000 description 1
- 229960002444 teduglutide Drugs 0.000 description 1
- 208000002918 testicular germ cell tumor Diseases 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 235000019157 thiamine Nutrition 0.000 description 1
- KYMBYSLLVAOCFI-UHFFFAOYSA-N thiamine Chemical compound CC1=C(CCO)SCN1CC1=CN=C(C)N=C1N KYMBYSLLVAOCFI-UHFFFAOYSA-N 0.000 description 1
- 229960003495 thiamine Drugs 0.000 description 1
- 239000011721 thiamine Substances 0.000 description 1
- 229960002898 threonine Drugs 0.000 description 1
- DSNBHJFQCNUKMA-SCKDECHMSA-N thromboxane A2 Chemical compound OC(=O)CCC\C=C/C[C@@H]1[C@@H](/C=C/[C@@H](O)CCCCC)O[C@@H]2O[C@H]1C2 DSNBHJFQCNUKMA-SCKDECHMSA-N 0.000 description 1
- 150000003595 thromboxanes Chemical class 0.000 description 1
- 210000001541 thymus gland Anatomy 0.000 description 1
- 230000017423 tissue regeneration Effects 0.000 description 1
- 101150037435 tnaB gene Proteins 0.000 description 1
- NGSWKAQJJWESNS-ZZXKWVIFSA-N trans-4-coumaric acid Chemical compound OC(=O)\C=C\C1=CC=C(O)C=C1 NGSWKAQJJWESNS-ZZXKWVIFSA-N 0.000 description 1
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 1
- 238000005891 transamination reaction Methods 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 101150118060 trxA gene Proteins 0.000 description 1
- 101150112435 trxC gene Proteins 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 230000036269 ulceration Effects 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 230000001018 virulence Effects 0.000 description 1
- 239000000304 virulence factor Substances 0.000 description 1
- 230000007923 virulence factor Effects 0.000 description 1
- 235000019163 vitamin B12 Nutrition 0.000 description 1
- 239000011715 vitamin B12 Substances 0.000 description 1
- 235000019168 vitamin K Nutrition 0.000 description 1
- 239000011712 vitamin K Substances 0.000 description 1
- 150000003721 vitamin K derivatives Chemical class 0.000 description 1
- 229940046010 vitamin k Drugs 0.000 description 1
- 239000002699 waste material Substances 0.000 description 1
- 101150024223 yaaA gene Proteins 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/70—Vectors or expression systems specially adapted for E. coli
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/185—Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
- A61K31/19—Carboxylic acids, e.g. valproic acid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/185—Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
- A61K31/19—Carboxylic acids, e.g. valproic acid
- A61K31/195—Carboxylic acids, e.g. valproic acid having an amino group
- A61K31/197—Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
- A61K31/198—Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/74—Bacteria
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/74—Bacteria
- A61K35/741—Probiotics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/2013—IL-2
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/2066—IL-10
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/22—Hormones
- A61K38/26—Glucagons
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/44—Oxidoreductases (1)
- A61K38/446—Superoxide dismutase (1.15)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/04—Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/14—Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/18—Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
- A61P11/06—Antiasthmatics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/02—Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/10—Drugs for disorders of the urinary system of the bladder
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/12—Drugs for disorders of the urinary system of the kidneys
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
- A61P15/08—Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
- A61P17/06—Antipsoriatics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
- A61P17/14—Drugs for dermatological disorders for baldness or alopecia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/08—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
- A61P21/04—Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/02—Drugs for disorders of the nervous system for peripheral neuropathies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/20—Hypnotics; Sedatives
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/02—Ophthalmic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/16—Otologicals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/02—Nutrients, e.g. vitamins, minerals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/06—Antihyperlipidemics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
- A61P33/02—Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/06—Immunosuppressants, e.g. drugs for graft rejection
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/14—Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/38—Drugs for disorders of the endocrine system of the suprarenal hormones
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/04—Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/06—Antianaemics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/10—Antioedematous agents; Diuretics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/08—Vasodilators for multiple indications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/74—Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/0004—Oxidoreductases (1.)
- C12N9/001—Oxidoreductases (1.) acting on the CH-CH group of donors (1.3)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/10—Transferases (2.)
- C12N9/12—Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
- C12N9/1217—Phosphotransferases with a carboxyl group as acceptor (2.7.2)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y103/00—Oxidoreductases acting on the CH-CH group of donors (1.3)
- C12Y103/08—Oxidoreductases acting on the CH-CH group of donors (1.3) with flavin as acceptor (1.3.8)
- C12Y103/08001—Butyryl-CoA dehydrogenase (1.3.8.1), i.e. short chain acyl-CoA dehydrogenase
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y115/00—Oxidoreductases acting on superoxide as acceptor (1.15)
- C12Y115/01—Oxidoreductases acting on superoxide as acceptor (1.15) with NAD or NADP as acceptor (1.15.1)
- C12Y115/01001—Superoxide dismutase (1.15.1.1)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y207/00—Transferases transferring phosphorus-containing groups (2.7)
- C12Y207/02—Phosphotransferases with a carboxy group as acceptor (2.7.2)
- C12Y207/02007—Butyrate kinase (2.7.2.7)
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2207/00—Modified animals
- A01K2207/20—Animals treated with compounds which are neither proteins nor nucleic acids
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2227/00—Animals characterised by species
- A01K2227/10—Mammal
- A01K2227/105—Murine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K2035/11—Medicinal preparations comprising living procariotic cells
- A61K2035/115—Probiotics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- compositions and therapeutic methods for inhibiting inflammatory mechanisms in the gut, restoring and tightening gut mucosal barrier function, and/or treating and preventing autoimmune disorders.
- the disclosure relates to genetically engineered bacteria that are capable of reducing inflammation in the gut and/or enhancing gut barrier function.
- the genetically engineered bacteria are capable of reducing gut inflammation and/or enhancing gut barrier function, thereby ameliorating or preventing an autoimmune disorder.
- the compositions and methods disclosed herein may be used for treating or preventing autoimmune disorders as well as diseases and conditions associated with gut inflammation and/or compromised gut barrier function, e.g., diarrheal diseases, inflammatory bowel diseases, and related diseases.
- IBDs Inflammatory bowel diseases
- TNF tumor necrosis factor
- Compromised gut barrier function also plays a central role in autoimmune diseases pathogenesis (Lerner et al., 2015a; Lerner et al., 2015b; Fasano et al., 2005; Fasano, 2012).
- a single layer of epithelial cells separates the gut lumen from the immune cells in the body.
- the epithelium is regulated by intercellular tight junctions and controls the equilibrium between tolerance and immunity to nonself-antigens (Fasano et al., 2005).
- Disrupting the epithelial layer can lead to pathological exposure of the highly immunoreactive subepithelium to the vast number of foreign antigens in the lumen (Lerner et al., 2015a) resulting in increased susceptibility to and both intestinal and extraintestinal autoimmune disorders can occur" (Fasano et al., 2005).
- Some foreign antigens are postulated to resemble self-antigens and can induce epitope-specific cross- reactivity that accelerates the progression of a pre-existing autoimmune disease or initiates an autoimmune disease (Fasano, 2012).
- Rheumatoid arthritis and celiac disease are autoimmune disorders that are thought to involve increased intestinal permeability (Lerner et al., 2015b).
- dysregulation of intercellular tight junctions can lead to disease onset (Fasano, 2012).
- the loss of protective function of mucosal barriers that interact with the environment is necessary for autoimmunity to develop (Lerner et al., 2015a).
- the genetically engineered bacteria disclosed herein are capable of producing therapeutic anti-inflammation and/or gut barrier enhancer molecules.
- the genetically engineered bacteria are functionally silent until they reach an inducing environment, e.g., a mammalian gut, wherein expression of the therapeutic molecule is induced.
- the genetically engineered bacteria are naturally nonpathogenic and may be introduced into the gut in order to reduce gut inflammation and/or enhance gut barrier function and may thereby further ameliorate or prevent an autoimmune disorder.
- the anti-inflammation and/or gut barrier enhancer molecule is stably produced by the genetically engineered bacteria, and/or the genetically engineered bacteria are stably maintained in vivo and/or in vitro.
- the invention also provides pharmaceutical compositions comprising the genetically engineered bacteria, and methods of treating diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier function, e.g., an inflammatory bowel disease or an autoimmune disorder.
- the genetically engineered bacteria of the invention produce one or more therapeutic molecule(s) under the control of one or more promoters induced by an environmental condition, e.g., an environmental condition found in the mammalian gut, such as an inflammatory condition or a low oxygen condition.
- an environmental condition e.g., an environmental condition found in the mammalian gut, such as an inflammatory condition or a low oxygen condition.
- the genetically engineered bacteria of the invention produce one or more therapeutic molecule(s) under the control of an oxygen level-dependent promoter, a reactive oxygen species (ROS)-dependent promoter, or a reactive nitrogen species (RNS)-dependent promoter, and a corresponding transcription factor.
- ROS reactive oxygen species
- RNS reactive nitrogen species
- the therapeutic molecule is butyrate; in an inducing environment, the butyrate biosynthetic gene cassette is activated, and butyrate is produced.
- Local production of butyrate induces the differentiation of regulatory T cells in the gut and/or promotes the barrier function of colonic epithelial cells.
- the genetically engineered bacteria of the invention produce their therapeutic effect only in inducing environments such as the gut, thereby lowering the safety issues associated with systemic exposure.
- Fig. 1 depicts a schematic of the eight-gene pathway from C. difficile for butyrate production.
- pLogic031 comprises the eight-gene pathway from C. difficile, bcd2- etfB3-etfA3-thiAl-hbd-crt2-pbt-buk, synthesized under the control of Tet-inducible promoters (pBR322 backbone).
- pLogic046 replaces the BCD/EFT complex, a potential rate-limiting step, with single gene from Treponema denticola, ter (frans-enoyl-2- reductase), and comprises ter-thiAl-hbd-crt2-pbt-buk.
- FIG. 2 depicts a schematic of a butyrate production pathway in which the circled genes (buk and pbt) may be deleted and replaced with tesB, which cleaves the CoA from butyryl-CoA.
- Fig. 3 depicts the gene organization of an exemplary recombinant bacterium of the invention and its derepression in the presence of nitric oxide (NO).
- NO nitric oxide
- the NsrR transcription factor (gray circle, "NsrR”) binds to and represses a corresponding regulatory region. Therefore, none of the butyrate biosynthesis enzymes (bcd2, etfB3, etfA3, thiAl, hbd, crt2, pbt, buk; black boxes) is expressed.
- the NsrR transcription factor interacts with NO, and no longer binds to or represses the regulatory sequence. This leads to expression of the butyrate biosynthesis enzymes (indicated by gray arrows and black squiggles) and ultimately to the production of butyrate.
- Fig. 4 depicts the gene organization of another exemplary recombinant bacterium of the invention and its derepression in the presence of NO.
- the NsrR transcription factor (gray circle, "NsrR”) binds to and represses a corresponding regulatory region. Therefore, none of the butyrate biosynthesis enzymes (ter, thiAl, hbd, crt2, pbt, buk; black boxes) is expressed.
- the NsrR transcription factor interacts with NO, and no longer binds to or represses the regulatory sequence. This leads to expression of the butyrate biosynthesis enzymes (indicated by gray arrows and black squiggles) and ultimately to the production of butyrate.
- FIG. 5 depicts the gene organization of an exemplary recombinant bacterium of the invention and its induction in the presence of H 2 0 2 .
- the OxyR transcription factor (gray circle, "OxyR") binds to, but does not induce, the oxyS promoter. Therefore, none of the butyrate biosynthesis enzymes (bcd2, etfB3, etfA3, thiAl, hbd, crt2, pbt, buk; black boxes) is expressed.
- the OxyR transcription factor interacts with H 2 0 2 and is then capable of inducing the oxyS promoter. This leads to expression of the butyrate biosynthesis enzymes (indicated by gray arrows and black squiggles) and ultimately to the production of butyrate.
- Fig. 6 depicts the gene organization of another exemplary recombinant bacterium of the invention and its induction in the presence of H 2 0 2 .
- the OxyR transcription factor (gray circle, "OxyR") binds to, but does not induce, the oxyS promoter. Therefore, none of the butyrate biosynthesis enzymes (ter, thiAl, hbd, crt2, pbt, buk; black boxes) is expressed.
- the OxyR transcription factor interacts with H 2 0 2 and is then capable of inducing the oxyS promoter. This leads to expression of the butyrate biosynthesis enzymes (indicated by gray arrows and black squiggles) and ultimately to the production of butyrate.
- FIG. 7 depicts the gene organization of an exemplary recombinant bacterium of the invention and its induction under low-oxygen conditions.
- FIG. 8 depicts the gene organization of an exemplary recombinant bacterium of the invention and its induction under low-oxygen conditions.
- relatively low butyrate production under aerobic conditions in which oxygen (0 2 ) prevents indicated by "X"
- FNR grey boxed "FNR”
- FNR promoter FNR promoter
- butyrate biosynthesis enzymes ter, thiAl, hbd, crt2, pbt, and buk; black boxes.
- FNR dimerizing two grey boxed "FNR"s
- binding to the FNR-responsive promoter binding to the FNR-responsive promoter, and inducing expression of the butyrate biosynthesis enzymes, which leads to the production of butyrate.
- FIG. 9 depicts the gene organization of an exemplary recombinant bacterium of the invention and its induction under low-oxygen conditions.
- Fig. 10 depicts an exemplary propionate biosynthesis gene cassette.
- FIG. 11 depicts the gene organization of an exemplary recombinant bacterium of the invention and its induction under low-oxygen conditions.
- Fig. 12 depicts an exemplary propionate biosynthesis gene cassette.
- Fig. 13 depicts the gene organization of an exemplary recombinant bacterium of the invention and its induction under low-oxygen conditions.
- Fig. 14 depicts an exemplary propionate biosynthesis gene cassette.
- FIG. 15 depicts a schematic of a butyrate gene cassette, pLogic031 comprising the eight-gene butyrate cassette.
- FIG. 16 depicts a schematic of a butyrate gene cassette, pLogic046 comprising the ter substitution (oval).
- FIG. 17 depicts a linear schematic of a butyrate gene cassette, pLogic046.
- Fig. 18 depicts a graph of butyrate production.
- pLOGIC031 (bcd)/+02 is Nissle containing plasmid pLOGIC031 grown aerobically.
- pLOGIC046 (ter)/+02 is Nissle containing plasmid pLOGIC046 grown aerobically.
- pLOGIC031 (bcd)/-02 is Nissle containing plasmid pLOGIC031 grown anaerobically.
- pLOGIC046 (ter)/-02 is Nissle containing plasmid pLOGIC046 grown anaerobically. The ter construct results in higher butyrate production.
- Fig. 19 depicts a graph of butyrate production using E. coli BW25113 butyrate-producing circuits comprising a nuoB gene deletion, which results in greater levels of butyrate production as compared to a wild-type parent control.
- nuoB is a main protein complex involved in the oxidation of NADH during respiratory growth.
- preventing the coupling of NADH oxidation to electron transport increases the amount of NADH being used to support butyrate production.
- Fig. 20 depicts a schematic of pLogic046-tesB, in which buk and pbt are deleted and tesB substituted.
- Fig. 21 depicts a linear schematic of a butyrate gene cassette, pLogic046- delta.ptb-buk-tesB+.
- Fig. 22 depicts butyrate production using pLOGIC046 (a Nissle strain comprising plasmid pLOGIC046, an ATC-inducible ter-comprising butyrate construct) and pLOGIC046-delta.pbt-buk/tesB+ (a Nissle strain comprising plasmid pLOGIC046-delta pbt.buk/tesB+, an ATC-inducible ter-comprising butyrate construct with a deletion in the pbt-buk genes and their replacement with the tesB gene).
- the tesB construct results in greater butyrate production.
- Fig. 23 depicts a schematic of a butyrate gene cassette, ydfZ-butyrate, comprising the ter substitution.
- Fig. 24 depicts SYN363 in the presence and absence of glucose and oxygen in vitro.
- SYN363 comprises a butyrate gene cassette comprising the ter-thiAl-hbd-crt2- tesB genes under the control of a ydfZ promoter.
- Fig. 25 depicts a graph measuring gut-barrier function in dextran sodium sulfate (DSS)-induced mouse models of IBD.
- DSS dextran sodium sulfate
- Fig. 26 depicts serum levels of FITC-dextran analyzed by
- FITC-dextran is a readout for gut barrier function in the DSS-induced mouse model of IBD.
- Fig. 27 depicts levels of mouse lipocalin 2 and calprotectin quantified by ELISA using the fecal samples in an in vivo model of IBD.
- SYN363 reduces inflammation and/or protects gut barrier function as compared to control SYN94.
- Fig. 28 depicts ATC or nitric oxide-inducible reporter constructs. These constructs, when induced by their cognate inducer, lead to expression of GFP. Nissle cells harboring plasmids with either the control, ATC-inducible P tet -GFP reporter construct or the nitric oxide inducible P nsr R-GFP reporter construct induced across a range of concentrations. Promoter activity is expressed as relative florescence units.
- Fig. 29 depicts a dot blot of bacteria harboring a plasmid expressing NsrR under control of a constitutive promoter and the reporter gene gfp (green fluorescent protein) under control of an NsrR-inducible promoter.
- IBD is induced in mice by supplementing drinking water with 2-3% dextran sodium sulfate (DSS).
- Chemiluminescent is shown for NsrR-regulated promoters induced in DSS-treated mice.
- Fig. 30 depicts the construction and gene organization of an exemplary plasmid comprising a gene encoding NsrR, a regulatory sequence from norB, and a butyrogenic gene cassette (pLogic031-nsrR-norB-butyrate construct).
- Fig. 31 depicts the construction and gene organization of another exemplary plasmid comprising a gene encoding NsrR, a regulatory sequence from norB, and a butyrogenic gene cassette (pLogic046- nsrR-norB-butyrogenic gene cassette).
- Fig. 32 depicts butyrate production using SYN001 + tet (control wild-type Nissle comprising no plasmid), SYN067 + tet (Nissle comprising the pLOGIC031 ATC- inducible butyrate plasmid), and SYN080 + tet (Nissle comprising the pLOGIC046 ATC- inducible butyrate plasmid).
- Fig. 33 depicts butyrate production by genetically engineered Nissle comprising the pLogic031-nsrR-norB-butyrate construct (SYN133) or the pLogic046-nsrR- norB-butyrate construct (SYN145), which produce more butyrate as compared to wild- type Nissle (SYN001).
- Fig. 34 depicts the construction and gene organization of an exemplary plasmid comprising an oxyS promoter and butyrogenic gene cassette (pLogic031-oxyS- butyrogenic gene cassette).
- Fig. 35 depicts the construction and gene organization of another exemplary plasmid comprising an oxyS promoter and butyrogenic gene cassette
- Fig. 36 depicts a schematic of an E. coli that is genetically engineered to express the essential gene tnaB, 5-methyltetrahydrofolate-homocysteine methyltransferase (mtr), tryptophan transporter, and the enzymes I DO and TDO to convert tryptophan into kynurenine.
- tnaB 5-methyltetrahydrofolate-homocysteine methyltransferase
- tryptophan transporter the enzymes I DO and TDO to convert tryptophan into kynurenine.
- Fig. 37 depicts a schematic of an E. coli that is genetically engineered to express interleukin under the control of a FN R-responsive promoter and further comprising a TAT secretion system.
- Fig. 38 depicts a schematic of an E. coli that is genetically engineered to express SOD under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 39 depicts a schematic of an E. coli that is genetically engineered to express GLP-2 under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 40 depicts a schematic of an E. coli that is genetically engineered to express a propionate gene cassette under the control of a FN R-responsive promoter.
- Fig. 41 depicts a schematic of an E. coli that is genetically engineered to express butyrate under the control of a FNR-responsive promoter.
- Fig. 42 depicts a schematic of an E. coli that is genetically engineered to express kynurenine, interleukin, SOD, GLP-2, a propionate gene cassette, and a butyrate gene cassette under the control of a FN R-responsive promoter and further comprising a TAT secretion system.
- Fig. 43 depicts a schematic of an E. coli that is genetically engineered to express interleukin, OSD, GLP-2, a propionate gene cassette, and a butyrate gene cassette under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 44 depicts a schematic of an E. coli that is genetically engineered to express SOD, a propionate gene cassette, and a butyrate gene cassette under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 45 depicts a schematic of an E. coli that is genetically engineered to express interleukin, a propionate gene cassette, and a butyrate gene cassette under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 46 depicts a schematic of an E. coli that is genetically engineered to express interleukin-10 (IL-10), a propionate gene cassette, and a butyrate gene cassette under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- IL-10 interleukin-10
- Fig. 47 depicts a schematic of an E. coli that is genetically engineered to express IL-2, IL-10, a propionate gene cassette, and a butyrate gene cassette under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 48 depicts a schematic of an E. coli that is genetically engineered to express IL-2, IL-10, a propionate gene cassette, a butyrate gene cassette, and SOD under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 49 depicts a schematic of an E. coli that is genetically engineered to express IL-2, IL-10, a propionate gene cassette, a butyrate gene cassette, SOD, and GLP-2 under the control of a FNR-responsive promoter and further comprising a TAT secretion system.
- Fig. 50 depicts a map of exemplary integration sites within the E. coli 1917 Nissle chromosome. These sites indicate regions where circuit components may be inserted into the chromosome without interfering with essential gene expression.
- Backslashes are used to show that the insertion will occur between divergently or convergently expressed genes. I nsertions within biosynthetic genes, such as thyA, can be useful for creating nutrient auxotrophies. In some embodiments, an individual circuit component is inserted into more than one of the indicated sites.
- Fig. 51 depicts an exemplary schematic of the E. coli 1917 Nissle chromosome comprising multiple mechanisms of action (MoAs).
- Fig. 52 depicts an exemplary schematic of the E. coli 1917 Nissle chromosome comprising multiple mechanisms of action for producing I L-2, I L-10, IL-22, I L-27, propionate, and butyrate.
- Fig. 53 depicts an exemplary schematic of the E. coli 1917 Nissle chromosome comprising multiple mechanisms of action for producing I L-10, I L-27, GLP-2, and butyrate.
- Fig. 54 depicts an exemplary schematic of the E. coli 1917 Nissle chromosome comprising multiple mechanisms of action for producing GLP-2, IL-10, I L-22, SOD, butyrate, and propionate.
- Fig. 55 depicts an exemplary schematic of the E. coli 1917 Nissle chromosome comprising multiple mechanisms of action for producing GLP-2, IL-2, IL-10, I L-22, I L-27, SOD, butyrate, and propionate.
- Fig. 56 depicts a table illustrating the survival of various amino acid auxotrophs in the mouse gut, as detected 24 hours and 48 hours post-gavage. These auxotrophs were generated using BW25113, a non-Nissle strain of E. coli.
- Fig. 57 depicts a schematic of a repression-based kill switch.
- I n a toxin- based system, the AraC transcription factor is activated in the presence of arabinose and induces expression of TetR and an anti-toxin. TetR prevents the expression of the toxin. When arabinose is removed, TetR and the anti-toxin do not get made and the toxin is produced which kills the cell.
- the AraC transcription factor is activated in the presence of arabinose and induces expression of an essential gene.
- Fig. 58 depicts another non-limiting embodiment of the disclosure, wherein the expression of a heterologous gene is activated by an exogenous
- the AraC transcription factor adopts a conformation that represses transcription. I n the presence of arabinose, the AraC transcription factor undergoes a conformational change that allows it to bind to and activate the araBAD promoter, which induces expression of TetR (tet repressor) and an anti-toxin.
- TetR tet repressor
- the anti-toxin builds up in the recombinant bacterial cell, while TetR prevents expression of a toxin (which is under the control of a promoter having a TetR binding site).
- both the anti-toxin and TetR are not expressed.
- Fig. 58 also depicts another non-limiting embodiment of the disclosure, wherein the expression of an essential gene not found in the recombinant bacteria is activated by an exogenous environmental signal.
- the AraC transcription factor adopts a conformation that represses transcription of the essential gene under the control of the araBAD promoter and the bacterial cell cannot survive.
- the AraC transcription factor undergoes a conformational change that allows it to bind to and activate the araBAD promoter, which induces expression of the essential gene and maintains viability of the bacterial cell.
- Fig. 59 depicts a non-limiting embodiment of the disclosure, where an anti-toxin is expressed from a constitutive promoter, and expression of a heterologous gene is activated by an exogenous environmental signal.
- the AraC transcription factor adopts a conformation that represses transcription.
- the AraC transcription factor undergoes a conformational change that allows it to bind to and activate the araBAD promoter, which induces expression of TetR, thus preventing expression of a toxin.
- TetR is not expressed, and the toxin is expressed, eventually overcoming the anti-toxin and killing the cell.
- the constitutive promoter regulating expression of the anti-toxin should be a weaker promoter than the promoter driving expression of the toxin.
- the araC gene is under the control of a constitutive promoter in this circuit.
- Fig. 60 depicts a schematic of a repression-based kill switch in which the AraC transcription factor is activated in the presence of arabinose and induces expression of TetR and an anti-toxin. TetR prevents the expression of the toxin. When arabinose is removed, TetR and the anti-toxin do not get made and the toxin is produced which kills the cell.
- Fig. 61 depicts another non-limiting embodiment of the disclosure, wherein the expression of a heterologous gene is activated by an exogenous
- the AraC transcription factor adopts a conformation that represses transcription.
- the AraC transcription factor undergoes a conformational change that allows it to bind to and activate the araBAD promoter, which induces expression of TetR (tet repressor) and an anti-toxin.
- TetR tet repressor
- the anti-toxin builds up in the recombinant bacterial cell, while TetR prevents expression of a toxin (which is under the control of a promoter having a TetR binding site).
- TetR prevents expression of a toxin (which is under the control of a promoter having a TetR binding site).
- both the anti-toxin and TetR are not expressed. Since TetR is not present to repress expression of the toxin, the toxin is expressed and kills the cell.
- the araC gene is under the control of a constitutive promoter in this circuit.
- Fig. 62 depicts one non-limiting embodiment of the disclosure, where an exogenous environmental condition, e.g., low-oxygen conditions, or one or more environmental signals activates expression of a heterologous gene and at least one recombinase from an inducible promoter or inducible promoters.
- the recombinase then flips a toxin gene into an activated conformation, and the natural kinetics of the recombinase create a time delay in expression of the toxin, allowing the heterologous gene to be fully expressed. Once the toxin is expressed, it kills the cell.
- Fig. 63 depicts another non-limiting embodiment of the disclosure, where an exogenous environmental condition, e.g., low-oxygen conditions, or one or more environmental signals activates expression of a heterologous gene, an anti-toxin, and at least one recombinase from an inducible promoter or inducible promoters.
- the recombinase then flips a toxin gene into an activated conformation, but the presence of the accumulated anti-toxin suppresses the activity of the toxin.
- expression of the anti-toxin is turned off.
- the toxin is constitutively expressed, continues to accumulate, and kills the bacterial cell.
- Fig. 64 depicts another non-limiting embodiment of the disclosure, where an exogenous environmental condition, e.g., low-oxygen conditions, or one or more environmental signals activates expression of a heterologous gene and at least one recombinase from an inducible promoter or inducible promoters.
- the recombinase then flips at least one excision enzyme into an activated conformation.
- the at least one excision enzyme then excises one or more essential genes, leading to senescence, and eventual cell death.
- Fig. 65 depicts a schematic of an activation-based kill switch, in which P, is any inducible promoter, e.g., a FNR-responsive promoter.
- the anti-toxin and recombinases are turned on, which results in the toxin being 'flipped' to the ON position after 4-6 hours, which results in a build-up of anti-toxin before the toxin is expressed. I n absence of the inducing signal, only toxin is made and the cell dies.
- Fig. 66 depicts a one non-limiting embodiment of the disclosure, in which the genetically engineered bacteria produces equal amount of a Hok toxin and a shortlived Sok anti-toxin.
- the cell loses the plasmid, the anti-toxin decays, and the cell dies.
- I n the upper panel the cell produces equal amounts of toxin and anti-toxin and is stable.
- I n the center panel the cell loses the plasmid and anti-toxin begins to decay. In the lower panel, the anti-toxin decays completely, and the cell dies.
- Fig. 67 depicts the use of GeneGuards as an engineered safety
- Fig. 68 depicts a modified type 3 secretion system (T3SS) to allow the bacteria to inject secreted therapeutic proteins into the gut lumen.
- An inducible promoter (small arrow, top), e.g. a FNR-responsive promoter, drives expression of the T3 secretion system gene cassette (3 large arrows, top) that produces the apparatus that secretes tagged peptides out of the cell.
- An inducible promoter small arrow, bottom
- a FNR-responsive promoter drives expression of a regulatory factor, e.g. T7 polymerase, that then activates the expression of the tagged therapeutic peptide (hexagons).
- Fig. 69 depicts a schematic of a secretion system based on the flagellar type I II secretion in which an incomplete flagellum is used to secrete a therapeutic peptide of interest (star) by recombinantly fusing the peptide to an N-terminal flagellar secretion signal of a native flagellar component so that the intracellularly expressed chimeric peptide ca n be mobilized across the inner and outer membranes into the surrounding host environment.
- Fig. 69 depicts a schematic of a secretion system based on the flagellar type I II secretion in which an incomplete flagellum is used to secrete a therapeutic peptide of interest (star) by recombinantly fusing the peptide to an N-terminal flagellar secretion signal of a native flagellar component so that the intracellularly expressed chimeric peptide ca n be mobilized across the inner and outer membranes into the surrounding host environment.
- Fig. 69 depicts a schematic
- a therapeutic peptide star
- the N-terminal signal sequence directs the protein to the SecA-YEG machinery which moves the protein across the inner membrane into the periplasm, followed by subsequent cleavage of the signal sequence.
- the beta-domain is recruited to the Bam complex where the beta-domain is folded and inserted into the outer membrane as a beta-barrel structure.
- the therapeutic peptide is then thread through the hollow pore of the beta-barrel structure ahead of the linker sequence.
- the therapeutic peptide is freed from the linker system by an autocatalytic cleavage or by targeting of a membrane-associated peptidase (scissors) to a complementary protease cut site in the linker.
- Fig. 71 depicts a schematic of a type I secretion system, which translocates a passenger peptide directly from the cytoplasm to the extracellular space using HlyB (an ATP-binding cassette transporter); HlyD (a membrane fusion protein); and TolC (an outer membrane protein) which form a channel through both the inner and outer membranes.
- HlyB an ATP-binding cassette transporter
- HlyD a membrane fusion protein
- TolC an outer membrane protein
- Fig. 72 depicts a schematic diagram of a wild-type clbA construct (upper panel) and a schematic diagram of a clbA knockout construct (lower panel).
- Fig. 73 depicts exemplary sequences of a wild-type clbA construct and a clbA knockout construct.
- Fig. 74 depicts a schematic for inflammatory bowel disease (IBD) therapies that target pro-inflammatory neutrophils and macrophages and regulatory T cells (Treg), restore epithelial barrier integrity, and maintain mucosal barrier function. Decreasing the pro-inflammatory action of neutrophils and macrophages and increasing Treg restores epithelial barrier integrity and the mucosal barrier.
- IBD inflammatory bowel disease
- Fig. 75 depicts a schematic of non-limiting processes for designing and producing the genetically engineered bacteria of the present disclosure: identifying diverse candidate approaches based on microbial physiology and disease biology, using bioinformatics to determine candidate metabolic pathways, prospective tools to determine performance targets required of optimized engineered synthetic biotics (A); cutting-edge DNA assembly to enable combinatorial testing of pathway organization, mathematical models to predict pathway efficiency, internal stable of proprietary switches and parts to permit control and tuning of engineered circuits (B); building core structures ("chassies”), stably integrating engineered circuits into optimal chromosomal locations for efficient expression, employing unique functional assays to assess genetic circuit fidelity and activity (C); chromosomal markers enabling monitoring of synthetic biotic localization and transit times in animal models, expert microbiome network and bioinformatics support expanding understanding of how specific disease states affect Gl microbial flora and the behaviors of synthetic biotics in that environment, activating process development research and optimization in-house during the discovery phase enables rapid and seamless transition
- Fig. 76 depicts a schematic of non-limiting manufacturing processes for upstream and downstream production of the genetically engineered bacteria of the present disclosure.
- A depicts the parameters for starter culture 1 (SCI): loop full - glycerol stock, duration overnight, temperature 37° C, shaking at 250 rpm.
- B depicts the parameters for starter culture 2 (SC2): 1/100 dilution from SCI, duration 1.5 hours, temperature 37° C, shaking at 250 rpm.
- C depicts the parameters for the production bioreactor: inoculum - SC2, temperature 37° C, pH set point 7.00, pH dead band 0.05, dissolved oxygen set point 50%, dissolved oxygen cascade agitation/gas FLO, agitation limits 300-1200 rpm, gas FLO limits 0.5-20 standard liters per minute, duration 24 hours.
- D depicts the parameters for harvest: centrifugation at speed 4000 rpm and duration 30 minutes, wash IX 10% glycerol/PBS, centrifugation, re-suspension 10% glycerol/PBS.
- E depicts the parameters for vial fill/storage: 1-2 mL aliquots, -80° C
- the present disclosure includes genetically engineered bacteria, pharmaceutical compositions thereof, and methods of reducing gut inflammation, enhancing gut barrier function, and/or treating or preventing autoimmune disorders.
- the genetically engineered bacteria comprise at least one non- native gene and/or gene cassette for producing a non-native anti-inflammation and/or gut barrier function enhancer molecule(s).
- the at least one gene and/or gene cassette is further operably linked to a regulatory region that is controlled by a transcription factor that is capable of sensing an inducing condition, e.g., a low-oxygen environment, the presence of ROS, or the presence of RNS.
- the genetically engineered bacteria are capable of producing the anti-inflammation and/or gut barrier function enhancer molecule(s) in inducing environments, e.g., in the gut.
- the genetically engineered bacteria are capable of producing the anti-inflammation and/or gut barrier function enhancer molecule(s) in inducing environments, e.g., in the gut.
- engineered bacteria and pharmaceutical compositions comprising those bacteria may be used to treat or prevent autoimmune disorders and/or diseases or conditions associated with gut inflammation and/or compromised gut barrier function, including IBD.
- gut inflammation and/or compromised gut barrier function include, but are not limited to, inflammatory bowel diseases, diarrheal diseases, and related diseases.
- IBD Inflammatory bowel diseases
- IBD ulcerative colitis
- collagenous colitis a group of diseases associated with gut inflammation, which include, but are not limited to, Crohn's disease, ulcerative colitis, collagenous colitis, lymphocytic colitis, diversion colitis, Behcet's disease, and
- diarrheal diseases include, but are not limited to, acute watery diarrhea, e.g., cholera; acute bloody diarrhea, e.g., dysentery; and persistent diarrhea.
- related diseases include, but are not limited to, short bowel syndrome, ulcerative proctitis, proctosigmoiditis, left-sided colitis, pancolitis, and fulminant colitis.
- Symptoms associated with the aforementioned diseases and conditions include, but are not limited to, one or more of diarrhea, bloody stool, mouth sores, perianal disease, abdominal pain, abdominal cramping, fever, fatigue, weight loss, iron deficiency, anemia, appetite loss, weight loss, anorexia, delayed growth, delayed pubertal development, inflammation of the skin, inflammation of the eyes, inflammation of the joints, inflammation of the liver, and inflammation of the bile ducts.
- a disease or condition associated with gut inflammation and/or compromised gut barrier function may be an autoimmune disorder.
- a disease or condition associated with gut inflammation and/or compromised gut barrier function may be co-morbid with an autoimmune disorder.
- autoimmune disorders include, but are not limited to, acute disseminated encephalomyelitis (ADEM), acute necrotizing hemorrhagic leukoencephalitis, Addison's disease,
- agammaglobulinemia alopecia areata, amyloidosis, ankylosing spondylitis, anti- GBM/anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune angioedema, autoimmune aplastic anemia, autoimmune dysautonomia, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune
- autoimmune inner ear disease AIED
- autoimmune myocarditis autoimmune myocarditis
- autoimmune oophoritis autoimmune pancreatitis
- autoimmune retinopathy autoimmune retinopathy
- autoimmune thrombocytopenic purpura ATP
- thyroid disease autoimmune thyroid disease
- autoimmune urticarial, axonal & neuronal neuropathies Balo disease, Behcet's disease, bullous pemphigoid, cardiomyopathy, Castleman disease, celiac disease, Chagas disease, chronic inflammatory demyelinating polyneuropathy (CIDP), chronic recurrent multifocal ostomyelitis (CRMO), Churg-Strauss syndrome, cicatricial pemphigoid/benign mucosal pemphigoid, Crohn's disease, Cogan's syndrome, cold agglutinin disease, congenital heart block, Coxsackie myocarditis, CREST disease, essential mixed cryoglobulinemia,
- encephalomyelitis Evans syndrome, fibrosing alveolitis, giant cell arteritis (temporal arteritis), giant cell myocarditis, glomerulonephritis, Goodpasture's syndrome, granulomatosis with polyangiitis (GPA), Graves' disease, Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, hemolytic anemia, Henoch-Schonlein purpura, herpes gestationis, hypogammaglobulinemia, idiopathic thrombocytopenic purpura (ITP), IgA nephropathy, lgG4-related sclerosing disease, immunoregulatory lipoproteins, inclusion body myositis, interstitial cystitis, juvenile arthritis, juvenile idiopathic arthritis, juvenile myositis, Kawasaki syndrome, Lambert-Eaton syndrome, leukocytoclastic vasculitis, lichen planus, lichen
- anti-inflammation molecules and/or “gut barrier function enhancer molecules” include, but are not limited to, short-chain fatty acids, butyrate, propionate, acetate, I L-2, IL-22, superoxide dismutase (SOD), kynurenine, GLP- 2, GLP-1, IL-10, I L-27, TGF- ⁇ , TGF- 2, N-acylphosphatidylethanolamines (NAPEs), elafin (also called peptidase inhibitor 3 and SKALP), trefoil factor, melatonin, PGD 2 , and kynurenic acid, as well as other molecules disclosed herein.
- SOD superoxide dismutase
- kynurenine GLP- 2, GLP-1, IL-10, I L-27, TGF- ⁇ , TGF- 2, N-acylphosphatidylethanolamines (NAPEs), elafin (also called peptidase inhibitor 3 and SKALP), tre
- Such molecules may also include compounds that inhibit pro-inflammatory molecules, e.g., a single-chain variable fragment (scFv), antisense RNA, siRNA, or shRNA that neutralizes TNF-a, IFN- ⁇ , IL- ⁇ , I L-6, I L-8, I L-17, a nd/or chemokines, e.g., CXCL-8 and CCL2.
- a molecule may be primarily antiinflammatory, e.g., IL-10, or primarily gut barrier function enhancing, e.g., GLP-2.
- a molecule may be both anti-inflammatory and gut barrier function enhancing.
- An anti- inflammation and/or gut barrier function enhancer molecule may be encoded by a single gene, e.g., elafin is encoded by the PI3 gene.
- an anti-inflammation and/or gut barrier function enhancer molecule may be synthesized by a biosynthetic pathway requiring multiple genes, e.g., butyrate. These molecules may also be referred to as therapeutic molecules.
- the term "gene” or “gene sequene” is meant to refer to a nucleic acid sequence encoding any of the anti-inflammatory and gut barrier function enhancing molecules described herein, e.g., I L-2, I L-22, superoxide dismutase (SOD), kynurenine, GLP-2, GLP-1, I L-10, IL-27, TGF- ⁇ , TGF- 2, N-acylphosphatidylethanolamines (NAPEs), elafin, and trefoil factor, as well as others.
- the nucleic acid sequence may comprise the entire gene sequence or a partial gene sequence encoding a functional molecule.
- the nucleic acid sequence may be a natural sequence or a synthetic sequence.
- the nucleic acid sequence may comprise a native or wild-type sequence or may comprise
- nucleic acid sequence may be codon-optimized.
- a "gene cassette” or “operon” encoding a biosynthetic pathway refers to the two or more genes that are required to produce an anti- inflammation and/or gut barrier function enhancer molecule, e.g., butyrate, propionate, and acetate.
- the gene cassette or operon may also comprise additional transcription and translation elements, e.g., a ribosome binding site.
- butyrogenic gene cassette and “butyrate biosynthesis gene cassette” are used interchangeably to refer to a set of genes capable of producing butyrate in a biosynthetic pathway.
- Unmodified bacteria that are capable of producing butyrate via an endogenous butyrate biosynthesis pathway include, but are not limited to, Clostridium, Peptoclostridium, Fusobacterium, Butyrivibrio, Eubacterium, and Treponema, and these endogenous butyrate biosynthesis pathways may be a source of genes for the genetically engineered bacteria of the invention.
- the genetically engineered bacteria of the invention may comprise butyrate biosynthesis genes from a different species, strain, or substrain of bacteria, or a combination of butyrate biosynthesis genes from different species, strains, and/or substrains of bacteria.
- a butyrogenic gene cassette may comprise, for example, the eight genes of the butyrate production pathway from Peptoclostridium difficile (also called Clostridium difficile): bcd2, etfB3, etfA3, thiAl, hbd, crt2, pbt, and buk, which encode butyryl-CoA
- dehydrogenase subunit electron transfer flavoprotein subunit beta, electron transfer flavoprotein subunit alpha, acetyl-CoA C-acetyltransferase, 3-hydroxybutyryl-CoA dehydrogenase, crotonase, phosphate butyryltransferase, and butyrate kinase, respectively (Aboulnaga et al., 2013).
- One or more of the butyrate biosynthesis genes may be functionally replaced or modified, e.g., codon optimized.
- Peptoclostridium difficile strain 630 and strain 1296 are both capable of producing butyrate, but comprise different nucleic acid sequences for etfA3, thiAl, hbd, crt2, pbt, and buk.
- a butyrogenic gene cassette may comprise bcd2, etfB3, etfA3, and thiAl from Peptoclostridium difficile strain 630, and hbd, crt2, pbt, and buk from Peptoclostridium difficile strain 1296.
- a single gene from Treponema denticola (ter, encoding trans-2-enoynl-CoA reductase) is capable of functionally replacing all three of the bcd2, etfB3, and etfA3 genes from Peptoclostridium difficile.
- a butyrogenic gene cassette may comprise thiAl, hbd, crt2, pbt, and buk from Peptoclostridium difficile and ter from Treponema denticola.
- addition of the tesB gene from Escherichia Coli is capable of functionally replacing pbt and buk genes from Peptoclostridium difficile.
- a butyrogenic gene cassette may comprise thiAl, hbd, and crt2 from Peptoclostridium difficile, ter from Treponema denticola, and tesS from Escherichia Coli, for example, thiAl from Peptoclostridium difficile strain 630, hbd and crt2 from Peptoclostridium difficile strain 1296, ter from Treponema denticola and tesB from Escherichia Coli.
- the butyrogenic gene cassette may comprise genes for the aerobic biosynthesis of butyrate and/or genes for the anaerobic or microaerobic biosynthesis of butyrate.
- One or more of the butyrate biosynthesis genes may be functionally replaced or modified, e.g., codon optimized.
- Exemplary butyrate gene cassettes are shown in Figs. 1, 3, 4, 5, 6, 7, and 8.
- propionate gene cassette and "propionate biosynthesis gene cassette” refer to a set of genes capable of producing propionate in a biosynthetic pathway.
- Unmodified bacteria that are capable of producing propionate via an endogenous propionate biosynthesis pathway include, but are not limited to, Clostridium propionicum, Megasphaera elsdenii, and Prevotella ruminicola, and these endogenous propionate biosynthesis pathways may be a source of genes for the genetically engineered bacteria of the invention.
- the genetically engineered bacteria of the invention may comprise propionate biosynthesis genes from a different species, strain, or substrain of bacteria, or a combination of propionate biosynthesis genes from different species, strains, and/or substrains of bacteria.
- the propionate gene cassette comprises acrylate pathway propionate biosynthesis genes, e.g., pet, IcdA, IcdB, IcdC, etfA, acrB, and acrC, which encode propionate CoA-transferase, lactoyl-CoA dehydratase A, lactoyl-CoA dehydratase B, lactoyl-CoA dehydratase C, electron transfer flavoprotein subunit A, acryloyl-CoA reductase B, and acryloyl-CoA reductase C, respectively (Hetzel et al., 2003, Selmer et al., 2002).
- the propionate gene cassette comprises acrylate pathway
- thrA f r thrB, thrC, ilvA f r , oceE, oceF, and Ipd, which encode homoserine dehydrogenase 1, homoserine kinase, L-threonine synthase, L- threonine dehydratase, pyruvate dehydrogenase, dihydrolipoamide acetyltransferase, and dihydrolipoyl dehydrogenase, respectively.
- the propionate gene cassette further comprises tesB, which encodes acyl-CoA thioesterase.
- the propionate gene cassette may comprise genes for the aerobic biosynthesis of propionate and/or genes for the anaerobic or microaerobic biosynthesis of propionate.
- One or more of the proprionate biosynthesis genes may be functionally replaced or modified, e.g., codon optimized.
- Exemplary propionic gene cassettes are shown in Figs. 9, 11, and 13.
- acetate gene cassette and "acetate biosynthesis gene cassette” refers to a set of genes capable of producing acetate in a biosynthetic pathway. Bacteria synthesize acetate from a number of carbon and energy sources, including a variety of substrates such as cellulose, lignin, and inorganic gases, and utilize different biosynthetic mechanisms and genes, which are known in the art (Ragsdale, 2008).
- Unmodified bacteria that are capable of producing acetate via an endogenous acetate biosynthesis pathway may be a source of acetate biosynthesis genes for the genetically engineered bacteria of the invention.
- the genetically engineered bacteria of the invention may comprise acetate biosynthesis genes from a different species, strain, or substrain of bacteria, or a combination of acetate biosynthesis genes from different species, strains, and/or substrains of bacteria.
- Escherichia coli are capable of consuming glucose and oxygen to produce acetate and carbon dioxide during aerobic growth (Kleman et al., 1994).
- Thermoacetogenium are acetogenic anaerobes that are capable of converting CO or C0 2 + H 2 into acetate, e.g., using the Wood-Ljungdahl pathway (Schiel- Bengelsdorf et al, 2012). Genes in the Wood-Ljungdahl pathway for various bacterial species are known in the art.
- the acetate gene cassette may comprise genes for the aerobic biosynthesis of acetate and/or genes for the anaerobic or microaerobic biosynthesis of acetate.
- One or more of the acetate biosynthesis genes may be
- Examples of acetate gene cassettes are described herein.
- Each gene sequence and/or gene cassette may be present on a plasmid or bacterial chromosome.
- the engineered bacteria comprise one or more gene sequence(s) and one or more gene cassettes
- the gene sequence(s) may be present on one or more plasmids and the gene cassette(s) may be present in the bacterial chromosome, and vice versa.
- multiple copies of any gene, gene cassette, or regulatory region may be present in the bacterium, wherein one or more copies of the gene, gene cassette, or regulatory region may be mutated or otherwise altered as described herein.
- the genetically engineered bacteria are engineered to comprise multiple copies of the same gene, gene cassette, or regulatory region in order to enhance copy number.
- the genetically engineered bacteria are engineered to comprise multiple different components of a gene cassette performing multiple different functions. In some embodiments, the genetically engineered bacteria are engineered to comprise one or more copies of different genes, gene cassettes, or regulatory regions to produce engineered bacteria that express more than one therapeutic molecule and/or perform more than one function.
- Each gene or gene cassette may be operably linked to an inducible promoter, e.g., an FNR-responsive promoter, an ROS-responsive promoter, and/or an RNS-responsive promoter.
- an inducible promoter refers to a regulatory region that is operably linked to one or more genes, wherein expression of the gene(s) is increased in the presence of an inducer of said regulatory region.
- a "directly inducible promoter” refers to a regulatory region, wherein the regulatory region is operably linked to a gene or a gene cassette encoding a biosynthetic pathway for producing an anti-inflammation and/or gut barrier function enhancer molecule, e.g. butyrate. In the presence of an inducer of said regulatory region, an anti-inflammation and/or gut barrier function enhancer molecule is expressed.
- An "indirectly inducible promoter” refers to a regulatory system comprising two or more regulatory regions, for example, a first regulatory region that is operably
- a gene encoding a first molecule e.g., a transcription factor
- a second regulatory region operably linked to a gene or a gene cassette encoding a biosynthetic pathway for producing an anti-inflammation and/or gut barrier function enhancer molecule, e.g. butyrate (or other anti-inflammation and/or gut barrier function enhancer molecule).
- the second regulatory region may be activated or repressed, thereby activating or repressing production of butyrate (or other anti-inflammation and/or gut barrier function enhancer molecule).
- Both a directly inducible promoter and an indirectly inducible promoter are encompassed by "inducible promoter.”
- operably linked refers a nucleic acid sequence, e.g., a gene or gene cassette for producing an anti-inflammation and/or gut barrier enhancer molecule, that is joined to a regulatory region sequence in a manner which allows expression of the nucleic acid sequence, e.g., acts in cis.
- a regulatory region is a nucleic acid that can direct transcription of a gene of interest and may comprise promoter sequences, enhancer sequences, response elements, protein recognition sites, inducible elements, promoter control elements, protein binding sequences, 5' and 3' untranslated regions, transcriptional start sites, termination sequences, polyadenylation sequences, and introns.
- exogenous environmental conditions refer to settings or circumstances under which the promoter described herein is directly or indirectly induced.
- exogenous environmental conditions is meant to refer to the environmental conditions external to the bacteria, but endogenous or native to a mammalian subject.
- exogenous and endogenous may be used
- the exogenous environmental conditions are specific to the gut of a mammal. In some embodiments, the exogenous environmental conditions are specific to the upper gastrointestinal tract of a mammal. I n some embodiments, the exogenous environmental conditions are specific to the lower gastrointestinal tract of a mammal. In some embodiments, the exogenous
- the exogenous environmental condition is an environment in which ROS is present. I n some embodiments, the exogenous environmental condition is an environment in which RNS is present.
- the exogenous environmental conditions are low- oxygen or anaerobic conditions such as the environment of the mammalian gut.
- exogenous environmental conditions refer to the presence of molecules or metabolites that are specific to the mammalian gut in a healthy or disease state, e.g., propionate.
- the gene or gene cassette for producing a therapeutic molecule is operably linked to an oxygen level-dependent promoter.
- an "oxygen level-dependent promoter” or “oxygen level-dependent regulatory region” refers to a nucleic acid sequence to which one or more oxygen level-sensing transcription factors is capable of binding, wherein the binding and/or activation of the corresponding transcription factor activates downstream gene expression.
- the gene or gene cassette for producing a therapeutic molecule is operably linked to an oxygen level-dependent regulatory region such that the therapeutic molecule is expressed in low-oxygen, microaerobic, or anaerobic conditions.
- the oxygen level-dependent regulatory region is operably linked to a butyrogenic or other gene cassette or gene sequence(s) (e.g., any of the genes described herein); in low-oxygen conditions, the oxygen level-dependent regulatory region is activated by a corresponding oxygen level-sensing transcription factor, thereby driving expression of the butyrogenic or other gene cassette or gene sequence(s).
- oxygen level-dependent transcription factors include, but are not limited to, FNR, AN R, and DNR.
- FNR-responsive promoters are known in the art (see, e.g., Castiglione et al., 2009; Eiglmeier et al., 1989; Galimand et al., 1991; Hasegawa et al.,
- RNS can cause deleterious cellular effects such as nitrosative stress.
- RNS include, but are not limited to, nitric oxide (NO), peroxynitrite or peroxynitrite anion (ONOO ), nitrogen dioxide ( ⁇ 0 2 ), dinitrogen trioxide (N 2 0 3 ), peroxynitrous acid (ONOOH), and nitroperoxycarbonate (ONOOC0 2 ⁇ ) (unpaired electrons denoted by ⁇ ).
- NO nitric oxide
- ONOO peroxynitrite or peroxynitrite anion
- N 2 0 3 dinitrogen trioxide
- ONOOH peroxynitrous acid
- ONOOC0 2 ⁇ unpaired electrons denoted by ⁇ .
- Bacteria have evolved transcription factors that are capable of sensing RNS levels.
- RNS-inducible regulatory region refers to a nucleic acid sequence to which one or more RNS-sensing transcription factors is capable of binding, wherein the binding and/or activation of the corresponding transcription factor activates downstream gene expression; in the presence of RNS, the transcription factor binds to and/or activates the regulatory region.
- the RNS-inducible regulatory region comprises a promoter sequence.
- the transcription factor senses RNS and subsequently binds to the RNS-inducible regulatory region, thereby activating downstream gene expression.
- the transcription factor is bound to the RNS-inducible regulatory region in the absence of RNS; in the presence of RNS, the transcription factor undergoes a conformational change, thereby activating downstream gene expression.
- the RNS-inducible regulatory region may be operatively linked to a gene or gene cassette, e.g., a butyrogenic or other gene
- a transcription factor senses RNS and activates a corresponding RNS-inducible regulatory region, thereby driving expression of an operatively linked gene sequence or gene cassette.
- RNS induces expression of the gene or gene cassette.
- RNS-derepressible regulatory region refers to a nucleic acid sequence to which one or more RNS-sensing transcription factors is capable of binding, wherein the binding of the corresponding transcription factor represses downstream gene expression; in the presence of RNS, the transcription factor does not bind to and does not repress the regulatory region.
- the RNS- derepressible regulatory region comprises a promoter sequence.
- the RNS-derepressible regulatory region may be operatively linked to a gene or gene cassette, e.g., a butyrogenic or other gene cassette or gene sequence(s).
- a transcription factor senses RNS and no longer binds to and/or represses the regulatory region, thereby derepressing an operatively linked gene sequence or gene cassette.
- RNS derepresses expression of the gene or gene cassette.
- RNS-repressible regulatory region refers to a nucleic acid sequence to which one or more RNS-sensing transcription factors is capable of binding, wherein the binding of the corresponding transcription factor represses downstream gene expression; in the presence of RNS, the transcription factor binds to and represses the regulatory region.
- the RNS-repressible regulatory region comprises a promoter sequence.
- the transcription factor that senses RNS is capable of binding to a regulatory region that overlaps with part of the promoter sequence.
- the transcription factor that senses RNS is capable of binding to a regulatory region that is upstream or downstream of the promoter sequence.
- the RNS-repressible regulatory region may be operatively linked to a gene sequence or gene cassette.
- a transcription factor senses RNS and binds to a corresponding RNS-repressible regulatory region, thereby blocking expression of an operatively linked gene sequence or gene cassette.
- RNS represses expression of the gene or gene cassette.
- a "RNS-responsive regulatory region” refers to a RNS- inducible regulatory region, a RNS-repressible regulatory region, and/or a RNS- derepressible regulatory region.
- the RNS-responsive regulatory region comprises a promoter sequence. Each regulatory region is capable of binding at least one corresponding RNS-sensing transcription factor. Examples of transcription factors that sense RNS and their corresponding RNS-responsive genes, promoters, and/or regulatory regions include, but are not limited to, those shown in Table 2.
- ROS can be produced as byproducts of aerobic respiration or metal- catalyzed oxidation and may cause deleterious cellular effects such as oxidative damage.
- ROS include, but are not limited to, hydrogen peroxide (H 2 0 2 ), organic peroxide (ROOH), hydroxyl ion (OH ), hydroxyl radical ( ⁇ ), superoxide or superoxide anion ( ⁇ 0 2 ⁇ ), singlet oxygen ( ⁇ 0 2 ), ozone (0 3 ), carbonate radical, peroxide or peroxyl radical ( ⁇ 0 2 ⁇ 2 ), hypochlorous acid (HOCI), hypochlorite ion (OCI ), sodium hypochlorite (NaOCI), nitric oxide ( ⁇ ), and peroxynitrite or peroxynitrite anion (ONOO ) (unpaired electrons denoted by ⁇ ).
- Bacteria have evolved transcription factors that are capable of sensing ROS levels. Different ROS signaling pathways are triggered by different ROS levels and occur with different kinetics (Marinho et al., 2014).
- ROS-inducible regulatory region refers to a nucleic acid sequence to which one or more ROS-sensing transcription factors is capable of binding, wherein the binding and/or activation of the corresponding transcription factor activates downstream gene expression; in the presence of ROS, the transcription factor binds to
- the ROS-inducible regulatory region comprises a promoter sequence.
- the transcription factor senses ROS and subsequently binds to the ROS-inducible regulatory region, thereby activating downstream gene expression.
- the transcription factor is bound to the ROS-inducible regulatory region in the absence of ROS; in the presence of ROS, the transcription factor undergoes a conformational change, thereby activating downstream gene expression.
- the ROS-inducible regulatory region may be operatively linked to a gene sequence or gene cassette, e.g., a butyrogenic gene cassette.
- a transcription factor e.g., OxyR
- OxyR senses ROS and activates a corresponding ROS-inducible regulatory region, thereby driving expression of an operatively linked gene sequence or gene cassette.
- ROS induces expression of the gene or gene cassette.
- ROS-derepressible regulatory region refers to a nucleic acid sequence to which one or more ROS-sensing transcription factors is capable of binding, wherein the binding of the corresponding transcription factor represses downstream gene expression; in the presence of ROS, the transcription factor does not bind to and does not repress the regulatory region.
- the ROS- derepressible regulatory region comprises a promoter sequence.
- the ROS-derepressible regulatory region may be operatively linked to a gene or gene cassette, e.g., a
- ROS butyrogenic or other gene cassette or gene sequence(s) described herein.
- a transcription factor e.g., OhrR
- ROS derepresses expression of the gene or gene cassette.
- ROS-repressible regulatory region refers to a nucleic acid sequence to which one or more ROS-sensing transcription factors is capable of binding, wherein the binding of the corresponding transcription factor represses downstream gene expression; in the presence of ROS, the transcription factor binds to and represses the regulatory region.
- the ROS-repressible regulatory region comprises a promoter sequence.
- ROS-31- senses ROS is capable of binding to a regulatory region that overlaps with part of the promoter sequence.
- the transcription factor that senses ROS is capable of binding to a regulatory region that is upstream or downstream of the promoter sequence.
- the ROS-repressible regulatory region may be operatively linked to a gene sequence or gene cassette.
- a transcription factor e.g., PerR
- PerR senses ROS and binds to a corresponding ROS-repressible regulatory region, thereby blocking expression of an operatively linked gene sequence or gene cassette.
- ROS represses expression of the gene or gene cassette.
- a "ROS-responsive regulatory region” refers to a ROS- inducible regulatory region, a ROS-repressible regulatory region, and/or a ROS- derepressible regulatory region.
- the ROS-responsive regulatory region comprises a promoter sequence. Each regulatory region is capable of binding at least one corresponding ROS-sensing transcription factor. Examples of transcription factors that sense ROS and their corresponding ROS-responsive genes, promoters, and/or regulatory regions include, but are not limited to, those shown in Table 3.
- a "tunable regulatory region” refers to a nucleic acid sequence under direct or indirect control of a transcription factor and which is capable of activating, repressing, derepressing, or otherwise controlling gene expression relative to levels of an inducer.
- the tunable regulatory region comprises a promoter sequence.
- the inducer may be RNS, or other inducer described herein, and the tunable regulatory region may be a RNS-responsive regulatory region or other responsive regulatory region described herein.
- the tunable regulatory region may be operatively linked to a gene sequence(s) or gene cassette, e.g., a butyrogenic or other gene cassette or gene sequence(s).
- the tunable regulatory region is a RNS-derepressible regulatory region, and when RNS is present, a RNS-sensing transcription factor no longer binds to and/or represses the regulatory region, thereby permitting expression of the operatively linked gene or gene cassette.
- the tunable regulatory region derepresses gene or gene cassette expression relative to RNS levels.
- Each gene or gene cassette may be operatively linked to a tunable regulatory region that is directly or indirectly controlled by a transcription factor that is capable of sensing at least one RNS.
- a "non-native" nucleic acid sequence refers to a nucleic acid sequence not normally present in a bacterium, e.g., an extra copy of an endogenous sequence, or a heterologous sequence such as a sequence from a different species, strain, or substrain of bacteria, or a sequence that is modified and/or mutated as compared to the unmodified sequence from bacteria of the same subtype.
- the non-native nucleic acid sequence is a synthetic, non-naturally occurring sequence (see, e.g., Purcell et al., 2013).
- the non-native nucleic acid sequence may be a regulatory region, a promoter, a gene, and/or one or more genes in gene cassette.
- “non-native” refers to two or more nucleic acid sequences that are not found in the same relationship to each other in nature.
- the non- native nucleic acid sequence may be present on a plasmid or chromosome. I n addition, multiple copies of any regulatory region, promoter, gene, and/or gene cassette may be present in the bacterium, wherein one or more copies of the regulatory region, promoter, gene, and/or gene cassette may be mutated or otherwise altered as described
- the genetically engineered bacteria are engineered to comprise multiple copies of the same regulatory region, promoter, gene, and/or gene cassette in order to enhance copy number or to comprise multiple different components of a gene cassette performing multiple different functions or to comprise one or more copies of different regulatory regions, promoters, genes, and/or gene cassette to produce engineered bacteria that express more than one therapeutic molecule and/or perform more than one function.
- the genetically engineered bacteria of the invention comprise a gene cassette that is operably linked to a directly or indirectly inducible promoter that is not associated with said gene cassette in nature, e.g., a FN R- responsive promoter operably linked to a butyrogenic gene cassette.
- Constant promoter refers to a promoter that is capable of facilitating continuous transcription of a coding sequence or gene under its control and/or to which it is operably linked.
- Constitutive promoters and functional variants are well known in the art and include, but are not limited to, BBa_J23100, a constitutive Escherichia coli o s promoter (e.g., an osmY promoter (I nternational Genetically Engineered Machine (iGEM) Registry of Standard Biological Parts Name BBa_J45992; BBa_J45993)), a constitutive Escherichia coli ⁇ 32 promoter (e.g., htpG heat shock promoter (BBa_J45504)), a constitutive Escherichia coli ⁇ 70 promoter (e.g., lacq promoter (BBa_J54200;
- a constitutive Escherichia coli o s promoter e.g., an osmY
- BBa_J56015) E. coli CreABCD phosphate sensing operon promoter (BBa_J64951), GlnRS promoter (BBa_K088007), lacZ promoter (BBa_K119000; BBa_K119001); M13K07 gene I promoter (BBa_M13101); M13K07 gene II promoter (BBa_M13102), M13K07 gene I II promoter (BBa_M13103), M13K07 gene IV promoter (BBa_M13104), M13K07 gene V promoter (BBa_M13105), M13K07 gene VI promoter (BBa_M13106), M13K07 gene VII I promoter (BBa_M13108), M13110 (BBa_M13110)), a constitutive Bacillus subtilis ⁇ ⁇ promoter (e.g., promoter veg (BBa_K143013), promoter 43 (BBa_K143013), P
- BBa_K823000 P
- a constitutive Bacillus subtilis ⁇ ⁇ promoter e.g., promoter etc (BBa_K143010), promoter gsiB (BBa_K143011)
- a Salmonella promoter e.g., Pspv2 from Salmonella (BBa_K112706), Pspv from Salmonella (BBa_K112707)
- a bacteriophage T7 promoter e.g., T7 promoter (BBa_l712074;
- BBa_J64997 BBa_K113010; BBa_K113011; BBa_K113012; BBa_R0085; BBa_R0180; BBa_R0181; BBa_R0182; BBa_R0183; BBa_Z0251; BBa_Z0252; BBa_Z0253)
- a bacteriophage SP6 promoter e.g., SP6 promoter (BBa_J64998)
- Gl gastrointestinal
- the gut also comprises accessory organs and glands, such as the spleen, liver, gallbladder, and pancreas.
- the upper gastrointestinal tract comprises the esophagus, stomach, and duodenum of the small intestine.
- the lower gastrointestinal tract comprises the remainder of the small intestine, i.e., the jejunum and ileum, and all of the large intestine, i.e., the cecum, colon, rectum, and anal canal.
- Bacteria can be found throughout the gut, e.g., in the gastrointestinal tract, and particularly in the intestines.
- Non-pathogenic bacteria refer to bacteria that are not capable of causing disease or harmful responses in a host.
- non-pathogenic bacteria are Gram-negative bacteria.
- non-pathogenic bacteria are Gram-positive bacteria.
- non-pathogenic bacteria are commensal bacteria, which are present in the indigenous microbiota of the gut.
- non-pathogenic bacteria examples include, but are not limited to Bacillus, Bacteroides, Bifidobacterium, Brevi bacteria, Clostridium, Enterococcus, Escherichia, Lactobacillus, Lactococcus, Saccharomyces, and Staphylococcus, e.g., Bacillus coagulans, Bacillus subtil is, Bacteroides fragilis, Bacteroides subtilis, Bacteroides thetaiotaomicron,
- Non-pathogenic bacteria also include
- Naturally pathogenic bacteria may be genetically engineered to reduce or eliminate pathogenicity.
- Probiotic is used to refer to live, non-pathogenic microorganisms, e.g., bacteria, which can confer health benefits to a host organism that contains an
- the host organism is a mammal. In some embodiments, the host organism is a human. Some species, strains, and/or subtypes of non-pathogenic bacteria are currently recognized as probiotic.
- probiotic bacteria examples include, but are not limited to, Bifidobacteria, Escherichia, Lactobacillus, and Saccharomyces, e.g., Bifidobacterium bifidum, Enterococcus faecium, Escherichia coli, Escherichia coli strain Nissle, Lactobacillus acidophilus, Lactobacillus bulgaricus, Lactobacillus paracasei, Lactobacillus plantarum, and Saccharomyces boulardii (Dinleyici et al., 2014; U.S. Patent No. 5,589,168; U.S. Patent No. 6,203,797; U.S. Patent 6,835,376).
- Bifidobacterium bifidum Enterococcus faecium
- Escherichia coli Escherichia coli strain Nissle
- Lactobacillus acidophilus Lactobacillus bulg
- the probiotic may be a variant or a mutant strain of bacterium (Arthur et al., 2012; Cuevas-Ramos et al., 2010; Olier et al., 2012; Nougayrede et al., 2006).
- Non-pathogenic bacteria may be genetically engineered to enhance or improve desired biological properties, e.g., survivability.
- Non-pathogenic bacteria may be genetically engineered to provide probiotic properties.
- Probiotic bacteria may be genetically engineered to enhance or improve probiotic properties.
- stable bacterium is used to refer to a bacterial host cell carrying non-native genetic material, e.g., a butyrogenic or other gene cassette or gene sequence(s), which is incorporated into the host genome or propagated on a self-replicating extra-chromosomal plasmid, such that the non-native genetic material is retained, expressed, and/or propagated.
- non-native genetic material e.g., a butyrogenic or other gene cassette or gene sequence(s)
- the stable bacterium is capable of survival and/or growth in vitro, e.g., in medium, and/or in vivo, e.g., in the gut.
- the stable bacterium may be a genetically modified bacterium comprising a butyrogenic or other gene cassette or gene sequence(s), in which the plasmid or chromosome carrying the butyrogenic or other gene cassette or gene sequence(s) is stably maintained in the host cell, such that the gene cassette or gene sequence(s) can be expressed in the host cell, and the host cell is capable of survival
- copy number affects the stability of expression of the non-native genetic material. In some embodiments, copy number affects the level of expression of the non-native genetic material.
- the term “treat” and its cognates refer to an amelioration of a disease or disorder, or at least one discernible symptom thereof. In another embodiment, “treat” refers to an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient. In another embodiment, “treat” refers to inhibiting the progression of a disease or disorder, either physically (e.g., stabilization of a discernible symptom), physiologically (e.g., stabilization of a physical parameter), or both. In another embodiment, “treat” refers to slowing the progression or reversing the progression of a disease or disorder. As used herein, “prevent” and its cognates refer to delaying the onset or reducing the risk of acquiring a given disease or disorder.
- Those in need of treatment may include individuals already having a particular medical disorder, as well as those at risk of having, or who may ultimately acquire the disorder.
- the need for treatment is assessed, for example, by the presence of one or more risk factors associated with the development of a disorder, the presence or progression of a disorder, or likely receptiveness to treatment of a subject having the disorder.
- Treating autoimmune disorders and/or diseases and conditions associated with gut inflammation and/or compromised gut barrier function may encompass reducing or eliminating excess inflammation and/or associated symptoms, and does not necessarily encompass the elimination of the underlying disease or disorder.
- the "initial colonization of the newborn intestine is particularly relevant to the proper development of the host's immune and metabolic functions and to determine disease risk in early and later life" (Sanz et al., 2015).
- early intervention e.g., prenatal, perinatal, neonatal
- using the genetically engineered bacteria of the invention may be sufficient to prevent or delay the onset of the disease or disorder.
- composition refers to a preparation of genetically engineered bacteria of the invention with other components such as a physiologically suitable carrier and/or excipient.
- physiologically acceptable carrier and “pharmaceutically acceptable carrier” which may be used interchangeably refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered bacterial compound.
- An adjuvant is included under these phrases.
- excipient refers to an inert substance added to a
- compositions to further facilitate administration of an active ingredient.
- examples include, but are not limited to, calcium bicarbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, polyethylene glycols, and surfactants, including, for example, polysorbate 20.
- terapéuticaally effective dose and "therapeutically effective amount” are used to refer to an amount of a compound that results in prevention, delay of onset of symptoms, or amelioration of symptoms of a condition, e.g., inflammation, diarrhea.
- a therapeutically effective amount may, for example, be sufficient to treat, prevent, reduce the severity, delay the onset, and/or reduce the risk of occurrence of one or more symptoms of an autoimmune disorder and/or a disease or condition associated with gut inflammation and/or compromised gut barrier function.
- a therapeutically effective amount, as well as a therapeutically effective frequency of administration can be determined by methods known in the art and discussed below.
- the genetically engineered bacteria of the invention are capable of producing a one or more non-native anti-inflammation and/or gut barrier function
- the genetically engineered bacteria are naturally non-pathogenic bacteria. In some embodiments, the genetically engineered bacteria are commensal bacteria. In some embodiments, the genetically engineered bacteria are probiotic bacteria. In some embodiments, the genetically engineered bacteria are naturally pathogenic bacteria that are modified or mutated to reduce or eliminate pathogenicity. In some embodiments, non-pathogenic bacteria are Gram- negative bacteria. In some embodiments, non-pathogenic bacteria are Gram-positive bacteria.
- Exemplary bacteria include, but are not limited to Bacillus, Bacteroides, Bifidobacterium, Brevibacteria, Clostridium, Enterococcus, Escherichia coli, Lactobacillus, Lactococcus, Saccharomyces, and Staphylococcus, e.g., Bacillus coagulans, Bacillus subtil is, Bacteroides fragilis, Bacteroides subtilis, Bacteroides thetaiotaomicron,
- Lactobacillus bulgaricus Lactobacillus casei, Lactobacillus johnsonii, Lactobacillus paracasei, Lactobacillus plantarum, Lactobacillus reuteri, Lactobacillus rhamnosus, Lactococcus lactis, Saccharomyces boulardii, Clostridium clusters IV and XlVa of
- the genetically engineered bacteria are selected from the group consisting of Bacteroides fragilis, Bacteroides thetaiotaomicron, Bacteroides subtilis, Bifidobacterium bifidum,
- the genetically engineered bacterium is a Gram-positive bacterium, e.g., Clostridium, that is naturally capable of producing high levels of butyrate.
- the genetically engineered bacterium is selected from the group consisting of C. butyricum ZJUCB, C butyricum S21, C
- thermobutyricum ATCC 49875 C beijerinckii, C. populeti ATCC 35295, C tyrobutyricum JM1, C tyrobutyricum CI P 1-776, C tyrobutyricum ATCC 25755, C tyrobutyricum CNRZ
- the genetically engineered bacterium is C. butyricum CBM588, a probiotic bacterium that is highly amenable to protein secretion and has demonstrated efficacy in treating IBD (Kanai et al., 2015).
- the genetically engineered bacterium is Bacillus, a probiotic bacterium that is highly genetically tractable and has been a popular chassis for industrial protein production; in some embodiments, the bacterium has highly active secretion and/or no toxic byproducts (Cutting, 2011).
- the genetically engineered bacteria are Escherichia coli strain Nissle 1917 (E. coli Nissle), a Gram-negative bacterium of the
- E. coli Nissle lacks prominent virulence factors (e.g., E. coli a-hemolysin, P-fimbrial adhesins) (Schultz, 2008). I n addition, it has been shown that E. coli Nissle does not carry pathogenic adhesion factors, does not produce any enterotoxins or cytotoxins, is not invasive, and is not uropathogenic.
- E. coli Nissle As early as in 1917, E. coli Nissle was packaged into medicinal capsules, called Mutaflor, for therapeutic use. E. coli Nissle has since been used to treat ulcerative colitis in humans in vivo (Rembacken et al., 1999), to treat inflammatory bowel disease, Crohn's disease, and pouchitis in humans in vivo (Schultz, 2008), and to inhibit enteroinvasive Salmonella, Legionella, Yersinia, and Shigella in vitro (Altenhoefer et al., 2004). It is commonly accepted that E. coli Nissle's therapeutic efficacy and safety have convincingly been proven (Ukena et al., 2007).
- the genetically engineered bacteria are E. coli Nissle and are naturally capable of promoting tight junctions and gut barrier function. I n some embodiments, the genetically engineered bacteria are E. coli and are highly amenable to recombinant protein technologies.
- genes from one or more different species of bacteria can be introduced into one another, e.g., the butyrogenic genes from Peptoclostridium difficile have been expressed in Escherichia coli (Aboulnaga et al., 2013).
- Unmodified E. coli Nissle and the genetically engineered bacteria of the invention may be destroyed, e.g., by defense factors in the gut or blood serum
- Residence time in vivo may be calculated for the genetically engineered bacteria. In some embodiments, the residence time is calculated for a human subject.
- the genetically engineered bacteria comprise one or more gene sequence(s) and/or gene cassette(s) for producing a non-native anti-inflammation and/or gut barrier function enhancer molecule.
- the genetically engineered bacteria comprise one or more gene sequence(s) for producing a non-native anti-inflammation and/or gut barrier function enhancer molecule.
- the genetically engineered bacteria may comprise two or more gene sequence(s) for producing a non-native anti-inflammation and/or gut barrier function enhancer molecule.
- the two or more gene sequences are multiple copies of the same gene.
- the two or more gene sequences are sequences encoding different genes.
- the two or more gene sequences are sequences encoding multiple copies of one or more different genes.
- the genetically engineered bacteria comprise one or more gene cassette(s) for producing a non-native anti-inflammation and/or gut barrier function enhancer molecule.
- the genetically engineered bacteria may comprise two or more gene cassette(s) for producing a non-native anti-inflammation and/or gut barrier function enhancer molecule.
- the two or more gene cassettes are multiple copies of the same gene cassette.
- the two or more gene cassettes are different gene cassettes for producing either the same or different anti- inflammation and/or gut barrier function enhancer molecule(s).
- the two or more gene cassettes are gene cassettes for producing multiple copies of one
- the anti-inflammation and/or gut barrier function enhancer molecule is selected from the group consisting of a short-chain fatty acid, butyrate, propionate, acetate, IL-2, IL-22, superoxide dismutase (SOD), GLP-2, GLP-1, IL-10, IL-27, TGF- ⁇ , TGF" 2, N-acylphosphatidylethanolamines (NAPEs), elafin (also known as peptidase inhibitor 3 or SKALP), trefoil factor, melatonin, PGD 2 , kynurenic acid, and kynurenine.
- a molecule may be primarily anti-inflammatory, e.g., IL-10, or primarily gut barrier function enhancing, e.g., GLP-2.
- a molecule may be both antiinflammatory and gut barrier function enhancing.
- the genetically engineered bacteria of the invention express one or more anti-inflammation and/or gut barrier function enhancer molecule(s) that is encoded by a single gene, e.g., the molecule is elafin and encoded by the PI3 gene, or the molecule is interleukin-10 and encoded by the IL10 gene.
- the genetically engineered bacteria of the invention encode one or more an anti-inflammation and/or gut barrier function enhancer molecule(s), e.g., butyrate, that is synthesized by a biosynthetic pathway requiring multiple genes.
- the one or more gene sequence(s) and/or gene cassette(s) may be expressed on a high-copy plasmid, a low-copy plasmid, or a chromosome.
- expression from the plasmid may be useful for increasing expression of the anti-inflammation and/or gut barrier function enhancer molecule(s).
- expression from the chromosome may be useful for increasing stability of expression of the anti-inflammation and/or gut barrier function enhancer molecule(s).
- the gene Sequence(s)or gene cassette(s) for producing the anti- inflammation and/or gut barrier function enhancer molecule(s) is integrated into the bacterial chromosome at one or more integration sites in the genetically engineered bacteria.
- one or more copies of the butyrate biosynthesis gene cassette may be integrated into the bacterial chromosome.
- the gene sequence(s) or gene cassette(s) for producing the anti-inflammation and/or gut barrier function enhancer molecule(s) is expressed from a plasmid in the genetically engineered bacteria.
- the gene sequence(s) or gene cassette(s) for producing the anti-inflammation and/or gut barrier function enhancer molecule(s) is expressed from a plasmid in the genetically engineered bacteria.
- the gene sequence(s) or gene cassette(s) for producing the anti-inflammation and/or gut barrier function enhancer molecule(s) is expressed from a plasmid in the genetically engineered bacteria.
- the gene sequence(s) or gene cassette(s) for producing the anti-inflammation and/or gut barrier function enhancer molecule(s) is expressed from a plasmid in the genetically engineered bacteria.
- the anti-inflammation and/or gut barrier function enhancer molecule(s) is inserted into the bacterial genome at one or more of the following insertion sites in E. coli Nissle: malE/K, araC/BAD, lacZ, thyA, malP/T. Any suitable insertion site may be used (see, e.g., Fig. 51 for exemplary insertion sites).
- the insertion site may be anywhere in the genome, e.g., in a gene required for survival and/or growth, such as thyA (to create an auxotroph); in an active area of the genome, such as near the site of genome replication; and/or in between divergent promoters in order to reduce the risk of unintended transcription, such as between AraB and AraC of the arabinose operon.
- a gene required for survival and/or growth such as thyA (to create an auxotroph)
- thyA to create an auxotroph
- an active area of the genome such as near the site of genome replication
- divergent promoters in order to reduce the risk of unintended transcription, such as between AraB and AraC of the arabinose operon.
- the genetically engineered bacteria of the invention comprise one or more butyrogenic gene cassette(s) and are capable of producing butyrate.
- the genetically engineered bacteria may include any suitable set of butyrogenic genes (see, e.g., Table 4). Unmodified bacteria comprising butyrate biosynthesis genes are known and include, but are not limited to, Peptoclostridium, Clostridium, Fusobacterium, Butyrivibrio, Eubacterium, and Treponema, and these endogenous butyrate biosynthesis pathways may be a source of genes for the genetically engineered bacteria of the invention.
- the genetically engineered bacteria of the invention comprise butyrate biosynthesis genes from a different species, strain, or substrain of bacteria.
- the genetically engineered bacteria comprise the eight genes of the butyrate biosynthesis pathway from
- Peptoclostridium difficile e.g., Peptoclostridium difficile strain 630: bcd2, etfB3, etfA3, thiAl, hbd, crt2, pbt, and buk (Aboulnaga et al., 2013), and are capable of producing butyrate under inducing conditions.
- Peptoclostridium difficile strain 630 and strain 1296 are both capable of producing butyrate, but comprise different nucleic acid sequences for etfA3, thiAl, hbd, crt2, pbt, and buk.
- the genetically engineered bacteria comprise a combination of butyrogenic genes from different species, strains, and/or substrains of bacteria, and are capable of producing butyrate under inducing conditions.
- the genetically engineered bacteria comprise bcd2, etfB3, etfA3, and thiAl from Peptoclostridium difficile strain 630, and hbd, crt2, pbt, and buk from Peptoclostridium difficile strain 1296.
- the gene products of the bcd2, etfA3, and etfB3 genes in Clostridium difficile form a complex that converts crotonyl-CoA to butyryl-CoA, which may function as an oxygen-dependent co-oxidant.
- the genetically engineered bacteria of the invention are designed to produce butyrate in a microaerobic or oxygen-limited environment, e.g., the mammalian gut, oxygen dependence could have a negative effect on butyrate production in the gut. It has been shown that a single gene from Treponema denticola [ter, encoding trans-2-enoynl-CoA reductase) can functionally replace this three-gene complex in an oxygen-independent manner.
- Treponema denticola encoding trans-2-enoynl-CoA reductase
- the genetically engineered bacteria comprise a ter gene, e.g., from
- Treponema denticola which can functionally replace all three of the bcd2, etfB3, and etfA3 genes, e.g., from Peptoclostridium difficile.
- the genetically engineered bacteria comprise thiAl, hbd, crt2, pbt, and buk, e.g., from Peptoclostridium difficile, and ter, e.g., from Treponema denticola, and are capable of producing butyrate in low-oxygen conditions (see, e.g., Table 4).
- the genetically engineered bacteria comprise genes for aerobic butyrate biosynthesis and/or genes for anaerobic or microaerobic butyrate biosynthesis.
- the genetically engineered bacteria of the invention comprise thiAl, hbd, crt2, pbt, and buk, e.g., from Peptoclostridium difficile; ter, e.g., from Treponema denticola; one or more of bcd2, etfB3, and etfA3, e.g., from Peptoclostridium difficile; and produce butyrate under inducing conditions.
- a butyrogenic gene cassette may comprise thiAl, hbd and crt2 from Peptoclostridium difficile, ter from Treponema denticola and tesBfrom E. coli.
- one or more of the butyrate biosynthesis genes may be functionally replaced or modified, e.g., codon optimized.
- the butyrogenic gene cassette comprises genes for the aerobic biosynthesis of butyrate and/or genes for the anaerobic or microaerobic biosynthesis of butyrate.
- one or more of the butyrate biosynthesis genes is functionally replaced, modified, and/or mutated in order to enhance stability and/or increase butyrate production in low-oxygen conditions.
- the local production of butyrate induces the
- the genetically engineered bacteria are capable of expressing the butyrate biosynthesis cassette and producing butyrate under inducing conditions.
- the genes may be codon-optimized, and translational and transcriptional elements may be added.
- Table 4 depicts the nucleic acid sequences of exemplary genes in the butyrate biosynthesis gene cassette.
- the genetically engineered bacteria comprise the nucleic acid sequence of any one of SEQ ID NOs: 1-10 or a functional fragment thereof.
- the genetically engineered bacteria comprise a nucleic acid sequence that, but for the redundancy of the genetic code, encodes the same
- the genetically engineered bacteria comprise a nucleic acid sequence that encodes a polypeptide of any one of SEQ I D NOs: 11-20 or a functional fragment thereof.
- genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to the DNA sequence of any one of SEQ I D NOs: 1-10 or a functional fragment thereof.
- genetically engineered bacteria comprise a nucleic acid that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to the nucleic acid sequence that encodes a polypeptide of any one of SEQ I D NOs: 11-20 or a functional fragment thereof.
- AAAC C T A T AAAAGG T GG T AC T TAT T CAG T AAG T GC T GC T AT GAT T G AAGAT T TAAAA G T G G GAG T T T TAG GAGAAC AC G C T T CAAAC C TAG G T G GAAT AAT AG C AAA
- inventions comprise a propionate gene cassette and are capable of producing propionate.
- the genetically engineered bacteria may express any suitable set of propionate
- propionate via an endogenous propionate biosynthesis pathway include, but are not limited to, Clostridium propionicum, Megasphaera elsdenii, and Prevotella
- the genetically engineered bacteria of the invention comprise propionate biosynthesis genes from a different species, strain, or substrain of bacteria.
- the genetically engineered bacteria comprise the genes pet, led, and aer from Clostridium propionicum.
- the genetically engineered bacteria comprise acrylate pathway genes for propionate biosynthesis, e.g., pet, IcdA, IcdB, IcdC, etfA, acrB, and acrC.
- the genetically engineered bacteria comprise pyruvate pathway genes for propionate biosynthesis, e.g., thrA fbr , thrB, thrC, ilvA fbr , oceE, aceF, and Ipd, and optionally further comprise tesB.
- the genes may be codon-optimized, and translational and transcriptional elements may be added. Table 6 depicts the nucleic acid sequences of exemplary genes in the propionate biosynthesis gene cassette.
- the genetically engineered bacteria comprise the nucleic acid sequence of any one of SEQ ID NOs: 21-34 and 10 or a functional fragment thereof. In some embodiments, the genetically engineered bacteria comprise a nucleic acid sequence that encodes a polypeptide of any one of SEQ ID NOs: 35-48 and 20 or a functional fragment thereof. In some embodiments, genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to the DNA sequence of any one of SEQ ID NOs: 21-34 and 10 or a functional fragment thereof. In some
- genetically engineered bacteria comprise a nucleic acid that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to the nucleic acid sequence that encodes a polypeptide of any one of SEQ ID NOs: 35-48 and 20 or a functional fragment thereof.
- SEQ ID NO: 48 TMEGKKAPAEPQRYDAVLVAIGRVPNGKNLDAGKAGVEVDDRGFIRVDKQ
- one or more of the propionate biosynthesis genes is a synthetic propionate biosynthesis gene. In some embodiments, one or more of the propionate biosynthesis genes is an E. coli propionate biosynthesis gene. In some
- one or more of the propionate biosynthesis genes is a C. glutamicum propionate biosynthesis gene. In some embodiments, one or more of the propionate biosynthesis genes is a C. propionicum propionate biosynthesis gene.
- the propionate gene cassette may comprise genes for the aerobic biosynthesis of propionate and/or genes for the anaerobic or microaerobic biosynthesis of propionate.
- One or more of the propionate biosynthesis genes may be functionally replaced or modified, e.g., codon optimized.
- the genetically engineered bacteria comprise a
- propionate biosynthesis genes is functionally replaced, modified, and/or mutated in order to enhance stability and/or increase propionate production under inducing conditions.
- the genetically engineered bacteria are capable of expressing the propionate biosynthesis cassette and producing propionate under inducing conditions.
- the genetically engineered bacteria of the invention comprise an acetate gene cassette and are capable of producing acetate.
- the genetically engineered bacteria may include any suitable set of acetate biosynthesis genes. Unmodified bacteria comprising acetate biosynthesis genes are known in the art and are capable of consuming various substrates to produce acetate under aerobic and/or anaerobic conditions (see, e.g., Ragsdale, 2008), and these endogenous acetate biosynthesis pathways may be a source of genes for the genetically engineered bacteria of the invention.
- the genetically engineered bacteria of the invention comprise acetate biosynthesis genes from a different species, strain, or substrain of bacteria.
- the native acetate biosynthesis genes in the genetically engineered bacteria are enhanced.
- the genetically engineered bacteria comprise aerobic acetate biosynthesis genes, e.g., from Escherichia coli.
- the genetically engineered bacteria comprise anaerobic acetate biosynthesis genes, e.g., from Acetitomaculum, Acetoanaerobium,
- the genetically engineered bacteria may comprise genes for aerobic acetate biosynthesis or genes for anaerobic or microaerobic acetate biosynthesis. In some embodiments, the genetically engineered bacteria comprise both aerobic and anaerobic or microaerobic acetate biosynthesis genes. In some embodiments, the genetically engineered bacteria comprise a combination of acetate biosynthesis genes from different species, strains, and/or substrains of bacteria, and are capable of producing acetate.
- one or more of the acetate biosynthesis genes is functionally replaced, modified, and/or mutated in order to enhance stability and/or acetate production.
- the genetically engineered bacteria are capable of expressing the acetate biosynthesis cassette and producing acetate under inducing conditions. In some embodiments, the genetically engineered bacteria are capable of producing an alternate short-chain fatty acid.
- the genetically engineered bacteria of the invention are capable of producing IL-10.
- IL-10 is a class 2 cytokine, a category which includes cytokines, interferons, and interferon-like molecules, such as IL-
- IL-10 is an anti-inflammatory cytokine that signals through two receptors, IL-10R1 and IL-10R2. Deficiencies in IL-10 and/or its receptors are associated with IBD and intestinal sensitivity (Nielsen, 2014). Bacteria expressing IL-10 or protease inhibitors may ameliorate conditions such as Crohn's disease and ulcerative colitis (Simpson et al., 2014).
- the genetically engineered bacteria may comprise any suitable gene encoding IL-10, e.g., human IL-10.
- the gene encoding IL-10 is modified and/or mutated, e.g., to enhance stability, increase IL-10 production, and/or increase anti-inflammatory potency under inducing conditions.
- the genetically engineered bacteria are capable of producing IL-10 under inducing conditions, e.g., under a condition(s) associated with inflammation.
- the genetically engineered bacteria are capable of producing IL-10 in low- oxygen conditions.
- the genetically engineered bacteria comprise a nucleic acid sequence that encodes IL-10.
- the genetically engineered bacteria comprise a nucleic acid sequence comprising SEQ ID NO: 49 or a functional fragment thereof.
- genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to a nucleic acid sequence comprising SEQ ID NO: 49 or a functional fragment thereof.
- IL-10 (SEQ ID NO: 49):
- the genetically engineered bacteria are capable of producing IL-2.
- Interleukin 2 (IL-2) mediates autoimmunity by preserving health of
- Treg cells including those expressing Foxp3, typically suppress effector T cells that are active against self-antigens, and in doing so, can dampen autoimmune activity.
- IL-2 functions as a cytokine to enhance Treg cell differentiation and activity while diminished IL-2 activity can promote autoimmunity events.
- IL-2 is generated by activated CD4+ T cells, and by other immune mediators including activated CD8+ T cells, activated dendritic cells, natural killer cells, and NK T cells.
- IL-2 binds to IL- 2R, which is composed of three chains including CD25, CD122, and CD132.
- IL-2 promotes growth of Treg cells in the thymus, while preserving their function and activity in systemic circulation. Treg cell activity plays an intricate role in the IBD setting, with murine studies suggesting a protective role in disease pathogenesis.
- IBD setting with murine studies suggesting a protective role in disease pathogenesis.
- the genetically engineered bacteria comprise a nucleic acid sequence encoding SEQ ID NO: 50 or a functional fragment thereof. In some embodiments, genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to a nucleic acid sequence encoding SEQ ID NO: 50 or a functional fragment thereof. In some embodiments, the genetically engineered bacteria are capable of producing IL-2 under inducing conditions, e.g., under a condition(s) associated with inflammation. In some embodiments, the genetically engineered bacteria are capable of producing IL-2 in low-oxygen conditions.
- the genetically engineered bacteria are capable of producing IL-22.
- Interleukin 22 (IL-22) cytokine can be produced by dendritic cells, lymphoid tissue inducer-like cells, natural killer cells and expressed on adaptive lymphocytes. Through initiation of Jak-STAT signaling pathways, IL-22 expression can trigger expression of antimicrobial compounds as well as a range of cell growth related pathways, both of which enhance tissue repair mechanisms. IL-22 is critical in promoting IL-22.
- Murine models have demonstrated improved intestinal inflammation states following
- the genetically engineered bacteria comprise a nucleic acid sequence encoding SEQ ID NO: 51 or a functional fragment thereof. In some embodiments, genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to a nucleic acid sequence encoding SEQ ID NO: 51 or a functional fragment thereof.
- the genetically engineered bacteria are capable of producing IL-22 under inducing conditions, e.g., under a condition(s) associated with inflammation. In some embodiments, the genetically engineered bacteria are capable of producing IL-22 in low- oxygen conditions.
- the genetically engineered bacteria are capable of producing IL-27.
- Interleukin 27 (IL-27) cytokine is predominately expressed by activated antigen presenting cells, while IL-27 receptor is found on a range of cells including T cells, NK cells, among others.
- IL-27 suppresses development of pro-inflammatory T helper 17 (Thl7) cells, which play a critical role in IBD pathogenesis.
- Thl7 pro-inflammatory T helper 17
- IL-27 can promote differentiation of IL-10 producing Trl cells and enhance IL-10 output, both of which have anti-inflammatory effects.
- IL-27 has protective effects on epithelial barrier function via activation of MAPK and STAT signaling within intestinal epithelial cells.
- IL-27 enhances production of antibacterial proteins that curb bacterial growth. Improvement in barrier function and reduction in bacterial growth suggest a favorable role for IL-27 in IBD pathogenesis.
- -70- engineered bacteria comprise a nucleic acid sequence encoding SEQ ID NO: 52 or a functional fragment thereof.
- genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to a nucleic acid sequence encoding SEQ ID NO: 52 or a functional fragment thereof.
- the genetically engineered bacteria are capable of producing IL-27 under inducing conditions, e.g., under a condition(s) associated with inflammation. In some embodiments, the genetically engineered bacteria are capable of producing IL-27 in low- oxygen conditions.
- the genetically engineered bacteria of the invention are capable of producing SOD. Increased ROS levels contribute to
- VCAM-1 vascular cell adhesion molecule 1
- SOD superoxide dismutase
- the genetically engineered bacteria comprise a nucleic acid sequence encoding SEQ ID NO: 52 or a functional fragment thereof. In some embodiments, genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least
- the genetically engineered bacteria are capable of producing SOD under inducing conditions, e.g., under a condition(s) associated with inflammation. In some embodiments, the genetically engineered bacteria are capable of producing SOD in low- oxygen conditions.
- the genetically engineered bacteria are capable of producing GLP-2 or proglucagon.
- Glucagon-like peptide 2 (GLP-2) is produced by intestinal endocrine cells and stimulates intestinal growth and enhances gut barrier function. GLP-2 administration has therapeutic potential in treating IBD, short bowel syndrome, and small bowel enteritis (Yazbeck et al., 2009).
- the genetically engineered bacteria may comprise any suitable gene encoding GLP-2 or proglucagon, e.g., human GLP-2 or proglucagon.
- a protease inhibitor e.g., an inhibitor of dipeptidyl peptidase, is also administered to decrease GLP-2 degradation.
- the genetically engineered bacteria express a degradation resistant GLP-2 analog, e.g., Teduglutide (Yazbeck et al., 2009).
- the gene encoding GLP-2 or proglucagon is modified and/or mutated, e.g., to enhance stability, increase GLP-2 production, and/or increase gut barrier enhancing potency under inducing conditions.
- the genetically engineered bacteria of the invention are capable of producing GLP-2 or proglucagon under inducing conditions.
- GLP-2 administration in a murine model of IBD is associated with reduced mucosal damage and inflammation, as well as a reduction in inflammatory mediators, such as TNF-a and IFN-y.
- the genetically engineered bacteria comprise a nucleic acid sequence encoding SEQ ID NO: 54 or a
- genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to a nucleic acid sequence encoding SEQ ID NO: 54 or a functional fragment thereof.
- the genetically engineered bacteria are capable of producing GLP-2 under inducing conditions, e.g., under a condition(s) associated with inflammation. In some embodiments, the genetically engineered bacteria are capable of producing GLP-2 in low- oxygen conditions.
- the genetically engineered bacteria are capable of producing kynurenine.
- Kynurenine is a metabolite produced in the first, rate-limiting step of tryptophan catabolism. This step involves the conversion of tryptophan to kynurenine, and may be catalyzed by the ubiquitously-expressed enzyme indoleamine 2,3-dioxygenase (IDO-1), or by tryptophan dioxygenase (TDO), an enzyme which is primarily localized to the liver (Alvarado et al., 2015).
- IDO-1 ubiquitously-expressed enzyme indoleamine 2,3-dioxygenase
- TDO tryptophan dioxygenase
- Biopsies from human patients with IBD show elevated levels of IDO-1 expression compared to biopsies from healthy individuals, particularly near sites of ulceration (Ferdinande et al., 2008; Wolf et al., 2004).
- IDO-1 enzyme expression is similarly upregulated in trinitrobenzene sulfonic acid- and dextran sodium sulfate-induced mouse models of IBD; inhibition of IDO-1
- the genetically engineered bacteria may comprise any suitable gene for producing
- the genetically engineered bacteria may comprise a gene or gene cassette for producing a tryptophan transporter, a gene or gene cassette for producing IDO-1, and a gene or gene cassette for producing TDO.
- the gene for producing kynurenine is modified and/or mutated, e.g., to enhance stability, increase kynurenine production, and/or increase anti-inflammatory
- the engineered bacteria have enhanced uptake or import of tryptophan, e.g., comprise a transporter or other mechanism for increasing the uptake of tryptophan into the bacterial cell.
- the genetically engineered bacteria are capable of producing kynurenine under inducing conditions, e.g., under a condition(s) associated with inflammation. In some embodiments, the genetically engineered bacteria are capable of producing kynurenine in low-oxygen conditions.
- the genetically engineered bacteria are capable of producing kynurenic acid.
- Kynurenic acid is produced from the irreversible
- Kynurenic acid acts as an antagonist of ionotropic glutamate receptors (Turski et al., 2013). While glutamate is known to be a major excitatory neurotransmitter in the central nervous system, there is now evidence to suggest an additional role for glutamate in the peripheral nervous system.
- the genetically engineered bacteria may comprise any suitable gene for producing kynurenic acid.
- the gene for producing kynurenic acid is modified and/or mutated, e.g., to enhance stability, increase kynurenic acid production, and/or increase anti-inflammatory potency under inducing conditions.
- the genetically engineered bacteria are capable of producing kynurenic acid under inducing conditions, e.g., under a condition(s) associated with inflammation.
- the genetically engineered bacteria are capable of producing kynurenic acid in low-oxygen conditions.
- the genetically engineered bacteria are capable of producing IL-19, IL-20, and/or IL-24. In some embodiments, the genetically engineered bacteria are capable of producing IL-19, IL-20, and/or IL-24 under inducing conditions,
- the genetically engineered bacteria are capable of producing I L-19, IL-20 and/or I L-24 in low- oxygen conditions.
- the genetically engineered bacteria of the invention are capable of producing a molecule that is capable of inhibiting a proinflammatory molecule.
- the genetically engineered bacteria may express any suitable inhibitory molecule, e.g., a single-chain variable fragment (scFv), antisense RNA, siRNA, or shRNA, that is capable of neutralizing one or more pro-inflammatory molecules, e.g., TN F, I FN-Y, IL- ⁇ , I L-6, I L-8, I L-17, IL-18, I L-21, IL-23, IL-26, I L-32, Arachidonic acid, prostaglandins (e.g., PGE 2 ), PGI 2 , serotonin, thromboxanes (e.g., TXA 2 ), leukotrienes (e.g., LTB 4 ), hepoxillin A 3 , or chemokines (Keates et al., 2008; Ahmad e
- scFv single
- the genetically engineered bacteria may inhibit one or more pro-inflammatory molecules, e.g., TNF, IL-17.
- the genetically engineered bacteria are capable of modulating one or more molecule(s) shown in Table 8. I n some embodiments, the genetically engineered bacteria are capable of inhibiting, removing, degrading, and/or metabolizing one or more inflammatory molecules.
- Urinary 4- hydroxybenzoic acid, 4 hydroxyphenylacetate, 4-cresol, and hydroxybenzoic acid, phenylacetate are elevated in colorectal 3hydroxyphenylpropionate, 4- cancer.
- Urinary 4-cresyl sulfate is hydroxyphenylpropionate, 3- elevated in children with severe autism. hydroxycinnamate, 4- methylphenol, tyrosine,
- Indole derivatives N- Clostridium Protect against stress-induced lesions in acetyltryptophan, indoleacetate, sporogenes, E. coli the Gl tract; modulate expression of indoleacetylglycine (IAG), indole, proinflammatory genes, increase indoxyl sulfate, indoles- expression of anti-inflammatory genes, propionate, melatonin, melatonin strengthen epithelial cell barrier 6-sulfate, serotonin, 5- properties. Implicated in Gl pathologies, hydroxyindole brain-gut axis, and a few neurological conditions.
- IAG indoleacetylglycine
- Vitamins vitamin K, vitamin B12, Bifidobacterium Provide complementary endogenous biotin, folate, sources of vitamins, strengthen immune thiamine, riboflavin, pyridoxine function, exert epigenetic effects to regulate cell proliferation.
- Polyamines putrescine, Campylobacter Exert genotoxic effects on the host, anticadaverine, jejuni, inflammatory and antitumoral effects. spermidine, spermine Clostridium Potential tumor markers.
- Lipids conjugated fatty acids, LPS, Bifidobacterium, Impact intestinal permeability, activate peptidoglycan, acylglycerols, oseburia, intestinebrain- liver neural axis to sphingomyelin, cholesterol, Lactobacillus, regulate glucose homeostasis; LPS phosphatidylcholines, Klebsiella, induces chronic systemic inflammation; phosphoethanolamines, Enterobacter, conjugated fatty acids improve triglycerides Citrobacter, hyperinsulinemia, enhance the immune
- lysine glucose, urea, a- Ruminococcus, compounds or modulation of linked ketoisovalerate, creatine, Faecalibacterium pathways including endocannabinoid creatinine, endocannabinoids, 2- system.
- the genetically engineered bacteria are capable of producing an anti-inflammation and/or gut barrier enhancer molecule and further
- the genetically engineered bacteria of the invention are capable of
- the genetically engineered bacteria of the invention are capable of expressing a gene cassette for producing butyrate, as well as a molecule or biosynthetic pathway for inhibiting, removing, degrading, and/or metabolizing an inflammatory molecule, e.g., PGE 2 .
- RNA interference is a post-transcriptional gene silencing mechanism in plants and animals. RNAi is activated when microRNA (miRNA), double- stranded RNA (dsRNA), or short hairpin RNA (shRNA) is processed into short interfering RNA (siRNA) duplexes (Keates et al., 2008). RNAi can be "activated in vitro and in vivo by non-pathogenic bacteria engineered to manufacture and deliver shRNA to target cells" such as mammalian cells (Keates et al., 2008). I n some embodiments, the genetically engineered bacteria of the invention induce RNAi-mediated gene silencing of one or more pro-inflammatory molecules in low-oxygen conditions. I n some embodiments, the genetically engineered bacteria produce siRNA targeting TNF in low-oxygen conditions.
- Single-chain variable fragments are "widely used antibody fragments... produced in prokaryotes" (Frenzel et al., 2013).
- scFv lacks the constant domain of a traditional antibody and expresses the antigen-binding domain as a single peptide.
- Bacteria such as Escherichia coli are capable of producing scFv that target proinflammatory cytokines, e.g., TNF (H ristodorov et al., 2014).
- the genetically engineered bacteria of the invention express a binding protein for neutralizing one or more pro-inflammatory molecules in low-oxygen conditions. I n some
- the genetically engineered bacteria produce scFv targeting TNF in low- oxygen conditions. In some embodiments, the genetically engineered bacteria produce both scFv and siRNA targeting one or more pro-inflammatory molecules in low-oxygen conditions (see, e.g., Xiao et al., 2014).
- genes and gene cassettes capable of producing anti-inflammation and/or gut barrier function enhancer molecules are known in the art and may be expressed by the genetically engineered bacteria of the invention.
- the gene or gene cassette for producing a therapeutic molecule also comprises additional transcription and translation
- -77- elements e.g., a ribosome binding site, to enhance expression of the therapeutic molecule.
- the genetically engineered bacteria produce two or more anti-inflammation and/or gut barrier function enhancer molecules. In certain embodiments, the two or more molecules behave synergistically to reduce gut inflammation and/or enhance gut barrier function. In some embodiments, the genetically engineered bacteria express at least one anti-inflammation molecule and at least one gut barrier function enhancer molecule. In certain embodiments, the genetically engineered bacteria express IL-10 and GLP-2. In alternate embodiments, the genetically engineered bacteria express IL-10 and butyrate.
- the genetically engineered bacteria are capable of producing IL-2, IL-10, IL-22, IL-27, propionate, and butyrate. In some embodiments, the genetically engineered bacteria are capable of producing IL-10, IL-27, GLP-2, and butyrate. In some embodiments, the genetically engineered bacteria are capable of producing GLP-2, IL-10, IL-22, SOD, butyrate, and propionate. In some embodiments, the genetically engineered bacteria are capable of GLP-2, IL-2, IL-10, IL-22, IL-27, SOD, butyrate, and propionate. Any suitable combination of therapeutic molecules may be produced by the genetically engineered bacteria.
- the genetically engineered bacteria of the invention comprise a promoter that is directly or indirectly induced by exogenous environmental conditions.
- a gene or gene cassette for producing an anti-inflammation and/or gut barrier function enhancer molecule is operably linked to an oxygen level-dependent promoter or regulatory region comprising said promoter.
- the gene or gene cassette is operably linked to an oxygen level-dependent promoter such that the therapeutic molecule is expressed in low-oxygen, microaerobic, or anaerobic conditions. For example, in low-oxygen conditions, the oxygen level-dependent promoter is activated by a corresponding oxygen level-sensing transcription factor, thereby driving production of the therapeutic molecule.
- the genetically engineered bacteria comprise a gene or a gene cassette for producing an anti-inflammation and/or gut barrier function enhancer molecule expressed under the control of a fumarate and nitrate reductase regulator (FN R)-responsive promoter, an anaerobic regulation of arginine deiminiase and nitrate reduction (AN R)-responsive promoter, or a dissimilatory nitrate respiration regulator (DN R)-responsive promoter, which are capable of being regulated by the transcription factors FNR, AN R, or DN R, respectively.
- FN R fumarate and nitrate reductase regulator
- AN R an anaerobic regulation of arginine deiminiase and nitrate reduction
- DN R dissimilatory nitrate respiration regulator
- the genetically engineered bacteria comprise a FNR-responsive promoter.
- FNR is a major transcriptional activator that controls the switch from aerobic to anaerobic metabolism (Unden et al., 1997).
- I n the anaerobic state FNR dimerizes into an active DNA binding protein that activates hundreds of genes responsible for adapting to anaerobic growth.
- I n the aerobic state FN R is prevented from dimerizing by oxygen and is inactive.
- multiple distinct FNR nucleic acid sequences are inserted in the genetically engineered bacteria.
- the promoter is an alternate oxygen level-dependent promoter, e.g., DNR (Trunk et al., 2010) or ANR (Ray et al., 1997).
- DNR Truenk et al., 2010
- ANR Ray et al., 1997
- aeruginosa the anaerobic regulation of AN R is "required for the expression of physiological functions which are inducible under oxygen-limiting or anaerobic conditions" (Sawers, 1991; Winteler et al., 1996).
- P. aeruginosa AN R is homologous with E. coli FNR, and "the consensus FN R site (TTGAT-— ATCAA) was recognized efficiently by ANR and FN R" (Winteler et al., 1996).
- FNR consensus FN R site
- AN R activates numerous genes responsible for adapting to anaerobic growth. I n the aerobic state, AN R is inactive.
- Pseudomonas fluorescens, Pseudomonas putida, Pseudomonas syringae, and Pseudomonas mendocina all have functional analogs of AN R (Zimmermann et al., 1991).
- Promoters that are regulated by ANR are known in the art, e.g., the promoter of the arcDABC operon (see, e.g., Hasegawa et al., 1998).
- the FNR family also includes the dissimilatory nitrate respiration regulator (DNR) (Arai et al., 1995), a transcription factor which is required in conjunction with ANR for "anaerobic nitrate respiration of Pseudomonas aeruginosa" (Hasegawa et al., 1998).
- DNR dissimilatory nitrate respiration regulator
- DN R DN R
- gene expression is further optimized by methods known in the art, e.g., by optimizing ribosomal binding sites and/or increasing mRNA stability.
- FNR promoter sequences are known in the art, and any suitable FNR promoter sequence(s) may be used in the genetically engineered bacteria of the invention. Any suitable FNR promoter(s) may be combined with any suitable gene or gene cassette for producing an anti-inflammation and/or gut barrier function enhancer molecule. Non-limiting FNR promoter sequences are provided in Table 9.
- the genetically engineered bacteria of the invention comprise one or more of: SEQ ID NO: 55, SEQ ID NO: 56, nirBl promoter (SEQ ID NO: 57), nirB2 promoter (SEQ ID NO: 58), nirB3 promoter (SEQ ID NO: 59), ydfZ promoter (SEQ ID NO: 60), nirB promoter fused to a strong ribosome binding site (SEQ ID NO: 61), ydfZ promoter fused to a strong ribosome binding site (SEQ ID NO: 62), fnrS, an anaerobically induced small RNA gene (fnrSl promoter SEQ ID NO: 63 or fnrS2 promoter SEQ ID NO: 64), nirB promoter fused to a crp binding site (SEQ ID NO: 65), and /nrS fused to a crp binding site (SEQ ID NO: 66).
- genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to the DNA sequence of SEQ ID NO: 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, or 66, or a functional fragment thereof.
- nirBl ATAAATCCGTTCAATTTGTCTGTTTTTTGCACAAACATGAAATATCAGAC SEQ ID NO: 57 AATTCCGTGACTTAAGAAAATTTATACAAATCAGCAATATACCCCTTAAG
- nirB+crp CCCGCTCATTTTGTCTATTTTTTGCACAAACATGAAATATCAGACAATTC SEQ ID NO: 65 CGTGACTTAAGAAAATTTATACAAATCAGCAATATACCCATTAAGGAGTA
- the gene or gene cassette for producing an anti- inflammation and/or gut barrier function enhancer molecule is expressed under the control of an oxygen level-dependent promoter fused to a binding site for a
- CRP transcriptional activator
- CRP cyclic AMP receptor protein or catabolite
- activator protein or CAP plays a major regulatory role in bacteria by repressing genes responsible for the uptake, metabolism, and assimilation of less favorable carbon sources when rapidly metabolizable carbohydrates, such as glucose, are present (Wu et al.,
- the gene or gene cassette for producing an anti-inflammation and/or gut barrier function enhancer molecule is controlled by an oxygen level-dependent promoter fused to a CRP binding site.
- the gene or gene cassette for producing an anti-inflammation and/or gut barrier function enhancer molecule is controlled by an oxygen level-dependent promoter fused to a CRP binding site.
- cyclic AMP binds to CRP when no glucose is present in the environment. This binding causes a conformational change in CRP, and allows CRP to bind tightly to its binding site. CRP binding then activates transcription of the gene or gene cassette by recruiting RNA polymerase to the FNR promoter via direct protein-protein interactions. In the presence of glucose, cyclic AMP does not bind to CRP and transcription of the gene or gene cassette for producing an anti-inflammation and/or gut barrier function enhancer molecule is repressed.
- an oxygen level-dependent promoter e.g., an FNR promoter fused to a binding site for a transcriptional activator is used to ensure that the gene or gene cassette for producing an anti-inflammation and/or gut barrier function enhancer molecule is not expressed under anaerobic conditions when sufficient amounts of glucose are present, e.g., by adding glucose to growth media in vitro.
- the genetically engineered bacteria comprise an oxygen level-dependent promoter from a different species, strain, or substrain of bacteria.
- the genetically engineered bacteria comprise an oxygen level-sensing transcription factor, e.g., FN R, ANR or DN R, from a different species, strain, or substrain of bacteria.
- the genetically engineered bacteria comprise an oxygen level-sensing transcription factor and corresponding promoter from a different species, strain, or substrain of bacteria.
- the heterologous oxygen level- dependent transcription factor and/or promoter may increase the production of the anti- inflammation and/or gut barrier enhancer molecule in low-oxygen conditions, as compared to the native transcription factor and promoter in the bacteria under the same conditions.
- the non-native oxygen level-dependent transcription factor and/or promoter may increase the production of the anti- inflammation and/or gut barrier enhancer molecule in low-oxygen conditions, as compared to the native transcription factor and promoter in the bacteria under the same conditions.
- the non-native oxygen level-dependent transcription factor and/or promoter may increase the production of the anti- inflammation and/or gut barrier enhancer molecule in low-oxygen conditions, as compared to the native transcription factor and promoter in the bacteria under the same conditions.
- transcription factor is a FNR protein from N. gonorrhoeae (see, e.g., Isabella et al., 2011).
- the corresponding wild-type transcription factor is deleted or mutated to reduce or eliminate wild-type activity.
- the corresponding wild-type transcription factor is left intact and retains wild-type activity.
- the heterologous transcription factor minimizes or eliminates off-
- the genetically engineered bacteria comprise a wild-type gene encoding an oxygen level-dependent transcription factor, such as FNR, ANR or DNR, and a corresponding promoter that is mutated relative to the wild-type promoter from bacteria of the same subtype.
- the mutated promoter increases the production of an anti-inflammation and/or gut barrier enhancer molecule in low-oxygen conditions, as compared to the wild-type promoter under the same conditions.
- the genetically engineered bacteria comprise a wild-type oxygen level- dependent promoter, e.g., a FN R-, AN R- or DNR-responsive promoter, and a
- the mutant transcription factor increases the expression of the anti-inflammation and/or gut barrier enhancer molecule in low-oxygen conditions, as compared to the wild-type transcription factor under the same conditions.
- the mutant oxygen level-dependent transcription factor is a FN R protein comprising amino acid substitutions that enhance dimerization and FNR activity (see, e.g., Moore et al., 2006).
- both the oxygen level-sensing transcription factor and corresponding promoter are mutated relative to the wild-type sequences from bacteria of the same subtype in order to increase expression of the anti- inflammation and/or gut barrier enhancer molecule in low-oxygen conditions.
- the genetically engineered bacteria of the invention comprise a gene encoding an oxygen level-sensing transcription factor, e.g., FNR, AN R or DNR, that is controlled by its native promoter, an inducible promoter, a promoter that is stronger than the native promoter, e.g., a GlnRS promoter, a P(Bla) promoter, or a constitutive promoter.
- an oxygen level-sensing transcription factor e.g., FNR, AN R or DNR
- an inducible promoter e.g., a promoter that is stronger than the native promoter, e.g., a GlnRS promoter, a P(Bla) promoter, or a constitutive promoter.
- expression of the oxygen level-dependent transcription factor is controlled by a different promoter than the promoter that controls expression of the therapeutic molecule
- the oxygen level-dependent transcription factor and therapeutic molecule are divergently transcribed from a promoter region.
- the genetically engineered bacteria of the invention comprise multiple copies of the endogenous gene encoding the oxygen level- sensing transcription factor, e.g., the /nr gene.
- the gene encoding the oxygen level-sensing transcription factor is present on a plasmid.
- the gene encoding the oxygen level-sensing transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on different plasmids. In some embodiments, the gene encoding the oxygen level-sensing
- the transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on the same plasmid.
- the gene encoding the oxygen level-sensing transcription factor is present on a chromosome.
- the gene encoding the oxygen level-sensing transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on different chromosomes.
- the gene encoding the oxygen level-sensing transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on the same chromosome.
- the gene or gene cassette for producing the anti- inflammation and/or gut barrier function enhancer molecule is present on a plasmid and operably linked to a promoter that is induced by low-oxygen conditions. In some embodiments, the gene or gene cassette for producing the anti-inflammation and/or gut barrier function enhancer molecule is present in the chromosome and operably linked to a promoter that is induced by low-oxygen conditions. In some embodiments, the gene or gene cassette for producing the anti-inflammation and/or gut barrier function enhancer molecule is present on a chromosome and operably linked to a promoter that is induced by exposure to tetracycline.
- the gene or gene cassette for producing the anti-inflammation and/or gut barrier function enhancer molecule is present on a plasmid and operably linked to a promoter that is induced by exposure to tetracycline.
- expression is further optimized by methods known
- the genetically engineered bacteria comprise a stably maintained plasmid or chromosome carrying the gene(s) or gene cassette(s) capable of producing an anti-inflammation and/or gut barrier function enhancer molecule, such that the gene(s) or gene cassette(s) can be expressed in the host cell, and the host cell is capable of survival and/or growth in vitro, e.g., in medium, and/or in vivo, e.g., in the gut.
- a bacterium may comprise multiple copies of the gene or gene cassette for producing the anti-inflammation and/or gut barrier function enhance molecule.
- the gene or gene cassette is expressed on a low-copy plasmid.
- the low-copy plasmid may be useful for increasing stability of expression. In some embodiments, the low-copy plasmid may be useful for decreasing leaky expression under non-inducing conditions. In some embodiments, the gene or gene cassette is expressed on a high-copy plasmid. In some embodiments, the high-copy plasmid may be useful for increasing gene or gene cassette expression. In some embodiments, gene or gene cassette is expressed on a
- the genetically engineered bacteria may comprise multiple copies of the gene(s) or gene cassette(s) capable of producing an anti- inflammation and/or gut barrier function enhancer molecule.
- the gene(s) or gene cassette(s) capable of producing an anti-inflammation and/or gut barrier function enhancer molecule is present on a plasmid and operably linked to an oxygen level-dependent promoter.
- the gene(s) or gene cassette(s) capable of producing an anti-inflammation and/or gut barrier function enhancer molecule is present in a chromosome and operably linked to an oxygen level-dependent promoter.
- the genetically engineered bacteria of the invention produce at least one anti-inflammation and/or gut barrier enhancer molecule in low-oxygen conditions to reduce local gut inflammation by at least about 1.5-fold, at least about 2-fold, at least about 10-fold, at least about 15-fold, at least about 20-fold, at
- I nflammation may be measured by methods known in the art, e.g., counting disease lesions using endoscopy; detecting T regulatory cell differentiation in peripheral blood, e.g., by fluorescence activated sorting; measuring T regulatory cell levels; measuring cytokine levels;
- inflammatory biomarkers e.g., by qPCR; PCR arrays; transcription factor phosphorylation assays; immunoassays; and/or cytokine assay kits (Mesoscale, Cayman Chemical, Qiagen).
- the genetically engineered bacteria produce at least about 1.5-fold, at least about 2-fold, at least about 10-fold, at least about 15-fold, at least about 20-fold, at least about 30-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 300-fold, at least about 400-fold, at least about 500-fold, at least about 600-fold, at least about 700-fold, at least about 800-fold, at least about 900-fold, at least about 1,000-fold, or at least about 1,500-fold more of an anti- inflammation and/or gut barrier enhancer molecule in low-oxygen conditions than unmodified bacteria of the same subtype under the same conditions.
- Certain unmodified bacteria will not have detectable levels of the anti-inflammation and/or gut barrier enhancer molecule. I n embodiments using genetically modified forms of these bacteria, the anti-inflammation and/or gut barrier enhancer molecule will be detectable in low- oxygen conditions.
- the anti-inflammation and/or gut barrier enhancer molecule is butyrate.
- Methods of measuring butyrate levels e.g., by mass spectrometry, gas chromatography, high-performance liquid chromatography (HPLC), are known in the art (see, e.g., Aboulnaga et al., 2013).
- butyrate is measured as butyrate level/bacteria optical density (OD).
- OD optical density
- butyrogenic gene cassette serves as a proxy measurement for butyrate production.
- the bacterial cells of the invention are harvested and lysed to measure butyrate production. I n alternate embodiments, butyrate production is measured in the bacterial cell medium.
- the genetically engineered bacteria produce at least about 1 nM/OD, at least about 10 nM/OD, at least about 100 nM/OD, at least about 500 nM/OD, at least about 1 ⁇ /OD, at least about 10 ⁇ /OD, at least about 100 ⁇ /OD, at least about 500 ⁇ /OD, at least about 1 mM/OD, at least about 2 mM/OD, at least about 3 mM/OD, at least about 5 mM/OD, at least about 10 mM/OD, at least about 20 mM/OD, at least about 30 mM/OD, or at least about 50 mM/OD of butyrate in low-oxygen conditions.
- the anti-inflammation and/or gut barrier enhancer molecule is propionate.
- Methods of measuring propionate levels e.g., by mass spectrometry, gas chromatography, high-performance liquid chromatography (HPLC), are known in the art (see, e.g., Hillman, 1978; Lukovac et al., 2014).
- measuring the activity and/or expression of one or more gene products in the propionate gene cassette serves as a proxy measurement for propionate production.
- the bacterial cells of the invention are harvested and lysed to measure propionate production.
- propionate production is measured in the bacterial cell medium.
- the genetically engineered bacteria produce at least about 1 ⁇ , at least about 10 ⁇ , at least about 100 ⁇ , at least about 500 ⁇ , at least about 1 mM, at least about 2 m M, at least about 3 mM, at least about 5 mM, at least about 10 mM, at least about 15 mM, at least about 20 mM, at least about 30 mM, at least about 40 mM, or at least about 50 mM of propionate in low-oxygen conditions.
- the genetically engineered bacteria of the invention comprise a tunable regulatory region that is directly or indirectly controlled by a transcription factor that is capable of sensing at least one reactive nitrogen species.
- the tunable regulatory region is operatively linked to a gene or gene cassette capable of directly or indirectly driving the expression of an anti-inflammation and/or gut barrier function enhancer molecule, thus controlling expression of the molecule relative to RNS
- the tunable regulatory region is a RNS-inducible regulatory region, and the molecule is butyrate; when RNS is present, e.g., in an inflamed tissue, a RNS- sensing transcription factor binds to and/or activates the regulatory region and drives expression of the butyrogenic gene cassette, thereby producing butyrate, which exerts anti-inflammation and/or gut barrier enhancing effects. Subsequently, when RNS is present, e.g., in an inflamed tissue, a RNS- sensing transcription factor binds to and/or activates the regulatory region and drives expression of the butyrogenic gene cassette, thereby producing butyrate, which exerts anti-inflammation and/or gut barrier enhancing effects. Subsequently, when RNS is present, e.g., in an inflamed tissue, a RNS- sensing transcription factor binds to and/or activates the regulatory region and drives expression of the butyrogenic gene cassette, thereby producing butyrate, which
- inflammation is ameliorated, RNS levels are reduced, and butyrate production is decreased or eliminated.
- the tunable regulatory region is a RNS-inducible regulatory region; in the presence of RNS, a transcription factor senses RNS and activates the RNS-inducible regulatory region, thereby driving expression of an operatively linked gene or gene cassette.
- the transcription factor senses RNS and subsequently binds to the RNS-inducible regulatory region, thereby activating downstream gene expression.
- the transcription factor is bound to the RNS-inducible regulatory region in the absence of RNS; when the transcription factor senses RNS, it undergoes a conformational change, thereby inducing downstream gene expression.
- the tunable regulatory region is a RNS-inducible regulatory region
- the transcription factor that senses RNS is NorR.
- NorR is an NO- responsive transcriptional activator that regulates expression of the nor VW genes encoding flavorubredoxin and an associated flavoprotein, which reduce NO to nitrous oxide.
- the genetically engineered bacteria of the invention may comprise any suitable RNS-responsive regulatory region from a gene that is activated by NorR. Genes that are capable of being activated by NorR are known in the art (see, e.g., Spiro, 2006; Vine et al., 2011; Karlinsey et al., 2012; Table 1).
- the genetically engineered bacteria of the invention comprise a RNS-inducible regulatory region from norVW that is operatively linked to a gene or gene cassette, e.g., a butyrogenic gene cassette.
- a NorR transcription factor senses RNS and activates to the norVW regulatory region, thereby driving expression of the operatively linked butyrogenic gene cassette and producing butyrate.
- the tunable regulatory region is a RNS-inducible regulatory region
- the transcription factor that senses RNS is DNR.
- the genetically engineered bacteria of the invention may comprise any suitable RNS- responsive regulatory region from a gene that is activated by DNR. Genes that are capable of being activated by DNR are known in the art (see, e.g., Castiglione et al., 2009; Giardina et al., 2008).
- the genetically engineered bacteria of the invention comprise a RNS-inducible regulatory region from norCB that is operatively linked to a gene or gene cassette, e.g., a butyrogenic gene cassette.
- a DN R transcription factor senses RNS and activates to the norCB regulatory region, thereby driving expression of the operatively linked butyrogenic gene cassette and producing butyrate.
- the DNR is Pseudomonas aeruginosa DNR.
- the tunable regulatory region is a RNS- derepressible regulatory region, and binding of a corresponding transcription factor represses downstream gene expression; in the presence of RNS, the transcription factor no longer binds to the regulatory region, thereby derepressing the operatively linked gene or gene cassette.
- the tunable regulatory region is a RNS- derepressible regulatory region, and the transcription factor that senses RNS is NsrR.
- NsrR is "an Rrf2-type transcriptional repressor [that] can sense NO and control the expression of genes responsible for NO metabolism" (Isabella et al., 2009).
- the genetically engineered bacteria of the invention may comprise any suitable RNS- responsive regulatory region from a gene that is repressed by NsrR.
- the NsrR is Neisseria gonorrhoeae NsrR. Genes that are capable of being repressed by NsrR are known in the art (see, e.g., Isabella et al., 2009; Dunn et al., 2010; Table 1).
- the genetically engineered bacteria of the invention comprise a RNS-derepressible regulatory region from norB that is operatively linked to a gene or gene cassette, e.g., a butyrogenic gene cassette. In the presence of RNS, an NsrR transcription factor senses RNS and no longer binds to the norB regulatory region,
- the genetically engineered bacteria it is advantageous for the genetically engineered bacteria to express a RNS-sensing transcription factor that does not regulate the expression of a significant number of native genes in the bacteria.
- the genetically engineered bacterium of the invention expresses a RNS- sensing transcription factor from a different species, strain, or substrain of bacteria, wherein the transcription factor does not bind to regulatory sequences in the genetically engineered bacterium of the invention.
- the genetically engineered bacterium of the invention is Escherichia coli
- the RNS-sensing transcription factor is NsrR, e.g., from is Neisseria gonorrhoeae, wherein the Escherichia coli does not comprise binding sites for said NsrR.
- the genetically engineered bacterium of the invention is Escherichia coli
- the RNS-sensing transcription factor is NsrR, e.g., from is Neisseria gonorrhoeae, wherein the Escherichia coli does not comprise binding sites for said NsrR.
- the genetically engineered bacterium of the invention is Escherichia coli
- heterologous transcription factor minimizes or eliminates off-target effects on endogenous regulatory regions and genes in the genetically engineered bacteria.
- the tunable regulatory region is a RNS-repressible regulatory region, and binding of a corresponding transcription factor represses downstream gene expression; in the presence of RNS, the transcription factor senses RNS and binds to the RNS-repressible regulatory region, thereby repressing expression of the operatively linked gene or gene cassette.
- the RNS-sensing transcription factor is capable of binding to a regulatory region that overlaps with part of the promoter sequence.
- the RNS-sensing transcription factor is capable of binding to a regulatory region that is upstream or downstream of the promoter sequence.
- the genetically engineered bacteria may comprise a two repressor activation regulatory circuit, which is used to express an anti- inflammation and/or gut barrier function enhancer molecule.
- the two repressor activation regulatory circuit comprises a first RNS-sensing repressor and a second repressor, which is operatively linked to a gene or gene cassette, e.g., a butyrogenic gene cassette.
- the RNS-sensing repressor inhibits transcription of the second repressor, which inhibits the transcription of the gene or gene
- second repressors useful in these embodiments include, but are not limited to, TetR, CI, and LexA.
- first repressor which occurs in the absence of RNS
- second repressor is transcribed, which represses expression of the gene or gene cassette, e.g., a butyrogenic gene cassette.
- expression of the second repressor is repressed, and the gene or gene cassette, e.g., a butyrogenic gene cassette, is expressed.
- a RNS-responsive transcription factor may induce, derepress, or repress gene expression depending upon the regulatory region sequence used in the genetically engineered bacteria.
- One or more types of RNS-sensing transcription factors and corresponding regulatory region sequences may be present in genetically engineered bacteria.
- the genetically engineered bacteria comprise one type of RNS-sensing transcription factor, e.g., NsrR, and one corresponding regulatory region sequence, e.g., from norB.
- the genetically engineered bacteria comprise one type of RNS-sensing transcription factor, e.g., NsrR, and two or more different corresponding regulatory region sequences, e.g., from norB and aniA.
- the genetically engineered bacteria comprise two or more types of RNS- sensing transcription factors, e.g., NsrR and NorR, and two or more corresponding regulatory region sequences, e.g., from norB and norR, respectively.
- One RNS-responsive regulatory region may be capable of binding more than one transcription factor.
- the genetically engineered bacteria comprise two or more types of RNS- sensing transcription factors and one corresponding regulatory region sequence.
- Nucleic acid sequences of several RNS-regulated regulatory regions are known in the art (see, e.g., Spiro, 2006; Isabella et al., 2009; Dunn et al., 2010; Vine et al., 2011; Karlinsey et al., 2012).
- the genetically engineered bacteria of the invention comprise a gene encoding a RNS-sensing transcription factor, e.g., the nsrR gene, that is controlled by its native promoter, an inducible promoter, a promoter that is stronger than the native promoter, e.g., the GlnRS promoter or the P(Bla) promoter, or a constitutive promoter.
- a RNS-sensing transcription factor e.g., the nsrR gene
- it may be advantageous to express the RNS-sensing transcription factor e.g., the nsrR gene, that is controlled by its native promoter, an inducible promoter, a promoter that is stronger than the native promoter, e.g., the GlnRS promoter or the P(Bla) promoter, or a constitutive promoter.
- expression of the RNS-sensing transcription factor is controlled by a different promoter than the promoter that controls expression of the therapeutic molecule.
- expression of the RNS- sensing transcription factor is controlled by the same promoter that controls expression of the therapeutic molecule.
- the RNS-sensing transcription factor and therapeutic molecule are divergently transcribed from a promoter region.
- the genetically engineered bacteria of the invention comprise a gene for a RNS-sensing transcription factor from a different species, strain, or substrain of bacteria.
- the genetically engineered bacteria comprise a RNS-responsive regulatory region from a different species, strain, or substrain of bacteria.
- the genetically engineered bacteria comprise a RNS-sensing transcription factor and corresponding RNS-responsive regulatory region from a different species, strain, or substrain of bacteria.
- the heterologous RNS-sensing transcription factor and regulatory region may increase the transcription of genes operatively linked to said regulatory region in the presence of RNS, as compared to the native transcription factor and regulatory region from bacteria of the same subtype under the same conditions.
- the genetically engineered bacteria comprise a RNS-sensing transcription factor, NsrR, and corresponding regulatory region, nsrR, from Neisseria gonorrhoeae.
- NsrR RNS-sensing transcription factor
- nsrR regulatory region
- the native RNS-sensing transcription factor, e.g., NsrR is left intact and retains wild-type activity.
- the native RNS-sensing transcription factor, e.g., NsrR is deleted or mutated to reduce or eliminate wild-type activity.
- the genetically engineered bacteria of the invention comprise multiple copies of the endogenous gene encoding the RNS-sensing transcription factor, e.g., the nsrR gene.
- the gene encoding the RNS-sensing transcription factor is present on a plasmid.
- the gene encoding the RNS-sensing transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on different plasmids.
- the gene encoding the RNS-sensing transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on the same plasmid.
- the gene encoding the RNS-sensing transcription factor is present on a chromosome.
- the gene encoding the RNS-sensing transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on different chromosomes.
- the gene encoding the RNS-sensing transcription factor and the gene or gene cassette for producing the therapeutic molecule are present on the same chromosome.
- the genetically engineered bacteria comprise a wild-type gene encoding a RNS-sensing transcription factor, e.g., the NsrR gene, and a corresponding regulatory region, e.g., a norB regulatory region, that is mutated relative to the wild-type regulatory region from bacteria of the same subtype.
- the mutated regulatory region increases the expression of the anti-inflammation and/or gut barrier enhancer molecule in the presence of RNS, as compared to the wild-type regulatory region under the same conditions.
- the genetically engineered bacteria comprise a wild-type RNS-responsive regulatory region, e.g., the norB regulatory region, and a corresponding transcription factor, e.g., NsrR, that is mutated relative to the wild-type transcription factor from bacteria of the same subtype.
- the mutant transcription factor increases the expression of the anti-inflammation and/or gut barrier enhancer molecule in the presence of RNS, as compared to the wild-type transcription factor under the same conditions.
- both the RNS-sensing transcription factor and corresponding regulatory region are mutated relative to the wild-type sequences from bacteria of the same subtype in order to increase expression of the anti-inflammation and/or gut barrier enhancer molecule in the presence of RNS.
- Nucleic acid sequences of exemplary RNS-regulated constructs comprising a gene encoding NsrR and a norB promoter are shown in Table 10 and Table 11.
- Table 10 depicts the nucleic acid sequence of an exemplary RNS-regulated construct comprising a gene encoding nsrR, a regulatory region of norB, and a butyrogenic gene cassette (pLogic031-nsrR-norB-butyrate construct; SEQ I D NO: 67).
- the sequence encoding NsrR is underlined and bolded, and the NsrR binding site, i.e., a regulatory
- Table 11 depicts the nucleic acid sequence of an exemplary RNS-regulated construct comprising a gene encoding nsrR, a regulatory region of norB, and a butyrogenic gene cassette (pLogic046-nsrR-norB-butyrate construct; SEQ ID NO: 68).
- the sequence encoding NsrR is underlined and bolded, and the NsrR binding site, i.e., a regulatory region of norB is poxeq.
- N ucleic acid sequences of tetracycline- regulated constructs comprising a tet promoter are shown in Table 12 and Table 13.
- Table 12 depicts the nucleic acid sequence of an exemplary tetracycline-regulated construct comprising a tet promoter and a butyrogenic gene cassette (pLogic031-tet- butyrate construct; SEQ ID NO: 69). The sequence encoding TetR is underlined, and the overlapping tetR/tetA promoters are poxeq.
- Table 13 depicts the nucleic acid sequence of an exemplary tetracycline-regulated construct comprising a tet promoter and a butyrogenic gene cassette (pLogic046-tet-butyrate construct; SEQ ID NO: 70).
- genetically engineered bacteria comprise a nucleic acid sequence that is at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% homologous to the DNA sequence of SEQ I D NO: 67, 68, 69, or 70, or a functional fragment thereof.
- the gene or gene cassette for producing the anti- inflammation and/or gut barrier function enhancer molecule is present on a plasmid and operably linked to a promoter that is induced by RNS. In some embodiments, the gene or gene cassette for producing the anti-inflammation and/or gut barrier function enhancer molecule is present in the chromosome and operably linked to a promoter that is induced by RNS. In some embodiments, the gene or gene cassette for producing the anti-inflammation and/or gut barrier function enhancer molecule is present on a chromosome and operably linked to a promoter that is induced by exposure to tetracycline.
- the gene or gene cassette for producing the anti- inflammation and/or gut barrier function enhancer molecule is present on a plasmid and operably linked to a promoter that is induced by exposure to tetracycline.
- expression is further optimized by methods known in the art, e.g., by optimizing ribosomal binding sites, manipulating transcriptional regulators, and/or increasing mRNA stability.
- the genetically engineered bacteria comprise a stably maintained plasmid or chromosome carrying the gene(s) or gene cassette(s) capable of producing an anti-inflammation and/or gut barrier function enhancer molecule, such that the gene(s) or gene cassette(s) can be expressed in the host cell, and the host cell is capable of survival and/or growth in vitro, e.g., in medium, and/or in vivo, e.g., in the gut.
- a bacterium may comprise multiple copies of the gene or gene cassette for producing the anti-inflammation and/or gut barrier function enhance molecule.
- gene or gene cassette is expressed on a low- copy plasmid.
- the low-copy plasmid may be useful for increasing stability of expression. In some embodiments, the low-copy plasmid may be useful for decreasing leaky expression under non-inducing conditions. In some embodiments, gene or gene cassette is expressed on a high-copy plasmid. In some embodiments, the high-
- -111- copy plasmid may be useful for increasing gene or gene cassette expression.
- gene or gene cassette is expressed on a chromosome.
- the genetically engineered bacteria may comprise multiple copies of the gene(s) or gene cassette(s) capable of producing an anti- inflammation and/or gut barrier function enhancer molecule.
- the gene(s) or gene cassette(s) capable of producing an anti-inflammation and/or gut barrier function enhancer molecule is present on a plasmid and operatively linked to a RNS- responsive regulatory region.
- the gene(s) or gene cassette(s) capable of producing an anti-inflammation and/or gut barrier function enhancer molecule is present in a chromosome and operatively linked to a RNS-responsive regulatory region.
- any of the gene(s) or gene cassette(s) of the present disclosure may be integrated into the bacterial chromosome at one or more integration sites.
- one or more copies of the butyrogenic gene cassette may be integrated into the bacterial chromosome. Having multiple copies of the butyrogenic gene cassette integrated into the chromosome allows for greater production of the butyrate and also permits fine-tuning of the level of expression.
- different circuits described herein, such as any of the kill-switch circuits, in addition to the therapeutic gene(s) or gene cassette(s) could be integrated into the bacterial chromosome at one or more different integration sites to perform multiple different functions.
- the genetically engineered bacteria of the invention produce at least one anti-inflammation and/or gut barrier enhancer molecule in the presence of RNS to reduce local gut inflammation by at least about 1.5-fold, at least about 2-fold, at least about 10-fold, at least about 15-fold, at least about 20-fold, at least about 30-fold, at least about 50-fold, at least about 100-fold, at least about 200- fold, at least about 300-fold, at least about 400-fold, at least about 500-fold, at least about 600-fold, at least about 700-fold, at least about 800-fold, at least about 900-fold, at least about 1,000-fold, or at least about 1,500-fold as compared to unmodified bacteria of the same subtype under the same conditions.
- Inflammation may be
- -112- measured by methods known in the art e.g., counting disease lesions using endoscopy; detecting T regulatory cell differentiation in peripheral blood, e.g., by fluorescence activated sorting; measuring T regulatory cell levels; measuring cytokine levels;
- inflammatory biomarkers e.g., by qPCR; PCR arrays; transcription factor phosphorylation assays; immunoassays; and/or cytokine assay kits (Mesoscale, Cayman Chemical, Qiagen).
- the genetically engineered bacteria produce at least about 1.5-fold, at least about 2-fold, at least about 10-fold, at least about 15-fold, at least about 20-fold, at least about 30-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 300-fold, at least about 400-fold, at least about 500-fold, at least about 600-fold, at least about 700-fold, at least about 800-fold, at least about 900-fold, at least about 1,000-fold, or at least about 1,500-fold more of an anti- inflammation and/or gut barrier enhancer molecule in the presence of RNS than unmodified bacteria of the same subtype under the same conditions.
- Certain unmodified bacteria will not have detectable levels of the anti-inflammation and/or gut barrier enhancer molecule. I n embodiments using genetically modified forms of these bacteria, the anti-inflammation and/or gut barrier enhancer molecule will be detectable in the presence of RNS.
- the anti-inflammation and/or gut barrier enhancer molecule is butyrate.
- Methods of measuring butyrate levels e.g., by mass spectrometry, gas chromatography, high-performance liquid chromatography (HPLC), are known in the art (see, e.g., Aboulnaga et al., 2013).
- butyrate is measured as butyrate level/bacteria optical density (OD).
- OD optical density
- butyrogenic gene cassette serves as a proxy measurement for butyrate production.
- the bacterial cells of the invention are harvested and lysed to measure butyrate production.
- butyrate production is measured in the bacterial cell medium.
- the genetically engineered bacteria produce at least about 1 nM/OD, at least about 10 nM/OD, at least about 100 nM/OD, at least about 500 nM/OD, at least about 1 ⁇ /OD, at least about 10
- -113- ⁇ /OD at least about 100 ⁇ /OD, at least about 500 ⁇ /OD, at least about 1 mM/OD, at least about 2 mM/OD, at least about 3 mM/OD, at least about 5 mM/OD, at least about 10 mM/OD, at least about 20 mM/OD, at least about 30 mM/OD, or at least about 50 mM/OD of butyrate in the presence of RNS.
- the genetically engineered bacteria of the invention comprise a tunable regulatory region that is directly or indirectly controlled by a transcription factor that is capable of sensing at least one reactive oxygen species.
- the tunable regulatory region is operatively linked to a gene or gene cassette capable of directly or indirectly driving the expression of an anti-inflammation and/or gut barrier function enhancer molecule, thus controlling expression of the molecule relative to ROS levels.
- the tunable regulatory region is a ROS-inducible regulatory region, and the molecule is butyrate; when ROS is present, e.g., in an inflamed tissue, a ROS- sensing transcription factor binds to and/or activates the regulatory region and drives expression of the butyrogenic gene cassette, thereby producing butyrate, which exerts anti-inflammation and/or gut barrier enhancing effects. Subsequently, when ROS is present, e.g., in an inflamed tissue, a ROS- sensing transcription factor binds to and/or activates the regulatory region and drives expression of the butyrogenic gene cassette, thereby producing butyrate, which exerts anti-inflammation and/or gut barrier enhancing effects. Subsequently, when ROS is present, e.g., in an inflamed tissue, a ROS- sensing transcription factor binds to and/or activates the regulatory region and drives expression of the butyrogenic gene cassette, thereby producing butyrate, which
- inflammation is ameliorated, ROS levels are reduced, and butyrate production is decreased or eliminated.
- the tunable regulatory region is a ROS-inducible regulatory region; in the presence of ROS, a transcription factor senses ROS and activates the ROS-inducible regulatory region, thereby driving expression of an operatively linked gene or gene cassette.
- the transcription factor senses ROS and subsequently binds to the ROS-inducible regulatory region, thereby activating downstream gene expression.
- the transcription factor is bound to the ROS-inducible regulatory region in the absence of ROS; when the transcription factor senses ROS, it undergoes a conformational change, thereby inducing downstream gene expression.
- the tunable regulatory region is a ROS-inducible regulatory region
- the transcription factor that senses ROS is OxyR.
- OxyR "functions primarily as a global regulator of the peroxide stress response" and is capable of
- genes involved in H 2 0 2 detoxification katE, ohpCF
- heme biosynthesis hemH
- reductant supply grxA, gor, trxC
- thiol-disulfide e.g., "genes involved in H 2 0 2 detoxification (katE, ohpCF), heme biosynthesis (hemH), reductant supply (grxA, gor, trxC), thiol-disulfide
- the genetically engineered bacteria of the invention may comprise any suitable ROS- responsive regulatory region from a gene that is activated by OxyR. Genes that are capable of being activated by OxyR are known in the art (see, e.g., Zheng et al., 2001; Dubbs et al., 2012; Table 1).
- the genetically engineered bacteria of the invention comprise a ROS-inducible regulatory region from oxyS that is operatively linked to a gene or gene cassette, e.g., a butyrogenic gene cassette.
- a gene or gene cassette e.g., a butyrogenic gene cassette.
- an OxyR transcription factor senses ROS and activates to the oxyS regulatory region, thereby driving expression of the operatively linked butyrogenic gene cassette and producing butyrate.
- OxyR is encoded by an E. coli oxyR gene.
- the oxyS regulatory region is an E. coli oxyS regulatory region.
- the ROS-inducible regulatory region is selected from the regulatory region of katG, dps, and ahpC.
- the tunable regulatory region is a ROS- inducible regulatory region, and the corresponding transcription factor that senses ROS is SoxR.
- SoxR When SoxR is "activated by oxidation of its [2Fe-2S] cluster, it increases the synthesis of SoxS, which then activates its target gene expression" (Koo et al., 2003).
- SoxR is known to respond primarily to superoxide and nitric oxide" (Koo et al., 2003), and is also capable of responding to H 2 0 2 .
- the genetically engineered bacteria of the invention may comprise any suitable ROS-responsive regulatory region from a gene that is activated by SoxR.
- the genetically engineered bacteria of the invention comprise a ROS-inducible regulatory region from soxS that is operatively linked to a gene or gene cassette, e.g., a butyrogenic gene cassette.
- a gene or gene cassette e.g., a butyrogenic gene cassette.
- the SoxR transcription factor senses ROS and activates the soxS regulatory region, thereby driving expression of the operatively linked butyrogenic gene cassette and producing butyrate.
- the tunable regulatory region is a ROS- derepressible regulatory region, and binding of a corresponding transcription factor represses downstream gene expression; in the presence of ROS, the transcription factor no longer binds to the regulatory region, thereby derepressing the operatively linked gene or gene cassette.
- the tunable regulatory region is a ROS- derepressible regulatory region
- the transcription factor that senses ROS is OhrR.
- OhrR "binds to a pair of inverted repeat DNA sequences overlapping the ohrA promoter site and thereby represses the transcription event," but oxidized OhrR is "unable to bind its DNA target" (Duarte et al., 2010).
- OhrR is a "transcriptional repressor [that] ...
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Diabetes (AREA)
- Wood Science & Technology (AREA)
- Immunology (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biotechnology (AREA)
- Biochemistry (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Hematology (AREA)
- Gastroenterology & Hepatology (AREA)
- Biophysics (AREA)
- Neurology (AREA)
- Physical Education & Sports Medicine (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Endocrinology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Urology & Nephrology (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Neurosurgery (AREA)
- Pulmonology (AREA)
Abstract
Description
Claims
Priority Applications (57)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
SG11201707025WA SG11201707025WA (en) | 2015-03-02 | 2016-03-02 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
BR112017018656-0A BR112017018656B1 (en) | 2015-03-02 | 2016-03-02 | GENETICALLY MODIFIED BACTERIA, PHARMACEUTICALLY ACCEPTABLE COMPOSITION COMPRISING SUCH BACTERIA AND USE OF SUCH COMPOSITION TO TREAT OR PREVENT A DISEASE OR CONDITION ASSOCIATED WITH INTESTINAL INFLAMMATION AND/OR INTESTINAL BARRIER FUNCTION COMPROMISED |
MX2017011037A MX2017011037A (en) | 2015-03-02 | 2016-03-02 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier. |
JP2017545655A JP7095993B2 (en) | 2015-03-02 | 2016-03-02 | Bacteria engineered for the treatment of diseases that benefit from reduced gastrointestinal inflammation and / or enhanced gastrointestinal mucosal barrier |
EP16710574.1A EP3265105A1 (en) | 2015-03-02 | 2016-03-02 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US15/301,230 US10273489B2 (en) | 2014-12-22 | 2016-03-02 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
CN201680025498.4A CN107636146A (en) | 2015-03-02 | 2016-03-02 | It is engineered to treat the bacterium of the disease for the gastrointestinal mucosal barrier benefited from the alimentary canal inflammation of reduction and/or tightened up |
AU2016226234A AU2016226234B2 (en) | 2015-03-02 | 2016-03-02 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
RU2017130462A RU2017130462A (en) | 2015-03-02 | 2016-03-02 | A BACTERIA CREATED FOR TREATING DISEASES EASILY DURED BY REDUCING THE INTESTINAL INFLAMMATION AND / OR STRENGTHENING THE INTESTINAL MUSCARY BARRIER |
CA2978315A CA2978315A1 (en) | 2015-03-02 | 2016-03-02 | Bacteria engineered to produce butyrate under low oxygen or anaerobic conditions and uses thereof |
KR1020177028200A KR20170121291A (en) | 2015-03-02 | 2016-03-02 | Engineered bacteria to treat diseases that benefit from reduced intestinal inflammation and / or enhanced intestinal mucosal barriers |
EP16724834.3A EP3294757B1 (en) | 2015-05-13 | 2016-05-13 | Bacteria engineered to treat a disease or disorder |
US15/319,564 US9889164B2 (en) | 2014-12-05 | 2016-05-13 | Bacteria engineered to treat a disease or disorder |
PCT/US2016/032565 WO2016183532A1 (en) | 2015-05-13 | 2016-05-13 | Bacteria engineered to treat a disease or disorder |
PCT/US2016/034200 WO2016200614A2 (en) | 2015-06-10 | 2016-05-25 | Bacteria engineered to treat diseases associated with hyperammonemia |
CA2988930A CA2988930A1 (en) | 2015-06-10 | 2016-05-25 | Bacteria engineered to treat diseases associated with hyperammonemia |
US15/164,828 US9688967B2 (en) | 2014-12-05 | 2016-05-25 | Bacteria engineered to treat diseases associated with hyperammonemia |
EP16731402.0A EP3307879A2 (en) | 2015-06-10 | 2016-05-25 | Bacteria engineered to treat diseases associated with hyperammonemia |
AU2016274311A AU2016274311A1 (en) | 2015-06-10 | 2016-05-25 | Bacteria engineered to treat diseases associated with hyperammonemia |
JP2017564379A JP6817966B2 (en) | 2015-06-10 | 2016-05-25 | Bacteria engineered to treat diseases associated with hyperammonemia |
US15/772,289 US11685925B2 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
AU2016346646A AU2016346646B2 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US15/260,319 US11384359B2 (en) | 2014-12-22 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
EP21192561.5A EP3988107A1 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
PCT/US2016/050836 WO2017074566A1 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
CA3002965A CA3002965A1 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
EP16771037.5A EP3368696A1 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
JP2018522565A JP2018532412A (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gastrointestinal inflammation and / or enhanced gastrointestinal mucosal barrier |
PCT/US2016/059518 WO2017075485A1 (en) | 2015-10-30 | 2016-10-28 | Bacteria engineered to treat disorders in which trimethylamine (tma) is detrimental |
US16/069,266 US20190010506A1 (en) | 2016-01-11 | 2016-12-28 | Bacteria engineered to treat metabolic diseases |
PCT/US2016/069052 WO2017123418A1 (en) | 2016-01-11 | 2016-12-28 | Bacteria engineered to treat metabolic diseases |
EP16823539.8A EP3402497A1 (en) | 2016-01-11 | 2016-12-28 | Bacteria engineered to treat metabolic diseases |
US16/069,199 US20190282628A1 (en) | 2016-01-11 | 2017-01-11 | Recombinant bacteria engineered to treat diseases and disorders associated with amino acid metabolism and methods of use thereof |
PCT/US2017/013074 WO2017123676A1 (en) | 2016-01-11 | 2017-01-11 | Recombinant bacteria engineered to treat diseases and disorders associated with amino acid metabolism and methods of use thereof |
PCT/US2017/012982 WO2017123610A2 (en) | 2016-01-11 | 2017-01-11 | Bacteria engineered to detoxify deleterious molecules |
PCT/US2017/016609 WO2017136795A1 (en) | 2016-02-04 | 2017-02-03 | Bacteria engineered to treat diseases associated with tryptophan metabolism |
PCT/US2017/016603 WO2017136792A2 (en) | 2016-02-04 | 2017-02-03 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
CA3013770A CA3013770A1 (en) | 2016-02-04 | 2017-02-03 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
AU2017213646A AU2017213646A1 (en) | 2016-02-04 | 2017-02-03 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
EP17705544.9A EP3411051A2 (en) | 2016-02-04 | 2017-02-03 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US16/074,559 US20210161976A1 (en) | 2016-02-04 | 2017-02-03 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
PCT/US2017/017552 WO2017139697A1 (en) | 2016-02-10 | 2017-02-10 | Bacteria engineered to treat diseases associated with hyperammonemia |
PCT/US2017/017563 WO2017139708A1 (en) | 2016-02-10 | 2017-02-10 | Bacteria engineered to treat nonalcoholic steatohepatitis (nash) |
US15/599,285 US11060073B2 (en) | 2014-12-05 | 2017-05-18 | Bacteria engineered to treat diseases associated with hyperammonemia |
ZA2017/05873A ZA201705873B (en) | 2015-03-02 | 2017-08-29 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
IL254226A IL254226B (en) | 2015-03-02 | 2017-08-30 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US15/852,762 US10933102B2 (en) | 2015-05-13 | 2017-12-22 | Bacteria engineered to treat a disease or disorder |
HK18109707.5A HK1250244A1 (en) | 2015-03-02 | 2018-07-26 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US17/123,469 US11845964B2 (en) | 2014-12-05 | 2020-12-16 | Bacteria engineered to treat diseases associated with hyperammonemia |
US17/124,661 US11883439B2 (en) | 2015-05-13 | 2020-12-17 | Bacteria engineered to treat a disease or disorder |
JP2020217391A JP2021061846A (en) | 2015-06-10 | 2020-12-25 | Engineered bacteria for treating diseases associated with hyperammonemia |
JP2021192519A JP2022033832A (en) | 2015-10-30 | 2021-11-26 | Bacteria engineered to treat diseases that benefit from reduced gastrointestinal gut inflammation and/or gastrointestinal gut mucosal barrier |
IL288870A IL288870A (en) | 2015-03-02 | 2021-12-09 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
AU2022203178A AU2022203178A1 (en) | 2015-03-02 | 2022-05-12 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US17/835,601 US20230043588A1 (en) | 2015-03-02 | 2022-06-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
AU2022275435A AU2022275435A1 (en) | 2015-10-30 | 2022-11-23 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US18/195,694 US20240110192A1 (en) | 2015-10-30 | 2023-05-10 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
Applications Claiming Priority (26)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201562127097P | 2015-03-02 | 2015-03-02 | |
US201562127131P | 2015-03-02 | 2015-03-02 | |
US62/127,097 | 2015-03-02 | ||
US62/127,131 | 2015-03-02 | ||
US201562184770P | 2015-06-25 | 2015-06-25 | |
US62/184,770 | 2015-06-25 | ||
US201562248814P | 2015-10-30 | 2015-10-30 | |
US201562248805P | 2015-10-30 | 2015-10-30 | |
US201562248825P | 2015-10-30 | 2015-10-30 | |
US62/248,825 | 2015-10-30 | ||
US62/248,805 | 2015-10-30 | ||
US62/248,814 | 2015-10-30 | ||
US201562256042P | 2015-11-16 | 2015-11-16 | |
US201562256044P | 2015-11-16 | 2015-11-16 | |
US201562256048P | 2015-11-16 | 2015-11-16 | |
US62/256,044 | 2015-11-16 | ||
US62/256,048 | 2015-11-16 | ||
US62/256,042 | 2015-11-16 | ||
US14/998,376 | 2015-12-22 | ||
US14/998,376 US20160206666A1 (en) | 2014-12-22 | 2015-12-22 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tighten gut mucosal barrier |
US201662291461P | 2016-02-04 | 2016-02-04 | |
US201662291470P | 2016-02-04 | 2016-02-04 | |
US201662291468P | 2016-02-04 | 2016-02-04 | |
US62/291,461 | 2016-02-04 | ||
US62/291,470 | 2016-02-04 | ||
US62/291,468 | 2016-02-04 |
Related Parent Applications (5)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US14/960,333 Continuation-In-Part US9487764B2 (en) | 2014-12-05 | 2015-12-04 | Bacteria engineered to treat diseases associated with hyperammonemia |
PCT/US2015/064140 Continuation-In-Part WO2016090343A1 (en) | 2014-12-05 | 2015-12-04 | Bacteria engineered to treat diseases associated with hyperammonemia |
US14/998,376 Continuation US20160206666A1 (en) | 2014-12-22 | 2015-12-22 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tighten gut mucosal barrier |
PCT/US2016/032562 Continuation-In-Part WO2016183531A1 (en) | 2014-12-05 | 2016-05-13 | Bacteria engineered to reduce hyperphenylalaninemia |
USPCT/US2016/093444 Continuation-In-Part | 2016-01-11 | 2016-06-24 |
Related Child Applications (7)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2015/064140 Continuation-In-Part WO2016090343A1 (en) | 2014-12-05 | 2015-12-04 | Bacteria engineered to treat diseases associated with hyperammonemia |
PCT/US2016/032565 Continuation-In-Part WO2016183532A1 (en) | 2014-12-05 | 2016-05-13 | Bacteria engineered to treat a disease or disorder |
PCT/US2016/032562 Continuation-In-Part WO2016183531A1 (en) | 2014-12-05 | 2016-05-13 | Bacteria engineered to reduce hyperphenylalaninemia |
US15/164,828 Continuation-In-Part US9688967B2 (en) | 2014-12-05 | 2016-05-25 | Bacteria engineered to treat diseases associated with hyperammonemia |
US15/260,319 Continuation-In-Part US11384359B2 (en) | 2014-12-22 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
PCT/US2016/050836 Continuation-In-Part WO2017074566A1 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US15/772,289 Continuation-In-Part US11685925B2 (en) | 2015-10-30 | 2016-09-08 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2016141108A1 true WO2016141108A1 (en) | 2016-09-09 |
WO2016141108A8 WO2016141108A8 (en) | 2017-12-21 |
Family
ID=56848760
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2016/020530 WO2016141108A1 (en) | 2014-12-05 | 2016-03-02 | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
Country Status (15)
Country | Link |
---|---|
US (1) | US20230043588A1 (en) |
EP (1) | EP3265105A1 (en) |
JP (1) | JP7095993B2 (en) |
KR (1) | KR20170121291A (en) |
CN (1) | CN107636146A (en) |
AU (2) | AU2016226234B2 (en) |
BR (1) | BR112017018656B1 (en) |
CA (1) | CA2978315A1 (en) |
HK (1) | HK1250244A1 (en) |
IL (2) | IL254226B (en) |
MX (1) | MX2017011037A (en) |
RU (1) | RU2017130462A (en) |
SG (1) | SG11201707025WA (en) |
WO (1) | WO2016141108A1 (en) |
ZA (1) | ZA201705873B (en) |
Cited By (25)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2017136792A3 (en) * | 2016-02-04 | 2017-09-08 | Synlogic, Inc. | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
WO2018183941A2 (en) | 2017-03-30 | 2018-10-04 | Progenity Inc. | Treatment of a disease of the gastrointestinal tract with live biotherapeutics |
WO2018204757A1 (en) * | 2017-05-04 | 2018-11-08 | Second Genome, Inc. | Proteins for the treatment of epithelial barrier function disorders |
WO2018223051A1 (en) * | 2017-06-02 | 2018-12-06 | Second Genome, Inc. | Proteins for the treatment of epithelial barrier function disorders |
WO2018237198A1 (en) | 2017-06-21 | 2018-12-27 | Synlogic Operating Company, Inc. | Bacteria for the treatment of disorders |
CN110709093A (en) * | 2017-04-17 | 2020-01-17 | 加利福尼亚大学董事会 | Engineered bacteria and methods of use |
WO2020223345A1 (en) | 2019-04-29 | 2020-11-05 | Antipov Eugene | Enumeration of genetically engineered microorganisms by live cell counting techniques |
JP2021505197A (en) * | 2017-12-05 | 2021-02-18 | バイオピーエルエックス,インコーポレイティド | Methods and compositions to prevent microbial infections |
EP3751008A4 (en) * | 2018-02-08 | 2021-11-17 | MD Healthcare Inc. | Nanovesicles derived from bacteria of lactococcus genus and use thereof |
EP3763830A4 (en) * | 2018-03-05 | 2021-12-08 | MD Healthcare Inc. | Nanovesicles derived from lactobacillus sp. bacteria, and use thereof |
WO2022120028A2 (en) | 2020-12-02 | 2022-06-09 | Synlogic Operating Company, Inc. | Engineered microorganisms |
WO2022144381A1 (en) * | 2020-12-30 | 2022-07-07 | Eligo Bioscience | Microbiome modulation of a host by delivery of dna payloads with minimized spread |
US11384359B2 (en) | 2014-12-22 | 2022-07-12 | Synlogic Operating Company, Inc. | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
WO2022204406A1 (en) * | 2021-03-24 | 2022-09-29 | Synlogic Operating Company, Inc. | Bacteria engineered to treat disorders in which oxalate is detrimental |
US11458172B2 (en) | 2017-09-08 | 2022-10-04 | New Portal Limited | Nucleic acid systems that enable bacteria to specifically target solid tumors via glucose-dependent viability |
EP3920908A4 (en) * | 2019-02-08 | 2022-10-05 | McMaster University | Use of activators of the aryl hydrocarbon receptor for treating gluten-induced gastrointestinal diseases |
WO2022221273A1 (en) | 2021-04-13 | 2022-10-20 | Synlogic Operating Company, Inc. | Bacteria engineered to secrete active proteins |
US11612617B2 (en) | 2015-05-06 | 2023-03-28 | Snipr Technologies Limited | Altering microbial populations and modifying microbiota |
US11612586B2 (en) * | 2016-03-18 | 2023-03-28 | The Texas A&M University System | Indole regulation of antigen presenting cells |
US11685925B2 (en) | 2015-10-30 | 2023-06-27 | Synlogic Operating Company, Inc. | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US11746352B2 (en) | 2019-12-30 | 2023-09-05 | Eligo Bioscience | Microbiome modulation of a host by delivery of DNA payloads with minimized spread |
WO2024081768A1 (en) | 2022-10-12 | 2024-04-18 | Synlogic Operating Company, Inc. | Bacteria engineered to produce active epidermal growth factor (egf) and their medical uses |
EP4125988A4 (en) * | 2020-03-26 | 2024-04-24 | The Regents of University of California | Detection and treatment of intestinal fibrosis |
US12076375B2 (en) | 2022-06-29 | 2024-09-03 | Snipr Biome Aps | Treating and preventing E coli infections |
US12098372B2 (en) | 2019-12-30 | 2024-09-24 | Eligo Bioscience | Microbiome modulation of a host by delivery of DNA payloads with minimized spread |
Families Citing this family (13)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111886333A (en) * | 2018-02-09 | 2020-11-03 | 学校法人庆应义塾 | Compositions and methods for inducing CD8+ T cells |
WO2019153902A1 (en) * | 2018-02-11 | 2019-08-15 | 中国科学院上海生命科学研究院 | Plant genome site-directed substitution method |
CN109771404A (en) * | 2019-02-28 | 2019-05-21 | 北京大学人民医院(北京大学第二临床医学院) | Application of the butyric acid in preparation prevention and/or treatment autoimmune disease drug |
JP2023516010A (en) * | 2020-02-25 | 2023-04-17 | シムビーボ コーポレイション | gene delivery system |
WO2022006748A1 (en) * | 2020-07-07 | 2022-01-13 | New Portal Limited | Genetically engineered live bacteria and methods of constructing the same |
KR102397299B1 (en) | 2020-07-13 | 2022-05-11 | 한국생명공학연구원 | Anaeroskeptrum propionigenes and uses thereof |
CN114073777A (en) * | 2020-08-18 | 2022-02-22 | 中国科学院深圳先进技术研究院 | Bacterial targeting vector carrying cytokine or polynucleotide thereof and application thereof in tumor treatment |
CN112322528B (en) * | 2020-11-03 | 2022-07-22 | 江南大学 | Lactobacillus rhamnosus capable of intervening metabolic syndrome and application thereof |
CN112877271B (en) * | 2021-02-05 | 2023-03-14 | 江西师范大学 | Method for improving L-arginine production of corynebacterium crenatum through anaerobic fermentation |
CN113150069B (en) * | 2021-02-24 | 2022-07-22 | 安杰利(重庆)生物科技有限公司 | Hyaluronidase inhibitor and preparation method thereof |
CN116369249B (en) * | 2023-01-13 | 2024-05-17 | 四川轻化工大学 | Construction method of zebra fish enteritis model |
WO2024194876A1 (en) * | 2023-03-23 | 2024-09-26 | Technion Research & Development Foundation Limited | Bacteriophages as a tool to manipulate gut commensal immune-modulation activity |
CN117701456B (en) * | 2023-12-22 | 2024-10-01 | 善恩康生物科技(苏州)有限公司 | Bifidobacterium lactis, preparation thereof and application thereof in reproductive injury or disease |
Citations (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5589168A (en) | 1991-04-08 | 1996-12-31 | Unilever Patent Holdings B.V. | Probiotic |
US5989463A (en) | 1997-09-24 | 1999-11-23 | Alkermes Controlled Therapeutics, Inc. | Methods for fabricating polymer-based controlled release devices |
US6203797B1 (en) | 1998-01-06 | 2001-03-20 | Stephen C. Perry | Dietary supplement and method for use as a probiotic, for alleviating the symptons associated with irritable bowel syndrome |
US20030166191A1 (en) | 1998-12-02 | 2003-09-04 | Timothy Gardner | Bistable genetic toggle switch |
EP1383897A2 (en) * | 2001-05-03 | 2004-01-28 | Vlaams Interuniversitair Instituut voor Biotechnologie vzw. | Self-containing lactococcus strain |
US6835376B1 (en) | 1999-03-11 | 2004-12-28 | Nestec S.A. | Lactobacillus paracasei strain for preventing diarrhea caused by pathogenic bacteria |
WO2006079790A2 (en) * | 2005-01-25 | 2006-08-03 | University Of Leeds | Controlled production and delivery of biologically active agents by gut bacteria |
US7731976B2 (en) | 2003-08-29 | 2010-06-08 | Cobb And Company, Llp | Treatment of irritable bowel syndrome using probiotic composition |
WO2013175358A1 (en) * | 2012-05-21 | 2013-11-28 | Institut National De La Recherche Agronomique | Stress-regulated prokaryotic expression cassettes |
Family Cites Families (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6551799B2 (en) * | 1999-12-07 | 2003-04-22 | Genentech, Inc. | Interleukin-22 polypeptides, nucleic acids encoding the same and methods for the treatment of pancreatic disorders |
CN1246463C (en) * | 2004-03-03 | 2006-03-22 | 清华大学 | Expression carrier of low oxygen inducing microorganusm and constructure process and application thereof |
CN101849969A (en) * | 2009-03-31 | 2010-10-06 | 青岛东海药业有限公司 | Application of butyric acid producing beneficial bacterium in preparing preparation for preventing and treating severe disease gut barrier injury and post-injury complication |
EP2623604B8 (en) * | 2012-02-02 | 2015-04-22 | Baylor College of Medicine | Adenoviral-based biological delivery and expression system for use in the treatment of osteoarthritis |
-
2016
- 2016-03-02 SG SG11201707025WA patent/SG11201707025WA/en unknown
- 2016-03-02 AU AU2016226234A patent/AU2016226234B2/en active Active
- 2016-03-02 KR KR1020177028200A patent/KR20170121291A/en not_active Application Discontinuation
- 2016-03-02 CN CN201680025498.4A patent/CN107636146A/en active Pending
- 2016-03-02 MX MX2017011037A patent/MX2017011037A/en unknown
- 2016-03-02 EP EP16710574.1A patent/EP3265105A1/en active Pending
- 2016-03-02 WO PCT/US2016/020530 patent/WO2016141108A1/en active Application Filing
- 2016-03-02 RU RU2017130462A patent/RU2017130462A/en unknown
- 2016-03-02 CA CA2978315A patent/CA2978315A1/en active Pending
- 2016-03-02 JP JP2017545655A patent/JP7095993B2/en active Active
- 2016-03-02 BR BR112017018656-0A patent/BR112017018656B1/en active IP Right Grant
-
2017
- 2017-08-29 ZA ZA2017/05873A patent/ZA201705873B/en unknown
- 2017-08-30 IL IL254226A patent/IL254226B/en unknown
-
2018
- 2018-07-26 HK HK18109707.5A patent/HK1250244A1/en unknown
-
2021
- 2021-12-09 IL IL288870A patent/IL288870A/en unknown
-
2022
- 2022-05-12 AU AU2022203178A patent/AU2022203178A1/en active Pending
- 2022-06-08 US US17/835,601 patent/US20230043588A1/en active Pending
Patent Citations (10)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5589168A (en) | 1991-04-08 | 1996-12-31 | Unilever Patent Holdings B.V. | Probiotic |
US5989463A (en) | 1997-09-24 | 1999-11-23 | Alkermes Controlled Therapeutics, Inc. | Methods for fabricating polymer-based controlled release devices |
US6203797B1 (en) | 1998-01-06 | 2001-03-20 | Stephen C. Perry | Dietary supplement and method for use as a probiotic, for alleviating the symptons associated with irritable bowel syndrome |
US20030166191A1 (en) | 1998-12-02 | 2003-09-04 | Timothy Gardner | Bistable genetic toggle switch |
US6835376B1 (en) | 1999-03-11 | 2004-12-28 | Nestec S.A. | Lactobacillus paracasei strain for preventing diarrhea caused by pathogenic bacteria |
EP1383897A2 (en) * | 2001-05-03 | 2004-01-28 | Vlaams Interuniversitair Instituut voor Biotechnologie vzw. | Self-containing lactococcus strain |
US7731976B2 (en) | 2003-08-29 | 2010-06-08 | Cobb And Company, Llp | Treatment of irritable bowel syndrome using probiotic composition |
WO2006079790A2 (en) * | 2005-01-25 | 2006-08-03 | University Of Leeds | Controlled production and delivery of biologically active agents by gut bacteria |
WO2013175358A1 (en) * | 2012-05-21 | 2013-11-28 | Institut National De La Recherche Agronomique | Stress-regulated prokaryotic expression cassettes |
US20150139940A1 (en) * | 2012-05-21 | 2015-05-21 | Institut National De La Recherche Agronomique (Inra) | Stress-regulated prokaryotic expression cassettes |
Non-Patent Citations (141)
Cited By (33)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11384359B2 (en) | 2014-12-22 | 2022-07-12 | Synlogic Operating Company, Inc. | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US11642363B2 (en) | 2015-05-06 | 2023-05-09 | Snipr Technologies Limited | Altering microbial populations and modifying microbiota |
US11844760B2 (en) | 2015-05-06 | 2023-12-19 | Snipr Technologies Limited | Altering microbial populations and modifying microbiota |
US11612617B2 (en) | 2015-05-06 | 2023-03-28 | Snipr Technologies Limited | Altering microbial populations and modifying microbiota |
US11685925B2 (en) | 2015-10-30 | 2023-06-27 | Synlogic Operating Company, Inc. | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
WO2017136792A3 (en) * | 2016-02-04 | 2017-09-08 | Synlogic, Inc. | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
US11612586B2 (en) * | 2016-03-18 | 2023-03-28 | The Texas A&M University System | Indole regulation of antigen presenting cells |
WO2018183941A2 (en) | 2017-03-30 | 2018-10-04 | Progenity Inc. | Treatment of a disease of the gastrointestinal tract with live biotherapeutics |
CN110709093A (en) * | 2017-04-17 | 2020-01-17 | 加利福尼亚大学董事会 | Engineered bacteria and methods of use |
WO2018204757A1 (en) * | 2017-05-04 | 2018-11-08 | Second Genome, Inc. | Proteins for the treatment of epithelial barrier function disorders |
US11273198B2 (en) | 2017-05-04 | 2022-03-15 | Second Genome, Inc. | Proteins for the treatment of epithelial barrier function disorders |
WO2018223051A1 (en) * | 2017-06-02 | 2018-12-06 | Second Genome, Inc. | Proteins for the treatment of epithelial barrier function disorders |
US11174293B2 (en) | 2017-06-02 | 2021-11-16 | Second Genome, Inc. | Proteins for the treatment of epithelial barrier function disorders |
WO2018237198A1 (en) | 2017-06-21 | 2018-12-27 | Synlogic Operating Company, Inc. | Bacteria for the treatment of disorders |
US11458172B2 (en) | 2017-09-08 | 2022-10-04 | New Portal Limited | Nucleic acid systems that enable bacteria to specifically target solid tumors via glucose-dependent viability |
US12042517B2 (en) | 2017-09-08 | 2024-07-23 | New Portal Limited | Nucleic acid systems that enable bacteria to specifically target solid tumors via glucose-dependent viability |
US11696931B2 (en) | 2017-09-08 | 2023-07-11 | New Portal Limited | Bacteria for targeting tumors and treating cancer |
JP2021505197A (en) * | 2017-12-05 | 2021-02-18 | バイオピーエルエックス,インコーポレイティド | Methods and compositions to prevent microbial infections |
JP7460531B2 (en) | 2017-12-05 | 2024-04-02 | バイオピーエルエックス,インコーポレイティド | Methods and compositions for preventing microbial infections - Patents.com |
US12097227B2 (en) | 2018-02-08 | 2024-09-24 | Md Healthcare Inc. | Nanovesicles derived from bacteria of Lactococcus genus and use thereof |
EP3751008A4 (en) * | 2018-02-08 | 2021-11-17 | MD Healthcare Inc. | Nanovesicles derived from bacteria of lactococcus genus and use thereof |
EP3763830A4 (en) * | 2018-03-05 | 2021-12-08 | MD Healthcare Inc. | Nanovesicles derived from lactobacillus sp. bacteria, and use thereof |
EP3920908A4 (en) * | 2019-02-08 | 2022-10-05 | McMaster University | Use of activators of the aryl hydrocarbon receptor for treating gluten-induced gastrointestinal diseases |
WO2020223345A1 (en) | 2019-04-29 | 2020-11-05 | Antipov Eugene | Enumeration of genetically engineered microorganisms by live cell counting techniques |
US12098372B2 (en) | 2019-12-30 | 2024-09-24 | Eligo Bioscience | Microbiome modulation of a host by delivery of DNA payloads with minimized spread |
US11746352B2 (en) | 2019-12-30 | 2023-09-05 | Eligo Bioscience | Microbiome modulation of a host by delivery of DNA payloads with minimized spread |
EP4125988A4 (en) * | 2020-03-26 | 2024-04-24 | The Regents of University of California | Detection and treatment of intestinal fibrosis |
WO2022120028A2 (en) | 2020-12-02 | 2022-06-09 | Synlogic Operating Company, Inc. | Engineered microorganisms |
WO2022144381A1 (en) * | 2020-12-30 | 2022-07-07 | Eligo Bioscience | Microbiome modulation of a host by delivery of dna payloads with minimized spread |
WO2022204406A1 (en) * | 2021-03-24 | 2022-09-29 | Synlogic Operating Company, Inc. | Bacteria engineered to treat disorders in which oxalate is detrimental |
WO2022221273A1 (en) | 2021-04-13 | 2022-10-20 | Synlogic Operating Company, Inc. | Bacteria engineered to secrete active proteins |
US12076375B2 (en) | 2022-06-29 | 2024-09-03 | Snipr Biome Aps | Treating and preventing E coli infections |
WO2024081768A1 (en) | 2022-10-12 | 2024-04-18 | Synlogic Operating Company, Inc. | Bacteria engineered to produce active epidermal growth factor (egf) and their medical uses |
Also Published As
Publication number | Publication date |
---|---|
US20230043588A1 (en) | 2023-02-09 |
SG11201707025WA (en) | 2017-09-28 |
JP7095993B2 (en) | 2022-07-05 |
CA2978315A1 (en) | 2016-09-09 |
IL254226A0 (en) | 2017-10-31 |
HK1250244A1 (en) | 2018-12-07 |
KR20170121291A (en) | 2017-11-01 |
BR112017018656B1 (en) | 2021-11-30 |
EP3265105A1 (en) | 2018-01-10 |
AU2016226234A1 (en) | 2017-09-21 |
RU2017130462A (en) | 2019-04-02 |
IL254226B (en) | 2022-01-01 |
IL288870A (en) | 2022-02-01 |
JP2018512841A (en) | 2018-05-24 |
WO2016141108A8 (en) | 2017-12-21 |
BR112017018656A2 (en) | 2018-04-17 |
AU2022203178A1 (en) | 2022-06-02 |
MX2017011037A (en) | 2018-03-02 |
ZA201705873B (en) | 2023-01-25 |
AU2016226234B2 (en) | 2022-02-17 |
RU2017130462A3 (en) | 2019-09-23 |
CN107636146A (en) | 2018-01-26 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2016226234B2 (en) | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier | |
US10273489B2 (en) | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier | |
US11685925B2 (en) | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier | |
US20240110192A1 (en) | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier | |
AU2018290278B2 (en) | Bacteria for the treatment of disorders | |
AU2016276973B2 (en) | Bacteria engineered to treat disorders involving the catabolism of a branched chain amino acid | |
JP7494345B2 (en) | Bacteria engineered to reduce hyperphenylalaninemia | |
WO2016210373A2 (en) | Recombinant bacteria engineered for biosafety, pharmaceutical compositions, and methods of use thereof | |
AU2017213646A1 (en) | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier | |
JP2018532412A5 (en) | ||
JP2018515106A (en) | Bacteria engineered to reduce hyperphenylalaninemia | |
WO2016210384A2 (en) | Bacteria engineered to treat metabolic diseases | |
JP2018535678A5 (en) | ||
WO2017123610A2 (en) | Bacteria engineered to detoxify deleterious molecules | |
US20220168362A1 (en) | Bacteria engineered to treat disorders involving the catabolism of a branched chain amino acid | |
JP2018521674A (en) | Bacteria engineered to treat disorders with propionate catabolism | |
US20210161976A1 (en) | Bacteria engineered to treat diseases that benefit from reduced gut inflammation and/or tightened gut mucosal barrier |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
WWE | Wipo information: entry into national phase |
Ref document number: 15301230 Country of ref document: US |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 16710574 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 11201707025W Country of ref document: SG Ref document number: MX/A/2017/011037 Country of ref document: MX |
|
ENP | Entry into the national phase |
Ref document number: 2017545655 Country of ref document: JP Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 2978315 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: 254226 Country of ref document: IL |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112017018656 Country of ref document: BR |
|
ENP | Entry into the national phase |
Ref document number: 2016226234 Country of ref document: AU Date of ref document: 20160302 Kind code of ref document: A |
|
REEP | Request for entry into the european phase |
Ref document number: 2016710574 Country of ref document: EP |
|
ENP | Entry into the national phase |
Ref document number: 20177028200 Country of ref document: KR Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2017130462 Country of ref document: RU |
|
ENP | Entry into the national phase |
Ref document number: 112017018656 Country of ref document: BR Kind code of ref document: A2 Effective date: 20170830 |