1
|
Bahramimehr F, Guthart A, Kurz S, Hai Y, Dawood M, Yücer R, Shahhamzehei N, Weiskirchen R, Roth W, Stremmel W, Bringmann G, Efferth T. Drug Repurposing to Inhibit Oncostatin M in Crohn's Disease. Molecules 2025; 30:1897. [PMID: 40363705 PMCID: PMC12073679 DOI: 10.3390/molecules30091897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Crohn's disease is an inflammatory bowel disease (IBD) that currently lacks satisfactory treatment options. Therefore, new targets for new drugs are urgently needed to combat this disease. In the present study, we investigated the transcriptomics-based mRNA expression of intestinal biopsies from patients with Crohn's disease. We compared the mRNA expression profiles of the ileum and colon of patients with those of healthy individuals. A total of 72 genes in the ileum and 33 genes in the colon were differentially regulated. Among these, six genes were overexpressed in both tissues, including IL1B, TCL1A, HCAR3, IGHG1, S100AB, and OSM. We further focused on OSM/oncostatin M. To confirm the responsiveness of intestinal tissues from patients with Crohn's disease to oncostatin M inhibition, we examined the expression of the oncostatin M using immunohistochemistry in patient biopsies as well as in kindlin-1-/- and kindlin-2-/- knockout mice, which exhibit an inflammatory bowel disease (IBD) phenotype, and found strong oncostatin M expression in all samples examined. Next, we conducted a drug-repurposing study using the supercomputer MOGON and bioinformatic methods. A total of 13 candidate compounds out of 1577 FDA-approved drugs were identified by PyRx-based virtual drug screening and AutoDock-based molecular docking. Their lowest binding energies (LBEs) ranged from -10.46 (±0.08) to -8.77 (±0.08) kcal/mol, and their predicted inhibition constants (pKi) ranged from 21.62 (±2.97) to 373.78 (±36.78) nM. Ecamsule has an interesting stereostructure with two C2-symmetric enantiomers (1S,4R-1'S,4'R and 1R,4S-1'R,4'S) (1a and 1b) and one meso diastereomer (1S,4R-1'R,4'S) (1c). These three stereoisomers showed strong, albeit differing, binding affinities in molecular docking. As examined by nuclear magnetic resonance and polarimetry, the 1S,4R-1'S,4'R isomer was the stereoisomer present in our commercially available preparations used for microscale thermophoresis. Ecamsule (1a) was chosen for in vitro validation using recombinant oncostatin M and microscale thermophoresis. Considerable dissociation constants were obtained for ecamsule after three repetitions with a Kd value of 11.36 ± 2.83 µM. Subsequently, we evaluated, by qRT-PCR, the efficacy of ecamsule (1a) as a potential drug that could prevent oncostatin M activation by inhibiting downstream inflammatory marker genes (IL6, TNFA, and CXCL11). In conclusion, we have identified oncostatin M as a promising new drug target for Crohn's disease through transcriptomics and ecamsule as a potential new drug candidate for Crohn's disease through a drug-repurposing approach both in silico and in vitro.
Collapse
Affiliation(s)
- Faranak Bahramimehr
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
| | - Axel Guthart
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
| | - Stefanie Kurz
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
| | - Yuanping Hai
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
| | - Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
- Department of Molecular Biology, Faculty of Medical Laboratory Science, Al-Neelain University, Khartoum 11121, Sudan
| | - Rümeysa Yücer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
| | - Nasim Shahhamzehei
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany;
| | - Wilfried Roth
- Institute of Pathology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Wolfgang Stremmel
- Clinic for Internal Medicine, Beethovenstraße 2, 76530 Baden-Baden, Germany;
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany;
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (F.B.); (A.G.); (S.K.); (Y.H.); (M.D.); (R.Y.); (N.S.)
| |
Collapse
|
2
|
Garrigue A, Kermasson L, Susini S, Fert I, Mahony CB, Sadek H, Luce S, Chouteau M, Cavazzana M, Six E, Le Bousse-Kerdilès MC, Anginot A, Souraud JB, Cormier-Daire V, Willems M, Sirvent A, Russello J, Callebaut I, André I, Bertrand JY, Lagresle-Peyrou C, Revy P. Human oncostatin M deficiency underlies an inherited severe bone marrow failure syndrome. J Clin Invest 2025; 135:e180981. [PMID: 39847438 PMCID: PMC11910226 DOI: 10.1172/jci180981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
Oncostatin M (OSM) is a cytokine with the unique ability to interact with both the OSM receptor (OSMR) and the leukemia inhibitory factor receptor (LIFR). On the other hand, OSMR interacts with IL31RA to form the interleukin-31 receptor. This intricate network of cytokines and receptors makes it difficult to understand the specific function of OSM. While monoallelic loss-of-function (LoF) mutations in OSMR underlie autosomal dominant familial primary localized cutaneous amyloidosis, the in vivo consequences of human OSM deficiency have never been reported so far. Here, we identified 3 young individuals from a consanguineous family presenting with inherited severe bone marrow failure syndromes (IBMFS) characterized by profound anemia, thrombocytopenia, and neutropenia. Genetic analysis revealed a homozygous 1 base-pair insertion in the sequence of OSM associated with the disease. Structural and functional analyses showed that this variant causes a frameshift that replaces the C-terminal portion of OSM, which contains the FxxK motif that interacts with both OSMR and LIFR, with a neopeptide. The lack of detection and signaling of the mutant OSM suggests a LoF mutation. Analysis of zebrafish models further supported the role of the OSM/OSMR signaling in erythroid progenitor proliferation and neutrophil differentiation. Our study provides the previously uncharacterized and unexpectedly limited in vivo consequence of OSM deficiency in humans.
Collapse
Affiliation(s)
- Alexandrine Garrigue
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Laëtitia Kermasson
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Équipe Labellisée LIGUE 2023, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Sandrine Susini
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Ingrid Fert
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Christopher B. Mahony
- University of Geneva, Faculty of Medicine, Department of Pathology and Immunology, Geneva, Switzerland; Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Hanem Sadek
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Sonia Luce
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Myriam Chouteau
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Marina Cavazzana
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
- Service de Biothérapie et d’Aphérèse, Hôpital Necker, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Emmanuelle Six
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | | | - Adrienne Anginot
- INSERM UMRS-MD 1197, Université de Paris-Saclay, Hôpital Paul Brousse, Villejuif, France
| | - Jean-Baptiste Souraud
- Service Anatomo-Pathologie, Hôpital d’Instruction des Armées Begin, Saint-Mandé, France
| | - Valérie Cormier-Daire
- Paris Cité University, Imagine Institute, Paris, France
- Reference Center for Skeletal Dysplasia, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marjolaine Willems
- Medical Genetics Department, CHU de Montpellier, Montpellier, France
- Department of Pediatric Oncology and Haematology, Montpellier Hospital, Montpellier, France
| | - Anne Sirvent
- Medical Genetics Department, CHU de Montpellier, Montpellier, France
| | - Jennifer Russello
- Service d’Hématologie Biologique, CHU de Montpellier, Montpellier, France
| | - Isabelle Callebaut
- Sorbonne Université, Muséum National d’Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, Paris 75005, France
| | - Isabelle André
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Julien Y. Bertrand
- University of Geneva, Faculty of Medicine, Department of Pathology and Immunology, Geneva, Switzerland; Geneva Centre for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Chantal Lagresle-Peyrou
- Institut Imagine, Université Paris Cité, INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
- Centre d’Investigation Clinique Biothérapie, Groupe Hospitalier Universitaire Ouest, AP-HP, Paris, France
| | - Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Équipe Labellisée LIGUE 2023, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
3
|
Mori T, Nagaraj NR, Surico PL, Zhou W, Parmar UPS, D’Esposito F, Gagliano C, Musa M, Zeppieri M. The therapeutic potential of targeting Oncostatin M and the interleukin-6 family in retinal diseases: A comprehensive review. Open Life Sci 2024; 19:20221023. [PMID: 39759107 PMCID: PMC11699559 DOI: 10.1515/biol-2022-1023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/29/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Retinal diseases, which can lead to significant vision loss, are complex conditions involving various cellular and molecular mechanisms. The interleukin-6 (IL-6) family, particularly Oncostatin M (OSM), has garnered attention for their roles in retinal inflammation, angiogenesis, and neuroprotection. This comprehensive review explores the dual nature of OSM and other IL-6 family members in retinal pathophysiology, highlighting their contribution to both degenerative and regenerative processes. The review also examines current research on OSM's interaction with key signaling pathways and discusses the potential of OSM and the IL-6 family as potential therapeutic targets. Understanding these mechanisms could lead to innovative treatments that modulate OSM activity, offering new avenues for managing retinal diseases and contributing to the development of more effective interventions.
Collapse
Affiliation(s)
- Tommaso Mori
- Department of Pathology, University of California, San Diego, La Jolla, CA, 92093, United States of America
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome, 00128, Italy
| | | | - Pier Luigi Surico
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome, 00128, Italy
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Wenjing Zhou
- Department of Pathology, University of California, San Diego, La Jolla, CA, 92093, United States of America
| | - Uday Pratap Singh Parmar
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, 02114, United States of America
- Department of Ophthalmology, Government Medical College and Hospital, Chandigarh, 160030, India
| | - Fabiana D’Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, London, NW1 5QH, United Kingdom
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, Piazza dell’Università, 94100, Enna, EN, Italy
- Eye Clinic Catania University San Marco Hospital, Viale Carlo Azeglio Ciampi, 95121, Catania, Italy
| | - Mutali Musa
- Department of Optometry, University of Benin, Benin City, 300238, Edo State, Nigeria
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100, Udine, Italy
| |
Collapse
|
4
|
Zhou Y, Stevis PE, Cao J, Ehrlich G, Jones J, Rafique A, Sleeman MW, Olson WC, Franklin MC. Structures of complete extracellular assemblies of type I and type II Oncostatin M receptor complexes. Nat Commun 2024; 15:9776. [PMID: 39532904 PMCID: PMC11557873 DOI: 10.1038/s41467-024-54124-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Oncostatin M (OSM) is a unique Interleukin 6 (IL-6) family cytokine that plays pivotal roles in numerous biological events by signaling via two types of receptor complexes. While type I OSM receptor complex is formed by glycoprotein 130 (gp130) heterodimerization with Leukemia Inhibitory Factor receptor (LIFR), type II OSM receptor complex is composed of gp130 and OSM receptor (OSMR). OSM is an important contributor to multiple inflammatory diseases and cancers while OSM inhibition has been shown to be effective at reducing symptoms, making OSM an attractive therapeutic target. Using cryogenic electron microscopy (cryo-EM), we characterize full extracellular assemblies of human type I OSM receptor complex and mouse type II OSM receptor complex. The juxtamembrane domains of both complexes are situated in close proximity due to acute bends of the receptors. The rigid N-terminal extension of OSM contributes to gp130 binding and OSM signaling. Neither glycosylation nor pro-domain cleavage of OSM affects its activity. Mutagenesis identifies multiple OSM and OSMR residues crucial for complex formation and signaling. Our data reveal the structural basis for the assemblies of both type I and type II OSM receptor complexes and provide insights for modulation of OSM signaling in therapeutics.
Collapse
Affiliation(s)
- Yi Zhou
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA.
| | | | - Jing Cao
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - George Ehrlich
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Jennifer Jones
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | - Mark W Sleeman
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | | |
Collapse
|
5
|
Rafii P, Cruz PR, Ettich J, Seibel C, Padrini G, Wittich C, Lang A, Petzsch P, Köhrer K, Moll JM, Floss DM, Scheller J. Engineered interleukin-6-derived cytokines recruit artificial receptor complexes and disclose CNTF signaling via the OSMR. J Biol Chem 2024; 300:107251. [PMID: 38569939 PMCID: PMC11039321 DOI: 10.1016/j.jbc.2024.107251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024] Open
Abstract
Ciliary neurotrophic factor (CNTF) activates cells via the non-signaling α-receptor CNTF receptor (CNTFR) and the two signaling β-receptors glycoprotein 130 (gp130) and leukemia inhibitory factor receptor (LIFR). The CNTF derivate, Axokine, was protective against obesity and insulin resistance, but clinical development was halted by the emergence of CNTF antibodies. The chimeric cytokine IC7 used the framework of interleukin (IL-)6 with the LIFR-binding site from CNTF to activate cells via IL-6R:gp130:LIFR complexes. Similar to CNTF/Axokine, IC7 protected mice from obesity and insulin resistance. Here, we developed CNTF-independent chimeras that specifically target the IL-6R:gp130:LIFR complex. In GIL-6 and GIO-6, we transferred the LIFR binding site from LIF or OSM to IL-6, respectively. While GIO-6 signals via gp130:IL-6R:LIFR and gp130:IL-6R:OSMR complexes, GIL-6 selectively activates the IL-6R:gp130:LIFR receptor complex. By re-evaluation of IC7 and CNTF, we discovered the Oncostatin M receptor (OSMR) as an alternative non-canonical high-affinity receptor leading to IL-6R:OSMR:gp130 and CNTFR:OSMR:gp130 receptor complexes, respectively. The discovery of OSMR as an alternative high-affinity receptor for IC7 and CNTF designates GIL-6 as the first truly selective IL-6R:gp130:LIFR cytokine, whereas GIO-6 is a CNTF-free alternative for IC7.
Collapse
Affiliation(s)
- Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patricia Rodrigues Cruz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christiane Seibel
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Giacomo Padrini
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christoph Wittich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Alexander Lang
- Division of Cardiology, Pulmonology, and Vascular Medicine, Cardiovascular Research Laboratory, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
6
|
Widjaja AA, Cook SA. Nonspecific Inhibition of IL6 Family Cytokine Signalling by Soluble gp130. Int J Mol Sci 2024; 25:1363. [PMID: 38338642 PMCID: PMC10855816 DOI: 10.3390/ijms25031363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
IL6 is a proinflammatory cytokine that binds to membrane-bound IL6 receptor (IL6R) or soluble IL6R to signal via gp130 in cis or trans, respectively. We tested the hypothesis that sgp130Fc, which is believed to be a selective IL6 trans-signalling inhibitor, is in fact a non-specific inhibitor of gp130 signalling. In human cancer and primary cells, sgp130Fc inhibited IL6, IL11, OSM and CT1 cis-signalling. The IC50 values of sgp130Fc for IL6 and OSM cis-signalling were markedly (20- to 200-fold) lower than the concentrations of sgp130Fc used in mouse studies and clinical trials. sgp130 inhibited IL6 and OSM signalling in the presence of an ADAM10/17 inhibitor and the absence of soluble IL6R or OSMR, with effects that were indistinguishable from those of a gp130 neutralising antibody. These data show that sgp130Fc does not exclusively block IL6 trans-signalling and reveal instead that broad inhibition of gp130 signalling likely underlies its therapeutic effects. This proposes global or modular inhibition of gp130 as a therapeutic approach for treating human disease.
Collapse
Affiliation(s)
- Anissa A. Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
| | - Stuart A. Cook
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore 169609, Singapore
- MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W6 8RF, UK
| |
Collapse
|
7
|
Han L, Yan J, Li T, Lin W, Huang Y, Shen P, Ba X, Huang Y, Qin K, Geng Y, Wang H, Zheng K, Liu Y, Wang Y, Chen Z, Tu S. Multifaceted oncostatin M: novel roles and therapeutic potential of the oncostatin M signaling in rheumatoid arthritis. Front Immunol 2023; 14:1258765. [PMID: 38022540 PMCID: PMC10654622 DOI: 10.3389/fimmu.2023.1258765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Rheumatoid arthritis (RA) is a self-immune inflammatory disease characterized by joint damage. A series of cytokines are involved in the development of RA. Oncostatin M (OSM) is a pleiotropic cytokine that primarily activates the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway, the mitogen-activated protein kinase (MAPK) signaling pathway, and other physiological processes such as cell proliferation, inflammatory response, immune response, and hematopoiesis through its receptor complex. In this review, we first describe the characteristics of OSM and its receptor, and the biological functions of OSM signaling. Subsequently, we discuss the possible roles of OSM in the development of RA from clinical and basic research perspectives. Finally, we summarize the progress of clinical studies targeting OSM for the treatment of RA. This review provides researchers with a systematic understanding of the role of OSM signaling in RA, which can guide the development of drugs targeting OSM for the treatment of RA.
Collapse
Affiliation(s)
- Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Yan
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiji Lin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Shen
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xin Ba
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinhong Geng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huanhuan Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaifeng Zheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yafei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Wolf CL, Pruett C, Lighter D, Jorcyk CL. The clinical relevance of OSM in inflammatory diseases: a comprehensive review. Front Immunol 2023; 14:1239732. [PMID: 37841259 PMCID: PMC10570509 DOI: 10.3389/fimmu.2023.1239732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/30/2023] [Indexed: 10/17/2023] Open
Abstract
Oncostatin M (OSM) is a pleiotropic cytokine involved in a variety of inflammatory responses such as wound healing, liver regeneration, and bone remodeling. As a member of the interleukin-6 (IL-6) family of cytokines, OSM binds the shared receptor gp130, recruits either OSMRβ or LIFRβ, and activates a variety of signaling pathways including the JAK/STAT, MAPK, JNK, and PI3K/AKT pathways. Since its discovery in 1986, OSM has been identified as a significant contributor to a multitude of inflammatory diseases, including arthritis, inflammatory bowel disease, lung and skin disease, cardiovascular disease, and most recently, COVID-19. Additionally, OSM has also been extensively studied in the context of several cancer types including breast, cervical, ovarian, testicular, colon and gastrointestinal, brain,lung, skin, as well as other cancers. While OSM has been recognized as a significant contributor for each of these diseases, and studies have shown OSM inhibition is effective at treating or reducing symptoms, very few therapeutics have succeeded into clinical trials, and none have yet been approved by the FDA for treatment. In this review, we outline the role OSM plays in a variety of inflammatory diseases, including cancer, and outline the previous and current strategies for developing an inhibitor for OSM signaling.
Collapse
Affiliation(s)
- Cody L. Wolf
- Department of Biomolecular Sciences, Boise State University, Boise, ID, United States
| | - Clyde Pruett
- Department of Biological Sciences, Boise State University, Boise, ID, United States
| | - Darren Lighter
- Department of Biological Sciences, Boise State University, Boise, ID, United States
| | - Cheryl L. Jorcyk
- Department of Biomolecular Sciences, Boise State University, Boise, ID, United States
- Department of Biological Sciences, Boise State University, Boise, ID, United States
| |
Collapse
|
9
|
Zheng Y, Zhang J, Guo T, Cao J, Wang L, Zhang J, Pang X, Gao F, Sun H, Xiao H. Canine interleukin-31 binds directly to OSMRβ with higher binding affinity than to IL-31RA. 3 Biotech 2023; 13:302. [PMID: 37588794 PMCID: PMC10425310 DOI: 10.1007/s13205-023-03724-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023] Open
Abstract
Interleukin-31 (IL-31) is a pro-inflammatory cytokine involved in skin inflammation and tumor progression. The IL-31 signaling cascade is initiated by its binding to two receptors, IL-31 receptor alpha (IL-31RA) and oncostatin M receptor subunit beta (OSMRβ). The previous study suggested that human IL-31 (hIL-31) directly interacts with IL-31RA and OSMRβ, independently, but the binding ability of hIL-31 to IL-31RA is stronger than to OSMRβ. In different to its human ortholog, feline IL-31 (fIL-31) has a higher binding affinity for feline OSMRβ. However, the binding pattern of canine IL-31 to its receptors remains to be elucidated. In this study, we purified the recombinant canine IL-31 (rcIL-31) protein and revealed its secondary structure to be mainly composed of alpha-helices. Moreover, in vitro studies show that rcIL-31 has the ability to induce the phosphorylation of signal transducer activator of transcription 3 (STAT3) and STAT5 in DH-82 cells. In the following, the binding efficacies of bioactive rcIL-31 for its individual receptor components have been measured using a flow cytometry assay. The result demonstrates that correctly refolded rcIL-31 binds independently with cIL-31RA and cOSMRβ which were expressed on the cell surface. Of note, rcIL-31 has a greater than tenfold higher affinity to OSMRβ than to IL-31RA. Additionally, we demonstrated that D1-D4, especially D4 of cOSMRβ, is crucial for its binding to cIL-31. Furthermore, this study proved that rcIL-31 has a high binding affinity to the soluble cOSMRβ with a KD value of 3.59 × 10-8 M. The results presented in the current study will have a significant implication in the development of drugs or antibodies against diseases induced by cIL-31 signaling.
Collapse
Affiliation(s)
- Yuxin Zheng
- College of Biological Engineering, Tianjin University of Science and Technology, Tianjin, 300457 China
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Jing Zhang
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Tianling Guo
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Jin Cao
- College of Biological Engineering, Tianjin University of Science and Technology, Tianjin, 300457 China
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Lixian Wang
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Jie Zhang
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Xuefei Pang
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Feng Gao
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| | - Hua Sun
- College of Biological Engineering, Tianjin University of Science and Technology, Tianjin, 300457 China
| | - Haixia Xiao
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308 China
- National Technology Innovation Center of Synthetic Biology, Tianjin, 300308 China
| |
Collapse
|
10
|
Toyama T, Xu S, Kanemitsu Y, Hasegawa T, Noguchi T, Lee JY, Matsuzawa A, Naganuma A, Hwang GW. Methylmercury directly modifies the 105th cysteine residue in oncostatin M to promote binding to tumor necrosis factor receptor 3 and inhibit cell growth. Arch Toxicol 2023; 97:1887-1897. [PMID: 37193757 DOI: 10.1007/s00204-023-03520-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/11/2023] [Indexed: 05/18/2023]
Abstract
We previously found that methylmercury induces expression of oncostatin M (OSM), which is released extracellularly and binds to tumor necrosis factor receptor 3 (TNFR3), possibly enhancing its own toxicity. However, the mechanism by which methylmercury causes OSM to bind to TNFR3 rather than to its known receptors, OSM receptor and LIFR, is unknown. In this study, we aimed to elucidate the effect of methylmercury modification of cysteine residues in OSM on binding to TNFR3. Immunostaining of TNFR3-V5-expressing cells suggested that methylmercury promoted binding of OSM to TNFR3 on the cell membrane. In an in vitro binding assay, OSM directly bound to the extracellular domain of TNFR3, and this binding was promoted by methylmercury. Additionally, the formation of a disulfide bond in the OSM molecule was essential for the binding of both proteins, and LC/MS analysis revealed that methylmercury directly modified the 105th cysteine residue (Cys105) in OSM. Next, mutant OSM, in which Cys105 was replaced by serine or methionine, increased the binding to TNFR3, and a similar effect was observed in immunoprecipitation using cultured cells. Furthermore, cell proliferation was inhibited by treatment with Cys105 mutant OSMs compared with wildtype OSM, and this effect was cancelled by TNFR3 knockdown. In conclusion, we revealed a novel mechanism of methylmercury toxicity, in which methylmercury directly modifies Cys105 in OSM, thereby inhibiting cell proliferation via promoting binding to TNFR3. This indicates a chemical disruption in the interaction between the ligand and the receptor is a part of methylmercury toxicity.
Collapse
Affiliation(s)
- Takashi Toyama
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Sidi Xu
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Yoshitomi Kanemitsu
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, 2-5274 Gakkochodori, Chuo-ku, Niigata, Niigata, 980-8574, Japan
| | - Takashi Hasegawa
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 951-8514, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Akira Naganuma
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Gi-Wook Hwang
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan.
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan.
| |
Collapse
|
11
|
Nada H, Sivaraman A, Lu Q, Min K, Kim S, Goo JI, Choi Y, Lee K. Perspective for Discovery of Small Molecule IL-6 Inhibitors through Study of Structure–Activity Relationships and Molecular Docking. J Med Chem 2023; 66:4417-4433. [PMID: 36971365 DOI: 10.1021/acs.jmedchem.2c01957] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Interleukin-6 (IL-6) is a proinflammatory cytokine that plays a key role in the pathogenesis and physiology of inflammatory and autoimmune diseases, such as coronary heart disease, cancer, Alzheimer's disease, asthma, rheumatoid arthritis, and most recently COVID-19. IL-6 and its signaling pathway are promising targets in the treatment of inflammatory and autoimmune diseases. Although, anti-IL-6 monoclonal antibodies are currently being used in clinics, huge unmet medical needs remain because of the high cost, administration-related toxicity, lack of opportunity for oral dosing, and potential immunogenicity of monoclonal antibody therapy. Furthermore, nonresponse or loss of response to monoclonal antibody therapy has been reported, which increases the importance of optimizing drug therapy with small molecule drugs. This work aims to provide a perspective for the discovery of novel small molecule IL-6 inhibitors by the analysis of the structure-activity relationships and computational studies for protein-protein inhibitors targeting the IL-6/IL-6 receptor/gp130 complex.
Collapse
|
12
|
Zhou Y, Stevis PE, Cao J, Saotome K, Wu J, Glatman Zaretsky A, Haxhinasto S, Yancopoulos GD, Murphy AJ, Sleeman MW, Olson WC, Franklin MC. Structural insights into the assembly of gp130 family cytokine signaling complexes. SCIENCE ADVANCES 2023; 9:eade4395. [PMID: 36930708 PMCID: PMC10022904 DOI: 10.1126/sciadv.ade4395] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
The interleukin-6 (IL-6) family cytokines signal through gp130 receptor homodimerization or heterodimerization with a second signaling receptor and play crucial roles in various cellular processes. We determined cryo-electron microscopy structures of five signaling complexes of this family, containing full receptor ectodomains bound to their respective ligands ciliary neurotrophic factor, cardiotrophin-like cytokine factor 1 (CLCF1), leukemia inhibitory factor, IL-27, and IL-6. Our structures collectively reveal similarities and differences in the assembly of these complexes. The acute bends at both signaling receptors in all complexes bring the membrane-proximal domains to a ~30 angstrom range but with distinct distances and orientations. We also reveal how CLCF1 engages its secretion chaperone cytokine receptor-like factor 1. Our data provide valuable insights for therapeutically targeting gp130-mediated signaling.
Collapse
Affiliation(s)
- Yi Zhou
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | - Jing Cao
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Kei Saotome
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | - Jiaxi Wu
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Du Q, Tu G, Qian Y, Yang J, Yao X, Xue W. Unbiased molecular dynamics simulation of a first-in-class small molecule inhibitor binds to oncostatin M. Comput Biol Med 2023; 155:106709. [PMID: 36854228 DOI: 10.1016/j.compbiomed.2023.106709] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/08/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023]
Abstract
Small molecule inhibitors (SMIs) targeting oncostatin M (OSM) signaling pathway represent new therapeutics to combat cancer, inflammatory bowel disease (IBD) and CNS disease. Recently, the first-in-class SMI named SMI-10B that target OSM and block its interaction with receptor (OSMR) were reported. However, the binding pocket and interaction mode of the compound on OSM remain poorly understood, which hampering the rational design of SMIs that target OSM. Here, using SMI-10B as a probe, the multiple pockets on OSM for small molecules binding were extensively explored by unbiased molecular dynamics (MD) simulations. Then, the near-native structure of the complex was identified by molecular mechanics generalized Born surface area (MM/GBSA) binding energy funnel. Moreover, the binding stabilities of the protein-ligand complexes in near- and non-native conformations were verified by additional independent MD runs and absolute free energy perturbation (FEP) calculation. In summary, the unique feature of SMI-10B spontaneously binds to OSM characterized here not only provide detailed information for understanding the molecular mechanism of SMI-10B binding to OSM, but also will facilitate the rational design of novel and more potent SMIs to block OSM signaling.
Collapse
Affiliation(s)
- Qingqing Du
- Depart of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Gao Tu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Yan Qian
- Depart of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Jingyi Yang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaojun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China.
| |
Collapse
|
14
|
Ben Boubaker R, Tiss A, Henrion D, Chabbert M. Homology Modeling in the Twilight Zone: Improved Accuracy by Sequence Space Analysis. Methods Mol Biol 2023; 2627:1-23. [PMID: 36959439 DOI: 10.1007/978-1-0716-2974-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
The analysis of the relationship between sequence and structure similarities during the evolution of a protein family has revealed a limit of sequence divergence for which structural conservation can be confidently assumed and homology modeling is reliable. Below this limit, the twilight zone corresponds to sequence divergence for which homology modeling becomes increasingly difficult and requires specific methods. Either with conventional threading methods or with recent deep learning methods, such as AlphaFold, the challenge relies on the identification of a template that shares not only a common ancestor (homology) but also a conserved structure with the query. As both homology and structural conservation are transitive properties, mining of sequence databases followed by multidimensional scaling (MDS) of the query sequence space can reveal intermediary sequences to infer homology and structural conservation between the query and the template. Here, as a case study, we studied the plethodontid receptivity factor isoform 1 (PRF1) from Plethodon jordani, a member of a pheromone protein family present only in lungless salamanders and weakly related to cytokines of the IL6 family. A variety of conventional threading methods led to the cytokine CNTF as a template. Sequence mining, followed by phylogenetic and MDS analysis, provided missing links between PRF1 and CNTF and allowed reliable homology modeling. In addition, we compared automated models obtained from web servers to a customized model to show how modeling can be improved by expert information.
Collapse
Affiliation(s)
- Rym Ben Boubaker
- UMR CNRS 6015 - INSERM 1083, Laboratoire MITOVASC, Université d'Angers, Angers, France
| | - Asma Tiss
- UMR CNRS 6015 - INSERM 1083, Laboratoire MITOVASC, Université d'Angers, Angers, France
| | - Daniel Henrion
- UMR CNRS 6015 - INSERM 1083, Laboratoire MITOVASC, Université d'Angers, Angers, France
| | - Marie Chabbert
- UMR CNRS 6015 - INSERM 1083, Laboratoire MITOVASC, Université d'Angers, Angers, France.
| |
Collapse
|
15
|
Rankouhi TR, Keulen DV, Tempel D, Venhorst J. Oncostatin M: Risks and Benefits of a Novel Therapeutic Target for Atherosclerosis. Curr Drug Targets 2022; 23:1345-1369. [PMID: 35959619 DOI: 10.2174/1389450123666220811101032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) is a leading cause of death worldwide. It is predicted that approximately 23.6 million people will die from CVDs annually by 2030. Therefore, there is a great need for an effective therapeutic approach to combat this disease. The European Cardiovascular Target Discovery (CarTarDis) consortium identified Oncostatin M (OSM) as a potential therapeutic target for atherosclerosis. The benefits of modulating OSM - an interleukin (IL)-6 family cytokine - have since been studied for multiple indications. However, as decades of high attrition rates have stressed, the success of a drug target is determined by the fine balance between benefits and the risk of adverse events. Safety issues should therefore not be overlooked. OBJECTIVE In this review, a risk/benefit analysis is performed on OSM inhibition in the context of atherosclerosis treatment. First, OSM signaling characteristics and its role in atherosclerosis are described. Next, an overview of in vitro, in vivo, and clinical findings relating to both the benefits and risks of modulating OSM in major organ systems is provided. Based on OSM's biological function and expression profile as well as drug intervention studies, safety concerns of inhibiting this target have been identified, assessed, and ranked for the target population. CONCLUSION While OSM may be of therapeutic value in atherosclerosis, drug development should also focus on de-risking the herein identified major safety concerns: tissue remodeling, angiogenesis, bleeding, anemia, and NMDA- and glutamate-induced neurotoxicity. Close monitoring and/or exclusion of patients with various comorbidities may be required for optimal therapeutic benefit.
Collapse
Affiliation(s)
- Tanja Rouhani Rankouhi
- Department of Risk Analysis for Products in Development, TNO, Utrechtseweg 48, 3704 HE, Zeist, The Netherlands
| | - Daniëlle van Keulen
- SkylineDx BV, Science and Clinical Development, 3062 ME Rotterdam, The Netherlands
| | - Dennie Tempel
- SkylineDx BV, Science and Clinical Development, 3062 ME Rotterdam, The Netherlands
| | - Jennifer Venhorst
- Department of Risk Analysis for Products in Development, TNO, Utrechtseweg 48, 3704 HE, Zeist, The Netherlands
| |
Collapse
|
16
|
McCollum S, Kalivas A, Kirkham M, Kunz K, Okojie J, Pavek A, Barrott J. Oncostatin M Receptor as a Therapeutic Target for Radioimmune Therapy in Synovial Sarcoma. Pharmaceuticals (Basel) 2022; 15:ph15060650. [PMID: 35745569 PMCID: PMC9228444 DOI: 10.3390/ph15060650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/10/2022] Open
Abstract
Synovial sarcoma (SS) is a pediatric muscle cancer that primarily affects adolescents and young adults and has few treatment options. Complicating the treatment of synovial sarcoma is the low mutational burden of SS. Inflammatory pathways have been identified as being upregulated in some SS, leading to the discovery of upregulated oncostatin M receptor (OSMR). It was found that OSMR is upregulated in SS by RNAseq analysis and quantitative PCR, highlighting its potential in the treatment of SS. Also, OSMR is upregulated in mouse models for synovial sarcoma as demonstrated by western blot and immunohistochemistry, and the protein is present in both primary and metastatic sites of disease. Using a radioimmune therapy drug model, targeted therapy was synthesized for use in OSMR expressing SS and it was demonstrated that this drug is stable, while capable of efficient OSMR binding and isotope capture. Finally, this antibody conjugate exhibited ideal pharmacokinetics and targeted sites of disease in our mouse model and was taken up in both primary and metastatic diseased tissue. This suggests OSMR as an ideal target for therapy and this radioimmune therapy provides a novel treatment option for a disease with few therapy choices.
Collapse
|
17
|
de Souza PPC, Henning P, Lerner UH. Stimulation of Osteoclast Formation by Oncostatin M and the Role of WNT16 as a Negative Feedback Regulator. Int J Mol Sci 2022; 23:3287. [PMID: 35328707 PMCID: PMC8953253 DOI: 10.3390/ijms23063287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Oncostatin M (OSM), which belongs to the IL-6 family of cytokines, is the most potent and effective stimulator of osteoclast formation in this family, as assessed by different in vitro assays. Osteoclastogenesis induced by the IL-6 type of cytokines is mediated by the induction and paracrine stimulation of the osteoclastogenic cytokine receptor activator of nuclear factor κ-B ligand (RANKL), expressed on osteoblast cell membranes and targeting the receptor activator of nuclear factor κ-B (RANK) on osteoclast progenitor cells. The potent effect of OSM on osteoclastogenesis is due to an unusually robust induction of RANKL in osteoblasts through the OSM receptor (OSMR), mediated by a JAK-STAT/MAPK signaling pathway and by unique recruitment of the adapter protein Shc1 to the OSMR. Gene deletion of Osmr in mice results in decreased numbers of osteoclasts and enhanced trabecular bone caused by increased trabecular thickness, indicating that OSM may play a role in physiological regulation of bone remodeling. However, increased amounts of OSM, either through administration of recombinant protein or of adenoviral vectors expressing Osm, results in enhanced bone mass due to increased bone formation without any clear sign of increased osteoclast numbers, a finding which can be reconciled by cell culture experiments demonstrating that OSM can induce osteoblast differentiation and stimulate mineralization of bone nodules in such cultures. Thus, in vitro studies and gene deletion experiments show that OSM is a stimulator of osteoclast formation, whereas administration of OSM to mice shows that OSM is not a strong stimulator of osteoclastogenesis in vivo when administered to adult animals. These observations could be explained by our recent finding showing that OSM is a potent stimulator of the osteoclastogenesis inhibitor WNT16, acting in a negative feedback loop to reduce OSM-induced osteoclast formation.
Collapse
Affiliation(s)
- Pedro P. C. de Souza
- The Innovation in Biomaterials Laboratory, School of Dentistry, Federal University of Goiás, Goiânia 74690-900, Brazil;
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| | - Ulf H. Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| |
Collapse
|
18
|
Felcher CM, Bogni ES, Kordon EC. IL-6 Cytokine Family: A Putative Target for Breast Cancer Prevention and Treatment. Int J Mol Sci 2022; 23:ijms23031809. [PMID: 35163731 PMCID: PMC8836921 DOI: 10.3390/ijms23031809] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022] Open
Abstract
The IL-6 cytokine family is a group of signaling molecules with wide expression and function across vertebrates. Each member of the family signals by binding to its specific receptor and at least one molecule of gp130, which is the common transmembrane receptor subunit for the whole group. Signal transduction upon stimulation of the receptor complex results in the activation of multiple downstream cascades, among which, in mammary cells, the JAK-STAT3 pathway plays a central role. In this review, we summarize the role of the IL-6 cytokine family—specifically IL-6 itself, LIF, OSM, and IL-11—as relevant players during breast cancer progression. We have compiled evidence indicating that this group of soluble factors may be used for early and more precise breast cancer diagnosis and to design targeted therapy to treat or even prevent metastasis development, particularly to the bone. Expression profiles and possible therapeutic use of their specific receptors in the different breast cancer subtypes are also described. In addition, participation of these cytokines in pathologies of the breast linked to lactation and involution of the gland, as post-partum breast cancer and mastitis, is discussed.
Collapse
Affiliation(s)
- Carla M. Felcher
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires—Consejo Nacional de Investigaciones Científicas y Técnicas (IFIBYNE-UBA-CONICET), Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina; (C.M.F.); (E.S.B.)
| | - Emilia S. Bogni
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires—Consejo Nacional de Investigaciones Científicas y Técnicas (IFIBYNE-UBA-CONICET), Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina; (C.M.F.); (E.S.B.)
| | - Edith C. Kordon
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires—Consejo Nacional de Investigaciones Científicas y Técnicas (IFIBYNE-UBA-CONICET), Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina; (C.M.F.); (E.S.B.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina
- Correspondence:
| |
Collapse
|
19
|
Liu J, Zhong Y, Liu H, Yang H, Lu P, Shi Y, Wang X, Zheng W, Yu X, Xu Y, Yang B. Oncostatin M sensitizes keratinocytes to UVB-induced inflammation via GSDME-mediated pyroptosis. J Dermatol Sci 2021; 104:95-103. [PMID: 34674925 DOI: 10.1016/j.jdermsci.2021.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Oncostatin M (OSM), an interleukin-6 (IL-6) family proinflammatory cytokine, plays a critical role in inflammatory skin diseases, but its mechanism of action is not well understood. OBJECTIVE To demonstrate the mechanism of OSM induced pyropotosis in normal human epidermal keratinocytes (NHEKs) and immortalized human keratinocytes (HaCaT cells). METHODS NHEKs and HaCaT cells were treated with OSM. Knockout of OSM receptor (OSMR) with CRISPR/Cas9 system, knockdown of GSDME with small interfering RNA and primary keratinocytes from Osmr-/- and Gsdme-/- mice were used to study the effect of OSMR and GSDME. After treatment of OSM, NHEKs and HaCaT cells were irradiated with UVB. The mRNA was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) and RNA sequencing, protein level was detected by Western Blotting, Elisa and immunofluorescence. Cell death was examined by lactate dehydrogenase (LDH) releasing. RESULTS Here we found that OSM induced pyropotosis in NHEKs and HaCaT cells, but knockout of OSMR abolished pyropotosis. RNA sequencing revealed an upregulation of several key genes involved in NLRP3 inflammasome activation following OSM treatment, among which NLRP3, GSDME, and IL-1β were confirmed by qRT-PCR and Western Blotting. Knockdown of GSDME alleviated OSM-induced pyropotosis. Pretreatment of OSM boosted UVB-induced pyroptosis and inflammation in NHEKs and HaCaT cells, and this priming function was lost in keratinocytes of Osmr-/- and Gsdme-/- mice. Similar results were obtained in a 3-dimensional culture of human epidermis. CONCLUSION OSM functions as a priming cytokine to enhance UVB-induced inflammation in keratinocytes, providing insight into the pathogenesis of inflammatory skin diseases.
Collapse
Affiliation(s)
- Jun Liu
- Institute of Dermatology and Venereology, Dermatology Hospital, Southern Medical University, Guangzhou, China; Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China.
| | - Yadan Zhong
- Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China; Department of Dermatology, The First People's Hospital of Foshan, Foshan, China
| | - Huiting Liu
- Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Huan Yang
- Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Ping Lu
- Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yanqiang Shi
- Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Wang
- Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Wen Zheng
- Department of Science & Education, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoling Yu
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yingping Xu
- Institute of Dermatology and Venereology, Dermatology Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yang
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
20
|
Mass OA, Tuccinardi J, Woodbury L, Wolf CL, Grantham B, Holdaway K, Pu X, King MD, Warner DL, Jorcyk CL, Warner LR. Bioactive recombinant human oncostatin M for NMR-based screening in drug discovery. Sci Rep 2021; 11:16174. [PMID: 34376712 PMCID: PMC8355150 DOI: 10.1038/s41598-021-95424-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Oncostatin M (OSM) is a pleiotropic, interleukin-6 family inflammatory cytokine that plays an important role in inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis, and cancer progression and metastasis. Recently, elevated OSM levels have been found in the serum of COVID-19 patients in intensive care units. Multiple anti-OSM therapeutics have been investigated, but to date no OSM small molecule inhibitors are clinically available. To pursue a high-throughput screening and structure-based drug discovery strategy to design a small molecule inhibitor of OSM, milligram quantities of highly pure, bioactive OSM are required. Here, we developed a reliable protocol to produce highly pure unlabeled and isotope enriched OSM from E. coli for biochemical and NMR studies. High yields (ca. 10 mg/L culture) were obtained in rich and minimal defined media cultures. Purified OSM was characterized by mass spectrometry and circular dichroism. The bioactivity was confirmed by induction of OSM/OSM receptor signaling through STAT3 phosphorylation in human breast cancer cells. Optimized buffer conditions yielded 1H, 15N HSQC NMR spectra with intense, well-dispersed peaks. Titration of 15N OSM with a small molecule inhibitor showed chemical shift perturbations for several key residues with a binding affinity of 12.2 ± 3.9 μM. These results demonstrate the value of bioactive recombinant human OSM for NMR-based small molecule screening.
Collapse
Affiliation(s)
- Olga A. Mass
- grid.184764.80000 0001 0670 228XBiomoleculer Research Center, Boise State University, Boise, ID 83725 USA
| | - Joseph Tuccinardi
- grid.184764.80000 0001 0670 228XDepartment of Chemistry and Biochemistry, Boise State University, 1910 University Dr., Boise, ID 83725 USA
| | - Luke Woodbury
- grid.184764.80000 0001 0670 228XBiomoleculer Research Center, Boise State University, Boise, ID 83725 USA
| | - Cody L. Wolf
- grid.184764.80000 0001 0670 228XBiomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725 USA ,grid.184764.80000 0001 0670 228XDepartment of Biological Sciences, Boise State University, Boise, ID 83725 USA
| | - Bri Grantham
- grid.184764.80000 0001 0670 228XBiomoleculer Research Center, Boise State University, Boise, ID 83725 USA
| | - Kelsey Holdaway
- grid.184764.80000 0001 0670 228XDepartment of Chemistry and Biochemistry, Boise State University, 1910 University Dr., Boise, ID 83725 USA
| | - Xinzhu Pu
- grid.184764.80000 0001 0670 228XBiomoleculer Research Center, Boise State University, Boise, ID 83725 USA ,grid.184764.80000 0001 0670 228XBiomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725 USA
| | - Matthew D. King
- grid.184764.80000 0001 0670 228XDepartment of Chemistry and Biochemistry, Boise State University, 1910 University Dr., Boise, ID 83725 USA ,grid.184764.80000 0001 0670 228XBiomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725 USA
| | - Don L. Warner
- grid.184764.80000 0001 0670 228XDepartment of Chemistry and Biochemistry, Boise State University, 1910 University Dr., Boise, ID 83725 USA ,grid.184764.80000 0001 0670 228XBiomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725 USA
| | - Cheryl L. Jorcyk
- grid.184764.80000 0001 0670 228XBiomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725 USA ,grid.184764.80000 0001 0670 228XDepartment of Biological Sciences, Boise State University, Boise, ID 83725 USA
| | - Lisa R. Warner
- grid.184764.80000 0001 0670 228XBiomoleculer Research Center, Boise State University, Boise, ID 83725 USA ,grid.184764.80000 0001 0670 228XDepartment of Chemistry and Biochemistry, Boise State University, 1910 University Dr., Boise, ID 83725 USA ,grid.184764.80000 0001 0670 228XBiomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725 USA
| |
Collapse
|
21
|
Scheller J, Berg A, Moll JM, Floss DM, Jungesblut C. Current status and relevance of single nucleotide polymorphisms in IL-6-/IL-12-type cytokine receptors. Cytokine 2021; 148:155550. [PMID: 34217594 DOI: 10.1016/j.cyto.2021.155550] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 01/06/2023]
Abstract
Cytokines control immune related events and are critically involved in a plethora of patho-physiological processes including autoimmunity and cancer development. In rare cases, single nucleotide polymorphisms (SNPs) or single nucleotide variations (SNVs) in cytokine receptors eventually cause detrimental ligand-independent, constitutive activation of signal transduction. Most SNPs have, however, no or only marginal influences on gene expression, protein stability, localization and function and thereby only slightly affecting pathogenesis probability. The SNP database (dbSNP) is an archive for a broad collection of polymorphisms in which SNPs are categorized and marked with a locus accession number "reference SNP" (rs). Here, we engineered an algorithm to directly align dbSNP information to DNA and protein sequence information to clearly illustrate a genetic SNP landscape exemplified for all tall cytokine receptors of the IL-6/IL-12 family, including IL-23R, IL-12Rβ1, IL-12Rβ2, gp130, LIFR, OSMR and WSX-1. This information was complemented by a comprehensive literature summary and structural insights of relevant disease-causing SNPs in cytokine/cytokine receptor interfaces. In summary, we present a general strategy with potential to apply to other cytokine receptor networks.
Collapse
Affiliation(s)
- Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| | - Anna Berg
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | |
Collapse
|
22
|
Du Q, Qian Y, Xue W. Cross-reactivity of two human IL-6 family cytokines OSM and LIF explored by protein-protein docking and molecular dynamics simulation. Biochim Biophys Acta Gen Subj 2021; 1865:129907. [PMID: 33845142 DOI: 10.1016/j.bbagen.2021.129907] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are two important pro-inflammatory cytokines of the interleukin-6 (IL-6) family. The two cytokines mediated signaling was recently found to be closely associated with cancer and chronic inflammation, which represent promising therapeutic targets for the treatment of many solid tumors and inflammatory disease. As the most closely related members, cross-reactivity of them may result in undesired activation of off-target cells, leading to toxicity or lack of efficacy of the therapeutic effects. However, the mechanism of the cross-reactivity of OSM and LIF is not well understood. METHODS In this work, protein-protein docking, molecular dynamics (MD) simulations with explicit solvent and post endpoints binding free energy (BFE) analysis were carried out to further understand the structural and energetic principles of interactions between the two cytokines and the shared receptor LIFR. RESULTS For the first time, the simulation given a computational model of OSM-LIFR interaction, and provided significant insights into the mechanism of OSM and LIF cross-react with LIFR. The identified common features shared by OSM and LIF bind to LIFR involving 10 "conserved" residues (90% similarity) distributed at the binding site III comprised of AB loop, BC loop and D helix. In addition, 11 shared residues were identified in LIFR contribute 77.85% and 84.63% energies for OSM and LIF binding, which play a critical role in the formation of the two cytokine-receptor complexes. Moreover, the "nonconserved" residues at the same position of cytokines such as Asp41 in OSM and Pro51 in LIF as well as the three residues (Glu338, Asn201 and Glu260) in LIFR were also discovered. CONCLUSIONS These important information may facilitate the rational design of novel chemical or biological agents with less toxicity and improved efficacy.
Collapse
Affiliation(s)
- Qingqing Du
- Depart of Pharmacy, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yan Qian
- Depart of Pharmacy, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
23
|
Oncostatin M: A mysterious cytokine in cancers. Int Immunopharmacol 2020; 90:107158. [PMID: 33187910 DOI: 10.1016/j.intimp.2020.107158] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/04/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Oncostatin M (OSM), as a member of the Interleukin-6 family cytokines, plays a significant role in inflammation, autoimmunity, and cancers. It is mainly secreted by T lymphocytes, neutrophils, and macrophages and was initially introduced as anti-cancer agent. However, in some cases, it promotes cancer progression. Overexpression of OSM and OSM receptor has been detected in various cancers including colon cancer, breast cancer, pancreatic cancer, myeloma, brain tumors, chronic lymphocytic leukemia, and hepatoblastoma. STAT3 is the main downstream signaling molecule of OSM, which operates the leading role in modifications of cancer cells and enhancing cell growth, invasion, survival, and all other hallmarks of cancer cells. However, due to the presence of multiple signaling pathways, it can act contradictory in some cancers. In this review, we will discuss the emerging roles of OSM in cancer and elucidate its function in tumor control or progression and finally discuss therapeutic approaches designed to manipulate this cytokine in cancer.
Collapse
|
24
|
Metcalfe RD, Putoczki TL, Griffin MDW. Structural Understanding of Interleukin 6 Family Cytokine Signaling and Targeted Therapies: Focus on Interleukin 11. Front Immunol 2020; 11:1424. [PMID: 32765502 PMCID: PMC7378365 DOI: 10.3389/fimmu.2020.01424] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Cytokines are small signaling proteins that have central roles in inflammation and cell survival. In the half-century since the discovery of the first cytokines, the interferons, over fifty cytokines have been identified. Amongst these is interleukin (IL)-6, the first and prototypical member of the IL-6 family of cytokines, nearly all of which utilize the common signaling receptor, gp130. In the last decade, there have been numerous advances in our understanding of the structural mechanisms of IL-6 family signaling, particularly for IL-6 itself. However, our understanding of the detailed structural mechanisms underlying signaling by most IL-6 family members remains limited. With the emergence of new roles for IL-6 family cytokines in disease and, in particular, roles of IL-11 in cardiovascular disease, lung disease, and cancer, there is an emerging need to develop therapeutics that can progress to clinical use. Here we outline our current knowledge of the structural mechanism of signaling by the IL-6 family of cytokines. We discuss how this knowledge allows us to understand the mechanism of action of currently available inhibitors targeting IL-6 family cytokine signaling, and most importantly how it allows for improved opportunities to pharmacologically disrupt cytokine signaling. We focus specifically on the need to develop and understand inhibitors that disrupt IL-11 signaling.
Collapse
Affiliation(s)
- Riley D Metcalfe
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Technology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Tracy L Putoczki
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Michael D W Griffin
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Technology Institute, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
25
|
Du Q, Qian Y, Xue W. Molecular Simulation of Oncostatin M and Receptor (OSM-OSMR) Interaction as a Potential Therapeutic Target for Inflammatory Bowel Disease. Front Mol Biosci 2020; 7:29. [PMID: 32195265 PMCID: PMC7064634 DOI: 10.3389/fmolb.2020.00029] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/11/2020] [Indexed: 01/06/2023] Open
Abstract
Therapeutics targeting cytokines such as the oncostatin M (OSM)-mediated inflammation represent a potential strategy for the treatment of inflammatory bowel disease (IBD). Despite the investigation of the specific role of the interactions between OSM and the receptor (OSMR) in IBD pathogenesis, the 3D structure of the OSM–OSMR complex remains elusive. In this work, the interaction mode between OSM and OSMR at atomic level was predicted by computational simulation approach. The interaction domain of the OSMR was built with the homology modeling method. The near-native structure of the OSM–OSMR complex was obtained by docking, and long-time scale molecular dynamics (MD) simulation in an explicit solvent was further performed to sample the conformations when OSM binds to the OSMR. After getting the equilibrated states of the simulation system, per-residue energy contribution was calculated to characterize the important residues for the OSM–OSMR complex formation. Based on these important residues, eight residues (OSM: Arg100, Leu103, Phe160, and Gln161; OSMR: Tyr214, Ser223, Asp262, and Trp267) were identified as the “hot spots” through computational alanine mutagenesis analysis and verified by additional MD simulation of R100A (one of the identified “hotspots”) mutant. Moreover, six cavities were detected at the OSM–OSMR interface through the FTMap analysis, and they were suggested as important binding sites. The predicted 3D structure of the OSM–OSMR complex and the identified “hot spots” constituting the core of the binding interface provide helpful information in understanding the OSM–OSMR interactions, and the detected sites serve as promising targets in designing small molecules to block the interactions.
Collapse
Affiliation(s)
- Qingqing Du
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Qian
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, China
| |
Collapse
|
26
|
Murakami M, Kamimura D, Hirano T. Pleiotropy and Specificity: Insights from the Interleukin 6 Family of Cytokines. Immunity 2019; 50:812-831. [DOI: 10.1016/j.immuni.2019.03.027] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
|
27
|
Adrian-Segarra JM, Sreenivasan K, Gajawada P, Lörchner H, Braun T, Pöling J. The AB loop of oncostatin M (OSM) determines species-specific signaling in humans and mice. J Biol Chem 2018; 293:20181-20199. [PMID: 30373773 DOI: 10.1074/jbc.ra118.004375] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/12/2018] [Indexed: 11/06/2022] Open
Abstract
The pleiotropic interleukin-6 (IL-6)-type cytokine oncostatin M (OSM) signals in multiple cell types, affecting processes such as cell differentiation, hematopoiesis, and inflammation. In humans, OSM exerts its effects through activation of either of two different heterodimeric receptor complexes, formed by glycoprotein 130 (gp130) and either OSM receptor (OSMR) or leukemia inhibitory factor receptor (LIFR). In contrast, the mouse OSM orthologue acts mainly through dimers containing OSMR and gp130 and shows limited activity through mouse LIFR. Despite their structural similarity, neither human nor mouse OSM signal through the other species' OSMR. The molecular basis for such species-specific signaling, however, remains poorly understood. To identify key molecular features of OSM that determine receptor activation in humans and mice, we generated chimeric mouse-human cytokines. Replacing regions within binding site III of murine OSM with the human equivalents showed that the cytokine's AB loop was critical for receptor selection. Substitutions of individual amino acids within this region demonstrated that residues Asn-37, Thr-40, and Asp-42 of the murine cytokine were responsible for limited LIFR activation and absence of human OSMR/LIFR signaling. In human OSM, Lys-44 appeared to be the main residue preventing mouse OSMR activation. Our data reveal that individual amino acids within the AB loop of OSM determine species-specific activities. These mutations might reflect a key step in the evolutionary process of this cytokine, in which receptor promiscuity gives way to ligand-receptor specialization.
Collapse
Affiliation(s)
- Juan M Adrian-Segarra
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and
| | - Krishnamoorthy Sreenivasan
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and
| | - Praveen Gajawada
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and
| | - Holger Lörchner
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and; the German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Thomas Braun
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and; the German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.
| | - Jochen Pöling
- From the Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany and; the German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
28
|
West NR, Owens BMJ, Hegazy AN. The oncostatin M-stromal cell axis in health and disease. Scand J Immunol 2018; 88:e12694. [DOI: 10.1111/sji.12694] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Nathaniel R. West
- Department of Cancer Immunology; Genentech; South San Francisco California
| | - Benjamin M. J. Owens
- Somerville College; University of Oxford; Oxford UK
- EUSA Pharma; Hemel Hempstead UK
| | - Ahmed N. Hegazy
- Division of Gastroenterology, Infectiology, and Rheumatology; Charité Universitätsmedizin; Berlin Germany
- Deutsches Rheuma-Forschungszentrum; ein Institut der Leibniz-Gemeinschaft; Berlin Germany
| |
Collapse
|
29
|
Adrian-Segarra JM, Schindler N, Gajawada P, Lörchner H, Braun T, Pöling J. The AB loop and D-helix in binding site III of human Oncostatin M (OSM) are required for OSM receptor activation. J Biol Chem 2018; 293:7017-7029. [PMID: 29511087 DOI: 10.1074/jbc.ra118.001920] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/16/2018] [Indexed: 01/11/2023] Open
Abstract
Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are closely related members of the interleukin-6 (IL-6) cytokine family. Both cytokines share a common origin and structure, and both interact through a specific region, termed binding site III, to activate a dimeric receptor complex formed by glycoprotein 130 (gp130) and LIF receptor (LIFR) in humans. However, only OSM activates the OSM receptor (OSMR)-gp130 complex. The molecular features that enable OSM to specifically activate the OSMR are currently unknown. To define specific sequence motifs within OSM that are critical for initiating signaling via OSMR, here we generated chimeric OSM-LIF cytokines and performed alanine-scanning experiments. Replacement of the OSM AB loop within OSM's binding site III with that of LIF abrogated OSMR activation, measured as STAT3 phosphorylation at Tyr-705, but did not compromise LIFR activation. Correspondingly, substitution of the AB loop and D-helix in LIF with their OSM counterparts was sufficient for OSMR activation. The alanine-scanning experiments revealed that residues Tyr-34, Gln-38, Gly-39, and Leu-45 (in the AB loop) and Pro-153 (in the D-helix) had specific roles in activating OSMR but not LIFR signaling, whereas Leu-40 and Cys-49 (in the AB loop), and Phe-160 and Lys-163 (in the D-helix) were required for activation of both receptors. Because most of the key amino acid residues identified here are conserved between LIF and OSM, we concluded that comparatively minor differences in a few amino acid residues within binding site III account for the differential biological effects of OSM and LIF.
Collapse
Affiliation(s)
- Juan M Adrian-Segarra
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Natalie Schindler
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Praveen Gajawada
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Holger Lörchner
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Braun
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jochen Pöling
- From the Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
30
|
Hermanns HM. Oncostatin M and interleukin-31: Cytokines, receptors, signal transduction and physiology. Cytokine Growth Factor Rev 2015. [DOI: 10.1016/j.cytogfr.2015.07.006] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Richards CD. The enigmatic cytokine oncostatin m and roles in disease. ISRN INFLAMMATION 2013; 2013:512103. [PMID: 24381786 PMCID: PMC3870656 DOI: 10.1155/2013/512103] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 09/29/2013] [Indexed: 12/11/2022]
Abstract
Oncostatin M is a secreted cytokine involved in homeostasis and in diseases involving chronic inflammation. It is a member of the gp130 family of cytokines that have pleiotropic functions in differentiation, cell proliferation, and hematopoetic, immunologic, and inflammatory networks. However, Oncostatin M also has activities novel to mediators of this cytokine family and others and may have fundamental roles in mechanisms of inflammation in pathology. Studies have explored Oncostatin M functions in cancer, bone metabolism, liver regeneration, and conditions with chronic inflammation including rheumatoid arthritis, lung and skin inflammatory disease, atherosclerosis, and cardiovascular disease. This paper will review Oncostatin M biology in a historical fashion and focus on its unique activities, in vitro and in vivo, that differentiate it from other cytokines and inspire further study or consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Carl D. Richards
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street, West, Hamilton, ON, Canada L8S 4K1
| |
Collapse
|
32
|
Drechsler J, Grötzinger J, Hermanns HM. Characterization of the rat oncostatin M receptor complex which resembles the human, but differs from the murine cytokine receptor. PLoS One 2012; 7:e43155. [PMID: 22937020 PMCID: PMC3425591 DOI: 10.1371/journal.pone.0043155] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/17/2012] [Indexed: 12/22/2022] Open
Abstract
Evaluation of a pathophysiological role of the interleukin-6-type cytokine oncostatin M (OSM) for human diseases has been complicated by the fact that mouse models of diseases targeting either OSM or the OSM receptor (OSMR) complex cannot fully reflect the human situation. This is due to earlier findings that human OSM utilizes two receptor complexes, glycoprotein 130 (gp130)/leukemia inhibitory factor receptor (LIFR) (type I) and gp130/OSMR (type II), both with wide expression profiles. Murine OSM on the other hand only binds to the gp130/OSMR (type II) receptor complex with high affinity. Here, we characterize the receptor usage for rat OSM. Using different experimental approaches (knock-down of the OSMR expression by RNA interference, blocking of the LIFR by LIF-05, an antagonistic LIF variant and stably transfected Ba/F3 cells) we can clearly show that rat OSM surprisingly utilizes both, the type I and type II receptor complex, therefore mimicking the human situation. Furthermore, it displays cross-species activities and stimulates cells of human as well as murine origin. Its signaling capacities closely mimic those of human OSM in cell types of different origin in the way that strong activation of the Jak/STAT, the MAP kinase as well as the PI3K/Akt pathways can be observed. Therefore, rat disease models would allow evaluation of the relevance of OSM for human biology.
Collapse
Affiliation(s)
- Johannes Drechsler
- From the Rudolf-Virchow-Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Joachim Grötzinger
- Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Heike M. Hermanns
- From the Rudolf-Virchow-Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
33
|
Chollangi S, Mather T, Rodgers KK, Ash JD. A unique loop structure in oncostatin M determines binding affinity toward oncostatin M receptor and leukemia inhibitory factor receptor. J Biol Chem 2012; 287:32848-59. [PMID: 22829597 DOI: 10.1074/jbc.m112.387324] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Oncostatin M (OSM) and leukemia inhibitory factor are pleiotropic cytokines that belong to the interleukin-6 (IL-6) family. These cytokines play a crucial role in diverse biological events like inflammation, neuroprotection, hematopoiesis, metabolism, and development. The family is grouped together based on structural similarities and their ability to activate the transmembrane receptor glycoprotein 130 (gp130). The common structure among these cytokines defines the spacing and the orientation of binding sites for cell surface receptors. OSM is unique in this family as it can signal using heterodimers of gp130 with either leukemia inhibitory factor receptor (LIFR) (type I) or oncostatin M receptor (OSMR) (type II). We have identified a unique helical loop on OSM between its B and C helices that is not found on other IL-6 family cytokines. This loop is located near the "FXXK" motif in active site III, which is essential for OSM's binding to both LIFR and OSMR. In this study, we show that the BC loop does not play a role in OSM's unique ability to bind OSMR. Shortening of the loop enhanced OSM's interaction with OSMR and LIFR as shown by kinetic and equilibrium binding analysis, suggesting the loop may hinder receptor interactions. As a consequence of improved binding, these structurally modified OSMs exhibited enhanced biological activity, including suppressed proliferation of A375 melanoma cells.
Collapse
Affiliation(s)
- Srinivas Chollangi
- Department of Bioengineering, University of Oklahoma, Norman, Oklahoma 73019, USA
| | | | | | | |
Collapse
|
34
|
Jones LL, Vignali DAA. Molecular interactions within the IL-6/IL-12 cytokine/receptor superfamily. Immunol Res 2012; 51:5-14. [PMID: 21553332 DOI: 10.1007/s12026-011-8209-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Production of cytokines by immune cells in response to stimuli and the binding of cytokines to specific receptors on target cells is a central feature of the immune response. The IL-12 cytokine family is particularly influential in determining the fate of T cells and is characterized by the sharing of cytokine and receptor subunits. A thorough understanding of the molecular interactions within this family will be a key to the development of therapeutic inhibitors or enhancers of IL-12 family function. While the current structural and molecular data for IL-12 family members is limited, there is ample information on the structurally related IL-6 cytokine family. This review will summarize the current structural and mutagenesis data within the IL-12 family and will attempt to utilize similarities between the IL-6 and IL-12 families to understand molecular interactions between IL-12 family subunits and with receptor components.
Collapse
Affiliation(s)
- Lindsay L Jones
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | |
Collapse
|
35
|
Le Saux S, Rousseau F, Barbier F, Ravon E, Grimaud L, Danger Y, Froger J, Chevalier S, Gascan H. Molecular dissection of human interleukin-31-mediated signal transduction through site-directed mutagenesis. J Biol Chem 2009; 285:3470-7. [PMID: 19920145 DOI: 10.1074/jbc.m109.049189] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interleukin (IL)-31 is a recently described cytokine, preferentially produced by T helper 2 lymphocytes and associated with skin diseases, such as atopic dermatitis. IL-31 is a member of the four alpha-helix bundle cytokine family and is related to the IL-6 subgroup. Its heterodimeric membrane receptor is composed of the gp130-like receptor (GPL) subunit associated to the oncostatin M receptor subunit. We identified critical amino acids implicated in the ligand receptor interaction by computational analysis combined with site-directed mutagenesis. Six IL-31 residues selected for their putative involvement in cytokine receptor contact sites were alanine-substituted, and the corresponding proteins were expressed in mammalian and bacterial systems. Biochemical, membrane binding, cell signaling, and cell proliferation analyses showed that mutation E44A, E106A, or H110A abolished IL-31 binding to GPL and the subsequent signaling events. A second ligand receptor-binding site involved Lys(134), with alanine substitution leading to a protein that still binds GPL, but is unable to recruit the second receptor subunit and the subsequent signaling pathways. The results indicate that IL-31 recognizes its receptor complex through two different binding sites, and we propose a three-dimensional model for IL-31.
Collapse
Affiliation(s)
- Sabine Le Saux
- Unité Mixte INSERM 564, Bâtiment Monteclair, 4 rue Larrey, 49933 Angers Cedex 09, France
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Recent structural information for complexes of cytokine receptor ectodomains bound to their ligands has significantly expanded our understanding of the macromolecular topology and ligand recognition mechanisms used by our three principal shared cytokine signaling receptors-gp130, gamma(c), and beta(c). The gp130 family receptors intricately coordinate three structurally unique cytokine-binding sites on their four-helix bundle cytokine ligands to assemble multimeric signaling complexes. These organizing principles serve as topological blueprints for the entire gp130 family of cytokines. Novel structures of gamma(c) and beta(c) complexes show us new twists, such as the use of a nonstandard sushi-type alpha receptors for IL-2 and IL-15 in assembling quaternary gamma(c) signaling complexes and an antiparallel interlocked dimer in the GM-CSF signaling complex with beta(c). Unlike gp130, which appears to recognize vastly different cytokine surfaces in chemically unique fashions for each ligand, the gamma(c)-dependent cytokines appear to seek out some semblance of a knobs-in-holes shape recognition code in order to engage gamma(c) in related fashions. We discuss the structural similarities and differences between these three shared cytokine receptors, as well as the implications for transmembrane signaling.
Collapse
Affiliation(s)
- Xinquan Wang
- Howard Hughes Medical Institute, Stanford University School of Medicine, Departments of Molecular and Cellular Physiology, and Structural Biology, Stanford, California 94305, USA.
| | | | | | | |
Collapse
|
37
|
Prisco U, Leung C, Xirouchaki C, Jones CH, Heath JK, Palmer RE. Residue-specific immobilization of protein molecules by size-selected clusters. J R Soc Interface 2006; 2:169-75. [PMID: 16849177 PMCID: PMC1629076 DOI: 10.1098/rsif.2005.0032] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The atomic force microscope (AFM), operating in contact mode, has been employed in buffer solution to study two proteins; (i) green fluorescent protein (GFP), from the hydromedusan jellyfish Aequorea victoria; and (ii) human oncostatin M (OSM), in the presence of size-selected gold nanoclusters pinned on to a highly oriented pyrolytic graphite substrate. The AFM images have revealed immobilization of single molecules of OSM, which are strongly bound to the gold nanoclusters. Conversely, no strong immobilization has been observed for the GFP, as these molecules were easily displaced by the scanning tip. The contrasting behaviour of the two proteins can be explained by the exposed molecular surface area of their cysteine residues as modelled on the basis of their respective X-ray crystallographic data structures. GFP contains two cysteine residues, but neither is readily available to chemisorb on the gold clusters, because the cysteines are largely inaccessible from the surface of the protein. In contrast, OSM has a total of five cysteine residues, with different degrees of accessibility, which make the protein amenable to anchoring on the nanoclusters. Statistical analysis of the height of the OSM molecules bound to the nanoclusters is in accordance with crystallographic data, and suggests various configurations of the proteins on the clusters, associated with the presence of different cysteine anchoring sites. These results suggest that the three-dimensional conformation of protein molecules is preserved when they are chemisorbed to size-selected gold clusters, thus opening a new route towards oriented immobilization of individual protein molecules.
Collapse
Affiliation(s)
- Umberto Prisco
- Nanoscale Physics Research Laboratory, School of Physics and Astronomy, The University of BirminghamEdgbaston, Birmingham B15 2TT, UK
| | - Carl Leung
- Nanoscale Physics Research Laboratory, School of Physics and Astronomy, The University of BirminghamEdgbaston, Birmingham B15 2TT, UK
| | - Chrisa Xirouchaki
- Nanoscale Physics Research Laboratory, School of Physics and Astronomy, The University of BirminghamEdgbaston, Birmingham B15 2TT, UK
| | - Celine H Jones
- School of Biosciences, The University of BirminghamEdgbaston, Birmingham B15 2TT, UK
| | - John K Heath
- School of Biosciences, The University of BirminghamEdgbaston, Birmingham B15 2TT, UK
| | - Richard E Palmer
- Nanoscale Physics Research Laboratory, School of Physics and Astronomy, The University of BirminghamEdgbaston, Birmingham B15 2TT, UK
- Author for correspondence ()
| |
Collapse
|
38
|
Boulanger MJ, Garcia KC. Shared cytokine signaling receptors: structural insights from the gp130 system. ACTA ACUST UNITED AC 2004; 68:107-46. [PMID: 15500860 DOI: 10.1016/s0065-3233(04)68004-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The vast majority of cytokine signaling is mediated by "shared" receptors that form central signaling components of higher-order complexes incorporating ligand-specific receptors. These include the common gamma chain (gamma(c)), common beta chain (beta(c)), and gp130, as well as others. These receptors have the dual tasks of cross-reactive cytokine recognition, and formation of precisely oriented multimeric signaling assemblies. Currently, detailed structural information on a shared receptor complex exists only for gp130, which is a highly pleiotropic shared cytokine signaling receptor essential for mammalian cell growth and homeostasis. To date, more than 10 different four-helix bundle ligands have been identified that incorporate gp130, or one of its close relatives such as LIF receptor, into functional oligomeric signaling complexes. In this review we summarize our current knowledge of shared receptor recognition and activation, with a focus on gp130. We discuss recent structural and functional information to analyze overall architectural assemblies of gp130 cytokine complexes and probe the basis for the extreme cross-reactivity of gp130 for its multiple cytokine ligands.
Collapse
Affiliation(s)
- Martin J Boulanger
- Department of Microbiology, Stanford University School of Medicine, Stanford, California 94305-5124, USA
| | | |
Collapse
|
39
|
Kamimura D, Ishihara K, Hirano T. IL-6 signal transduction and its physiological roles: the signal orchestration model. Rev Physiol Biochem Pharmacol 2004; 149:1-38. [PMID: 12687404 DOI: 10.1007/s10254-003-0012-2] [Citation(s) in RCA: 343] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin (IL)-6 is a pleiotropic cytokine that not only affects the immune system, but also acts in other biological systems and many physiological events in various organs. In a target cell, IL-6 can simultaneously generate functionally distinct or sometimes contradictory signals through its receptor complex, IL-6Ralpha and gp130. One good illustration is derived from the in vitro observations that IL-6 promotes the growth arrest and differentiation of M1 cells through gp130-mediated STAT3 activation, whereas the Y759/SHP-2-mediated cascade by gp130 stimulation has growth-enhancing effects. The final physiological output can be thought of as a consequence of the orchestration of the diverse signaling pathways generated by a given ligand. This concept, the signal orchestration model, may explain how IL-6 can elicit proinflammatory or anti-inflammatory effects, depending on the in vivo environmental circumstances. Elucidation of the molecular mechanisms underlying this issue is a challenging subject for future research. Intriguingly, recent in vivo studies indicated that the SHP-2-binding site- and YXXQ-mediated pathways through gp130 are not mutually exclusive but affect each other: a mutation at the SHP-2-binding site prolongs STAT3 activation, and a loss of STAT activation by gp130 truncation leads to sustained SHP-2/ERK MAPK phosphorylation. Although IL-6/gp130 signaling is a promising target for drug discovery for many human diseases, the interdependence of each signaling pathway may be an obstacle to the development of a nonpeptide orally active small molecule to inhibit one of these IL-6 signaling cascades, because it would disturb the signal orchestration. In mice, a consequence of the imbalanced signals causes unexpected results such as gastrointestinal disorders, autoimmune diseases, and/or chronic inflammatory proliferative diseases. However, lessons learned from IL-6 KO mice indicate that IL-6 is not essential for vital biological processes, but a significant impact on disease progression in many experimental models for human disorders. Thus, IL-6/gp130 signaling will become a more attractive therapeutic target for human inflammatory diseases when a better understanding of IL-6 signaling, including the identification of the conductor for gp130 signal transduction, is achieved.
Collapse
Affiliation(s)
- D Kamimura
- Department of Molecular Oncology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | |
Collapse
|
40
|
Boulanger MJ, Bankovich AJ, Kortemme T, Baker D, Garcia KC. Convergent mechanisms for recognition of divergent cytokines by the shared signaling receptor gp130. Mol Cell 2003; 12:577-89. [PMID: 14527405 DOI: 10.1016/s1097-2765(03)00365-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Gp130 is a shared cell-surface signaling receptor for at least ten different hematopoietic cytokines, but the basis of its degenerate recognition properties is unknown. We have determined the crystal structure of human leukemia inhibitory factor (LIF) bound to the cytokine binding region (CHR) of gp130 at 2.5 A resolution. Strikingly, we find that the shared binding site on gp130 has an entirely rigid core, while the LIF binding interface diverges sharply in structure and chemistry from that of other gp130 ligands. Dissection of the LIF-gp130 interface, along with comparative studies of other gp130 cytokines, reveal that gp130 has evolved a "thermodynamic plasticity" that is relatively insensitive to ligand structure, to enable crossreactivity. These observations reveal a novel and alternative mechanism for degenerate recognition from that of structural plasticity.
Collapse
Affiliation(s)
- Martin J Boulanger
- Department of Microbiology and Immunology, Stanford University School of Medicine, Fairchild D319, 299 Campus Drive, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
The IL (interleukin)-6-type cytokines IL-6, IL-11, LIF (leukaemia inhibitory factor), OSM (oncostatin M), ciliary neurotrophic factor, cardiotrophin-1 and cardiotrophin-like cytokine are an important family of mediators involved in the regulation of the acute-phase response to injury and infection. Besides their functions in inflammation and the immune response, these cytokines play also a crucial role in haematopoiesis, liver and neuronal regeneration, embryonal development and fertility. Dysregulation of IL-6-type cytokine signalling contributes to the onset and maintenance of several diseases, such as rheumatoid arthritis, inflammatory bowel disease, osteoporosis, multiple sclerosis and various types of cancer (e.g. multiple myeloma and prostate cancer). IL-6-type cytokines exert their action via the signal transducers gp (glycoprotein) 130, LIF receptor and OSM receptor leading to the activation of the JAK/STAT (Janus kinase/signal transducer and activator of transcription) and MAPK (mitogen-activated protein kinase) cascades. This review focuses on recent progress in the understanding of the molecular mechanisms of IL-6-type cytokine signal transduction. Emphasis is put on the termination and modulation of the JAK/STAT signalling pathway mediated by tyrosine phosphatases, the SOCS (suppressor of cytokine signalling) feedback inhibitors and PIAS (protein inhibitor of activated STAT) proteins. Also the cross-talk between the JAK/STAT pathway with other signalling cascades is discussed.
Collapse
|
42
|
Heinrich PC, Behrmann I, Haan S, Hermanns HM, Müller-Newen G, Schaper F. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J 2003; 374:1-20. [PMID: 12773095 PMCID: PMC1223585 DOI: 10.1042/bj20030407] [Citation(s) in RCA: 2390] [Impact Index Per Article: 108.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2003] [Revised: 04/30/2003] [Accepted: 05/29/2003] [Indexed: 12/11/2022]
Abstract
The IL (interleukin)-6-type cytokines IL-6, IL-11, LIF (leukaemia inhibitory factor), OSM (oncostatin M), ciliary neurotrophic factor, cardiotrophin-1 and cardiotrophin-like cytokine are an important family of mediators involved in the regulation of the acute-phase response to injury and infection. Besides their functions in inflammation and the immune response, these cytokines play also a crucial role in haematopoiesis, liver and neuronal regeneration, embryonal development and fertility. Dysregulation of IL-6-type cytokine signalling contributes to the onset and maintenance of several diseases, such as rheumatoid arthritis, inflammatory bowel disease, osteoporosis, multiple sclerosis and various types of cancer (e.g. multiple myeloma and prostate cancer). IL-6-type cytokines exert their action via the signal transducers gp (glycoprotein) 130, LIF receptor and OSM receptor leading to the activation of the JAK/STAT (Janus kinase/signal transducer and activator of transcription) and MAPK (mitogen-activated protein kinase) cascades. This review focuses on recent progress in the understanding of the molecular mechanisms of IL-6-type cytokine signal transduction. Emphasis is put on the termination and modulation of the JAK/STAT signalling pathway mediated by tyrosine phosphatases, the SOCS (suppressor of cytokine signalling) feedback inhibitors and PIAS (protein inhibitor of activated STAT) proteins. Also the cross-talk between the JAK/STAT pathway with other signalling cascades is discussed.
Collapse
Affiliation(s)
- Peter C Heinrich
- Institut für Biochemie, RWTH Aachen, Universitätsklinikum, Pauwelsstrasse 30, D-52074 Aachen, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Plun-Favreau H, Perret D, Diveu C, Froger J, Chevalier S, Lelièvre E, Gascan H, Chabbert M. Leukemia inhibitory factor (LIF), cardiotrophin-1, and oncostatin M share structural binding determinants in the immunoglobulin-like domain of LIF receptor. J Biol Chem 2003; 278:27169-79. [PMID: 12707269 DOI: 10.1074/jbc.m303168200] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Leukemia inhibitory factor (LIF), cardiotrophin-1 (CT-1), and oncostatin M (OSM) are four helix bundle cytokines acting through a common heterodimeric receptor composed of gp130 and LIF receptor (LIFR). Binding to LIFR occurs through a binding site characterized by an FXXK motif located at the N terminus of helix D (site III). The immunoglobulin (Ig)-like domain of LIFR was modeled, and the physico-chemical properties of its Connolly surface were analyzed. This analysis revealed an area displaying properties complementary to those of the LIF site III. Two residues of the Ig-like domain of LIFR, Asp214 and Phe284, formed a mirror image of the FXXK motif. Engineered LIFR mutants in which either or both of these two residues were mutated to alanine were transfected in Ba/F3 cells already containing gp130. The F284A mutation impaired the biological response induced by LIF and CT-1, whereas the response to OSM remained unchanged. The Asp214 mutation did not alter the functional responses. The D214A/F284A double mutation, however, totally impaired cellular proliferation to LIF and CT-1 and partially impaired OSM-induced proliferation with a 20-fold increase in EC50. These results were corroborated by the analysis of STAT3 phosphorylation and Scatchard analysis of cytokine binding to Ba/F3 cells. Molecular modeling of the complex of LIF with the Ig-like domain of LIFR provides a clue for the superadditivity of the D214A/F284A double mutation. Our results indicate that LIF, CT-1, and OSM share an overlapping binding site located in the Ig-like domain of LIFR. The different behaviors of LIF and CT-1, on one side, and of OSM, on the other side, can be related to the different affinity of their site III for LIFR.
Collapse
Affiliation(s)
- Hélène Plun-Favreau
- INSERM U564, Centre Hospitalier Universitaire d'Angers, 4 rue Larrey, 49033 Angers, France
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Human prolactin is a 199-residue (23 kDa) protein closely related to growth hormone and placental lactogen with properties and functions resembling both a hormone and a cytokine. As a traditional hormone, prolactin is produced by lactotrophic cells in the pituitary and secreted into the bloodstream where it acts distally to regulate reproduction and promote lactation. Pituitary cells store prolactin in secretory granules organized around large prolactin aggregates, which are produced within the trans layer of the Golgi complex. Extrapituitary prolactin is synthesized by a wide variety of cells but is not stored in secretory granules. Extrapituitary prolactin displays immunomodulatory activities and acts as a growth factor for cancers of the breast, prostate and tissues of the female reproductive system. We have determined the tertiary structure of human prolactin using three-dimensional (3D) and four-dimensional (4D) heteronuclear NMR spectroscopy. As expected, prolactin adopts an "up-up-down-down" four-helical bundle topology and resembles other members of the family of hematopoietic cytokines. Prolactin displays three discrete structural differences from growth hormone: (1) a structured N-terminal loop in contact with the first helix, (2) a missing mini-helix in the loop between the first and second helices, and (3) a shorter loop between the second and third helices lacking the perpendicular mini-helix observed in growth hormone. Residues necessary for functional binding to the prolactin receptor are clustered on the prolactin surface in a position similar to growth hormone. The backbone dynamics of prolactin were investigated by analysis of 15N NMR relaxation phenomena and demonstrated a rigid four-helical bundle with relatively mobile interconnecting loops. Comparison of global macromolecular tumbling at 0.1mM and 1.0mM prolactin revealed reversible oligomerization, which was correlated to dynamic light scattering experiments. The existence of a reversible oligomerization reaction in solution provides insight into previous results describing the in vitro and in vivo aggregation properties of human prolactin.
Collapse
Affiliation(s)
- Camille Keeler
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520-8035, USA
| | | | | |
Collapse
|
45
|
Bitard J, Daburon S, Duplomb L, Blanchard F, Vuisio P, Jacques Y, Godard A, Heath JK, Moreau JF, Taupin JL. Mutations in the immunoglobulin-like domain of gp190, the leukemia inhibitory factor (LIF) receptor, increase or decrease its affinity for LIF. J Biol Chem 2003; 278:16253-61. [PMID: 12601009 DOI: 10.1074/jbc.m207193200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The leukemia inhibitory factor (LIF) receptor comprises the low affinity binding chain gp190 and the high affinity converter gp130. The ectodomain of gp190 is among the most complex in the hematopoietin receptor family, because it contains two typical cytokine receptor homology domains separated by an immunoglobulin-like (Ig-like) domain. Human and murine gp190 proteins share 76% homology, but murine gp190 binds human LIF with a much higher affinity, a property attributed to the Ig-like domain. Using alanine-scanning mutagenesis of the Ig-like domain, we mapped a LIF binding site at its carboxyl terminus, mainly involving residue Phe-328. Mutation of selected residues into their orthologs in the murine receptor (Q251E and N321D) significantly increased the affinity for human LIF. Interestingly, these residues, although localized at both the amino and carboxyl terminus, make a spatially unique LIF binding site in a structural model of the Ig-like module. These results demonstrate definitively the role of the Ig-like domain in LIF binding and the potential to modulate receptor affinity in this family with very limited amino acid changes.
Collapse
Affiliation(s)
- Juliette Bitard
- CNRS UMR 5540, Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Grötzinger J. Molecular mechanisms of cytokine receptor activation. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1592:215-23. [PMID: 12421667 DOI: 10.1016/s0167-4889(02)00316-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cytokine receptors are transmembrane proteins that transmit a signal into the cell upon ligand binding. Commonly, these molecules have one hydrophobic segment of about 20-26 amino acids that is believed to span the membrane as a helix and this divides these receptors into extra- and intracellular components. By utilizing two different epitopes, the cytokines bridge two receptor chains, resulting in a close proximity of the intracellular component and thereby initiating the intracellular signalling cascade. The dimerization event is believed to be the mechanism by which the signal is transmitted across a membrane. In the light of new results obtained for the erythropoietin receptor, James A. Wells questioned whether any dimer would be sufficient. This review will expand upon the above question by discussing the more complex signal-transducing receptor subunits of the Interleukin-6 type family of cytokines. Based on the recently solved quaternary structure of the Insulin receptor, possible analogies will be confronted.
Collapse
Affiliation(s)
- Joachim Grötzinger
- Department of Biochemistry, Christian-Albrechts-Universität zu Kiel, Germany.
| |
Collapse
|
47
|
Chow DC, Brevnova L, He XL, Martick MM, Bankovich A, Garcia KC. A structural template for gp130-cytokine signaling assemblies. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1592:225-35. [PMID: 12421668 DOI: 10.1016/s0167-4889(02)00317-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The gp130-cytokine system has been fertile ground for protein structure-function studies aimed at elucidating the basis of ligand recognition and receptor activation. A number of longstanding questions involve the mechanism of the stepwise assembly of the active signaling complexes, as well as the structure of the gp130-cytokine complexes. It has been clear from functional studies that the paradigm of gp130-cyokine recognition will differ substantially from the classical homo-dimeric systems, typified by human growth hormone (hGH) and its receptor. Recently, a crystal structure of a viral interleukin-6 (vIL-6), complexed with the D1D2D3 domains of the gp130 extracellular domain, has resolved many of these questions, and reconciled much of the functional and mutagenesis data which have existed for a variety of gp130-cytokines. In this review, we discuss the structure of the vIL-6/gp130 complex in some detail and suggest that the geometry of this complex will be a common structural template utilized by other gp130-cytokines, as well as cytokines from distinct signaling systems.
Collapse
Affiliation(s)
- Dar-chone Chow
- Deparment of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305-5124, USA
| | | | | | | | | | | |
Collapse
|
48
|
Hill EE, Morea V, Chothia C. Sequence conservation in families whose members have little or no sequence similarity: the four-helical cytokines and cytochromes. J Mol Biol 2002; 322:205-33. [PMID: 12215425 DOI: 10.1016/s0022-2836(02)00653-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Proteins for which there are good structural, functional and genetic similarities that imply a common evolutionary origin, can have sequences whose similarities are low or undetectable by conventional sequence comparison procedures. Do these proteins have sequence conservation beyond the simple conservation of hydrophobic and hydrophilic character at specific sites and if they do what is its nature? To answer these questions we have analysed the structures and sequences of two superfamilies: the four-helical cytokines and cytochromes c'-b(562). Members of these superfamilies have sequence similarities that are either very low or not detectable. The cytokine superfamily has within it a long chain family and a short chain family. The sequences of known representative structures of the two families were aligned using structural information. From these alignments we identified the regions that conserve the same main-chain conformation: the common core (CC). For members of the same family, the CC comprises some 50% of the individual structures; for the combination of both families it is 30%. We added homologous sequences to the structural alignment. Analysis of the residues occurring at sites within the CCs showed that 30% have little or no conservation, whereas about 40% conserve the polar/neutral or hydrophobic/neutral character of their residues. The remaining 30% conserve hydrophobic residues with strong or medium limitations on their volume variations. Almost all of these residues are found at sites that form the "buried spine" of each helix (at sites i, i+3, i+7, i+10, etc., or i, i+4, i+7, i+11, etc.) and they pack together at the centre of each structure to give a pattern of residue-residue contacts that is almost absolutely conserved. These CC conserved hydrophobic residues form only 10-15% of all the residues in the individual structures.A similar analysis of the cytochromes c'-b(562), which bind haem and have a very different function to that of the cytokines, gave very similar results. Again some 30% of the CC residues have hydrophobic residues with strong or medium conservation. Most of these form the buried spine of each helix and play the same role as those in the cytokines. The others, and some spine residues bind the haem co-factor.
Collapse
Affiliation(s)
- Emma E Hill
- MRC Laboratory of Molecular Biology, Cambridge, UK.
| | | | | |
Collapse
|
49
|
Aasland D, Oppmann B, Grötzinger J, Rose-John S, Kallen KJ. The upper cytokine-binding module and the Ig-like domain of the leukaemia inhibitory factor (LIF) receptor are sufficient for a functional LIF receptor complex. J Mol Biol 2002; 315:637-46. [PMID: 11812136 DOI: 10.1006/jmbi.2001.5282] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To elucidate the function of the two cytokine-binding modules (CBM) of the leukemia inhibitory factor receptor (LIFR), receptor chimeras of LIFR and the interleukin-6 receptor (IL-6R) were constructed. Either the NH(2)-terminal (chimera RILLIFdeltaI) or the COOH-terminal LIFR CBM (chimera RILLIFdeltaII) were replaced by the structurally related CBM of the IL-6R which does not bind LIF. Chimera RILLIFdeltaI is functionally inactive, whereas RILLIFdeltaII binds LIF and mediates signalling as efficiently as the wild-type LIFR. Deletion mutants of the LIFR revealed that both the NH(2)-terminal CBM and the Ig-like domain of the LIFR are involved in LIF binding, presumably via the LIF site III epitope. The main function of the COOH-terminal CBM of the LIFR is to position the NH(2)-terminal CBM and the Ig-like domain, so that these can bind to LIF. In analogy to a recently published model of the IL-6R complex, a model of the active LIFR complex is suggested which positions the COOH-terminal CBM at LIF site I and the NH(2)-terminal CBM and the Ig-like domain at site III. An additional contact is postulated between the Ig-like domain of gp130 and the NH(2)-terminal CBM of the LIFR.
Collapse
Affiliation(s)
- Dorthe Aasland
- Biochemisches Institut, Christian Albrechts Universität Kiel, Ohlshausenstr. 40, Kiel, D-24098, Germany
| | | | | | | | | |
Collapse
|
50
|
Chow D, He X, Snow AL, Rose-John S, Garcia KC. Structure of an extracellular gp130 cytokine receptor signaling complex. Science 2001; 291:2150-5. [PMID: 11251120 DOI: 10.1126/science.1058308] [Citation(s) in RCA: 203] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The activation of gp130, a shared signal-transducing receptor for a family of cytokines, is initiated by recognition of ligand followed by oligomerization into a higher order signaling complex. Kaposi's sarcoma-associated herpesvirus encodes a functional homolog of human interleukin-6 (IL-6) that activates human gp130. In the 2.4 angstrom crystal structure of the extracellular signaling assembly between viral IL-6 and human gp130, two complexes are cross-linked into a tetramer through direct interactions between the immunoglobulin domain of gp130 and site III of viral IL-6, which is necessary for receptor activation. Unlike human IL-6 (which uses many hydrophilic residues), the viral cytokine largely uses hydrophobic amino acids to contact gp130, which enhances the complementarity of the viral IL-6-gp130 binding interfaces. The cross-reactivity of gp130 is apparently due to a chemical plasticity evident in the amphipathic gp130 cytokine-binding sites.
Collapse
Affiliation(s)
- D Chow
- Department of Microbiology and Immunology, Stanford University School of Medicine, Fairchild D319, 299 Campus Drive, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|