1
|
Maldonado H, Dreger M, Bedgood LD, Kyriakou T, Wolanska KI, Rigby ME, Marotta VE, Webster JM, Wang J, Rusilowicz-Jones EV, Marshall JF, Coulson JM, Macpherson IR, Hurlstone A, Morgan MR. A trafficking regulatory subnetwork governs α Vβ 6 integrin-HER2 cross-talk to control breast cancer invasion and drug resistance. SCIENCE ADVANCES 2024; 10:eadk9944. [PMID: 39630893 PMCID: PMC11616693 DOI: 10.1126/sciadv.adk9944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/31/2024] [Indexed: 12/07/2024]
Abstract
HER2 and αVβ6 integrin are independent predictors of breast cancer survival and metastasis. We identify an αVβ6/HER2 cross-talk mechanism driving invasion, which is dysregulated in drug-resistant HER2+ breast cancer cells. Proteomic analyses reveal ligand-bound αVβ6 recruits HER2 and a trafficking subnetwork, comprising guanosine triphosphatases RAB5 and RAB7A and the Rab regulator guanine nucleotide dissociation inhibitor 2 (GDI2). The RAB5/RAB7A/GDI2 functional module mediates direct cross-talk between αVβ6 and HER2, affecting receptor trafficking and signaling. Acute exposure to trastuzumab increases recruitment of the subnetwork to αVβ6, but trastuzumab resistance decouples GDI2 recruitment. GDI2, RAB5, and RAB7A cooperate to regulate migration and transforming growth factor-β activation to promote invasion. However, these mechanisms are dysregulated in trastuzumab-resistant cells. In patients, RAB5A, RAB7A, and GDI2 expression correlates with patient survival and αVβ6 expression predicts relapse following trastuzumab treatment. Thus, the RAB5/RAB7A/GDI2 subnetwork regulates αVβ6-HER2 cross-talk to drive breast cancer invasion but is subverted in trastuzumab-resistant cells to drive αVβ6-independent and HER2-independent tumor progression.
Collapse
Affiliation(s)
- Horacio Maldonado
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Marcel Dreger
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Lara D. Bedgood
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Theano Kyriakou
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Katarzyna I. Wolanska
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Megan E. Rigby
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Valeria E. Marotta
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Justine M. Webster
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Jun Wang
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Emma V. Rusilowicz-Jones
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Judy M. Coulson
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Iain R. Macpherson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| | - Adam Hurlstone
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Mark R. Morgan
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| |
Collapse
|
2
|
Subrahmanian S, Varshney R, Subramani K, Murphy B, Woolington S, Ahamed J. N-Acetylcysteine Inhibits Aortic Stenosis Progression in a Murine Model by Blocking Shear-Induced Activation of Platelet Latent Transforming Growth Factor Beta 1. Antioxid Redox Signal 2024; 41:e1187-e1196. [PMID: 34619980 PMCID: PMC11693965 DOI: 10.1089/ars.2021.0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/08/2023]
Abstract
Objective: Aortic stenosis (AS) is characterized by narrowing of the aortic valve opening, resulting in peak blood flow velocity that induces high wall shear stress (WSS) across the valve. Severe AS leads to heart failure and death. There is no treatment available for AS other than valve replacement. Platelet-derived transforming growth factor beta 1 (TGF-β1) partially contributes to AS progression in mice, and WSS is a potent activator of latent TGF-β1. N-acetylcysteine (NAC) inhibits WSS-induced TGF-β1 activation in vitro. We hypothesize that NAC will inhibit AS progression by inhibiting WSS-induced TGF-β1 activation. Approach: We treated a cohort of Ldlr(-/-)Apob(100/100) low density lipoprotein receptor (LDLR) mice fed a high-fat diet with NAC (2% in drinking water) at different stages of disease progression and measured its effect on AS progression and TGF-β1 activation. Results: Short-term NAC treatment inhibited AS progression in mice with moderate and severe AS relative to controls, but not in LDLR mice lacking platelet-derived TGF-β1 (TGF-β1platlet-KO-LDLR). NAC treatment reduced TGF-β signaling, p-Smad2 and collagen levels, and mesenchymal transition from isolectin B4 and CD45-positive cells in LDLR mice. Mechanistically, NAC treatment resulted in plasma NAC concentrations ranging from 75.5 to 449.2 ng/mL, which were sufficient to block free thiol labeling of plasma proteins and reduce active TGF-β1 levels without substantially affecting reactive oxygen species-modified products in valvular cells. Conclusions: Short-term treatment with NAC inhibits AS progression by inhibiting WSS-induced TGF-β1 activation in the LDLR mouse model of AS, motivating a clinical trial of NAC and/or other thiol-reactive agent(s) as a potential therapy for AS.
Collapse
Affiliation(s)
- Sandeep Subrahmanian
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Rohan Varshney
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Kumar Subramani
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Brennah Murphy
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Sean Woolington
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
3
|
Corbalan JJ, Jagadeesan P, Frietze KK, Taylor R, Gao GL, Gallagher G, Nickels JT. Humanized monoacylglycerol acyltransferase 2 mice develop metabolic dysfunction-associated steatohepatitis. J Lipid Res 2024; 65:100695. [PMID: 39505262 DOI: 10.1016/j.jlr.2024.100695] [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: 08/19/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Mice lacking monoacylglycerol acyltransferase 2 (mMGAT21) are resistant to diet-induced fatty liver, suggesting hMOGAT2 inhibition is a viable option for treating metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH). We generated humanized hMOGAT2 mice (HuMgat2) for use in pre-clinical studies testing the efficacy of hMOGAT2 inhibitors for treating MASLD/MASH. HuMgat2 mice developed MASH when fed a steatotic diet. Computer-aided histology revealed the presence of hepatocyte cell ballooning, immune cell infiltration, and fibrosis. Hepatocytes accumulated Mallory-Denk bodies containing phosphorylated p62/sequestosome-1-ubiquitinated protein aggregates likely caused by defects in autophagy. Metainflammation and apoptotic cell death were seen in the livers of HuMgat2 mice. Treating HuMgat2 mice with elafibranor reduced several MASH phenotypes. RNASeq analysis predicted changes in bile acid transporter expression that correlated with altered bile acid metabolism indicative of cholestasis. Our results suggest that HuMgat2 mice will serve as a pre-clinical model for testing hMOGAT2 inhibitor efficacy and toxicity and allow for the study of hMOGAT2 in the context of MASH.
Collapse
Affiliation(s)
- J Jose Corbalan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Pranavi Jagadeesan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Karla K Frietze
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Grace L Gao
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA
| | - Grant Gallagher
- Oncoveda, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Joseph T Nickels
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
4
|
Wang T, Kim SY, Peng Y, Zheng J, Layne MD, Murphy-Ullrich JE, Albro MB. Autoinduction-Based Quantification of In Situ TGF-β Activity in Native and Engineered Cartilage. Tissue Eng Part C Methods 2024; 30:522-532. [PMID: 39311474 DOI: 10.1089/ten.tec.2024.0190] [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] [Indexed: 10/10/2024] Open
Abstract
Transforming growth factor beta (TGF-β) is a potent growth factor that regulates the homeostasis of native cartilage and is administered as an anabolic supplement for engineered cartilage growth. The quantification of TGF-β activity in live tissues in situ remains a significant challenge, as conventional activity assessments (e.g., Western blotting of intracellular signaling molecules or reporter cell assays) are unable to measure absolute levels of TGF-β activity in three-dimensional tissues. In this study, we develop a quantification platform established on TGF-β's autoinduction response, whereby active TGF-β (aTGF-β) signaling in cells induces their biosynthesis and secretion of new TGF-β in its latent form (LTGF-β). As such, cell-secreted LTGF-β can serve as a robust, non-destructive, label-free biomarker for quantifying in situ activity of TGF-β in live cartilage tissues. Here, we detect LTGF-β1 secretion levels for bovine native tissue explants and engineered tissue constructs treated with varying doses of media-supplemented aTGF-β3 using an isoform-specific ELISA. We demonstrate that: 1) LTGF-β secretion levels increase proportionally to aTGF-β exposure, reaching 7.4- and 6.6-fold increases in native and engineered cartilage, respectively; 2) synthesized LTGF-β exhibits low retention in both native and engineered cartilage tissue; and 3) secreted LTGF-β is stable in conditioned media for 2 weeks, thus enabling a reliable biological standard curve between LTGF-β secretion and exposed TGF-β activity. Accordingly, we perform quantifications of TGF-β activity in bovine native cartilage, demonstrating up to 0.59 ng/mL in response to physiological dynamic loading. We further quantify the in situ TGF-β activity in aTGF-β-conjugated scaffolds for engineered tissue, which exhibits 1.81 ng/mL of TGF-β activity as a result of a nominal 3 μg/mL loading dose. Overall, cell-secreted LTGF-β can serve as a robust biomarker to quantify in situ activity of TGF-β in live cartilage tissue and can be potentially applied for a wide range of applications, including multiple tissue types and tissue engineering platforms with different cell populations and scaffolds.
Collapse
Affiliation(s)
- Tianbai Wang
- Division of Materials Science & Engineering, Boston University, Boston, Massachusetts, USA
| | - Sung Yeon Kim
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, USA
| | - Yifan Peng
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Jane Zheng
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Matthew D Layne
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | | | - Michael B Albro
- Division of Materials Science & Engineering, Boston University, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Jin M, Seed RI, Cai G, Shing T, Wang L, Ito S, Cormier A, Wankowicz SA, Jespersen JM, Baron JL, Carey ND, Campbell MG, Yu Z, Tang PK, Cossio P, Wen W, Lou J, Marks J, Nishimura SL, Cheng Y. Dynamic allostery drives autocrine and paracrine TGF-β signaling. Cell 2024; 187:6200-6219.e23. [PMID: 39288764 PMCID: PMC11531391 DOI: 10.1016/j.cell.2024.08.036] [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: 01/20/2024] [Revised: 06/10/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
TGF-β, essential for development and immunity, is expressed as a latent complex (L-TGF-β) non-covalently associated with its prodomain and presented on immune cell surfaces by covalent association with GARP. Binding to integrin αvβ8 activates L-TGF-β1/GARP. The dogma is that mature TGF-β must physically dissociate from L-TGF-β1 for signaling to occur. Our previous studies discovered that αvβ8-mediated TGF-β autocrine signaling can occur without TGF-β1 release from its latent form. Here, we show that mice engineered to express TGF-β1 that cannot release from L-TGF-β1 survive without early lethal tissue inflammation, unlike those with TGF-β1 deficiency. Combining cryogenic electron microscopy with cell-based assays, we reveal a dynamic allosteric mechanism of autocrine TGF-β1 signaling without release where αvβ8 binding redistributes the intrinsic flexibility of L-TGF-β1 to expose TGF-β1 to its receptors. Dynamic allostery explains the TGF-β3 latency/activation mechanism and why TGF-β3 functions distinctly from TGF-β1, suggesting that it broadly applies to other flexible cell surface receptor/ligand systems.
Collapse
Affiliation(s)
- Mingliang Jin
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Robert I Seed
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Guoqing Cai
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Tiffany Shing
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Li Wang
- Department of Pathology, UCSF, San Francisco, CA, USA
| | - Saburo Ito
- Department of Pathology, UCSF, San Francisco, CA, USA
| | | | | | | | - Jody L Baron
- Department of Medicine and UCSF Liver Center, UCSF, San Francisco, CA, USA
| | - Nicholas D Carey
- Department of Medicine and UCSF Liver Center, UCSF, San Francisco, CA, USA
| | - Melody G Campbell
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Zanlin Yu
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Phu K Tang
- Center for Computational Mathematics, Flatiron Institute, New York, NY, USA
| | - Pilar Cossio
- Center for Computational Mathematics, Flatiron Institute, New York, NY, USA; Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Weihua Wen
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA
| | - James Marks
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA
| | | | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, CA, USA; Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA.
| |
Collapse
|
6
|
Tharp KM, Kersten K, Maller O, Timblin GA, Stashko C, Canale FP, Menjivar RE, Hayward MK, Berestjuk I, Ten Hoeve J, Samad B, Ironside AJ, di Magliano MP, Muir A, Geiger R, Combes AJ, Weaver VM. Tumor-associated macrophages restrict CD8 + T cell function through collagen deposition and metabolic reprogramming of the breast cancer microenvironment. NATURE CANCER 2024; 5:1045-1062. [PMID: 38831058 DOI: 10.1038/s43018-024-00775-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Tumor progression is accompanied by fibrosis, a condition of excessive extracellular matrix accumulation, which is associated with diminished antitumor immune infiltration. Here we demonstrate that tumor-associated macrophages (TAMs) respond to the stiffened fibrotic tumor microenvironment (TME) by initiating a collagen biosynthesis program directed by transforming growth factor-β. A collateral effect of this programming is an untenable metabolic milieu for productive CD8+ T cell antitumor responses, as collagen-synthesizing macrophages consume environmental arginine, synthesize proline and secrete ornithine that compromises CD8+ T cell function in female breast cancer. Thus, a stiff and fibrotic TME may impede antitumor immunity not only by direct physical exclusion of CD8+ T cells but also through secondary effects of a mechano-metabolic programming of TAMs, which creates an inhospitable metabolic milieu for CD8+ T cells to respond to anticancer immunotherapies.
Collapse
Affiliation(s)
- Kevin M Tharp
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Kelly Kersten
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
| | - Ori Maller
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Greg A Timblin
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Connor Stashko
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Fernando P Canale
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Rosa E Menjivar
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Mary-Kate Hayward
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ilona Berestjuk
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Johanna Ten Hoeve
- UCLA Metabolomics Center, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bushra Samad
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | | | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, Cancer Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Alexis J Combes
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences and Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and The Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Sabat M, Carney DW, Hernandez-Torres G, Gibson TS, Balakrishna D, Zou H, Xu R, Chen CH, de Jong R, Dougan DR, Qin L, Bigi-Botterill SV, Chambers A, Miura J, Johnson LK, Ermolieff J, Johns D, Selimkhanov J, Kwok L, DeMent K, Proffitt C, Vu P, Lindsey EA, Ivetac T, Jennings A, Wang H, Manam P, Santos C, Fullenwider C, Manohar R, Flick AC. Design and Discovery of a Potent and Selective Inhibitor of Integrin αvβ1. J Med Chem 2024; 67:10306-10320. [PMID: 38872300 DOI: 10.1021/acs.jmedchem.4c00743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Selective inhibition of the RGD (Arg-Gly-Asp) integrin αvβ1 has been recently identified as an attractive therapeutic approach for the treatment of liver fibrosis given its function, target expression, and safety profile. Our identification of a non-RGD small molecule lead followed by focused, systematic changes to the core structure utilizing a crystal structure, in silico modeling, and a tractable synthetic approach resulted in the identification of a potent small molecule exhibiting a remarkable affinity for αvβ1 relative to several other integrin isoforms measured. Azabenzimidazolone 25 demonstrated antifibrotic efficacy in an in vivo rat liver fibrosis model and represents a tool compound capable of further exploring the biological consequences of selective αvβ1 inhibition.
Collapse
Affiliation(s)
- Mark Sabat
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Daniel W Carney
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Gloria Hernandez-Torres
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony S Gibson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deepika Balakrishna
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Hua Zou
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rui Xu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chien-Hung Chen
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ron de Jong
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Douglas R Dougan
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ling Qin
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Simone V Bigi-Botterill
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Alison Chambers
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Joanne Miura
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lucas K Johnson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jacques Ermolieff
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deidre Johns
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jangir Selimkhanov
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lily Kwok
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Kevin DeMent
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chris Proffitt
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Phong Vu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Erick A Lindsey
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony Ivetac
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andy Jennings
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Haixia Wang
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Padma Manam
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cipriano Santos
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cody Fullenwider
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rohan Manohar
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andrew C Flick
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| |
Collapse
|
8
|
Sachan N, Phoon CK, Bu L, Zilberberg L, Ahamed J, Rifkin DB. Binding requirements for latent transforming growth factor Beta2 activation. Matrix Biol Plus 2024; 22:100149. [PMID: 38831847 PMCID: PMC11145359 DOI: 10.1016/j.mbplus.2024.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/15/2024] [Accepted: 05/11/2024] [Indexed: 06/05/2024] Open
Abstract
Although the mechanism for activation of latent TGFβ1 and TGFβ3 is understood to involve the binding of the TGFβ propeptide (LAP) to both an integrin and an insoluble substrate, the activation of latent TGFβ2 has been unclear because the TGFβ2 LAP does not have the classical integrin binding sequence found in the other two TGFβ isoform LAPs. To assess the potential requirement for covalent linkage with a matrix or cell surface protein for the activation of latent TGFβ2, we generated mice in which the TGFβ2 Cys residue predicted to be involved in binding was mutated to Ser (Tgfb2C24S). We reasoned that, if covalent interaction with a second molecule is required for latent TGFβ2 activation, mutant mice should display a Tgfb2 null (Tgfb2-/-)-like phenotype. Tgfb2C24S mice closely phenocopy Tgfb2-/- mice with death in utero between E18 and P1 and with congenital heart and kidney defects similar to those described for Tgfb2-/- mice. The mutant latent TGFβ2 is secreted at levels similar to WT, yet TGFβ signaling monitored as nuclear pSmad2 is suppressed. We conclude that, like latent TGFβ1, latent TGFβ2 activation requires binding to an immobilized matrix or plasma membrane molecule.
Collapse
Affiliation(s)
- Nalani Sachan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Colin K.L. Phoon
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Lei Bu
- Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Lior Zilberberg
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Daniel B. Rifkin
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
9
|
Sun M, Acosta AC, Emerick V, Adams S, Avila MY, Margo CE, Espana EM. Dysfunctional latent transforming growth factor β activation after corneal injury in a classical Ehlers-Danlos model. Matrix Biol 2024; 128:21-30. [PMID: 38340967 PMCID: PMC10996040 DOI: 10.1016/j.matbio.2024.02.004] [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: 08/23/2023] [Revised: 12/17/2023] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Patients with classical Ehlers Danlos syndrome (cEDS) suffer impaired wound healing and from scars formed after injuries that are atrophic and difficult to close surgically. Haploinsufficiency in COL5A1 creates systemic morphological and functional alterations in the entire body. We investigated mechanisms that impair wound healing from corneal lacerations (full thickness injuries) in a mouse model of cEDS (Col5a1+/-). We found that collagen V reexpression in this model is upregulated during corneal tissue repair and that wound healing is delayed, impaired, and results in large atrophic corneal scars. We noted that in a matrix with a 50 % content of collagen V, activation of latent Transforming Growth Factor (TGF) β is dysregulated. Corneal myofibroblasts with a haploinsufficiency of collagen V failed to mechanically activate latent TGF β. Second harmonic imaging microscopy showed a disorganized, undulated, and denser collagen matrix in our Col5a1+/- model that suggested alterations in the extracellular matrix structure and function. We hypothesize that a regenerated collagen matrix with only 50 % content of collagen V is not resistant enough mechanically to allow adequate activation of latent TGF β by fibroblasts and myofibroblasts.
Collapse
Affiliation(s)
- Mei Sun
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Ana Carolina Acosta
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Victoria Emerick
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Sheila Adams
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Marcel Y Avila
- Departament of Ophthalmology, Universidad Nacional de Colombia, Bogota, Colombia
| | - Curtis E Margo
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA; Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Edgar M Espana
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA; Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
10
|
Trinh-Minh T, Chen CW, Tran Manh C, Li YN, Zhu H, Zhou X, Chakraborty D, Zhang Y, Rauber S, Dees C, Lin NY, Kah D, Gerum R, Bergmann C, Kreuter A, Reuter C, Groeber-Becker F, Eckes B, Distler O, Fabry B, Ramming A, Schambony A, Schett G, Distler JH. Noncanonical WNT5A controls the activation of latent TGF-β to drive fibroblast activation and tissue fibrosis. J Clin Invest 2024; 134:e159884. [PMID: 38747285 PMCID: PMC11093613 DOI: 10.1172/jci159884] [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: 03/04/2022] [Accepted: 03/20/2024] [Indexed: 05/19/2024] Open
Abstract
Transforming growth factor β (TGF-β) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-β remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-β in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-β. The activation of latent TGF-β requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-β, rebalanced TGF-β signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-β in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.
Collapse
Affiliation(s)
- Thuong Trinh-Minh
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Chih-Wei Chen
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Cuong Tran Manh
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Yi-Nan Li
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Honglin Zhu
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiang Zhou
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Debomita Chakraborty
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Yun Zhang
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Simon Rauber
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Clara Dees
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Neng-Yu Lin
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Delf Kah
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Richard Gerum
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Alexander Kreuter
- Clinic for Dermatology, Venereology and Allergology, HELIOS St. Elisabeth Clinic Oberhausen, North-Rhine-Westphalia, Germany
| | - Christiane Reuter
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research (ISC) Würzburg, Bavaria, Germany
| | - Florian Groeber-Becker
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research (ISC) Würzburg, Bavaria, Germany
| | - Beate Eckes
- Translational Matrix Biology, University of Cologne, Cologne, North-Rhine-Westphalia, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, North-Rhine-Westphalia, Germany
| | - Oliver Distler
- Rheumaklinik, University Hospital Zurich, Zurich, Switzerland
| | - Ben Fabry
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Andreas Ramming
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Alexandra Schambony
- Division of Developmental Biology, Biology Department, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Georg Schett
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Jörg H.W. Distler
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| |
Collapse
|
11
|
Taber A, Konecny A, Oda SK, Scott-Browne J, Prlic M. TGF-β broadly modifies rather than specifically suppresses reactivated memory CD8 T cells in a dose-dependent manner. Proc Natl Acad Sci U S A 2023; 120:e2313228120. [PMID: 37988468 PMCID: PMC10691214 DOI: 10.1073/pnas.2313228120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/16/2023] [Indexed: 11/23/2023] Open
Abstract
Transforming growth factor β (TGF-β) directly acts on naive, effector, and memory T cells to control cell fate decisions, which was shown using genetic abrogation of TGF-β signaling. TGF-β availability is altered by infections and cancer; however, the dose-dependent effects of TGF-β on memory CD8 T cell (Tmem) reactivation are still poorly defined. We examined how activation and TGF-β signals interact to shape the functional outcome of Tmem reactivation. We found that TGF-β could suppress cytotoxicity in a manner that was inversely proportional to the strength of the activating TCR or proinflammatory signals. In contrast, even high doses of TGF-β had a comparatively modest effect on IFN-γ expression in the context of weak and strong reactivation signals. Since CD8 Tmem may not always receive TGF-β signals concurrently with reactivation, we also explored whether the temporal order of reactivation versus TGF-β signals is of importance. We found that exposure to TGF-β before or after an activation event were both sufficient to reduce cytotoxic effector function. Concurrent ATAC-seq and RNA-seq analysis revealed that TGF-β altered ~10% of the regulatory elements induced by reactivation and also elicited transcriptional changes indicative of broadly modulated functional properties. We confirmed some changes on the protein level and found that TGF-β-induced expression of CCR8 was inversely proportional to the strength of the reactivating TCR signal. Together, our data suggest that TGF-β is not simply suppressing CD8 Tmem but modifies functional and chemotactic properties in context of their reactivation signals and in a dose-dependent manner.
Collapse
Affiliation(s)
- Alexis Taber
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA98109
| | - Andrew Konecny
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA98109
- Department of Immunology, University of Washington, Seattle, WA98195
| | - Shannon K. Oda
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA98101
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA98105
| | - James Scott-Browne
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO80206
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO80045
| | - Martin Prlic
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA98109
- Department of Immunology, University of Washington, Seattle, WA98195
| |
Collapse
|
12
|
Nagesh PT, Nishi H, Rawal S, Zahr T, Miano JM, Sorci-Thomas M, Xu H, Akbar N, Choudhury RP, Misra A, Fisher EA. HDL regulates TGFß-receptor lipid raft partitioning, restoring contractile features of cholesterol-loaded vascular smooth muscle cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.562786. [PMID: 37905061 PMCID: PMC10614922 DOI: 10.1101/2023.10.19.562786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background Cholesterol-loading of mouse aortic vascular smooth muscle cells (mVSMCs) downregulates miR-143/145, a master regulator of the contractile state downstream of TGFβ signaling. In vitro, this results in transitioning from a contractile mVSMC to a macrophage-like state. This process likely occurs in vivo based on studies in mouse and human atherosclerotic plaques. Objectives To test whether cholesterol-loading reduces VSMC TGFβ signaling and if cholesterol efflux will restore signaling and the contractile state in vitro and in vivo. Methods Human coronary artery (h)VSMCs were cholesterol-loaded, then treated with HDL (to promote cholesterol efflux). For in vivo studies, partial conditional deletion of Tgfβr2 in lineage-traced VSMC mice was induced. Mice wild-type for VSMC Tgfβr2 or partially deficient (Tgfβr2+/-) were made hypercholesterolemic to establish atherosclerosis. Mice were then treated with apoA1 (which forms HDL). Results Cholesterol-loading of hVSMCs downregulated TGFβ signaling and contractile gene expression; macrophage markers were induced. TGFβ signaling positively regulated miR-143/145 expression, increasing Acta2 expression and suppressing KLF4. Cholesterol-loading localized TGFβ receptors into lipid rafts, with consequent TGFβ signaling downregulation. Notably, in cholesterol-loaded hVSMCs HDL particles displaced receptors from lipid rafts and increased TGFβ signaling, resulting in enhanced miR-145 expression and decreased KLF4-dependent macrophage features. ApoA1 infusion into Tgfβr2+/- mice restored Acta2 expression and decreased macrophage-marker expression in plaque VSMCs, with evidence of increased TGFβ signaling. Conclusions Cholesterol suppresses TGFβ signaling and the contractile state in hVSMC through partitioning of TGFβ receptors into lipid rafts. These changes can be reversed by promotion of cholesterol efflux, consistent with evidence in vivo.
Collapse
Affiliation(s)
- Prashanth Thevkar Nagesh
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Hitoo Nishi
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Shruti Rawal
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Tarik Zahr
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Mary Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Hao Xu
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Oxford University Hospitals, NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Oxford University Hospitals, NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, NSW, Australia
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, and Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, United States of America
| |
Collapse
|
13
|
Chong DLW, Mikolasch TA, Sahota J, Rebeyrol C, Garthwaite HS, Booth HL, Heightman M, Denneny EK, José RJ, Khawaja AA, Duckworth A, Labelle M, Scotton CJ, Porter JC. Investigating the role of platelets and platelet-derived transforming growth factor-β in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2023; 325:L487-L499. [PMID: 37643008 PMCID: PMC10639018 DOI: 10.1152/ajplung.00227.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Transforming growth factor-β1 (TGFβ1) is the key profibrotic cytokine in idiopathic pulmonary fibrosis (IPF), but the primary source of this cytokine in this disease is unknown. Platelets have abundant stores of TGFβ1, although the role of these cells in IPF is ill-defined. In this study, we investigated whether platelets, and specifically platelet-derived TGFβ1, mediate IPF disease progression. Patients with IPF and non-IPF patients were recruited to determine platelet reactivity, and separate cohorts of patients with IPF were followed for mortality. To study whether platelet-derived TGFβ1 modulates pulmonary fibrosis (PF), mice with a targeted deletion of TGFβ1 in megakaryocytes and platelets (TGFβ1fl/fl.PF4-Cre) were used in the well-characterized bleomycin-induced pulmonary fibrosis (PF) animal model. In a discovery cohort, we found significantly higher mortality in patients with IPF who had elevated platelet counts within the normal range. However, our validation cohort did not confirm this observation, despite significantly increased platelets, neutrophils, active TGFβ1, and CCL5, a chemokine produced by inflammatory cells, in the blood, lung, and bronchoalveolar lavage (BAL) of patients with IPF. In vivo, we showed that despite platelets being readily detected within the lungs of bleomycin-treated mice, neither the degree of pulmonary inflammation nor fibrosis was significantly different between TGFβ1fl/fl.PF4-Cre and control mice. Our results demonstrate for the first time that platelet-derived TGFβ1 does not significantly mediate inflammation or fibrosis in a PF animal model. Furthermore, our human studies revealed blood platelet counts do not consistently predict mortality in IPF but other platelet-derived mediators, such as C-C chemokine ligand 5 (CCL5), may promote neutrophil recruitment and human IPF.NEW & NOTEWORTHY Platelets are a rich source of profibrotic TGFβ; however, the role of platelets in idiopathic pulmonary fibrosis (IPF) is unclear. We identified that patients with IPF have significantly more platelets, neutrophils, and active TGFβ in their airways than control patients. Using an animal model of IPF, we demonstrated that platelet-derived TGFβ does not significantly drive lung fibrosis or inflammation. Our findings offer a better understanding of platelets in both human and animal studies of IPF.
Collapse
Affiliation(s)
- Deborah L W Chong
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| | - Theresia A Mikolasch
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Jagdeep Sahota
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Carine Rebeyrol
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Helen S Garthwaite
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Helen L Booth
- Interstitial Lung Disease Service, University College London Hospital, London, United Kingdom
| | - Melissa Heightman
- Interstitial Lung Disease Service, University College London Hospital, London, United Kingdom
| | - Emma K Denneny
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Ricardo J José
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Akif A Khawaja
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Anna Duckworth
- Department of Clinical and Biomedical Science, University of Exeter, Exeter, United Kingdom
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Chris J Scotton
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
- Department of Clinical and Biomedical Science, University of Exeter, Exeter, United Kingdom
| | - Joanna C Porter
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
14
|
Roy A, Shi L, Chang A, Dong X, Fernandez A, Kraft JC, Li J, Le VQ, Winegar RV, Cherf GM, Slocum D, Poulson PD, Casper GE, Vallecillo-Zúniga ML, Valdoz JC, Miranda MC, Bai H, Kipnis Y, Olshefsky A, Priya T, Carter L, Ravichandran R, Chow CM, Johnson MR, Cheng S, Smith M, Overed-Sayer C, Finch DK, Lowe D, Bera AK, Matute-Bello G, Birkland TP, DiMaio F, Raghu G, Cochran JR, Stewart LJ, Campbell MG, Van Ry PM, Springer T, Baker D. De novo design of highly selective miniprotein inhibitors of integrins αvβ6 and αvβ8. Nat Commun 2023; 14:5660. [PMID: 37704610 PMCID: PMC10500007 DOI: 10.1038/s41467-023-41272-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
The RGD (Arg-Gly-Asp)-binding integrins αvβ6 and αvβ8 are clinically validated cancer and fibrosis targets of considerable therapeutic importance. Compounds that can discriminate between homologous αvβ6 and αvβ8 and other RGD integrins, stabilize specific conformational states, and have high thermal stability could have considerable therapeutic utility. Existing small molecule and antibody inhibitors do not have all these properties, and hence new approaches are needed. Here we describe a generalized method for computationally designing RGD-containing miniproteins selective for a single RGD integrin heterodimer and conformational state. We design hyperstable, selective αvβ6 and αvβ8 inhibitors that bind with picomolar affinity. CryoEM structures of the designed inhibitor-integrin complexes are very close to the computational design models, and show that the inhibitors stabilize specific conformational states of the αvβ6 and the αvβ8 integrins. In a lung fibrosis mouse model, the αvβ6 inhibitor potently reduced fibrotic burden and improved overall lung mechanics, demonstrating the therapeutic potential of de novo designed integrin binding proteins with high selectivity.
Collapse
Affiliation(s)
- Anindya Roy
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Lei Shi
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Encodia Inc, 5785 Oberlin Drive, San Diego, CA, 92121, USA
| | - Ashley Chang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Xianchi Dong
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, Nanjing, China
| | - Andres Fernandez
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - John C Kraft
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - Viet Q Le
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - Rebecca Viazzo Winegar
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Gerald Maxwell Cherf
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Denali Therapeutics, South San Francisco, CA, USA
| | - Dean Slocum
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - P Daniel Poulson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Garrett E Casper
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | | | - Jonard Corpuz Valdoz
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Marcos C Miranda
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Hua Bai
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Yakov Kipnis
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Audrey Olshefsky
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Tanu Priya
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Lauren Carter
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Rashmi Ravichandran
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Cameron M Chow
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Max R Johnson
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Suna Cheng
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - McKaela Smith
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Catherine Overed-Sayer
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Donna K Finch
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Alchemab Therapeutics Ltd, Cambridge, UK
| | - David Lowe
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Evox Therapeutics Limited, Oxford Science Park, Medawar Centre, East Building, Robert Robinson Avenue, Oxford, OX4 4HG, England
| | - Asim K Bera
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Gustavo Matute-Bello
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, USA
| | - Timothy P Birkland
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Frank DiMaio
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Ganesh Raghu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Lance J Stewart
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
| | - Pam M Van Ry
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA.
| | - Timothy Springer
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA.
| | - David Baker
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
15
|
Cirella A, Bolaños E, Luri-Rey C, Di Trani CA, Olivera I, Gomis G, Glez-Vaz J, Pinci B, Garasa S, Sánchez-Gregorio S, Azpilikueta A, Eguren-Santamaria I, Valencia K, Palencia B, Alvarez M, Ochoa MC, Teijeira Á, Berraondo P, Melero I. Intratumoral immunotherapy with mRNAs encoding chimeric protein constructs encompassing IL-12, CD137 agonists, and TGF-β antagonists. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:668-682. [PMID: 37650116 PMCID: PMC10462790 DOI: 10.1016/j.omtn.2023.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023]
Abstract
Intratumoral immunotherapy strategies for cancer based on interleukin-12 (IL-12)-encoding cDNA and mRNA are under clinical development in combination with anti-PD-(L)1 monoclonal antibodies. To make the most of these approaches, we have constructed chimeric mRNAs encoding single-chain IL-12 fused to single-chain fragment variable (scFv) antibodies that bind to transforming growth factor β (TGF-β) and CD137 (4-1BB). Several neutralizing TGF-β agents and CD137 agonists are also undergoing early-phase clinical trials. To attain TGF-β and CD137 binding by the constructions, we used bispecific tandem scFv antibodies (taFvs) derived from the specific 1D11 and 1D8 monoclonal antibodies (mAbs), respectively. Transfection of mRNAs encoding the chimeric constructs achieved functional expression of the proteins able to act on their targets. Upon mRNA intratumoral injections in the transplantable mouse cancer models CT26, MC38, and B16OVA, potent therapeutic effects were observed following repeated injections into the tumors. Efficacy was dependent on the number of CD8+ T cells able to recognize tumor antigens that infiltrated the malignant tissue. Although the abscopal effects on concomitant uninjected lesions were modest, such distant effects on untreated lesions were markedly increased when combined with systemic PD-1 blockade.
Collapse
Affiliation(s)
- Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Gabriel Gomis
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Beatrice Pinci
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Saray Garasa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Sandra Sánchez-Gregorio
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Arantza Azpilikueta
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Iñaki Eguren-Santamaria
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Karmele Valencia
- Program of Solid Tumors, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Belén Palencia
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Maite Alvarez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Maria C. Ochoa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Álvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Department of Oncology, Clínica Universidad de Navarra, 28027 Madrid, Spain
- Centro Del Cancer de La Universidad de Navarra (CCUN), 31008 Pamplona, Spain
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
16
|
Steinberg T, Dieterle MP, Ramminger I, Klein C, Brossette J, Husari A, Tomakidi P. On the Value of In Vitro Cell Systems for Mechanobiology from the Perspective of Yes-Associated Protein/Transcriptional Co-Activator with a PDZ-Binding Motif and Focal Adhesion Kinase and Their Involvement in Wound Healing, Cancer, Aging, and Senescence. Int J Mol Sci 2023; 24:12677. [PMID: 37628858 PMCID: PMC10454169 DOI: 10.3390/ijms241612677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Mechanobiology comprises how cells perceive different mechanical stimuli and integrate them into a process called mechanotransduction; therefore, the related mechanosignaling cascades are generally important for biomedical research. The ongoing discovery of key molecules and the subsequent elucidation of their roles in mechanobiology are fundamental to understanding cell responses and tissue conditions, such as homeostasis, aging, senescence, wound healing, and cancer. Regarding the available literature on these topics, it becomes abundantly clear that in vitro cell systems from different species and tissues have been and are extremely valuable tools for enabling the discovery and functional elucidation of key mechanobiological players. Therefore, this review aims to discuss the significant contributions of in vitro cell systems to the identification and characterization of three such key players using the selected examples of yes-associated protein (YAP), its paralog transcriptional co-activator with a PDZ-binding motif (TAZ), and focal adhesion kinase (FAK) and their involvement in wound healing, cancer, aging, and senescence. In addition, the reader is given suggestions as to which future prospects emerge from the in vitro studies discussed herein and which research questions still remain open.
Collapse
Affiliation(s)
- Thorsten Steinberg
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany
| | - Martin Philipp Dieterle
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany
| | - Imke Ramminger
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104 Freiburg, Germany
| | - Charlotte Klein
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany
| | - Julie Brossette
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104 Freiburg, Germany
| | - Ayman Husari
- Center for Dental Medicine, Department of Orthodontics, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany
| | - Pascal Tomakidi
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany
| |
Collapse
|
17
|
Taber A, Konecny A, Scott-Browne J, Prlic M. TGF-β broadly modifies rather than specifically suppresses reactivated memory CD8 T cells in a dose-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.27.550871. [PMID: 37546887 PMCID: PMC10402134 DOI: 10.1101/2023.07.27.550871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Transforming growth factor β (TGF-β) directly acts on naïve, effector and memory T cells to control cell fate decisions, which was shown using genetic abrogation of TGF-β signaling. TGF-β availability is altered by infections and cancer, however the dose-dependent effects of TGF-β on memory CD8 T cell (Tmem) reactivation are still poorly defined. We examined how activation and TGF-β signals interact to shape the functional outcome of Tmem reactivation. We found that TGF-β could suppress cytotoxicity in a manner that was inversely proportional to the strength of the activating TCR or pro-inflammatory signals. In contrast, even high doses of TGF-β had a comparatively modest effect on IFN-γ expression in the context of weak and strong reactivation signals. Since CD8 Tmem may not always receive TGF-β signals concurrently with reactivation, we also explored whether the temporal order of reactivation versus TGF-β signals is of importance. We found that exposure to TGF-β prior to as well as after an activation event were both sufficient to reduce cytotoxic effector function. Concurrent ATAC-seq and RNA-seq analysis revealed that TGF-β altered ~10% of the regulatory elements induced by reactivation and also elicited transcriptional changes indicative of broadly modulated functional properties. We confirmed some changes on the protein level and found that TGF-β-induced expression of CCR8 was inversely proportional to the strength of the reactivating TCR signal. Together, our data suggest that TGF-β is not simply suppressing CD8 Tmem, but modifies functional and chemotactic properties in context of their reactivation signals and in a dose-dependent manner.
Collapse
Affiliation(s)
- Alexis Taber
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA 98109, USA
| | - Andrew Konecny
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98195
| | - James Scott-Browne
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO 80206
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Martin Prlic
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98195
| |
Collapse
|
18
|
Ruan Q, Lin X, Wang L, Wang N, Zhao Y, Wang H, Tian FY, Hu N, Li Y, Zhao B. An engineered (CAGA)12-EGFP cell-based biosensor for high-content and accurate detection of active TGF-β. Biosens Bioelectron 2022; 220:114884. [DOI: 10.1016/j.bios.2022.114884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/10/2022]
|
19
|
Radwanska A, Cottage CT, Piras A, Overed-Sayer C, Sihlbom C, Budida R, Wrench C, Connor J, Monkley S, Hazon P, Schluter H, Thomas MJ, Hogaboam CM, Murray LA. Increased expression and accumulation of GDF15 in IPF extracellular matrix contribute to fibrosis. JCI Insight 2022; 7:153058. [PMID: 35993367 PMCID: PMC9462497 DOI: 10.1172/jci.insight.153058] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unmet medical need. It is characterized by formation of scar tissue leading to a progressive and irreversible decline in lung function. IPF is associated with repeated injury, which may alter the composition of the extracellular matrix (ECM). Here, we demonstrate that IPF patient–derived pulmonary ECM drives profibrotic response in normal human lung fibroblasts (NHLF) in a 3D spheroid assay. Next, we reveal distinct alterations in composition of the diseased ECM, identifying potentially novel associations with IPF. Growth differentiation factor 15 (GDF15) was identified among the most significantly upregulated proteins in the IPF lung–derived ECM. In vivo, GDF15 neutralization in a bleomycin-induced lung fibrosis model led to significantly less fibrosis. In vitro, recombinant GDF15 (rGDF15) stimulated α smooth muscle actin (αSMA) expression in NHLF, and this was mediated by the activin receptor-like kinase 5 (ALK5) receptor. Furthermore, in the presence of rGDF15, the migration of NHLF in collagen gel was reduced. In addition, we observed a cell type–dependent effect of GDF15 on the expression of cell senescence markers. Our data suggest that GDF15 mediates lung fibrosis through fibroblast activation and differentiation, implicating a potential direct role of this matrix-associated cytokine in promoting aberrant cell responses in disease.
Collapse
Affiliation(s)
- Agata Radwanska
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christopher Travis Cottage
- Bioscience COPD/IPF, Research and Early Development, R&I, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Antonio Piras
- Bioscience In Vivo, Research and Early Development, R&I, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine Overed-Sayer
- Bioscience COPD/IPF, Research and Early Development, R&I, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Carina Sihlbom
- Proteomics Core Facility of Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Ramachandramouli Budida
- Translational Science and Experimental Medicine, Research and Early Development, R&I, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine Wrench
- Bioscience COPD/IPF, Research and Early Development, R&I, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Jane Connor
- Bioscience COPD/IPF, Research and Early Development, R&I, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Susan Monkley
- Translational Science and Experimental Medicine, Research and Early Development, R&I, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Petra Hazon
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Holger Schluter
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Matthew J. Thomas
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Lynne A. Murray
- Bioscience COPD/IPF, Research and Early Development, R&I, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
20
|
Duxfield A, Munkley J, Briggs MD, Dennis EP. CRELD2 is a novel modulator of calcium release and calcineurin-NFAT signalling during osteoclast differentiation. Sci Rep 2022; 12:13884. [PMID: 35974042 PMCID: PMC9381524 DOI: 10.1038/s41598-022-17347-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022] Open
Abstract
Cysteine rich with epidermal growth factor (EGF)-like domains 2 (CRELD2) is an endoplasmic reticulum (ER) resident chaperone protein with calcium binding properties. CRELD2 is an ER-stress regulated gene that has been implicated in the pathogenesis of skeletal dysplasias and has been shown to play an important role in the differentiation of chondrocytes and osteoblasts. Despite CRELD2 having an established role in skeletal development and bone formation, its role in osteoclasts is currently unknown. Here we show for the first time that CRELD2 plays a novel role in trafficking transforming growth factor beta 1 (TGF-β1), which is linked to an upregulation in the expression of Nfat2, the master regulator of osteoclast differentiation in early osteoclastogenesis. Despite this finding, we show that overexpressing CRELD2 impaired osteoclast differentiation due to a reduction in the activity of the calcium-dependant phosphatase, calcineurin. This in turn led to a subsequent block in the dephosphorylation of nuclear factor of activated T cells 1 (NFATc1), preventing its nuclear localisation and activation as a pro-osteoclastogenic transcription factor. Our exciting results show that the overexpression of Creld2 in osteoclasts impaired calcium release from the ER which is essential for activating calcineurin and promoting osteoclastogenesis. Therefore, our data proposes a novel inhibitory role for this calcium-binding ER-resident chaperone in modulating calcium flux during osteoclast differentiation which has important implications in our understanding of bone remodelling and the pathogenesis of skeletal diseases.
Collapse
Affiliation(s)
- Adam Duxfield
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK
| | - Jennifer Munkley
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK
| | - Michael D Briggs
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK
| | - Ella P Dennis
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK.
| |
Collapse
|
21
|
Singh K, Sachan N, Ene T, Dabovic B, Rifkin D. Latent Transforming Growth Factor β Binding Protein 3 Controls Adipogenesis. Matrix Biol 2022; 112:155-170. [PMID: 35933071 DOI: 10.1016/j.matbio.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022]
Abstract
Transforming growth factor-beta (TGFβ) is released from cells as part of a trimeric latent complex consisting of TGFβ, the TGFβ propeptides, and either a latent TGFβ binding protein (LTBP) or glycoprotein-A repetitions predominant (GARP) protein. LTBP1 and 3 modulate latent TGFβ function with respect to secretion, matrix localization, and activation and, therefore, are vital for the proper function of the cytokine in a number of tissues. TGFβ modulates stem cell differentiation into adipocytes (adipogenesis), but the potential role of LTBPs in this process has not been studied. We observed that 72 h post adipogenesis initiation Ltbp1, 2, and 4 expression levels decrease by 74-84%, whereas Ltbp3 expression levels remain constant during adipogenesis. We found that LTBP3 silencing in C3H/10T1/2 cells reduced adipogenesis, as measured by the percentage of cells with lipid vesicles and the expression of the transcription factor peroxisome proliferator-activated receptor gamma (PPARγ). Lentiviral mediated expression of an Ltbp3 mRNA resistant to siRNA targeting rescued the phenotype, validating siRNA specificity. Knockdown (KD) of Ltbp3 expression in 3T3-L1, M2, and primary bone marrow stromal cells (BMSC) indicated a similar requirement for Ltbp3. Epididymal and inguinal white adipose tissue fat pad weights of Ltbp3-/- mice were reduced by 62% and 57%, respectively, compared to wild-type mice. Inhibition of adipogenic differentiation upon LTBP3 loss is mediated by TGFβ, as TGFβ neutralizing antibody and TGFβ receptor I kinase blockade rescue the LTBP3 KD phenotype. These results indicate that LTBP3 has a TGFβ-dependent function in adipogenesis both in vitro and possibly in vivo.
Collapse
Affiliation(s)
- Karan Singh
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nalani Sachan
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Taylor Ene
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Branka Dabovic
- Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel Rifkin
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA; Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
22
|
The Role of the Fibronectin Synergy Site for Skin Wound Healing. Cells 2022; 11:cells11132100. [PMID: 35805184 PMCID: PMC9265582 DOI: 10.3390/cells11132100] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 12/03/2022] Open
Abstract
Skin is constantly exposed to injuries that are repaired with different outcomes, either regeneration or scarring. Scars result from fibrotic processes modulated by cellular physical forces transmitted by integrins. Fibronectin (FN) is a major component in the provisional matrix assembled to repair skin wounds. FN enables cell adhesion binding of α5β1/αIIbβ3 and αv-class integrins to an RGD-motif. An additional linkage for α5/αIIb is the synergy site located in close proximity to the RGD motif. The mutation to impair the FN synergy region (Fn1syn/syn) demonstrated that its absence permits complete development. However, only with the additional engagement to the FN synergy site do cells efficiently resist physical forces. To test how the synergy site-mediated adhesion affects the course of wound healing fibrosis, we used a mouse model of skin injury and in-vitro migration studies with keratinocytes and fibroblasts on FNsyn. The loss of FN synergy site led to normal re-epithelialization caused by two opposing migratory defects of activated keratinocytes and, in the dermis, induced reduced fibrotic responses, with lower contents of myofibroblasts and FN deposition and diminished TGF-β1-mediated cell signalling. We demonstrate that weakened α5β1-mediated traction forces on FNsyn cause reduced TGF-β1 release from its latent complex.
Collapse
|
23
|
Ko MK, Woo JI, Gonzalez JM, Kim G, Sakai L, Peti-Peterdi J, Kelber JA, Hong YK, Tan JC. Fibrillin-1 mutant mouse captures defining features of human primary open glaucoma including anomalous aqueous humor TGF beta-2. Sci Rep 2022; 12:10623. [PMID: 35739142 PMCID: PMC9226129 DOI: 10.1038/s41598-022-14062-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Primary open angle glaucoma (POAG) features an optic neuropathy, elevated aqueous humor (AH) TGFβ2, and major risk factors of central corneal thickness (CCT), increasing age and intraocular pressure (IOP). We examined Tight skin (Tsk) mice to see if mutation of fibrillin-1, a repository for latent TGFβ, is associated with characteristics of human POAG. We measured: CCT by ocular coherence tomography (OCT); IOP; retinal ganglion cell (RGC) and optic nerve axon counts by microscopic techniques; visual electrophysiologic scotopic threshold responses (STR) and pattern electroretinogram (PERG); and AH TGFβ2 levels and activity by ELISA and MINK epithelial cell-based assays respectively. Tsk mice had open anterior chamber angles and compared with age-matched wild type (WT) mice: 23% thinner CCT (p < 0.003); IOP that was higher (p < 0.0001), more asymmetric (p = 0.047), rose with age (p = 0.04) and had a POAG-like frequency distribution. Tsk mice also had RGCs that were fewer (p < 0.04), declined with age (p = 0.0003) and showed increased apoptosis and glial activity; fewer optic nerve axons (p = 0.02); abnormal axons and glia; reduced STR (p < 0.002) and PERG (p < 0.007) visual responses; and higher AH TGFβ2 levels (p = 0.0002) and activity (p = 1E-11) especially with age. Tsk mice showed defining features of POAG, implicating aberrant fibrillin-1 homeostasis as a pathogenic contributor to emergence of a POAG phenotype.
Collapse
Affiliation(s)
| | | | | | | | - Lynn Sakai
- Department of Medical and Molecular Genetics, Oregon Health Sciences University, Portland, OR, USA
| | - Janos Peti-Peterdi
- Departments of Physiology, Biophysics and Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jonathan A Kelber
- Developmental Oncogene Laboratory, California State University Northridge, Northridge, CA, USA
| | - Young-Kwon Hong
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - James C Tan
- Doheny Eye Institute, Los Angeles, CA, USA.
- Department of Ophthalmology, University of California Los Angeles, Los Angeles, CA, USA.
- Sightgene, Inc., 9227 Reseda Blvd, #182, Northridge, CA, 91324-3137, USA.
| |
Collapse
|
24
|
Wang C, Dong L, Zhao Z, Zhang Z, Sun Y, Li C, Li G, You X, Yang X, Wang H, Hong W. Design and Synthesis of Novel PRMT1 Inhibitors and Investigation of Their Effects on the Migration of Cancer Cell. Front Chem 2022; 10:888727. [PMID: 35755248 PMCID: PMC9214036 DOI: 10.3389/fchem.2022.888727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Protein arginine methyltransferase 1 (PRMT1) can catalyze the protein arginine methylation by transferring the methyl group from S-adenosyl-L-methionine (SAM) to the guanidyl nitrogen atom of protein arginine, which influences a variety of biological processes including epithelial-mesenchymal transition (EMT) and EMT-mediated mobility of cancer cells. The upregulation of PRMT1 is involved in a diverse range of cancer, such as lung cancer, and there is an urgent need to develop novel and potent PRMT1 inhibitors. In this article, a series of 2,5-substituted furan derivatives and 2,4-substituted thiazole derivatives were designed and synthesized by targeting at the substrate arginine-binding site on PRMT1, and 10 compounds demonstrated significant inhibitory effects against PRMT1. Among them, the most potent inhibitor, compound 1r (WCJ-394), significantly affected the expression of PRMT1-related proteins in A549 cells and downregulated the expression of mesenchymal markers, by which WCJ-394 inhibited the TGF-β1-induced EMT in A549 cells and prevented the cancer cell migration. The current study demonstrated that WCJ-394 was a potent PRMT1 inhibitor, which could be used as the leading compound for further drug discovery.
Collapse
Affiliation(s)
- Caijiao Wang
- School of Chemistry and Chemical Engineering, North Minzu University, Yinchuan, China
| | - Luyao Dong
- Beijing Key Laboratory of Antimicrobial Agents/Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziqi Zhao
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Zeqing Zhang
- School of Chemistry and Chemical Engineering, North Minzu University, Yinchuan, China
| | - Yutong Sun
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Chonglong Li
- School of Chemistry and Chemical Engineering, North Minzu University, Yinchuan, China
| | - Guoqing Li
- Beijing Key Laboratory of Antimicrobial Agents/Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuefu You
- Beijing Key Laboratory of Antimicrobial Agents/Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyi Yang
- Beijing Key Laboratory of Antimicrobial Agents/Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Wang
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine, Minzu University of China, Ministry of Education, Beijing, China
- Institute of National Security, Minzu University of China, Beijing, China
| | - Wei Hong
- School of Chemistry and Chemical Engineering, North Minzu University, Yinchuan, China
- Jingjinji National Center of Technology Innovation, Beijing, China
| |
Collapse
|
25
|
Parente-Pereira AC, Beatson RE, Davies DM, Hull C, Whilding LM, Porter JC, Maher J. Generation and application of TGFβ-educated human Vγ9Vδ2 T cells. STAR Protoc 2022; 3:101319. [PMID: 35496793 PMCID: PMC9043756 DOI: 10.1016/j.xpro.2022.101319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Clinical trials that tested the antitumor activity of γδ T cells have been mostly unsuccessful. To address this, we expanded human Vγ9Vδ2 T cells in TGFβ1, a cytokine which enhances their viability, trafficking properties, and intrinsic antitumor activity. This protocol summarizes the production and in vitro functional characterization of TGFβ1 educated human Vγ9Vδ2 cells and highlights their compatibility with chimeric antigen receptor (CAR) engineering. We also describe in vivo testing of the antitumor activity of these CAR T cells in mice. For complete details on the use and execution of this protocol, please refer to Beatson et al. (2021). TGFβ promotes viability and retards differentiation of Vγ9Vδ2 cells TGFβ-educated Vγ9Vδ2 cells achieve greater invasion and anticancer activity Specificity of these cells can be reprogrammed using chimeric antigen receptors
Collapse
|
26
|
Seed RI, Nishimura SL. Measurement of Cell Intrinsic TGF-β Activation Mediated by the Integrin αvβ8. Bio Protoc 2022; 12:e4385. [PMID: 35800099 PMCID: PMC9081475 DOI: 10.21769/bioprotoc.4385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/03/2021] [Accepted: 02/28/2022] [Indexed: 12/29/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a multi-functional cytokine that plays a significant role in multiple diseases, including fibrosis and tumor progression. Whilst the biologic effects of TGF-β are well characterized, it is unclear how TGF-β signaling is regulated to impart specific responses within certain cell types. One mechanism of regulation may be through TGF-β activation, since TGF-β is always expressed in a latent form (L-TGF-β). Campbell et al.(2020) recently presented a new structural model to demonstrate how the integrin αvβ8 might specifically control TGF-β activation and signaling. In this model, αvβ8 binds to cell surface L-TGF-β1 to induce a conformational change, which exposes mature TGF-β peptide to TGF-β receptors (TGF-βRs), allowing initiation of TGF-β signaling from within the latent complex. This model also predicts that TGF-β signaling would be directed specifically towards the TGF-β expressing cell surface. We sought to test the validity of the new structural model by creating a cell-based assay which utilizes luciferase TGF-β reporter cells (TMLC). TMLC cells express high levels of TGF-βRs, but do not express cell surface L-TGF-β. We modified TMLC reporter cells to express cell surface L-TGF-β1 in a mutant form, which prevents the release of mature TGF-β from the latent complex. The newly generated cell lines were then used in a novel functional assay to investigate whether integrin αvβ8 could potentiate cell intrinsic TGF-β signaling from within the latent complex in vitro.
Collapse
Affiliation(s)
- Robert Ian Seed
- Department of Pathology, University of California, San Francisco, San Francisco, USA,*For correspondence:
| | - Stephen Lloyd Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, USA,ImmunoX initiative, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
27
|
Hargest V, Bub T, Neale G, Schultz-Cherry S. Astrovirus-induced epithelial-mesenchymal transition via activated TGF-β increases viral replication. PLoS Pathog 2022; 18:e1009716. [PMID: 35452499 PMCID: PMC9067694 DOI: 10.1371/journal.ppat.1009716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 05/04/2022] [Accepted: 03/30/2022] [Indexed: 11/18/2022] Open
Abstract
Human astroviruses (HAstV), positive sense single-stranded RNA viruses, are one of the leading causes of diarrhea worldwide. Despite their high prevalence, the cellular mechanisms of astrovirus pathogenesis remain ill-defined. Previous studies showed HAstV increased epithelial barrier permeability by causing a re-localization of the tight junction protein, occludin. In these studies, we demonstrate that HAstV replication induces epithelial-mesenchymal transition (EMT), by upregulating the transcription of EMT-related genes within 8 hours post-infection (hpi), followed by the loss of cell-cell contacts and disruption of polarity by 24 hpi. While multiple classical HAstV serotypes, including clinical isolates, induce EMT, the non-classical genotype HAstV-VA1 and two strains of reovirus are incapable of inducing EMT. Unlike the re-localization of tight junction proteins, HAstV-induced EMT requires productive replication and is dependent transforming growth factor-β (TGF-β) activity. Finally, inhibiting TGF-β signaling and EMT reduces viral replication, highlighting its importance in the viral life cycle. This finding puts classical strains of HAstV-1 in an exclusive group of non-oncogenic viruses triggering EMT.
Collapse
Affiliation(s)
- Virginia Hargest
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Theresa Bub
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Integrated Program of Biomedical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
28
|
Li Y, Fan W, Link F, Wang S, Dooley S. Transforming growth factor β latency: A mechanism of cytokine storage and signalling regulation in liver homeostasis and disease. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 4:100397. [PMID: 35059619 PMCID: PMC8760520 DOI: 10.1016/j.jhepr.2021.100397] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) is a potent effector in the liver, which is involved in a plethora of processes initiated upon liver injury. TGF-β affects parenchymal, non-parenchymal, and inflammatory cells in a highly context-dependent manner. Its bioavailability is critical for a fast response to various insults. In the liver – and probably in other organs – this is made possible by the deposition of a large portion of TGF-β in the extracellular matrix as an inactivated precursor form termed latent TGF-β (L-TGF-β). Several matrisomal proteins participate in matrix deposition, latent complex stabilisation, and activation of L-TGF-β. Extracellular matrix protein 1 (ECM1) was recently identified as a critical factor in maintaining the latency of deposited L-TGF-β in the healthy liver. Indeed, its depletion causes spontaneous TGF-β signalling activation with deleterious effects on liver architecture and function. This review article presents the current knowledge on intracellular L-TGF-β complex formation, secretion, matrix deposition, and activation and describes the proteins and processes involved. Further, we emphasise the therapeutic potential of toning down L-TGF-β activation in liver fibrosis and liver cancer.
Collapse
Affiliation(s)
- Yujia Li
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Weiguo Fan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford CA, USA
| | - Frederik Link
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sai Wang
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Tel.: 06213835595.
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Corresponding authors. Addresses: Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Tel.: 06213833768;
| |
Collapse
|
29
|
Sun M, Koudouna E, Cogswell D, Avila MY, Koch M, Espana EM. Collagen XII Regulates Corneal Stromal Structure by Modulating Transforming Growth Factor-β Activity. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:308-319. [PMID: 34774848 PMCID: PMC8908044 DOI: 10.1016/j.ajpath.2021.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 02/03/2023]
Abstract
Collagen XII is a regulator of corneal stroma structure and function. The current study examined the role of collagen XII in regulating corneal stromal transforming growth factor (TGF)-β activation and latency. Specifically, with the use of conventional collagen XII null mouse model, the role of collagen XII in the regulation of TGF-β latency and activity in vivo was investigated. Functional quantification of latent TGF-β in stromal matrix was performed by using transformed mink lung reporter cells that produce luciferase as a function of active TGF-β. Col12a1 knockdown with shRNA was used to test the role of collagen XII in TGF-β activation. Col12a1-/- hypertrophic stromata were observed with keratocyte hyperplasia. Increased collagen fibril forward signal was found by second harmonic generation microscopy in the absence of collagen XII. Collagen XII regulated mRNA synthesis of Serpine1, Col1a1, and Col5a1 and deposition of collagens in the extracellular matrix. A functional plasminogen activator inhibitor luciferase assay showed that collagen XII is necessary for latent TGF-β storage in the extracellular matrix and that collagen XII down-regulates active TGF-β. Collagen XII dictates stromal structure and function by regulating TGF-β activity. A hypertrophic phenotype in Col12a1-/- corneal tissue can be explained by abnormal up-regulation of TGF-β activation and decreased latent storage.
Collapse
Affiliation(s)
- Mei Sun
- Cornea and External Disease, Department of Ophthalmology, Department of Molecular Pharmacology and Physiology, Tampa, Florida
| | - Elena Koudouna
- Structural Biophysics, School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Devon Cogswell
- Cornea and External Disease, Department of Ophthalmology, Department of Molecular Pharmacology and Physiology, Tampa, Florida
| | - Marcel Y. Avila
- Department of Ophthalmology, Universidad Nacional de Colombia, Bogota, Colombia
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, University of Cologne, Cologne, Germany
| | - Edgar M. Espana
- Cornea and External Disease, Department of Ophthalmology, Department of Molecular Pharmacology and Physiology, Tampa, Florida,Morsani College of Medicine, University of South Florida, Tampa, Florida,Address correspondence to Edgar M. Espana, M.D., Ophthalmology, University of South Florida, Morsani College of Medicine, 13330 USF Laurel Dr., 4th Floor, MDC11, Tampa, FL 33612.
| |
Collapse
|
30
|
Lainé A, Labiad O, Hernandez-Vargas H, This S, Sanlaville A, Léon S, Dalle S, Sheppard D, Travis MA, Paidassi H, Marie JC. Regulatory T cells promote cancer immune-escape through integrin αvβ8-mediated TGF-β activation. Nat Commun 2021; 12:6228. [PMID: 34711823 PMCID: PMC8553942 DOI: 10.1038/s41467-021-26352-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Presence of TGFβ in the tumor microenvironment is one of the most relevant cancer immune-escape mechanisms. TGFβ is secreted in an inactive form, and its activation within the tumor may depend on different cell types and mechanisms than its production. Here we show in mouse melanoma and breast cancer models that regulatory T (Treg) cells expressing the β8 chain of αvβ8 integrin (Itgβ8) are the main cell type in the tumors that activates TGFβ, produced by the cancer cells and stored in the tumor micro-environment. Itgβ8 ablation in Treg cells impairs TGFβ signalling in intra-tumoral T lymphocytes but not in the tumor draining lymph nodes. Successively, the effector function of tumor infiltrating CD8+ T lymphocytes strengthens, leading to efficient control of tumor growth. In cancer patients, anti-Itgβ8 antibody treatment elicits similar improved cytotoxic T cell activation. Thus, this study reveals that Treg cells work in concert with cancer cells to produce bioactive-TGFβ and to create an immunosuppressive micro-environment.
Collapse
Affiliation(s)
- Alexandra Lainé
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Université Lyon 1, 69373, Lyon, France
| | - Ossama Labiad
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Université Lyon 1, 69373, Lyon, France
| | - Hector Hernandez-Vargas
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Université Lyon 1, 69373, Lyon, France
| | - Sébastien This
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, 69007, Lyon, France
| | - Amélien Sanlaville
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Université Lyon 1, 69373, Lyon, France
| | - Sophie Léon
- Plateforme Ex-Vivo, Département de Recherche Translationnelle et d'Innovation, Centre Léon Bérard, Lyon, France
| | - Stéphane Dalle
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Université Lyon 1, 69373, Lyon, France
- Department of Dermatology, Claude Bernard Université Lyon 1, Centre Hospitalier Lyon Sud, 69495, Pierre Bénite, France
| | - Dean Sheppard
- University of California San Francisco, San Francisco, CA, USA
| | - Mark A Travis
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Helena Paidassi
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, 69007, Lyon, France
| | - Julien C Marie
- Tumor Escape Resistance and Immunity department, Cancer Research Center of Lyon INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Claude Bernard Université Lyon 1, 69373, Lyon, France.
| |
Collapse
|
31
|
Decaris ML, Schaub JR, Chen C, Cha J, Lee GG, Rexhepaj M, Ho SS, Rao V, Marlow MM, Kotak P, Budi EH, Hooi L, Wu J, Fridlib M, Martin SP, Huang S, Chen M, Muñoz M, Hom TF, Wolters PJ, Desai TJ, Rock F, Leftheris K, Morgans DJ, Lepist EI, Andre P, Lefebvre EA, Turner SM. Dual inhibition of α vβ 6 and α vβ 1 reduces fibrogenesis in lung tissue explants from patients with IPF. Respir Res 2021; 22:265. [PMID: 34666752 PMCID: PMC8524858 DOI: 10.1186/s12931-021-01863-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022] Open
Abstract
RATIONALE αv integrins, key regulators of transforming growth factor-β activation and fibrogenesis in in vivo models of pulmonary fibrosis, are expressed on abnormal epithelial cells (αvβ6) and fibroblasts (αvβ1) in fibrotic lungs. OBJECTIVES We evaluated multiple αv integrin inhibition strategies to assess which most effectively reduced fibrogenesis in explanted lung tissue from patients with idiopathic pulmonary fibrosis. METHODS Selective αvβ6 and αvβ1, dual αvβ6/αvβ1, and multi-αv integrin inhibitors were characterized for potency, selectivity, and functional activity by ligand binding, cell adhesion, and transforming growth factor-β cell activation assays. Precision-cut lung slices generated from lung explants from patients with idiopathic pulmonary fibrosis or bleomycin-challenged mouse lungs were treated with integrin inhibitors or standard-of-care drugs (nintedanib or pirfenidone) and analyzed for changes in fibrotic gene expression or TGF-β signaling. Bleomycin-challenged mice treated with dual αvβ6/αvβ1 integrin inhibitor, PLN-74809, were assessed for changes in pulmonary collagen deposition and Smad3 phosphorylation. MEASUREMENTS AND MAIN RESULTS Inhibition of integrins αvβ6 and αvβ1 was additive in reducing type I collagen gene expression in explanted lung tissue slices from patients with idiopathic pulmonary fibrosis. These data were replicated in fibrotic mouse lung tissue, with no added benefit observed from inhibition of additional αv integrins. Antifibrotic efficacy of dual αvβ6/αvβ1 integrin inhibitor PLN-74809 was confirmed in vivo, where dose-dependent inhibition of pulmonary Smad3 phosphorylation and collagen deposition was observed. PLN-74809 also, more potently, reduced collagen gene expression in fibrotic human and mouse lung slices than clinically relevant concentrations of nintedanib or pirfenidone. CONCLUSIONS In the fibrotic lung, dual inhibition of integrins αvβ6 and αvβ1 offers the optimal approach for blocking fibrogenesis resulting from integrin-mediated activation of transforming growth factor-β.
Collapse
Affiliation(s)
| | | | - Chun Chen
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Jacob Cha
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Gail G Lee
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Steve S Ho
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Vikram Rao
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Prerna Kotak
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Erine H Budi
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Lisa Hooi
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Jianfeng Wu
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | | | - Shaoyi Huang
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Ming Chen
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Manuel Muñoz
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Tushar J Desai
- Department of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - David J Morgans
- Pliant Therapeutics, South San Francisco, CA, USA
- Maze Therapeutics, South San Francisco, CA, USA
| | | | - Patrick Andre
- Pliant Therapeutics, South San Francisco, CA, USA
- Acceleron Pharma, Cambridge, MA, USA
| | | | | |
Collapse
|
32
|
De La Cruz Diaz JS, Hirai T, Anh-Thu Nguyen B, Zenke Y, Yang Y, Li H, Nishimura S, Kaplan DH. TNF-α and IL-1β Do Not Induce Langerhans Cell Migration by Inhibiting TGFβ Activation. JID INNOVATIONS 2021; 1:100028. [PMID: 34909727 PMCID: PMC8659779 DOI: 10.1016/j.xjidi.2021.100028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/26/2021] [Accepted: 05/03/2021] [Indexed: 11/24/2022] Open
Abstract
In the skin, Langerhans cells (LCs) require autocrine latent TGFβ that is transactivated by the integrins ανβ6 and ανβ8 expressed by keratinocytes (KCs) for long-term epidermal retention. Selective expression of a ligand-independent, constitutively active form of TGFβR1 inhibits LC migration during homeostasis and in response to UVB exposure. In this study, we found that LC migration in response to inflammatory stimuli was also inhibited by ligand-independent TGFβR1 signaling. Contrary to UVB stimulation, which reduced KC expression of ανβ6, in vitro and in vivo exposure to TNF-α or IL-1β increased ανβ6 transcript and protein expression by KCs. This resulted in increased KC-mediated transactivation of latent TGFβ. Expression of ανβ8 was largely unchanged. These findings show that ligand-independent TGFβR1 signaling in LCs can overcome inflammatory migration stimuli, but reduced KC-mediated transactivation of latent TGFβ by KCs may only drive LC migration during homeostasis and in response to UV stimulation.
Collapse
Key Words
- DMBA, 7,12-dimethylbenz[a]anthracene
- EpCAM, epithelial cell adhesion molecule
- IFE, interfollicular
- IM, infundibulum/isthmus
- KC, keratinocyte
- LAP, latency associated peptide
- LC, Langerhans cell
- LN, lymph node
- MHC, major histocompatibility complex
- pKC, primary keratinocyte
Collapse
Affiliation(s)
- Jacinto S. De La Cruz Diaz
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Toshiro Hirai
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Breanna Anh-Thu Nguyen
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yukari Zenke
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Dermatology, St. Luke’s International Hospital, Tokyo, Japan
| | - Yi Yang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiyue Li
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- School of Medicine, Tsinghua University, Beijing, China
| | - Stephen Nishimura
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Daniel H. Kaplan
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
33
|
Sun T, Huang Z, Liang WC, Yin J, Lin WY, Wu J, Vernes JM, Lutman J, Caplazi P, Jeet S, Wong T, Wong M, DePianto DJ, Morshead KB, Sun KH, Modrusan Z, Vander Heiden JA, Abbas AR, Zhang H, Xu M, N'Diaye EN, Roose-Girma M, Wolters PJ, Yadav R, Sukumaran S, Ghilardi N, Corpuz R, Emson C, Meng YG, Ramalingam TR, Lupardus P, Brightbill HD, Seshasayee D, Wu Y, Arron JR. TGFβ2 and TGFβ3 isoforms drive fibrotic disease pathogenesis. Sci Transl Med 2021; 13:13/605/eabe0407. [PMID: 34349032 DOI: 10.1126/scitranslmed.abe0407] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/19/2020] [Accepted: 06/06/2021] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-β (TGFβ) is a key driver of fibrogenesis. Three TGFβ isoforms (TGFβ1, TGFβ2, and TGFβ3) in mammals have distinct functions in embryonic development; however, the postnatal pathological roles and activation mechanisms of TGFβ2 and TGFβ3 have not been well characterized. Here, we show that the latent forms of TGFβ2 and TGFβ3 can be activated by integrin-independent mechanisms and have lower activation thresholds compared to TGFβ1. Unlike TGFB1, TGFB2 and TGFB3 expression is increased in human lung and liver fibrotic tissues compared to healthy control tissues. Thus, TGFβ2 and TGFβ3 may play a pathological role in fibrosis. Inducible conditional knockout mice and anti-TGFβ isoform-selective antibodies demonstrated that TGFβ2 and TGFβ3 are independently involved in mouse fibrosis models in vivo, and selective TGFβ2 and TGFβ3 inhibition does not lead to the increased inflammation observed with pan-TGFβ isoform inhibition. A cocrystal structure of a TGFβ2-anti-TGFβ2/3 antibody complex reveals an allosteric isoform-selective inhibitory mechanism. Therefore, inhibiting TGFβ2 and/or TGFβ3 while sparing TGFβ1 may alleviate fibrosis without toxicity concerns associated with pan-TGFβ blockade.
Collapse
Affiliation(s)
- Tianhe Sun
- Department of Immunology Discovery, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Zhiyu Huang
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wei-Ching Liang
- Department of Antibody Engineering, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jianping Yin
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wei Yu Lin
- Department of Antibody Engineering, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jia Wu
- Department of Antibody Engineering, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jean-Michel Vernes
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jeff Lutman
- Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Patrick Caplazi
- Department of Pathology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tiffany Wong
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Manda Wong
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Daryle J DePianto
- Department of Immunology Discovery, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Katrina B Morshead
- Department of Immunology Discovery, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kai-Hui Sun
- Department of Protein Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Department of Protein Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason A Vander Heiden
- Department of OMNI Bioinformatics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alexander R Abbas
- Department of OMNI Bioinformatics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hua Zhang
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Min Xu
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Elsa-Noah N'Diaye
- Department of Immunology Discovery, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Meron Roose-Girma
- Department of Molecular Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Rajbharan Yadav
- Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Siddharth Sukumaran
- Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Nico Ghilardi
- Department of Immunology Discovery, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Racquel Corpuz
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Claire Emson
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Y Gloria Meng
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Thirumalai R Ramalingam
- Department of Biomarker Discovery OMNI, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Patrick Lupardus
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hans D Brightbill
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dhaya Seshasayee
- Department of Antibody Engineering, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yan Wu
- Department of Antibody Engineering, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joseph R Arron
- Department of Immunology Discovery, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
34
|
Zehender A, Li YN, Lin NY, Stefanica A, Nüchel J, Chen CW, Hsu HH, Zhu H, Ding X, Huang J, Shen L, Györfi AH, Soare A, Rauber S, Bergmann C, Ramming A, Plomann M, Eckes B, Schett G, Distler JHW. TGFβ promotes fibrosis by MYST1-dependent epigenetic regulation of autophagy. Nat Commun 2021; 12:4404. [PMID: 34285225 PMCID: PMC8292318 DOI: 10.1038/s41467-021-24601-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
Activation of fibroblasts is essential for physiological tissue repair. Uncontrolled activation of fibroblasts, however, may lead to tissue fibrosis with organ dysfunction. Although several pathways capable of promoting fibroblast activation and tissue repair have been identified, their interplay in the context of chronic fibrotic diseases remains incompletely understood. Here, we provide evidence that transforming growth factor-β (TGFβ) activates autophagy by an epigenetic mechanism to amplify its profibrotic effects. TGFβ induces autophagy in fibrotic diseases by SMAD3-dependent downregulation of the H4K16 histone acetyltransferase MYST1, which regulates the expression of core components of the autophagy machinery such as ATG7 and BECLIN1. Activation of autophagy in fibroblasts promotes collagen release and is both, sufficient and required, to induce tissue fibrosis. Forced expression of MYST1 abrogates the stimulatory effects of TGFβ on autophagy and re-establishes the epigenetic control of autophagy in fibrotic conditions. Interference with the aberrant activation of autophagy inhibits TGFβ-induced fibroblast activation and ameliorates experimental dermal and pulmonary fibrosis. These findings link uncontrolled TGFβ signaling to aberrant autophagy and deregulated epigenetics in fibrotic diseases and may contribute to the development of therapeutic interventions in fibrotic diseases.
Collapse
Affiliation(s)
- Ariella Zehender
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Yi-Nan Li
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Neng-Yu Lin
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Adrian Stefanica
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Julian Nüchel
- Center for Biochemistry, University of Cologne, Faculty of Medicine, Cologne, Germany
| | - Chih-Wei Chen
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Hsiao-Han Hsu
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Honglin Zhu
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao Ding
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Jingang Huang
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Lichong Shen
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Andrea-Hermina Györfi
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Alina Soare
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Simon Rauber
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Andreas Ramming
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Markus Plomann
- Center for Biochemistry, University of Cologne, Faculty of Medicine, Cologne, Germany
| | - Beate Eckes
- Translational Matrix Biology, University of Cologne, Faculty of Medicine, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany.
- Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
35
|
Dodagatta-Marri E, Ma HY, Liang B, Li J, Meyer DS, Chen SY, Sun KH, Ren X, Zivak B, Rosenblum MD, Headley MB, Pinzas L, Reed NI, Del Cid JS, Hann BC, Yang S, Giddabasappa A, Noorbehesht K, Yang B, Dal Porto J, Tsukui T, Niessen K, Atakilit A, Akhurst RJ, Sheppard D. Integrin αvβ8 on T cells suppresses anti-tumor immunity in multiple models and is a promising target for tumor immunotherapy. Cell Rep 2021; 36:109309. [PMID: 34233193 PMCID: PMC8321414 DOI: 10.1016/j.celrep.2021.109309] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/17/2021] [Accepted: 06/04/2021] [Indexed: 01/18/2023] Open
Abstract
αvβ8 integrin, a key activator of transforming growth factor β (TGF-β), inhibits anti-tumor immunity. We show that a potent blocking monoclonal antibody against αvβ8 (ADWA-11) causes growth suppression or complete regression in syngeneic models of squamous cell carcinoma, mammary cancer, colon cancer, and prostate cancer, especially when combined with other immunomodulators or radiotherapy. αvβ8 is expressed at the highest levels in CD4+CD25+ T cells in tumors, and specific deletion of β8 from T cells is as effective as ADWA-11 in suppressing tumor growth. ADWA-11 increases expression of a suite of genes in tumor-infiltrating CD8+ T cells normally inhibited by TGF-β and involved in tumor cell killing, including granzyme B and interferon-γ. The in vitro cytotoxic effect of tumor CD8 T cells is inhibited by CD4+CD25+ cells, and this suppressive effect is blocked by ADWA-11. These findings solidify αvβ8 integrin as a promising target for cancer immunotherapy. TGF-β suppresses anti-tumor immunity. Dodagatta-Marri, Ma et al. show that the TGF-β-activating integrin αvβ8 is expressed on CD25+CD4+ tumor T cells and suppresses anti-tumor immunity by CD8+ T cells. Blocking this integrin enhances tumor cell killing and synergizes with multiple immune modulators or radiotherapy to induce long-term anti-tumor immunity.
Collapse
Affiliation(s)
- Eswari Dodagatta-Marri
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Hsiao-Yen Ma
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Benjia Liang
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China
| | - John Li
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Dominique S Meyer
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Szu-Ying Chen
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kai-Hui Sun
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Xin Ren
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Bahar Zivak
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Mark B Headley
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Pinzas
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Nilgun I Reed
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Joselyn S Del Cid
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Byron C Hann
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Sharon Yang
- Comparative Medicine, Pfizer Inc., San Diego, CA, USA
| | | | | | - Bing Yang
- Oncology Research Unit, Pfizer Inc., Pearl River, NY, USA
| | - Joseph Dal Porto
- Pfizer Centers for Therapeutic Innovation, San Francisco, CA, USA
| | - Tatsuya Tsukui
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kyle Niessen
- Pfizer Centers for Therapeutic Innovation, San Francisco, CA, USA
| | - Amha Atakilit
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Rosemary J Akhurst
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
| | - Dean Sheppard
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
36
|
Identification of chlorophyll a-b binding protein AB96 as a novel TGFβ1 neutralizing agent. Sci Rep 2021; 11:7740. [PMID: 33833368 PMCID: PMC8032758 DOI: 10.1038/s41598-021-87454-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 03/15/2021] [Indexed: 11/12/2022] Open
Abstract
The discovery of compounds and proteins from plants has greatly contributed to modern medicine. Vernonia amygdalina Del. (Compositae) is used by humans and primates for a variety of conditions including parasitic infection. This paper describes the serendipitous discovery that V. amygdalina extract was able to bind to, and functionally inhibit, active TGFβ1. The binding agent was isolated and identified as chlorophyll a-b binding protein AB96. Given that active TGFβ1 contributes to the pathology of many infectious diseases, inhibiting these processes may explain some of the benefits associated with the ingestion of this species. This is the first plant-derived cytokine-neutralizing protein to be described and paves the way for further such discoveries.
Collapse
|
37
|
Seed RI, Kobayashi K, Ito S, Takasaka N, Cormier A, Jespersen JM, Publicover J, Trilok S, Combes AJ, Chew NW, Chapman J, Krummel MF, Lou J, Marks J, Cheng Y, Baron JL, Nishimura SL. A tumor-specific mechanism of T reg enrichment mediated by the integrin αvβ8. Sci Immunol 2021; 6:6/57/eabf0558. [PMID: 33771888 DOI: 10.1126/sciimmunol.abf0558] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/06/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022]
Abstract
Regulatory T cells (Tregs) that promote tumor immune evasion are enriched in certain tumors and correlate with poor prognosis. However, mechanisms for Treg enrichment remain incompletely understood. We described a mechanism for Treg enrichment in mouse and human tumors mediated by the αvβ8 integrin. Tumor cell αvβ8 bound to latent transforming growth factor-β (L-TGF-β) presented on the surface of T cells, resulting in TGF-β activation and immunosuppressive Treg differentiation in vitro. In vivo, tumor cell αvβ8 expression correlated with Treg enrichment, immunosuppressive Treg gene expression, and increased tumor growth, which was reduced in mice by αvβ8 inhibition or Treg depletion. Structural modeling and cell-based studies suggested a highly geometrically constrained complex forming between αvβ8-expressing tumor cells and L-TGF-β-expressing T cells, facilitating TGF-β activation, independent of release and diffusion, and providing limited access to TGF-β inhibitors. These findings suggest a highly localized tumor-specific mechanism for Treg enrichment.
Collapse
Affiliation(s)
- Robert I Seed
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Kenji Kobayashi
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Saburo Ito
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Naoki Takasaka
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Anthony Cormier
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Jillian M Jespersen
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jean Publicover
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Suprita Trilok
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nayvin W Chew
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jocelyne Chapman
- Department of Gynecology and Oncology, University of California, San Francisco San Francisco, CA 94110, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - James Marks
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jody L Baron
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA. .,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
38
|
Brunner G, Roux M, Böhm V, Meiners T. Cellular and molecular changes that predispose skin in chronic spinal cord injury to pressure ulcer formation. Int Wound J 2021; 18:728-737. [PMID: 33723924 PMCID: PMC8450792 DOI: 10.1111/iwj.13575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/04/2022] Open
Abstract
Patients with spinal cord injury have a predisposition to develop pressure ulcers. Specific characteristics of the patients' skin potentially involved have not yet been identified. The purpose of this investigation was to determine whether loss of neuronal control affects cellular and molecular homeostasis in the skin. Intact afflicted skin, wound edge of pressure ulcers, and control skin were analysed. Platelets, transforming growth factor‐β1, and activin A were identified by immunohistochemistry. Transforming growth factor‐β‐like activity was determined by bioassay, and gene expression by DNA microarray analysis or RT‐PCR. In afflicted skin, enhanced platelet extravasation was detected. Transforming growth factor‐β1 and activin A accumulated in the dermal‐epidermal junction zone. Transforming growth factor‐β‐like activity and activin A expression were increased in intact afflicted skin (compared to control skin) and were further enhanced in pressure ulcers. In vitro, activity was generated by fibroblast‐epithelial cell interactions, which also induced activin A. Thus, loss of neuronal control in spinal cord injury appears to trigger inappropriate wound healing processes in the patients' skin. Plasma leakage and increased transforming growth factor‐β‐like activity combined with shear forces potentially enhance the risk for pressure ulcer formation.
Collapse
Affiliation(s)
- Georg Brunner
- Center for Spinal Cord Injuries, Werner Wicker Hospital, Bad Wildungen, Germany.,Department of Cancer Research, Skin Cancer Center Hornheide, Münster, Germany
| | - Meike Roux
- Department of Cancer Research, Skin Cancer Center Hornheide, Münster, Germany
| | - Volker Böhm
- Center for Spinal Cord Injuries, Werner Wicker Hospital, Bad Wildungen, Germany
| | - Thomas Meiners
- Center for Spinal Cord Injuries, Werner Wicker Hospital, Bad Wildungen, Germany
| |
Collapse
|
39
|
Modulation of α Vβ 6 integrin in osteoarthritis-related synovitis and the interaction with VTN (381-397 a.a.) competing for TGF-β1 activation. Exp Mol Med 2021; 53:210-222. [PMID: 33526813 PMCID: PMC8080589 DOI: 10.1038/s12276-021-00558-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis is characterized by structural alteration of joints. Fibrosis of the synovial tissue is often detected and considered one of the main causes of joint stiffness and pain. In our earlier proteomic study, increased levels of vitronectin (VTN) fragment (amino acids 381–397) were observed in the serum of osteoarthritis patients. In this work, the affinity of this fragment for integrins and its putative role in TGF-β1 activation were investigated. A competition study determined the interaction of VTN(381–397 a.a.) with αVβ6 integrin. Subsequently, the presence of αVβ6 integrin was substantiated on primary human fibroblast-like synoviocytes (FLSs) by western blot and flow cytometry. By immunohistochemistry, β6 was detected in synovial membranes, and its expression showed a correlation with tissue fibrosis. Moreover, β6 expression was increased under TGF-β1 stimulation; hence, a TGF-β bioassay was applied. We observed that αVβ6 could mediate TGF-β1 bioavailability and that VTN(381–397 a.a.) could prevent TGF-β1 activation by interacting with αVβ6 in human FLSs and increased α-SMA. Finally, we analyzed serum samples from healthy controls and patients with osteoarthritis and other rheumatic diseases by nano-LC/Chip MS–MS, confirming the increased expression of VTN(381–397 a.a.) in osteoarthritis as well as in lupus erythematosus and systemic sclerosis. These findings corroborate our previous observations concerning the overexpression of VTN(381–397 a.a.) in osteoarthritis but also in other rheumatic diseases. This fragment interacts with αVβ6 integrin, a receptor whose expression is increased in FLSs from the osteoarthritic synovial membrane and that can mediate the activation of the TGF-β1 precursor in human FLSs. Insights into a mechanism underlying the formation of fibrotic tissue within joints in osteoarthritis may also prove relevant to other rheumatological disorders. The general mechanisms underlying fibrosis are reasonably well understood, but it remains unclear what triggers these processes in osteoarthitis. Researchers of the University of Liège in Belgium have uncovered a possible explanation based on experiments with cultured primary synovial fibroblasts from patients. Osteoarthitis is characterized by increased levels of a fragment of the protein vitronectin, and the researchers demonstrated that this in turn binds to a protein called αVβ6, potentially promoting initiation of fibrosis. They also observed elevated levels of the same vitronectin fragment in two other rheumatoid disorders, lupus and systemic sclerosis, and concluded that further research is needed to characterize this protein’s role in inflammation and fibrosis.
Collapse
|
40
|
Roper JA, Wilkinson AL, Gower E, Slack RJ. Downregulation of the αv β6 Integrin via RGD Engagement Is Affinity and Time Dependent. J Pharmacol Exp Ther 2021; 376:273-280. [PMID: 33318076 DOI: 10.1124/jpet.120.000379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
The arginyl-glycinyl-aspartic acid (RGD) integrin alpha-v beta-6 (αvβ6) has been identified as playing a key role in the activation of transforming growth factor-β (TGFβ) that is hypothesized to be pivotal in the development of fibrosis and other diseases. In this study, αvβ6 small molecule inhibitors were characterized in a range of in vitro systems to determine affinity, kinetics, and duration of TGFβ inhibition. High αvβ6 binding affinity was shown to be correlated with slow dissociation kinetics. Compound 1 (high αvβ6 affinity, slow dissociation) and SC-68448 (low αvβ6 affinity, fast dissociation) induced concentration- and time-dependent internalization of αvβ6 in normal human bronchial epithelial (NHBE) cells. After washout, the αvβ6 cell surface repopulation was faster for SC-68448 compared with compound 1 In addition, αvβ6-dependent release of active TGFβ from NHBE cells was inhibited by compound 1 and SC-68448. After washout of SC-68448, release of active TGFβ was restored, whereas after washout of compound 1 the inhibition of TGFβ activation was maintained and only reversible in the presence of a lysosomal inhibitor (chloroquine). However, SC-68448 was able to reduce total levels of αvβ6 in NHBE cells if present continuously. These observations suggest αvβ6 can be degraded after high affinity RGD binding that sorts the integrin for lysosomal degradation after internalization, likely due to sustained engagement as a result of slow dissociation kinetics. In addition, the αvβ6 integrin can also be downregulated after sustained engagement of the RGD binding site with low affinity ligands that do not sort the integrin for immediate lysosomal degradation. SIGNIFICANCE STATEMENT: The fate of RGD integrin after ligand binding has not been widely investigated. Using the αvβ6 integrin as a case study, we have demonstrated that RGD-induced downregulation of αvβ6 is both affinity and time dependent. High affinity ligands induced downregulation via lysosomal degradation, likely due to slow dissociation, whereas sustained low affinity ligand engagement was only able to decrease αvβ6 expression over longer periods of time. Our study provides a potential unique mechanism for obtaining duration of action for drugs targeting integrins.
Collapse
Affiliation(s)
- James A Roper
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Alex L Wilkinson
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Elaine Gower
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | - Robert J Slack
- Fibrosis Discovery Performance Unit (DPU), Respiratory Therapy Area Unit (TAU), GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| |
Collapse
|
41
|
Fell S, Wang Z, Blanchard A, Nanthakumar C, Griffin M. Transglutaminase 2: a novel therapeutic target for idiopathic pulmonary fibrosis using selective small molecule inhibitors. Amino Acids 2021; 53:205-217. [PMID: 33474654 PMCID: PMC7910249 DOI: 10.1007/s00726-020-02938-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/26/2020] [Indexed: 01/29/2023]
Abstract
This study investigates the effects of a site-directed TG2-selective inhibitor on the lung myofibroblast phenotype and ECM deposition to elucidate TG2 as a novel therapeutic target in idiopathic pulmonary fibrosis (IPF)-an incurable progressive fibrotic disease. IPF fibroblasts showed increased expression of TG2, α smooth muscle actin (αSMA) and fibronectin (FN) with increased extracellular TG2 and transforming growth factor β1 (TGFβ1) compared to normal human lung fibroblasts (NHLFs) which do not express αSMA and express lower levels of FN. The myofibroblast phenotype shown by IPF fibroblasts could be reversed by selective TG2 inhibition with a reduction in matrix FN and TGFβ1 deposition. TG2 transduction or TGFβ1 treatment of NHLFs led to a comparable phenotype to that of IPF fibroblasts which was reversible following selective TG2 inhibition. Addition of exogenous TG2 to NHLFs also induced the myofibroblast phenotype by a mechanism involving TGFβ1 activation which could be ameliorated by selective TG2 inhibition. SMAD3-deleted IPF fibroblasts via CRISPR-cas9 genome editing, showed reduced TG2 protein levels following TGFβ1 stimulation. This study demonstrates a key role for TG2 in the induction of the myofibroblast phenotype and shows the potential for TG2-selective inhibitors as therapeutic agents for the treatment of fibrotic lung diseases like IPF.
Collapse
Affiliation(s)
- Shaun Fell
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Zhuo Wang
- School of Life and Health Sciences, Aston University, Birmingham, UK.
| | - Andy Blanchard
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R and D, Stevenage, UK
| | - Carmel Nanthakumar
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R and D, Stevenage, UK
| | - Martin Griffin
- School of Life and Health Sciences, Aston University, Birmingham, UK.
| |
Collapse
|
42
|
Abstract
Myofibroblasts are critical to processes involved in normal wound healing and during pathological fibrosis. They transdifferentiate from fibroblasts, and in doing so become contractile and capable of secreting large amounts of extracellular matrix proteins. Transforming growth factor-beta (TGFβ) is a key cytokine involved in wound healing and fibrogenesis. TGFβ signaling has long been the subject of experimental therapeutic approaches to inhibit fibrosis in a variety of organ systems. Inhibition of TGFβ can reduce myofibroblast transdifferentiation, contractility, and matrix production. Importantly, TGFβ is released from cells and sequestered in the extracellular matrix in a latent form that requires activation for biological function. There have been multiple mechanisms of TGFβ activation described in a variety of cell types and in cell free systems; however, myofibroblasts have previously been shown to activate TGFβ via cell surface integrins, particularly αvβ5 integrins. This chapter will provide detailed protocols for accurately measuring activation of TGFβ by myofibroblasts in vitro. Levels of active TGFβ usually represent a small proportion of the total amount of latent TGFβ present in the matrix. Methods to measure active TGFβ therefore need to be sensitive and specific to detect the active cytokine only.
Collapse
Affiliation(s)
- Joanne Porte
- Division of Respiratory Medicine, NIHR Nottingham Respiratory Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Gisli Jenkins
- Division of Respiratory Medicine, NIHR Nottingham Respiratory Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Amanda L Tatler
- Division of Respiratory Medicine, NIHR Nottingham Respiratory Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
43
|
Taki T, Shiraki Y, Enomoto A, Weng L, Chen C, Asai N, Murakumo Y, Yokoi K, Takahashi M, Mii S. CD109 regulates in vivo tumor invasion in lung adenocarcinoma through TGF-β signaling. Cancer Sci 2020; 111:4616-4628. [PMID: 33007133 PMCID: PMC7734007 DOI: 10.1111/cas.14673] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 09/09/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022] Open
Abstract
Stromal invasion is considered an important prognostic factor in patients with lung adenocarcinoma. The mechanisms underlying the formation of tumor stroma and stromal invasion have been studied in the lung; however, they are still unclear. CD109 is a glycosylphosphatidylinositol-anchored glycoprotein highly expressed in several types of human malignant tumors including lung cancers. In this study, we investigated the in vivo functions of CD109 protein in malignant lung tumors. Initially, we identified an association between higher expression of CD109 protein in human lung adenocarcinoma and a significantly worse prognosis, according to immunohistochemical analysis. We also showed that CD109 deficiency significantly reduced the area of stromal invasive lesions in a genetically engineered CD109-deficient lung adenocarcinoma mouse model, which correlated with the results observed in human lung adenocarcinoma. Furthermore, we identified latent TGF-β binding protein-1 (LTBP1) as a CD109-interacting protein using mass spectrometry and confirmed their interaction by co-immunoprecipitation. Importantly, increased CD109 expression enhanced stromal TGF-β activation in the presence of LTBP1. Therefore, these data suggest the significance of the regulation of TGF-β signaling through CD109 and LTBP1 interaction in tumor stroma and also reveal the importance of CD109 expression levels in promoting lung cancer cell proliferation, migration, and invasion, and thus predicting the outcome of patients suffering from lung adenocarcinoma. Therefore, CD109 protein could be a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Tetsuro Taki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yukihiro Shiraki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Molecular Pathology, Center for Neurological Disease and CancerNagoya University Graduate School of MedicineNagoyaJapan
| | - Atsushi Enomoto
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Liang Weng
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Chen Chen
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Naoya Asai
- Department of Molecular Pathology, Graduate School of MedicineFujita Health UniversityToyoakeJapan
| | - Yoshiki Murakumo
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Kohei Yokoi
- Department of Thoracic SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | - Masahide Takahashi
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Molecular Pathology, Center for Neurological Disease and CancerNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinji Mii
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Molecular Pathology, Center for Neurological Disease and CancerNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
44
|
Full-length IL-33 regulates Smad3 phosphorylation and gene transcription in a distinctive AP2-dependent manner. Cell Immunol 2020; 357:104203. [DOI: 10.1016/j.cellimm.2020.104203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/30/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
|
45
|
Alonso-Herranz L, Sahún-Español Á, Paredes A, Gonzalo P, Gkontra P, Núñez V, Clemente C, Cedenilla M, Villalba-Orero M, Inserte J, García-Dorado D, Arroyo AG, Ricote M. Macrophages promote endothelial-to-mesenchymal transition via MT1-MMP/TGFβ1 after myocardial infarction. eLife 2020; 9:57920. [PMID: 33063665 PMCID: PMC7609061 DOI: 10.7554/elife.57920] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022] Open
Abstract
Macrophages (Mφs) produce factors that participate in cardiac repair and remodeling after myocardial infarction (MI); however, how these factors crosstalk with other cell types mediating repair is not fully understood. Here we demonstrated that cardiac Mφs increased the expression of Mmp14 (MT1-MMP) 7 days post-MI. We selectively inactivated the Mmp14 gene in Mφs using a genetic strategy (Mmp14f/f:Lyz2-Cre). This conditional KO (MAC-Mmp14 KO) resulted in attenuated post-MI cardiac dysfunction, reduced fibrosis, and preserved cardiac capillary network. Mechanistically, we showed that MT1-MMP activates latent TGFβ1 in Mφs, leading to paracrine SMAD2-mediated signaling in endothelial cells (ECs) and endothelial-to-mesenchymal transition (EndMT). Post-MI MAC-Mmp14 KO hearts contained fewer cells undergoing EndMT than their wild-type counterparts, and Mmp14-deficient Mφs showed a reduced ability to induce EndMT in co-cultures with ECs. Our results indicate the contribution of EndMT to cardiac fibrosis and adverse remodeling post-MI and identify Mφ MT1-MMP as a key regulator of this process.
Collapse
Affiliation(s)
- Laura Alonso-Herranz
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Álvaro Sahún-Español
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Paredes
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Pilar Gonzalo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Polyxeni Gkontra
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Vanessa Núñez
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina Clemente
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Marta Cedenilla
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María Villalba-Orero
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Vall d'Hebron University Hospital and Research Institute (VHIR), Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - David García-Dorado
- Cardiovascular Diseases Research Group, Vall d'Hebron University Hospital and Research Institute (VHIR), Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Alicia G Arroyo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Mercedes Ricote
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
46
|
Zhou Y, Ng DYE, Richards AM, Wang P. microRNA-221 Inhibits Latent TGF-β1 Activation through Targeting Thrombospondin-1 to Attenuate Kidney Failure-Induced Cardiac Fibrosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:803-814. [PMID: 33230477 PMCID: PMC7645417 DOI: 10.1016/j.omtn.2020.09.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 09/30/2020] [Indexed: 12/21/2022]
Abstract
Kidney failure (KF) is associated with cardiac fibrosis and significantly increased mortality in heart failure. Thrombospondin-1 (TSP1), a key regulator of latent transforming growth factor-β1 (L-TGF-β1) activation, is a predicted target of miR-221. We hypothesized miR-221 attenuates severe KF-associated cardiac fibrosis via targeting of Thbs1 with subsequent inhibition of L-TGF-β1 activation. Rat cardiac fibroblasts (cFB) were isolated and transfected with microRNA-221 (miR-221) mimics or mimic control (miR-221 and MC) with or without exposure to L-TGF-β1. We demonstrate miR-221 downregulates Thbs1 via direct 3′ untranslated region (3′ UTR) targeting with consequent inhibition of L-TGF-β1 activation in cFB as proven by the significant reduction of myofibroblast activation, collagen secretion, TGF-β1 signaling, TSP1 secretion, and TGF-β1 bioactivity measured by Pai1 promoter reporter. The 5/6 nephrectomy (Nx) model of cardiac fibrosis was used to test the in vivo therapeutic efficacy of miR-221 (i.v. 1 mg/kg ×3). miR-221 significantly inhibited Nx-induced upregulation of TSP1 and p-SMAD3 in the heart at day-7 and reduced cardiac fibrosis (picro-sirius), improved cardiac function (±dP/dt), and improved 8-week survival rate (60% versus 36%; p = 0.038). miR-221 mimic treatment improved survival and reduced cardiac fibrosis in a model of severe KF. miR-221 is a therapeutic target to address cardiac fibrosis originating from renal disease and other causes.
Collapse
Affiliation(s)
- Yue Zhou
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599 Singapore.,Department of Medicine, National University Health System, Singapore 119228, Singapore
| | - Denise Yu En Ng
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599 Singapore.,Department of Medicine, National University Health System, Singapore 119228, Singapore
| | - Arthur Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599 Singapore.,Department of Medicine, National University Health System, Singapore 119228, Singapore.,Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Peipei Wang
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599 Singapore.,Department of Medicine, National University Health System, Singapore 119228, Singapore
| |
Collapse
|
47
|
Kumar R, Mickael C, Kassa B, Sanders L, Hernandez-Saavedra D, Koyanagi DE, Kumar S, Pugliese SC, Thomas S, McClendon J, Maloney JP, Janssen WJ, Stenmark KR, Tuder RM, Graham BB. Interstitial macrophage-derived thrombospondin-1 contributes to hypoxia-induced pulmonary hypertension. Cardiovasc Res 2020; 116:2021-2030. [PMID: 31710666 PMCID: PMC7519884 DOI: 10.1093/cvr/cvz304] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/06/2019] [Accepted: 11/08/2019] [Indexed: 01/05/2023] Open
Abstract
AIMS Transforming growth factor-β (TGF-β) signalling is required for chronic hypoxia-induced pulmonary hypertension (PH). The activation of TGF-β by thrombospondin-1 (TSP-1) contributes to the pathogenesis of hypoxia-induced PH. However, neither the cellular source of pathologic TSP-1 nor the downstream signalling pathway that link activated TGF-β to PH have been determined. In this study, we hypothesized that circulating monocytes, which are recruited to become interstitial macrophages (IMs), are the major source of TSP-1 in hypoxia-exposed mice, and TSP-1 activates TGF-β with increased Rho-kinase signalling, causing vasoconstriction. METHODS AND RESULTS Flow cytometry revealed that a specific subset of IMs is the major source of pathologic TSP-1 in hypoxia. Intravenous depletion and parabiosis experiments demonstrated that these cells are circulating prior to recruitment into the interstitium. Rho-kinase-mediated vasoconstriction was a major downstream target of active TGF-β. Thbs1 deficient bone marrow (BM) protected against hypoxic-PH by blocking TGF-β activation and Rho-kinase-mediated vasoconstriction. CONCLUSION In hypoxia-challenged mice, BM derived and circulating monocytes are recruited to become IMs which express TSP-1, resulting in TGF-β activation and Rho-kinase-mediated vasoconstriction.
Collapse
Affiliation(s)
- Rahul Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, Building 100, 3rd floor, 1001 Potrero Ave, San Francisco, CA 94110, USA
| | - Claudia Mickael
- Department of Medicine, Program in Translational Lung Research, Anschutz Medical Campus, Building RC2, 9th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - Biruk Kassa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, Building 100, 3rd floor, 1001 Potrero Ave, San Francisco, CA 94110, USA
| | - Linda Sanders
- Department of Medicine, Program in Translational Lung Research, Anschutz Medical Campus, Building RC2, 9th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - Daniel Hernandez-Saavedra
- Department of Medicine, Program in Translational Lung Research, Anschutz Medical Campus, Building RC2, 9th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - Daniel E Koyanagi
- Department of Medicine, Program in Translational Lung Research, Anschutz Medical Campus, Building RC2, 9th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - Sushil Kumar
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratory, Anschutz Medical Campus, Building RC2, 8th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - Steve C Pugliese
- Department of Medicine, University of Pennsylvania, 831 Gates building, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Stacey Thomas
- Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Jazalle McClendon
- Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - James P Maloney
- Department of Medicine, Anschutz Medical Campus, Building RC2, 9th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - William J Janssen
- Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Kurt R Stenmark
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratory, Anschutz Medical Campus, Building RC2, 8th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - Rubin M Tuder
- Department of Medicine, Program in Translational Lung Research, Anschutz Medical Campus, Building RC2, 9th floor, 12700 E 19th Ave, Aurora, CO 80045, USA
| | - Brian B Graham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, Building 100, 3rd floor, 1001 Potrero Ave, San Francisco, CA 94110, USA
| |
Collapse
|
48
|
John AE, Graves RH, Pun KT, Vitulli G, Forty EJ, Mercer PF, Morrell JL, Barrett JW, Rogers RF, Hafeji M, Bibby LI, Gower E, Morrison VS, Man Y, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Le J, Flint DJ, Pyne S, Habgood A, Organ LA, Joseph C, Edwards-Pritchard RC, Maher TM, Fisher AJ, Gudmann NS, Leeming DJ, Chambers RC, Lukey PT, Marshall RP, Macdonald SJF, Jenkins RG, Slack RJ. Translational pharmacology of an inhaled small molecule αvβ6 integrin inhibitor for idiopathic pulmonary fibrosis. Nat Commun 2020; 11:4659. [PMID: 32938936 PMCID: PMC7494911 DOI: 10.1038/s41467-020-18397-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
The αvβ6 integrin plays a key role in the activation of transforming growth factor-β (TGFβ), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvβ6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvβ6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFβ signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvβ6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvβ6, induces prolonged inhibition of TGFβ signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy. The αvβ6 integrin is key in activating the pro-fibrotic cytokine TGFβ in idiopathic pulmonary fibrosis. Here, the authors show an inhaled small molecule αvβ6 inhibitor GSK3008348 induces prolonged inhibition of TGFβ signaling pathways in human and murine models of lung fibrosis via αvβ6 degradation.
Collapse
Affiliation(s)
- Alison E John
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Rebecca H Graves
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - K Tao Pun
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Giovanni Vitulli
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Ellen J Forty
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Paul F Mercer
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Josie L Morrell
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - John W Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Rebecca F Rogers
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Maryam Hafeji
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Lloyd I Bibby
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Elaine Gower
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Valerie S Morrison
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Yim Man
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - James A Roper
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Jeni C Luckett
- Radiological Sciences, University of Nottingham, Nottingham, UK
| | - Lee A Borthwick
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Ben S Barksby
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rachel A Burgoyne
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rory Barnes
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Joelle Le
- Drug Design and Selection - Molecular Design, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - David J Flint
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Anthony Habgood
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Louise A Organ
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Chitra Joseph
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | | | - Toby M Maher
- NIHR Respiratory Clinical Research Facility, Royal Brompton Hospital, London, UK.,Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, UK
| | - Andrew J Fisher
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK.,Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS, Foundation Trust, Newcastle upon Tyne, UK
| | - Natasja Stæhr Gudmann
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Diana J Leeming
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Pauline T Lukey
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Richard P Marshall
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Simon J F Macdonald
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - R Gisli Jenkins
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| | - Robert J Slack
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
49
|
Insight into Molecular Mechanism for Activin A-Induced Bone Morphogenetic Protein Signaling. Int J Mol Sci 2020; 21:ijms21186498. [PMID: 32899497 PMCID: PMC7555472 DOI: 10.3390/ijms21186498] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/30/2020] [Accepted: 09/03/2020] [Indexed: 01/14/2023] Open
Abstract
Activins transduce the TGF-β pathway through a heteromeric signaling complex consisting of type I and type II receptors, and activins also inhibit bone morphogenetic protein (BMP) signaling mediated by type I receptor ALK2. Recent studies indicated that activin A cross-activates the BMP pathway through ALK2R206H, a mutation associated with Fibrodysplasia Ossificans Progressiva (FOP). How activin A inhibits ALK2WT-mediated BMP signaling but activates ALK2R206H-mediated BMP signaling is not well understood, and here we offer some insights into its molecular mechanism. We first demonstrated that among four BMP type I receptors, ALK2 is the only subtype able to mediate the activin A-induced BMP signaling upon the dissociation of FKBP12. We further showed that BMP4 does not cross-signal TGF-β pathway upon FKBP12 inhibition. In addition, although the roles of type II receptors in the ligand-independent BMP signaling activated by FOP-associated mutant ALK2 have been reported, their roles in activin A-induced BMP signaling remains unclear. We demonstrated in this study that the known type II BMP receptors contribute to activin A-induced BMP signaling through their kinase activity. Together, the current study provided important mechanistic insights at the molecular level into further understanding physiological and pathophysiological BMP signaling.
Collapse
|
50
|
Nyazika TK, Law A, Swarthout TD, Sibale L, Ter Braake D, French N, Heyderman RS, Everett D, Kadioglu A, Jambo KC, Neill DR. Influenza-like illness is associated with high pneumococcal carriage density in Malawian children. J Infect 2020; 81:549-556. [PMID: 32711042 PMCID: PMC7375306 DOI: 10.1016/j.jinf.2020.06.079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/25/2020] [Accepted: 06/27/2020] [Indexed: 01/29/2023]
Abstract
Influenza-like illness (ILI) in children is associated with high pneumococcal carriage density. Children with ILI harboured more viral organisms than asymptomatic healthy children. Children with ILI patients had higher IL-8 levels in nasal aspirates than asymptomatic healthy children.
Background High pneumococcal carriage density is a risk factor for invasive pneumococcal disease (IPD) and transmission, but factors that increase pneumococcal carriage density are still unclear. Methods We undertook a cross-sectional study to evaluate the microbial composition, cytokine levels and pneumococcal carriage densities in samples from children presenting with an influenza-like illness (ILI) and asymptomatic healthy controls (HC). Results The proportion of children harbouring viral organisms (Relative risk (RR) 1.4, p = 0.0222) or ≥ 4 microbes at a time (RR 1.9, p < 0.0001), was higher in ILI patients than HC. ILI patients had higher IL-8 levels in nasal aspirates than HC (median [IQR], 265.7 [0 – 452.3] vs. 0 [0 – 127.3] pg/ml; p = 0.0154). Having an ILI was associated with higher pneumococcal carriage densities compared to HC (RR 4.2, p < 0.0001). Conclusion These findings suggest that children with an ILI have an increased propensity for high pneumococcal carriage density. This could in part contribute to increased susceptibility to IPD and transmission in the community.
Collapse
Affiliation(s)
- Tinashe K Nyazika
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Department of Pathology, College of Health Sciences, University of Malawi, Blantyre, Malawi.
| | - Alice Law
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Todd D Swarthout
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi; Division of Infection and Immunity, NIHR Global Health Research Unit on Mucosal Pathogens, University College London, London, United Kingdom
| | - Lusako Sibale
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Danielle Ter Braake
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom; Department of Biomolecular Health Sciences, Division of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht, the Netherlands
| | - Neil French
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Robert S Heyderman
- Division of Infection and Immunity, NIHR Global Health Research Unit on Mucosal Pathogens, University College London, London, United Kingdom
| | - Dean Everett
- The Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Aras Kadioglu
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Kondwani C Jambo
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.
| | - Daniel R Neill
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|