1
|
Zhang M, Wu X, Zhu H, Fu C, Yang W, Jing X, Liu W, Cheng Y. Construction and Bioinformatics Analysis of ceRNA Regulatory Networks in Idiopathic Pulmonary Fibrosis. Biochem Genet 2024:10.1007/s10528-024-10853-y. [PMID: 38871957 DOI: 10.1007/s10528-024-10853-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive form of pulmonary fibrosis of unknown etiology. Despite ongoing research, there is currently no cure for this disease. Recent studies have highlighted the significance of competitive endogenous RNA (ceRNA) regulatory networks in IPF development. Therefore, this study investigated the ceRNA network associated with IPF pathogenesis. We obtained gene expression datasets (GSE32538, GSE32537, GSE47460, and GSE24206) from the Gene Expression Omnibus (GEO) database and analyzed them using bioinformatics tools to identify differentially expressed messenger RNAs (DEmRNAs), microRNAs (DEmiRNAs), and long non-coding RNAs (DElncRNA). For DEmRNAs, we conducted an enrichment analysis, constructed protein-protein interaction networks, and identified hub genes. Additionally, we predicted the target genes of differentially expressed mRNAs and their interacting long non-coding RNAs using various databases. Subsequently, we screened RNA molecules with ceRNA regulatory relations in the lncACTdb database based on the screening results. Furthermore, we performed disease and functional enrichment analyses and pathway prediction for miRNAs in the ceRNA network. We also validated the expression levels of candidate DEmRNAs through quantitative real-time reverse transcriptase polymerase chain reaction and analyzed the correlation between the expression of these candidate DEmRNAs and the percent predicted pre-bronchodilator forced vital capacity [%predicted FVC (pre-bd)]. We found that three ceRNA regulatory axes, specifically KCNQ1OT1/XIST/NEAT1-miR-20a-5p-ITGB8, XIST-miR-146b-5p/miR-31-5p- MMP16, and NEAT1-miR-31-5p-MMP16, have the potential to significantly affect IPF progression. Further examination of the underlying regulatory mechanisms within this network enhances our understanding of IPF pathogenesis and may aid in the identification of diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Menglin Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
- Department of Respiratory and Critical Care Medicine, People's Hospital of Anshun City Guizhou Province, Anshun, 561000, China
| | - Xiao Wu
- Department of Critical Care Medicine, The Second People's Hospital of Guiyang, Guiyang, 550004, China
| | - Honglan Zhu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Chenkun Fu
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Wenting Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Xiaoting Jing
- Department of Respiratory and Critical Care Medicine, Guiyang Public Health Clinical Center, Guiyang, 550002, China.
| | - Wenqu Liu
- Department of Respiratory and Critical Care Medicine, People's Hospital of Anshun City Guizhou Province, Anshun, 561000, China.
| | - Yiju Cheng
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
- Department of Respiratory and Critical Care Medicine, The Fourth People's Hospital of Guiyang, Guiyang, 550002, China.
| |
Collapse
|
2
|
Ahuja S, Zaheer S. Multifaceted TGF-β signaling, a master regulator: From bench-to-bedside, intricacies, and complexities. Cell Biol Int 2024; 48:87-127. [PMID: 37859532 DOI: 10.1002/cbin.12097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Physiological embryogenesis and adult tissue homeostasis are regulated by transforming growth factor-β (TGF-β), an evolutionarily conserved family of secreted polypeptide factors, acting in an autocrine and paracrine manner. The role of TGF-β in inflammation, fibrosis, and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, especially fibrosis and cancer, overexpressed TGF-β causes extracellular matrix deposition, epithelial-mesenchymal transition, cancer-associated fibroblast formation, and/or angiogenesis. In this review article, we have tried to dive deep into the mechanism of action of TGF-β in inflammation, fibrosis, and carcinogenesis. As TGF-β and its downstream signaling mechanism are implicated in fibrosis and carcinogenesis blocking this signaling mechanism appears to be a promising avenue. However, targeting TGF-β carries substantial risk as this pathway is implicated in multiple homeostatic processes and is also known to have tumor-suppressor functions. There is a need for careful dosing of TGF-β drugs for therapeutic use and patient selection.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
3
|
Li S, Sampson C, Liu C, Piao HL, Liu HX. Integrin signaling in cancer: bidirectional mechanisms and therapeutic opportunities. Cell Commun Signal 2023; 21:266. [PMID: 37770930 PMCID: PMC10537162 DOI: 10.1186/s12964-023-01264-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
Integrins are transmembrane receptors that possess distinct ligand-binding specificities in the extracellular domain and signaling properties in the cytoplasmic domain. While most integrins have a short cytoplasmic tail, integrin β4 has a long cytoplasmic tail that can indirectly interact with the actin cytoskeleton. Additionally, 'inside-out' signals can induce integrins to adopt a high-affinity extended conformation for their appropriate ligands. These properties enable integrins to transmit bidirectional cellular signals, making it a critical regulator of various biological processes.Integrin expression and function are tightly linked to various aspects of tumor progression, including initiation, angiogenesis, cell motility, invasion, and metastasis. Certain integrins have been shown to drive tumorigenesis or amplify oncogenic signals by interacting with corresponding receptors, while others have marginal or even suppressive effects. Additionally, different α/β subtypes of integrins can exhibit opposite effects. Integrin-mediated signaling pathways including Ras- and Rho-GTPase, TGFβ, Hippo, Wnt, Notch, and sonic hedgehog (Shh) are involved in various stages of tumorigenesis. Therefore, understanding the complex regulatory mechanisms and molecular specificities of integrins are crucial to delaying cancer progression and suppressing tumorigenesis. Furthermore, the development of integrin-based therapeutics for cancer are of great importance.This review provides an overview of integrin-dependent bidirectional signaling mechanisms in cancer that can either support or oppose tumorigenesis by interacting with various signaling pathways. Finally, we focus on the future opportunities for emergent therapeutics based on integrin agonists. Video Abstract.
Collapse
Affiliation(s)
- Siyi Li
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Chibuzo Sampson
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Changhao Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Hai-Long Piao
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
4
|
Pang X, He X, Qiu Z, Zhang H, Xie R, Liu Z, Gu Y, Zhao N, Xiang Q, Cui Y. Targeting integrin pathways: mechanisms and advances in therapy. Signal Transduct Target Ther 2023; 8:1. [PMID: 36588107 PMCID: PMC9805914 DOI: 10.1038/s41392-022-01259-6] [Citation(s) in RCA: 436] [Impact Index Per Article: 218.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 01/03/2023] Open
Abstract
Integrins are considered the main cell-adhesion transmembrane receptors that play multifaceted roles as extracellular matrix (ECM)-cytoskeletal linkers and transducers in biochemical and mechanical signals between cells and their environment in a wide range of states in health and diseases. Integrin functions are dependable on a delicate balance between active and inactive status via multiple mechanisms, including protein-protein interactions, conformational changes, and trafficking. Due to their exposure on the cell surface and sensitivity to the molecular blockade, integrins have been investigated as pharmacological targets for nearly 40 years, but given the complexity of integrins and sometimes opposite characteristics, targeting integrin therapeutics has been a challenge. To date, only seven drugs targeting integrins have been successfully marketed, including abciximab, eptifibatide, tirofiban, natalizumab, vedolizumab, lifitegrast, and carotegrast. Currently, there are approximately 90 kinds of integrin-based therapeutic drugs or imaging agents in clinical studies, including small molecules, antibodies, synthetic mimic peptides, antibody-drug conjugates (ADCs), chimeric antigen receptor (CAR) T-cell therapy, imaging agents, etc. A serious lesson from past integrin drug discovery and research efforts is that successes rely on both a deep understanding of integrin-regulatory mechanisms and unmet clinical needs. Herein, we provide a systematic and complete review of all integrin family members and integrin-mediated downstream signal transduction to highlight ongoing efforts to develop new therapies/diagnoses from bench to clinic. In addition, we further discuss the trend of drug development, how to improve the success rate of clinical trials targeting integrin therapies, and the key points for clinical research, basic research, and translational research.
Collapse
Affiliation(s)
- Xiaocong Pang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Xu He
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiwei Qiu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Hanxu Zhang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Ran Xie
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiyan Liu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Yanlun Gu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Nan Zhao
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Qian Xiang
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| |
Collapse
|
5
|
Gringhuis SI, Kaptein TM, Remmerswaal EBM, Drewniak A, Wevers BA, Theelen B, D'Haens GRAM, Boekhout T, Geijtenbeek TBH. Fungal sensing by dectin-1 directs the non-pathogenic polarization of T H17 cells through balanced type I IFN responses in human DCs. Nat Immunol 2022; 23:1735-1748. [PMID: 36456734 DOI: 10.1038/s41590-022-01348-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/29/2022] [Indexed: 12/05/2022]
Abstract
The non-pathogenic TH17 subset of helper T cells clears fungal infections, whereas pathogenic TH17 cells cause inflammation and tissue damage; however, the mechanisms controlling these distinct responses remain unclear. Here we found that fungi sensing by the C-type lectin dectin-1 in human dendritic cells (DCs) directed the polarization of non-pathogenic TH17 cells. Dectin-1 signaling triggered transient and intermediate expression of interferon (IFN)-β in DCs, which was mediated by the opposed activities of transcription factors IRF1 and IRF5. IFN-β-induced signaling led to integrin αvβ8 expression directly and to the release of the active form of the cytokine transforming growth factor (TGF)-β indirectly. Uncontrolled IFN-β responses as a result of IRF1 deficiency induced high expression of the IFN-stimulated gene BST2 in DCs and restrained TGF-β activation. Active TGF-β was required for polarization of non-pathogenic TH17 cells, whereas pathogenic TH17 cells developed in the absence of active TGF-β. Thus, dectin-1-mediated modulation of type I IFN responses allowed TGF-β activation and non-pathogenic TH17 cell development during fungal infections in humans.
Collapse
Affiliation(s)
- Sonja I Gringhuis
- Department of Experimental Immunology, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands. .,Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.
| | - Tanja M Kaptein
- Department of Experimental Immunology, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.,Renal Transplant Unit, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Agata Drewniak
- Department of Experimental Immunology, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Brigitte A Wevers
- Department of Experimental Immunology, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Bart Theelen
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Geert R A M D'Haens
- Gastroenterology and Hepatology, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Teun Boekhout
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands.,Institute for Biodiversity and Ecosystem Dynamics (IBED), University of Amsterdam, Amsterdam, The Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands. .,Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Song G, Meng F, Luo B. The β
8
integrin EGF domains support a constitutive extended conformation, and the cytoplasmic domain impairs outside‐in signaling. J Cell Physiol 2022; 237:4251-4261. [DOI: 10.1002/jcp.30871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/28/2022] [Accepted: 08/27/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Guannan Song
- Department of Biological Sciences Louisiana State University Baton Rouge Louisiana USA
| | - Fei Meng
- Department of Biological Sciences Louisiana State University Baton Rouge Louisiana USA
| | - Bing‐Hao Luo
- Department of Biological Sciences Louisiana State University Baton Rouge Louisiana USA
| |
Collapse
|
7
|
This S, Paidassi H. New perspectives on the regulation of germinal center reaction via αvβ8- mediated activation of TGFβ. Front Immunol 2022; 13:942468. [PMID: 36072589 PMCID: PMC9441935 DOI: 10.3389/fimmu.2022.942468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor-β (TGFβ) is a long-known modulator of immune responses but has seemingly contradictory effects on B cells. Among cytokines, TGFβ has the particularity of being produced and secreted in a latent form and must be activated before it can bind to its receptor and induce signaling. While the concept of controlled delivery of TGFβ signaling via αvβ8 integrin-mediated activation has gained some interest in the field of mucosal immunity, the role of this molecular mechanism in regulating T-dependent B cell responses is just emerging. We review here the role of TGFβ and its activation, in particular by αvβ8 integrin, in the regulation of mucosal IgA responses and its demonstrated and putative involvement in regulating germinal center (GC) B cell responses. We examine both the direct effect of TGFβ on GC B cells and its ability to modulate the functions of helper cells, namely follicular T cells (Tfh and Tfr) and follicular dendritic cells. Synthetizing recently published works, we reconcile apparently conflicting data and propose an innovative and unified view on the regulation of the GC reaction by TGFβ, highlighting the role of its activation by αvβ8 integrin.
Collapse
Affiliation(s)
- Sébastien This
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de microbiologie, immunologie et infectiologie, Université de Montréal, Montréal, QC, Canada
| | - Helena Paidassi
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| |
Collapse
|
8
|
Rahman SR, Roper JA, Grove JI, Aithal GP, Pun KT, Bennett AJ. Integrins as a drug target in liver fibrosis. Liver Int 2022; 42:507-521. [PMID: 35048542 DOI: 10.1111/liv.15157] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023]
Abstract
As the worldwide prevalence of chronic liver diseases is high and continuing to increase, there is an urgent need for treatment to prevent cirrhosis-related morbidity and mortality. Integrins are heterodimeric cell-surface proteins that are promising targets for therapeutic intervention. αv integrins are central in the development of fibrosis as they activate latent TGFβ, a known profibrogenic cytokine. The αv subunit can form heterodimers with β1, β3, β5, β6 or β8 subunits and one or more of these integrins are central to the development of liver fibrosis, however, their relative importance is not understood. This review summarises the current knowledge of αv integrins and their respective β subunits in different organs, with a focus on liver fibrosis and the emerging preclinical and clinical data with regards to αv integrin inhibitors.
Collapse
Affiliation(s)
- Syedia R Rahman
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,FRAME Alternatives Laboratory, Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| | - James A Roper
- Novel Human Genetics Research Unit, GlaxoSmithKline, Stevenage, UK
| | - Jane I Grove
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| | - K Tao Pun
- Novel Human Genetics Research Unit, GlaxoSmithKline, Stevenage, UK
| | - Andrew J Bennett
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,FRAME Alternatives Laboratory, Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
9
|
Suppression of the fibrotic encapsulation of silicone implants by inhibiting the mechanical activation of pro-fibrotic TGF-β. Nat Biomed Eng 2021; 5:1437-1456. [PMID: 34031559 DOI: 10.1038/s41551-021-00722-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
The fibrotic encapsulation of implants involves the mechanical activation of myofibroblasts and of pro-fibrotic transforming growth factor beta 1 (TGF-β1). Here, we show that both softening of the implant surfaces and inhibition of the activation of TGF-β1 reduce the fibrotic encapsulation of subcutaneous silicone implants in mice. Conventionally stiff silicones (elastic modulus, ~2 MPa) coated with a soft silicone layer (elastic modulus, ~2 kPa) reduced collagen deposition as well as myofibroblast activation without affecting the numbers of macrophages and their polarization states. Instead, fibroblasts around stiff implants exhibited enhanced intracellular stress, increased the recruitment of αv and β1 integrins, and activated TGF-β1 signalling. In vitro, the recruitment of αv integrin to focal adhesions and the activation of β1 integrin and of TGF-β were higher in myofibroblasts grown on latency-associated peptide (LAP)-coated stiff silicones than on soft silicones. Antagonizing αv integrin binding to LAP through the small-molecule inhibitor CWHM-12 suppressed active TGF-β signalling, myofibroblast activation and the fibrotic encapsulation of stiff subcutaneous implants in mice.
Collapse
|
10
|
Garlatti V, Lovisa S, Danese S, Vetrano S. The Multiple Faces of Integrin-ECM Interactions in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:10439. [PMID: 34638778 PMCID: PMC8508809 DOI: 10.3390/ijms221910439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/03/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) comprises a series of chronic and relapsing intestinal diseases, with Crohn's disease and ulcerative colitis being the most common. The abundant and uncontrolled deposition of extracellular matrix, namely fibrosis, is one of the major hallmarks of IBD and is responsible for the progressive narrowing and closure of the intestine, defined as stenosis. Although fibrosis is usually considered the product of chronic inflammation, the substantial failure of anti-inflammatory therapies to target and reduce fibrosis in IBD suggests that fibrosis might be sustained in an inflammation-independent manner. Pharmacological therapies targeting integrins have recently shown great promise in the treatment of IBD. The efficacy of these therapies mainly relies on their capacity to target the integrin-mediated recruitment and functionality of the immune cells at the damage site. However, by nature, integrins also act as mechanosensitive molecules involved in the intracellular transduction of signals and modifications originating from the extracellular matrix. Therefore, understanding integrin signaling in the context of IBD may offer important insights into mechanisms of matrix remodeling, which are uncoupled from inflammation and could underlie the onset and persistency of intestinal fibrosis. In this review, we present the currently available knowledge on the role of integrins in the etiopathogenesis of IBD, highlighting their role in the context of immune-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Valentina Garlatti
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Pharmaceutical Sciences, University of Piemonte Orientale ‘A. Avogadro’, 28100 Novara, Italy
| | - Sara Lovisa
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Silvio Danese
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Stefania Vetrano
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
11
|
Martin CJ, Datta A, Littlefield C, Kalra A, Chapron C, Wawersik S, Dagbay KB, Brueckner CT, Nikiforov A, Danehy FT, Streich FC, Boston C, Simpson A, Jackson JW, Lin S, Danek N, Faucette RR, Raman P, Capili AD, Buckler A, Carven GJ, Schürpf T. Selective inhibition of TGFβ1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape. Sci Transl Med 2021; 12:12/536/eaay8456. [PMID: 32213632 DOI: 10.1126/scitranslmed.aay8456] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/17/2019] [Accepted: 03/03/2020] [Indexed: 12/12/2022]
Abstract
Despite breakthroughs achieved with cancer checkpoint blockade therapy (CBT), many patients do not respond to anti-programmed cell death-1 (PD-1) due to primary or acquired resistance. Human tumor profiling and preclinical studies in tumor models have recently uncovered transforming growth factor-β (TGFβ) signaling activity as a potential point of intervention to overcome primary resistance to CBT. However, the development of therapies targeting TGFβ signaling has been hindered by dose-limiting cardiotoxicities, possibly due to nonselective inhibition of multiple TGFβ isoforms. Analysis of mRNA expression data from The Cancer Genome Atlas revealed that TGFΒ1 is the most prevalent TGFβ isoform expressed in many types of human tumors, suggesting that TGFβ1 may be a key contributor to primary CBT resistance. To test whether selective TGFβ1 inhibition is sufficient to overcome CBT resistance, we generated a high-affinity, fully human antibody, SRK-181, that selectively binds to latent TGFβ1 and inhibits its activation. Coadministration of SRK-181-mIgG1 and an anti-PD-1 antibody in mice harboring syngeneic tumors refractory to anti-PD-1 treatment induced profound antitumor responses and survival benefit. Specific targeting of TGFβ1 was also effective in tumors expressing more than one TGFβ isoform. Combined SRK-181-mIgG1 and anti-PD-1 treatment resulted in increased intratumoral CD8+ T cells and decreased immunosuppressive myeloid cells. No cardiac valvulopathy was observed in a 4-week rat toxicology study with SRK-181, suggesting that selectively blocking TGFβ1 activation may avoid dose-limiting toxicities previously observed with pan-TGFβ inhibitors. These results establish a rationale for exploring selective TGFβ1 inhibition to overcome primary resistance to CBT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Susan Lin
- Scholar Rock, Inc., Cambridge, MA 02139, USA
| | | | | | - Pichai Raman
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
12
|
Budi EH, Schaub JR, Decaris M, Turner S, Derynck R. TGF-β as a driver of fibrosis: physiological roles and therapeutic opportunities. J Pathol 2021; 254:358-373. [PMID: 33834494 DOI: 10.1002/path.5680] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
Many chronic diseases are marked by fibrosis, which is defined by an abundance of activated fibroblasts and excessive deposition of extracellular matrix, resulting in loss of normal function of the affected organs. The initiation and progression of fibrosis are elaborated by pro-fibrotic cytokines, the most critical of which is transforming growth factor-β1 (TGF-β1). This review focuses on the fibrogenic roles of increased TGF-β activities and underlying signaling mechanisms in the activated fibroblast population and other cell types that contribute to progression of fibrosis. Insight into these roles and mechanisms of TGF-β as a universal driver of fibrosis has stimulated the development of therapeutic interventions to attenuate fibrosis progression, based on interference with TGF-β signaling. Their promise in preclinical and clinical settings will be discussed. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Erine H Budi
- Pliant Therapeutics Inc, South San Francisco, CA, USA
| | | | | | - Scott Turner
- Pliant Therapeutics Inc, South San Francisco, CA, USA
| | - Rik Derynck
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
14
|
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
|
15
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Carvacho I, Piesche M. RGD-binding integrins and TGF-β in SARS-CoV-2 infections - novel targets to treat COVID-19 patients? Clin Transl Immunology 2021; 10:e1240. [PMID: 33747508 PMCID: PMC7971943 DOI: 10.1002/cti2.1240] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
The new coronavirus SARS-CoV-2 is a global pandemic and a severe public health crisis. SARS-CoV-2 is highly contagious and shows high mortality rates, especially in elderly and patients with pre-existing medical conditions. At the current stage, no effective drugs are available to treat these patients. In this review, we analyse the rationale of targeting RGD-binding integrins to potentially inhibit viral cell infection and to block TGF-β activation, which is involved in the severity of several human pathologies, including the complications of severe COVID-19 cases. Furthermore, we demonstrate the correlation between ACE2 and TGF-β expression and the possible consequences for severe COVID-19 infections. Finally, we list approved drugs or drugs in clinical trials for other diseases that also target the RGD-binding integrins or TGF-β. These drugs have already shown a good safety profile and, therefore, can be faster brought into a trial to treat COVID-19 patients.
Collapse
Affiliation(s)
- Ingrid Carvacho
- Department of Biology and ChemistryFaculty of Basic SciencesUniversidad Católica del MauleTalcaChile
| | - Matthias Piesche
- Biomedical Research Laboratories, Medicine FacultyUniversidad Católica del MauleTalcaChile
- Oncology Center, Medicine FacultyUniversidad Católica del MauleTalcaChile
| |
Collapse
|
17
|
Song G, Luo BH. Effects of the association of the α v β 8 lower legs on integrin ligand binding. J Cell Biochem 2021; 122:801-813. [PMID: 33619784 DOI: 10.1002/jcb.29912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/18/2022]
Abstract
Many integrins transmit signals through global conformational changes. However, it is unclear whether integrin αv β8 adopts a similar mechanism during integrin activation and signaling on the cell surface. Here, we showed that disulfide-bonded mutants, which prevented integrin αv β8 lower leg dissociation, bound ligands with similar level as the wild-type protein, suggesting that αv β8 ligand binding did not require lower leg disassociation. We further showed that the N-glycosylation mutant at the interface between the β I and hybrid domains did not affect ligand binding, suggesting that the αv β8 open headpiece was not present on the cell surface. We proposed that αv β8 integrin may adopt only one state, that is, the extended conformation with a closed headpiece. Our results showed that two lower legs retained heterodimeric interfaces, and this association might be important for stabilizing integrin in the extended conformation. Therefore, αv β8 may not transmit bidirectional signals across the plasma membrane but instead may serve as an anchoring site with high affinity and high accessibility for extracellular ligands.
Collapse
Affiliation(s)
- Guannan Song
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bing-Hao Luo
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
18
|
Cui Y, Ji J, Hou J, Tan Y, Han X. Identification of Key Candidate Genes Involved in the Progression of Idiopathic Pulmonary Fibrosis. Molecules 2021; 26:molecules26041123. [PMID: 33672678 PMCID: PMC7924352 DOI: 10.3390/molecules26041123] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal, agnogenic interstitial lung disease with limited therapeutic options. To investigate vital genes involved in the development of IPF, we integrated and compared four expression profiles (GSE110147, GSE53845, GSE24206, and GSE10667), including 87 IPF samples and 40 normal samples. By reanalyzing these datasets, we managed to identify 62 upregulated genes and 20 downregulated genes in IPF samples compared with normal samples. Differentially expressed genes (DEGs) were analyzed by gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis to illustrate relevant pathways of IPF, biological processes, molecular function, and cell components. The DEGs were then subjected to protein-protein interaction (PPI) for network analysis, serving to find 11 key candidate genes (ANXA3, STX11, THBS2, MMP1, MMP9, MMP7, MMP10, SPP1, COL1A1, ITGB8, IGF1). The result of RT-qPCR and immunohistochemical staining verified our finding as well. In summary, we identified 11 key candidate genes related to the process of IPF, which may contribute to novel treatments of IPF.
Collapse
Affiliation(s)
- Yu Cui
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing 210093, China; (Y.C.); (J.J.); (J.H.); (Y.T.)
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Jie Ji
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing 210093, China; (Y.C.); (J.J.); (J.H.); (Y.T.)
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Jiwei Hou
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing 210093, China; (Y.C.); (J.J.); (J.H.); (Y.T.)
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Yi Tan
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing 210093, China; (Y.C.); (J.J.); (J.H.); (Y.T.)
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing 210093, China; (Y.C.); (J.J.); (J.H.); (Y.T.)
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
- Correspondence:
| |
Collapse
|
19
|
Zhang J, Wang T, Saigal A, Johnson J, Morrisson J, Tabrizifard S, Hollingsworth SA, Eddins MJ, Mao W, O'Neill K, Garcia-Calvo M, Carballo-Jane E, Liu D, Ham T, Zhou Q, Dong W, Meng HW, Hicks J, Cai TQ, Akiyama T, Pinto S, Cheng AC, Greshock T, Marquis JC, Ren Z, Talukdar S, Shaheen HH, Handa M. Discovery of a new class of integrin antibodies for fibrosis. Sci Rep 2021; 11:2118. [PMID: 33483531 PMCID: PMC7822819 DOI: 10.1038/s41598-021-81253-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
Lung fibrosis, or the scarring of the lung, is a devastating disease with huge unmet medical need. There are limited treatment options and its prognosis is worse than most types of cancer. We previously discovered that MK-0429 is an equipotent pan-inhibitor of αv integrins that reduces proteinuria and kidney fibrosis in a preclinical model. In the present study, we further demonstrated that MK-0429 significantly inhibits fibrosis progression in a bleomycin-induced lung injury model. In search of newer integrin inhibitors for fibrosis, we characterized monoclonal antibodies discovered using Adimab's yeast display platform. We identified several potent neutralizing integrin antibodies with unique human and mouse cross-reactivity. Among these, Ab-31 blocked the binding of multiple αv integrins to their ligands with IC50s comparable to those of MK-0429. Furthermore, both MK-0429 and Ab-31 suppressed integrin-mediated cell adhesion and latent TGFβ activation. In IPF patient lung fibroblasts, TGFβ treatment induced profound αSMA expression in phenotypic imaging assays and Ab-31 demonstrated potent in vitro activity at inhibiting αSMA expression, suggesting that the integrin antibody is able to modulate TGFβ action though mechanisms beyond the inhibition of latent TGFβ activation. Together, our results highlight the potential to develop newer integrin therapeutics for the treatment of fibrotic lung diseases.
Collapse
Affiliation(s)
- Ji Zhang
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA.
| | - Tao Wang
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Ashmita Saigal
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Josephine Johnson
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Jennifer Morrisson
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Sahba Tabrizifard
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Scott A Hollingsworth
- Computational & Structural Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Michael J Eddins
- Computational & Structural Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Wenxian Mao
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Kim O'Neill
- In Vitro Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Margarita Garcia-Calvo
- In Vitro Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Ester Carballo-Jane
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - DingGang Liu
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Taewon Ham
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Qiong Zhou
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Weifeng Dong
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Hsien-Wei Meng
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Jacqueline Hicks
- Discovery Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Tian-Quan Cai
- In Vivo Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Taro Akiyama
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Shirly Pinto
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Alan C Cheng
- Computational & Structural Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Thomas Greshock
- Discovery Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - John C Marquis
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Zhao Ren
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Saswata Talukdar
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Hussam Hisham Shaheen
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Masahisa Handa
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA.
| |
Collapse
|
20
|
Song G, Luo BH. Atypical structure and function of integrin α V β 8. J Cell Physiol 2020; 236:4874-4887. [PMID: 33368230 DOI: 10.1002/jcp.30242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022]
Abstract
Integrins are heterodimeric transmembrane proteins that play important roles in various biological processes. Most integrins serve as adhesion molecules and transmit bidirectional signaling across the cell membrane through global conformational changes from the bent closed to the extended open conformation. However, integrin β8 is distinctive in structure and function. Its cytoplasmic domain lacks the conserved protein-binding sequence, which is important in transmitting inside-out signals, suggesting that integrin β8 may have a different activation mechanism or lack such signaling. In addition, the ligand-binding or activating metal ion Mn2+ does not induce a global conformational change in integrin β8 . It may have only one conformation, that is, an extended, closed conformation, but with high affinity for ligands under physiological conditions, and is, therefore, considered an atypical integrin member. The extended structure and high ligand-binding affinity of integrin αv β8 make it ideal for encountering and binding ligands expressed on an opposing cell or in the extracellular matrix. In this review, we summarize the progress in integrin β8 research with a focus on its distinctive function and structure among integrin members.
Collapse
Affiliation(s)
- Guannan Song
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bing-Hao Luo
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
21
|
Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020; 587:555-566. [PMID: 33239795 DOI: 10.1038/s41586-020-2938-9] [Citation(s) in RCA: 1066] [Impact Index Per Article: 213.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
Fibrosis can affect any organ and is responsible for up to 45% of all deaths in the industrialized world. It has long been thought to be relentlessly progressive and irreversible, but both preclinical models and clinical trials in various organ systems have shown that fibrosis is a highly dynamic process. This has clear implications for therapeutic interventions that are designed to capitalize on this inherent plasticity. However, despite substantial progress in our understanding of the pathobiology of fibrosis, a translational gap remains between the identification of putative antifibrotic targets and conversion of this knowledge into effective treatments in humans. Here we discuss the transformative experimental strategies that are being leveraged to dissect the key cellular and molecular mechanisms that regulate fibrosis, and the translational approaches that are enabling the emergence of precision medicine-based therapies for patients with fibrosis.
Collapse
Affiliation(s)
- Neil C Henderson
- University of Edinburgh Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK.,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Thomas A Wynn
- Inflammation & Immunology Research Unit, Pfizer Worldwide Research, Development & Medical, Cambridge, MA, USA.
| |
Collapse
|
22
|
Minagawa S, Yoshida M, Araya J, Hara H, Imai H, Kuwano K. Regulated Necrosis in Pulmonary Disease. A Focus on Necroptosis and Ferroptosis. Am J Respir Cell Mol Biol 2020; 62:554-562. [PMID: 32017592 DOI: 10.1165/rcmb.2019-0337tr] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
To date, increasing evidence suggests the possible involvement of various types of cell death in lung diseases. The recognized regulated cell death includes necrotic cell death that is immunogenic, releasing damage-associated molecular patterns and driving tissue inflammation. Necroptosis is a well-understood form of regulated necrosis that is executed by RIPK3 (receptor-interacting protein kinase 3) and the pseudokinase MLKL (mixed lineage kinase domain-like protein). Ferroptosis is a newly described caspase-independent form of regulated necrosis that is characterized by the increase of detrimental lipid reactive oxygen species produced via iron-dependent lipid peroxidation. The role of these two cell death pathways differs depending on the disease, cell type, and microenvironment. Moreover, some experimental cell death models have demonstrated shared ferroptotic and necroptotic cell death and the synergistic effect of simultaneous inhibition. This review examines the role of regulated necrotic cell death, particularly necroptosis and ferroptosis, in lung disease pathogenesis in the context of recent insights into the roles of the key effector molecules of these two cell death pathways.
Collapse
Affiliation(s)
- Shunsuke Minagawa
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan; and
| | - Masahiro Yoshida
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan; and
| | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan; and
| | - Hiromichi Hara
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan; and
| | - Hirotaka Imai
- Laboratory of Hygienic Chemistry and Medicinal Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan; and
| |
Collapse
|
23
|
McCarty JH. αvβ8 integrin adhesion and signaling pathways in development, physiology and disease. J Cell Sci 2020; 133:133/12/jcs239434. [PMID: 32540905 DOI: 10.1242/jcs.239434] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cells must interpret a complex milieu of extracellular cues to modulate intracellular signaling events linked to proliferation, differentiation, migration and other cellular processes. Integrins are heterodimeric transmembrane proteins that link the extracellular matrix (ECM) to the cytoskeleton and control intracellular signaling events. A great deal is known about the structural and functional properties for most integrins; however, the adhesion and signaling pathways controlled by αvβ8 integrin, which was discovered nearly 30 years ago, have only recently been characterized. αvβ8 integrin is a receptor for ECM-bound forms of latent transforming growth factor β (TGFβ) proteins and promotes the activation of TGFβ signaling pathways. Studies of the brain, lung and immune system reveal that the αvβ8 integrin-TGFβ axis mediates cell-cell contact and communication within complex multicellular structures. Perturbing components of this axis results in aberrant cell-cell adhesion and signaling leading to the initiation of various pathologies, including neurodegeneration, fibrosis and cancer. As discussed in this Review, understanding the functions for αvβ8 integrin, its ECM ligands and intracellular effector proteins is not only an important topic in cell biology, but may lead to new therapeutic strategies to treat human pathologies related to integrin dysfunction.
Collapse
Affiliation(s)
- Joseph H McCarty
- Department of Neurosurgery, Brain Tumor Center, M.D. Anderson Cancer Center, 6767 Bertner Avenue, Unit 1004, Houston, TX 77030, USA
| |
Collapse
|
24
|
He D, Li J, Zhou B, Chen Y, Hui Q, Ye F, Zhang L, He X, Niu W, Zhang Q. A correlational meta-analytical study of transforming growth factor-β genetic polymorphisms as a risk factor for chronic obstructive pulmonary disease. Gene 2020; 744:144633. [PMID: 32240778 DOI: 10.1016/j.gene.2020.144633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Several studies have examined the association between transforming growth factor-β (TGF-β) genetic polymorphisms and chronic obstructive pulmonary disease (COPD) risk, but the results remained inconclusive and controversial. AIMS We aimed to examine the correlation between TGF-β genetic polymorphisms and COPD risk through a comprehensive meta-analysis. Additionally, changes in circulating TGF-β concentrations across genotypes of TGF-β genetic polymorphisms were analyzed. METHODS Literature search, quality assessment, and data extraction were completed independently and in duplicate. Data are expressed in odds ratio (OR) or weighted mean difference (WMD) with 95% confidence interval (CI). RESULTS A total of 12 articles, involving 14 independent studies and 7 170 participants, were meta-analyzed for the correlation of five polymorphisms (rs2241712, rs1800469, rs1982073, rs6957, and rs2241718) in TGF-β gene with COPD risk. Under the allele model, no statistical significance was observed for all polymorphisms associated with COPD risk. Subsidiary analyses indicated that country, COPD stage, and diagnosis of COPD were potential sources of between-study heterogeneity. Filled full plots revealed no missing studies for all studied polymorphisms, except rs1982073. Genotype-phenotype analyses showed that carriers of rs1800469 CT genotype had significantly higher concentrations of circulating TGF-β than those with CC genotype in COPD patients (WMD: 0.28 pg/ml, 95% CI: 0.01 to 0.56). CONCLUSION Our findings failed to support the candidacy of TGF-β gene in the development of COPD, whereas the contribution of TGF-β gene to COPD might be ethnicity- and stage-dependent.
Collapse
Affiliation(s)
- Danni He
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China; Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Jialin Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Bo Zhou
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanmei Chen
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Qin Hui
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Fang Ye
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Lipeng Zhang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China; Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xuge He
- Department of Urology, Anshan Cancer Hospital, Anshan, Liaoning Province, China.
| | - Wenquan Niu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China.
| | - Qi Zhang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China.
| |
Collapse
|
25
|
Xiao H, Huang X, Wang S, Liu Z, Dong R, Song D, Dai H. Metformin ameliorates bleomycin-induced pulmonary fibrosis in mice by suppressing IGF-1. Am J Transl Res 2020; 12:940-949. [PMID: 32269725 PMCID: PMC7137034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/15/2020] [Indexed: 06/11/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease, which is characterized by the progressive deterioration in lung function. In the pathogenesis of IPF, insulin-like growth factor-1 (IGF-1) has been found to be heavily involved. Metformin, a commonly used oral antidiabetic agent, is known to inhibit IGF-1 by the reversal of hyperinsulinemia. In this study, we evaluated the effects of metformin in pulmonary fibrosis in C57/BL6J mice, and further understand the role of IGF-1 signaling pathway involving in this process. Pulmonary fibrosis was induced experimentally in these mice by the intratracheal injection of bleomycin (BLM). Metformin was given orally the day before or 14 days after bleomycin injection, while pirfenidone was used as the positive control. Our study showed that intratracheal injection of bleomycin induced pulmonary fibrosis in mice, with observed elevation in collagen, fibronectin and α-SMA level, characterized by the enhanced IGF-1 and PI3K expression. Metformin was able to inhibit these effects significantly, and its antifibrotic effect had no marked difference with pirfenidone. Our results show that metformin attenuates bleomycin-induced pulmonary fibrosis via IGF-1 pathway.
Collapse
Affiliation(s)
- Huijuan Xiao
- Department of Respiratory Medicine, Capital Medical UniversityBeijing 100054, P. R. China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship HospitalBeijing 100029, P. R. China
- National Clinical Research Center for Respiratory DiseasesBeijing 100029, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical SciencesBeijing 100029, P. R. China
| | - Xiaoxi Huang
- Department of Medical Research, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijing 100020, P. R. China
| | - Shiyao Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship HospitalBeijing 100029, P. R. China
- National Clinical Research Center for Respiratory DiseasesBeijing 100029, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical SciencesBeijing 100029, P. R. China
| | - Zheng Liu
- Department of Medical Research, Beijing Chao-Yang Hospital, Capital Medical UniversityBeijing 100020, P. R. China
| | - Run Dong
- Department of Respiratory Medicine, Zhengzhou Central HospitalZhengzhou, P. R. China
| | - Dingyun Song
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship HospitalBeijing 100029, P. R. China
- National Clinical Research Center for Respiratory DiseasesBeijing 100029, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical SciencesBeijing 100029, P. R. China
| | - Huaping Dai
- Department of Respiratory Medicine, Capital Medical UniversityBeijing 100054, P. R. China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship HospitalBeijing 100029, P. R. China
- National Clinical Research Center for Respiratory DiseasesBeijing 100029, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical SciencesBeijing 100029, P. R. China
| |
Collapse
|
26
|
Campbell MG, Cormier A, Ito S, Seed RI, Bondesson AJ, Lou J, Marks JD, Baron JL, Cheng Y, Nishimura SL. Cryo-EM Reveals Integrin-Mediated TGF-β Activation without Release from Latent TGF-β. Cell 2020; 180:490-501.e16. [PMID: 31955848 PMCID: PMC7238552 DOI: 10.1016/j.cell.2019.12.030] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/15/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
Integrin αvβ8 binds with exquisite specificity to latent transforming growth factor-β (L-TGF-β). This binding is essential for activating L-TGF-β presented by a variety of cell types. Inhibiting αvβ8-mediated TGF-β activation blocks immunosuppressive regulatory T cell differentiation, which is a potential therapeutic strategy in cancer. Using cryo-electron microscopy, structure-guided mutagenesis, and cell-based assays, we reveal the binding interactions between the entire αvβ8 ectodomain and its intact natural ligand, L-TGF-β, as well as two different inhibitory antibody fragments to understand the structural underpinnings of αvβ8 binding specificity and TGF-β activation. Our studies reveal a mechanism of TGF-β activation where mature TGF-β signals within the confines of L-TGF-β and the release and diffusion of TGF-β are not required. The structural details of this mechanism provide a rational basis for therapeutic strategies to inhibit αvβ8-mediated L-TGF-β activation.
Collapse
Affiliation(s)
- Melody G Campbell
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Anthony Cormier
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Saburo Ito
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Robert I Seed
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew J Bondesson
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - James D Marks
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Jody L Baron
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
27
|
Rare copy number variants in over 100,000 European ancestry subjects reveal multiple disease associations. Nat Commun 2020; 11:255. [PMID: 31937769 PMCID: PMC6959272 DOI: 10.1038/s41467-019-13624-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/14/2019] [Indexed: 01/05/2023] Open
Abstract
Copy number variants (CNVs) are suggested to have a widespread impact on the human genome and phenotypes. To understand the role of CNVs across human diseases, we examine the CNV genomic landscape of 100,028 unrelated individuals of European ancestry, using SNP and CGH array datasets. We observe an average CNV burden of ~650 kb, identifying a total of 11,314 deletion, 5625 duplication, and 2746 homozygous deletion CNV regions (CNVRs). In all, 13.7% are unreported, 58.6% overlap with at least one gene, and 32.8% interrupt coding exons. These CNVRs are significantly more likely to overlap OMIM genes (2.94-fold), GWAS loci (1.52-fold), and non-coding RNAs (1.44-fold), compared with random distribution (P < 1 × 10−3). We uncover CNV associations with four major disease categories, including autoimmune, cardio-metabolic, oncologic, and neurological/psychiatric diseases, and identify several drug-repurposing opportunities. Our results demonstrate robust frequency definition for large-scale rare variant association studies, identify CNVs associated with major disease categories, and illustrate the pleiotropic impact of CNVs in human disease. Associations of copy number variations (CNVs) with complex traits are challenging to study because of their low frequency. Here, the authors analyse SNP array and array comparative genomic hybridization data of 100,028 individuals and report their associations with immune-related, cardiometabolic and neuropsychiatric diseases as well as cancer.
Collapse
|
28
|
Activation and suppression of hematopoietic integrins in hemostasis and immunity. Blood 2020; 135:7-16. [DOI: 10.1182/blood.2019003336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Nolte and Margadant review the current understanding of the activation and inactivation of integrin receptors expressed by hematopoietic cells and the role of these conformational changes in modulating platelet and leukocyte function.
Collapse
|
29
|
Nolte M, Margadant C. Controlling Immunity and Inflammation through Integrin-Dependent Regulation of TGF-β. Trends Cell Biol 2020; 30:49-59. [DOI: 10.1016/j.tcb.2019.10.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022]
|
30
|
McEntee CP, Gunaltay S, Travis MA. Regulation of barrier immunity and homeostasis by integrin-mediated transforming growth factor β activation. Immunology 2019; 160:139-148. [PMID: 31792952 PMCID: PMC7218408 DOI: 10.1111/imm.13162] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor β (TGF‐β) is a multifunctional cytokine that regulates cell growth, differentiation, adhesion, migration and death dependent on cell type, developmental stage, or tissue conditions. Various cell types secrete TGF‐β, but always as an inactive complex. Hence, for TGF‐β to function, this latent complex must somehow be activated. Work in recent years has highlighted a critical role for members of the αv integrin family, including αvβ1, αvβ3, αvβ5, αvβ6 and αvβ8 that are involved in TGF‐β activation in various contexts, particularly at barrier sites such as the gut, lung and skin. The integrins facilitating this context‐ and location‐specific regulation can be dysregulated in certain diseases, so are potential therapeutic targets in a number of disorders. In this review, we discuss the role of TGF‐β at these barrier sites with a focus on how integrin‐mediated TGF‐β activation regulates tissue and immune homeostasis, and how this is altered in disease.
Collapse
Affiliation(s)
- Craig P McEntee
- Lydia Becker Institute for Immunology and Inflammation, Manchester, UK.,Wellcome Trust Centre for Cell-Matrix Research, Manchester, UK.,Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research (MCCIR), Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Sezin Gunaltay
- Lydia Becker Institute for Immunology and Inflammation, Manchester, UK.,Wellcome Trust Centre for Cell-Matrix Research, Manchester, UK.,Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research (MCCIR), Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Mark A Travis
- Lydia Becker Institute for Immunology and Inflammation, Manchester, UK.,Wellcome Trust Centre for Cell-Matrix Research, Manchester, UK.,Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research (MCCIR), Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| |
Collapse
|
31
|
Lodyga M, Hinz B. TGF-β1 - A truly transforming growth factor in fibrosis and immunity. Semin Cell Dev Biol 2019; 101:123-139. [PMID: 31879265 DOI: 10.1016/j.semcdb.2019.12.010] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022]
Abstract
'Jack of all trades, master of everything' is a fair label for transforming growth factor β1 (TGF-β) - a cytokine that controls our life at many levels. In the adult organism, TGF-β1 is critical for the development and maturation of immune cells, maintains immune tolerance and homeostasis, and regulates various aspects of immune responses. Following acute tissue damages, TGF-β1 becomes a master regulator of the healing process with impacts on about every cell type involved. Divergence from the tight control of TGF-β1 actions, for instance caused by chronic injury, severe trauma, or infection can tip the balance from regulated physiological to excessive pathological repair. This condition of fibrosis is characterized by accumulation and stiffening of collagenous scar tissue which impairs organ functions to the point of failure. Fibrosis and dysregulated immune responses are also a feature of cancer, in which tumor cells escape immune control partly by manipulating TGF-β1 regulation and where immune cells are excluded from the tumor by fibrotic matrix created during the stroma 'healing' response. Despite the obvious potential of TGF-β-signalling therapies, globally targeting TGF-β1 receptor, downstream pathways, or the active growth factor have proven to be extremely difficult if not impossible in systemic treatment regimes. However, TGF-β1 binding to cell receptors requires prior activation from latent complexes that are extracellularly presented on the surface of immune cells or within the extracellular matrix. These different locations have led to some divergence in the field which is often either seen from the perspective of an immunologists or a fibrosis/matrix researcher. Despite these human boundaries, there is considerable overlap between immune and tissue repair cells with respect to latent TGF-β1 presentation and activation. Moreover, the mechanisms and proteins employed by different cells and spatiotemporal control of latent TGF-β1 activation provide specificity that is amenable to drug development. This review aims at synthesizing the knowledge on TGF-β1 extracellular activation in the immune system and in fibrosis to further stimulate cross talk between the two research communities in solving the TGF-β conundrum.
Collapse
Affiliation(s)
- Monika Lodyga
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, M5G1G6, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, M5G1G6, Canada.
| |
Collapse
|
32
|
Sevoflurane Prevents Airway Remodeling via Downregulation of VEGF and TGF-β1 in Mice with OVA-Induced Chronic Airway Inflammation. Inflammation 2019; 42:1015-1022. [PMID: 30680697 DOI: 10.1007/s10753-019-00963-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Asthma is characterized by chronic airway inflammation, which is the underlying cause of airway remodeling featured by goblet cell hyperplasia, subepithelial fibrosis, and proliferation of smooth muscle. Sevoflurane has been used to treat life-threatening asthma and our previous study shows that sevoflurane inhibits acute lung inflammation in ovalbumin (OVA)-induced allergic mice. However, the effect of sevoflurane on airway remodeling in the context of chronic airway inflammation and the underlying mechanism are still unknown. Here, female C57BL/6 mice were used to establish chronic airway inflammation model. Hematoxylin and eosin (H&E), periodic acid-Schiff (PAS), and Sirius red (SR) staining were used to evaluate airway remodeling. Protein levels of α-SMA, VEGF, and TGF-β1 in lung tissues were detected by western blotting analyses and immunohistochemistry staining. Results showed that inhalation of sevoflurane inhibited chronic airway inflammation including inflammatory cell infiltration and pro-inflammatory cytokine production in BALF of the OVA-challenged mice. Meanwhile, sevoflurane suppressed airway thickening, goblet cell hyperplasia, smooth muscle hyperplasia, collagen deposition, and fiber hyperplasia in the lung tissues of the mice with airway remodeling. Most notably, sevoflurane inhibited the OVA-induced expressions of VEGF and TGF-β1. These results suggested that sevoflurane effectively inhibits airway remodeling in mouse model of chronic airway inflammation, which may be due to the downregulation of VEGF and TGF-β1in lung tissues. Therefore, our results indicate a potential role of sevoflurane in inhibiting airway remodeling besides its known suppression effect on airway inflammation, and support the use of sevoflurane in treating severe asthma in ICU.
Collapse
|
33
|
Wang J, Su Y, Iacob RE, Engen JR, Springer TA. General structural features that regulate integrin affinity revealed by atypical αVβ8. Nat Commun 2019; 10:5481. [PMID: 31792290 PMCID: PMC6889490 DOI: 10.1038/s41467-019-13248-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/22/2019] [Indexed: 01/16/2023] Open
Abstract
Integrin αVβ8, which like αVβ6 functions to activate TGF-βs, is atypical. Its β8 subunit binds to a distinctive cytoskeleton adaptor and does not exhibit large changes in conformation upon binding to ligand. Here, crystal structures, hydrogen-deuterium exchange dynamics, and affinity measurements on mutants are used to compare αVβ8 and αVβ6. Lack of a binding site for one of three βI domain divalent cations and a unique β6-α7 loop conformation in β8 facilitate movements of the α1 and α1’ helices at the ligand binding pocket toward the high affinity state, without coupling to β6-α7 loop reshaping and α7-helix pistoning that drive large changes in βI domain-hybrid domain orientation seen in other integrins. Reciprocal swaps between β6 and β8 βI domains increase affinity of αVβ6 and decrease affinity of αVβ8 and define features that regulate affinity of the βI domain and its coupling to the hybrid domain. The activation mechanism of integrin αVβ8 differs from other integrins. Combining X-ray crystallography, hydrogen deuterium exchange mass spectrometry and mutation, the authors reveal structural features responsible for these differences and provide insights into how typical integrins are regulated.
Collapse
Affiliation(s)
- Jianchuan Wang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Yang Su
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Roxana E Iacob
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - John R Engen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA. .,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
34
|
Liang X, He X, Li Y, Wang J, Wu D, Yuan X, Wang X, Li G. Lyn regulates epithelial-mesenchymal transition in CS-exposed model through Smad2/3 signaling. Respir Res 2019; 20:201. [PMID: 31477108 PMCID: PMC6720409 DOI: 10.1186/s12931-019-1166-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/15/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized by airflow limitation that is progressive and not fully reversible. Cigarette smoking is one of the most commonly and important risk factors for COPD, which contributes to airway remodeling, the outstanding pathological changes in COPD. One potential mechanism which might be important for airway remodeling is the process called epithelial-mesenchymal transition (EMT). However, the underlying molecular mechanisms of EMT in CS-induced COPD are still poorly understood. METHODS Two Gene Expression Omnibus (GEO) datasets (GSE108134 and GSE5058) were combined to identify the key genes involved in COPD. Then, single-gene analysis of Lyn was performed. Lyn expression was confirmed in patients with COPD. 16HBE cells were treated with cigarette smoking extracts (CSE). Wild type (WT) C57BL/6 J mice and Lyn+/+ transgenic mice were exposed to CSE to establish CS-exposed model. Pathological changes were observed by hematoxylin-eosin staining. The expression levels of EMT markers were examined by using western blot and immunofluorescence. The expression and phosphorylation levels of Lyn and Smad2/3 were detected as well. RESULTS The gain of mesenchymal markers vimentin and α-SMA with a concomitant loss of E-cadherin was observed in both in vivo and in vitro studies. Meanwhile, cigarette smoking extracts (CSE) induced EMT in 16HBE cells in a time- and dose- dependent manner. Furthermore, by analyzing GEO datasets and using molecular methods, we explored a kinase, Lyn, its expression correlated with the expression of E-cadherin, vimentin and α-SMA in CS-exposed model. Moreover, we found that EMT induced by CSE was regulated by activated Lyn through phosphorylation of Smad2/3. CONCLUSIONS In summary, we found that Lyn regulates epithelial-mesenchymal transition in CS-exposed model through Smad2/3 signaling. As a kinase, Lyn is "druggable", and might provide a therapeutic opportunity for targeting EMT. Therefore, our research might provide a new method to treat COPD by targeting Lyn kinase specifically.
Collapse
Affiliation(s)
- Xiaobo Liang
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
- First Department of Respiratory Disease, Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Xiang He
- Laboratory of Allergy and Inflammation of Allergy Department, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
- Department of Respiratory Disease, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
| | - Yin Li
- Department of Thoracic Surgery, Zhongshan Hospital, Affiliated Hospital of Fudan University, Shanghai, 200032 China
| | - Junyi Wang
- Laboratory of Allergy and Inflammation of Allergy Department, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
- Department of Respiratory Disease, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
| | - Dehong Wu
- Laboratory of Allergy and Inflammation of Allergy Department, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
- Department of Respiratory Disease, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
| | - Xiefang Yuan
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Xiaoyun Wang
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Guoping Li
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
- Laboratory of Allergy and Inflammation of Allergy Department, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
- Department of Respiratory Disease, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031 China
| |
Collapse
|
35
|
Yoshida M, Minagawa S, Araya J, Sakamoto T, Hara H, Tsubouchi K, Hosaka Y, Ichikawa A, Saito N, Kadota T, Sato N, Kurita Y, Kobayashi K, Ito S, Utsumi H, Wakui H, Numata T, Kaneko Y, Mori S, Asano H, Yamashita M, Odaka M, Morikawa T, Nakayama K, Iwamoto T, Imai H, Kuwano K. Involvement of cigarette smoke-induced epithelial cell ferroptosis in COPD pathogenesis. Nat Commun 2019; 10:3145. [PMID: 31316058 PMCID: PMC6637122 DOI: 10.1038/s41467-019-10991-7] [Citation(s) in RCA: 382] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/14/2019] [Indexed: 01/20/2023] Open
Abstract
Ferroptosis is a necrotic form of regulated cell death (RCD) mediated by phospholipid peroxidation in association with free iron-mediated Fenton reactions. Disrupted iron homeostasis resulting in excessive oxidative stress has been implicated in the pathogenesis of chronic obstructive pulmonary disease (COPD). Here, we demonstrate the involvement of ferroptosis in COPD pathogenesis. Our in vivo and in vitro models show labile iron accumulation and enhanced lipid peroxidation with concomitant non-apoptotic cell death during cigarette smoke (CS) exposure, which are negatively regulated by GPx4 activity. Treatment with deferoxamine and ferrostatin-1, in addition to GPx4 knockdown, illuminate the role of ferroptosis in CS-treated lung epithelial cells. NCOA4-mediated ferritin selective autophagy (ferritinophagy) is initiated during ferritin degradation in response to CS treatment. CS exposure models, using both GPx4-deficient and overexpressing mice, clarify the pivotal role of GPx4-regulated cell death during COPD. These findings support a role for cigarette smoke-induced ferroptosis in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Masahiro Yoshida
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Shunsuke Minagawa
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan.
| | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Taro Sakamoto
- Laboratory of Hygienic Chemistry and Medicinal Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, 108-8641, Tokyo, Japan
| | - Hiromichi Hara
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Kazuya Tsubouchi
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Yusuke Hosaka
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Akihiro Ichikawa
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Nayuta Saito
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Tsukasa Kadota
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Nahoko Sato
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Yusuke Kurita
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Kenji Kobayashi
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Saburo Ito
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Hirohumi Utsumi
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Hiroshi Wakui
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Takanori Numata
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Yumi Kaneko
- Division of Chest Diseases, Department of Surgery, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Shohei Mori
- Division of Chest Diseases, Department of Surgery, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Hisatoshi Asano
- Division of Chest Diseases, Department of Surgery, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Makoto Yamashita
- Division of Chest Diseases, Department of Surgery, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Makoto Odaka
- Division of Chest Diseases, Department of Surgery, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Toshiaki Morikawa
- Division of Chest Diseases, Department of Surgery, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Katsutoshi Nakayama
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Takeo Iwamoto
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| | - Hirotaka Imai
- Laboratory of Hygienic Chemistry and Medicinal Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, 108-8641, Tokyo, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 105-8461, Tokyo, Japan
| |
Collapse
|
36
|
Greenhalgh SN, Matchett KP, Taylor RS, Huang K, Li JT, Saeteurn K, Donnelly MC, Simpson EEM, Pollack JL, Atakilit A, Simpson KJ, Maher JJ, Iredale JP, Sheppard D, Henderson NC. Loss of Integrin αvβ8 in Murine Hepatocytes Accelerates Liver Regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:258-271. [PMID: 30448409 PMCID: PMC6360354 DOI: 10.1016/j.ajpath.2018.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/14/2018] [Accepted: 10/10/2018] [Indexed: 02/08/2023]
Abstract
Recent fate-mapping studies in mice have provided substantial evidence that mature adult hepatocytes are a major source of new hepatocytes after liver injury. In other systems, integrin αvβ8 has a major role in activating transforming growth factor (TGF)-β, a potent inhibitor of hepatocyte proliferation. We hypothesized that depletion of hepatocyte integrin αvβ8 would increase hepatocyte proliferation and accelerate liver regeneration after injury. Using Itgb8flox/flox;Alb-Cre mice to deplete hepatocyte αvβ8, after partial hepatectomy, hepatocyte proliferation and liver-to-body weight ratio were significantly increased in Itgb8flox/flox;Alb-Cre mice compared with control mice. Antibody-mediated blockade of hepatocyte αvβ8 in vitro, with assessment of TGF-β signaling pathways by real-time quantitative PCR array, supported the hypothesis that integrin αvβ8 inhibition alters hepatocyte TGF-β signaling toward a pro-regenerative phenotype. A diethylnitrosamine-induced model of hepatocellular carcinoma, used to examine the possibility that this pro-proliferative phenotype might be oncogenic, revealed no difference in either tumor number or size between Itgb8flox/flox;Alb-Cre and control mice. Immunohistochemistry for integrin αvβ8 in healthy and injured human liver demonstrated that human hepatocytes express integrin αvβ8. Depletion of hepatocyte integrin αvβ8 results in increased hepatocyte proliferation and accelerated liver regeneration after partial hepatectomy in mice. These data demonstrate that targeting integrin αvβ8 may represent a promising therapeutic strategy to drive liver regeneration in patients with a broad range of liver diseases.
Collapse
Affiliation(s)
- Stephen N Greenhalgh
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Kylie P Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard S Taylor
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Katherine Huang
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - John T Li
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Koy Saeteurn
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Mhairi C Donnelly
- Department of Hepatology, Scottish Liver Transplant Unit and University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Eilidh E M Simpson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Joshua L Pollack
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Amha Atakilit
- Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Kenneth J Simpson
- Department of Hepatology, Scottish Liver Transplant Unit and University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Jacquelyn J Maher
- Liver Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - John P Iredale
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Senate House, University of Bristol, Bristol, United Kingdom
| | - Dean Sheppard
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
37
|
Sciurba JC, Gieseck RL, Jiwrajka N, White SD, Karmele EP, Redes J, Vannella KM, Henderson NC, Wynn TA, Hart KM. Fibroblast-specific integrin-alpha V differentially regulates type 17 and type 2 driven inflammation and fibrosis. J Pathol 2019; 248:16-29. [PMID: 30536905 DOI: 10.1002/path.5215] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/08/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023]
Abstract
Fibroproliferative diseases affect a significant proportion of the world's population. Despite this, core mechanisms driving organ fibrosis of diverse etiologies remain ill defined. Recent studies suggest that integrin-alpha V serves as a master driver of fibrosis in multiple organs. Although diverse mechanisms contribute to the progression of fibrosis, TGF-β and IL-13 have emerged as central mediators of fibrosis during type 1/type 17, and type 2 polarized inflammatory responses, respectively. To investigate if integrin-alpha V interactions or signaling is critical to the development of type 2 fibrosis, we analyzed fibroblast-specific integrin-alpha V knockout mice in three type 2-driven inflammatory disease models. While we confirmed a role for integrin-alpha V in type 17-associated fibrosis, integrin-alpha V was not critical to the development of type 2-driven fibrosis. Additionally, our studies support a novel mechanism through which fibroblasts, via integrin-alpha V expression, are capable of regulating immune polarization. We show that when integrin-alpha V is deleted on fibroblasts, initiation of type 17 inflammation is inhibited leading to a deregulation of type 2 inflammation. This mechanism is most evident in a model of severe asthma, which is characterized by a mixed type 2/type 17 inflammatory response. Together, these findings suggest dual targeting of integrin-alpha V and type 2 pathways may be needed to ameliorate fibrosis and prevent rebound of opposing pro-fibrotic and inflammatory mechanisms. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Joshua C Sciurba
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard L Gieseck
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nikhil Jiwrajka
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandra D White
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Erik P Karmele
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Redes
- MRC Centre for Inflammation Research, Centre for Multiple Sclerosis Research, BHF Centre for Cardiovascular Science, and Centre for Immunity Infection and Evolution, University of Edinburgh, Edinburgh, UK
| | - Kevin M Vannella
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neil C Henderson
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thomas A Wynn
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin M Hart
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
38
|
Takasaka N, Seed RI, Cormier A, Bondesson AJ, Lou J, Elattma A, Ito S, Yanagisawa H, Hashimoto M, Ma R, Levine MD, Publicover J, Potts R, Jespersen JM, Campbell MG, Conrad F, Marks JD, Cheng Y, Baron JL, Nishimura SL. Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight 2018; 3:122591. [PMID: 30333313 DOI: 10.1172/jci.insight.122591] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022] Open
Abstract
TGF-β is a promising immunotherapeutic target. It is expressed ubiquitously in a latent form that must be activated to function. Determination of where and how latent TGF-β (L-TGF-β) is activated in the tumor microenvironment could facilitate cell- and mechanism-specific approaches to immunotherapeutically target TGF-β. Binding of L-TGF-β to integrin αvβ8 results in activation of TGF-β. We engineered and used αvβ8 antibodies optimized for blocking or detection, which - respectively - inhibit tumor growth in syngeneic tumor models or sensitively and specifically detect β8 in human tumors. Inhibition of αvβ8 potentiates cytotoxic T cell responses and recruitment of immune cells to tumor centers - effects that are independent of PD-1/PD-L1. β8 is expressed on the cell surface at high levels by tumor cells, not immune cells, while the reverse is true of L-TGF-β, suggesting that tumor cell αvβ8 serves as a platform for activating cell-surface L-TGF-β presented by immune cells. Transcriptome analysis of tumor-associated lymphoid cells reveals macrophages as a key cell type responsive to β8 inhibition with major increases in chemokine and tumor-eliminating genes. High β8 expression in tumor cells is seen in 20%-80% of various cancers, which rarely coincides with high PD-L1 expression. These data suggest tumor cell αvβ8 is a PD-1/PD-L1-independent immunotherapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yifan Cheng
- Department of Biochemistry and Biophysics, and.,Howard Hughes Medical Institute, UCSF, San Francisco, California, USA
| | | | | |
Collapse
|
39
|
Araya J, Tsubouchi K, Sato N, Ito S, Minagawa S, Hara H, Hosaka Y, Ichikawa A, Saito N, Kadota T, Yoshida M, Fujita Y, Utsumi H, Kobayashi K, Yanagisawa H, Hashimoto M, Wakui H, Ishikawa T, Numata T, Kaneko Y, Asano H, Yamashita M, Odaka M, Morikawa T, Nishimura SL, Nakayama K, Kuwano K. PRKN-regulated mitophagy and cellular senescence during COPD pathogenesis. Autophagy 2018; 15:510-526. [PMID: 30290714 DOI: 10.1080/15548627.2018.1532259] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Cigarette smoke (CS)-induced accumulation of mitochondrial damage has been widely implicated in chronic obstructive pulmonary disease (COPD) pathogenesis. Mitophagy plays a crucial role in eliminating damaged mitochondria, and is governed by the PINK1 (PTEN induced putative protein kinase 1)-PRKN (parkin RBR E3 ubiquitin protein ligase) pathway. Although both increased PINK1 and reduced PRKN have been implicated in COPD pathogenesis in association with mitophagy, there are conflicting reports for the role of mitophagy in COPD progression. To clarify the involvement of PRKN-regulated mitophagy in COPD pathogenesis, prkn knockout (KO) mouse models were used. To illuminate how PINK1 and PRKN regulate mitophagy in relation to CS-induced mitochondrial damage and cellular senescence, overexpression and knockdown experiments were performed in airway epithelial cells (AEC). In comparison to wild-type mice, prkn KO mice demonstrated enhanced airway wall thickening with emphysematous changes following CS exposure. AEC in CS-exposed prkn KO mice showed accumulation of damaged mitochondria and increased oxidative modifications accompanied by accelerated cellular senescence. In vitro experiments showed PRKN overexpression was sufficient to induce mitophagy during CSE exposure even in the setting of reduced PINK1 protein levels, resulting in attenuation of mitochondrial ROS production and cellular senescence. Conversely PINK1 overexpression failed to recover impaired mitophagy caused by PRKN knockdown, indicating that PRKN protein levels can be the rate-limiting factor in PINK1-PRKN-mediated mitophagy during CSE exposure. These results suggest that PRKN levels may play a pivotal role in COPD pathogenesis by regulating mitophagy, suggesting that PRKN induction could mitigate the progression of COPD. Abbreviations: AD: Alzheimer disease; AEC: airway epithelial cells; BALF: bronchoalveolar lavage fluid; AKT: AKT serine/threonine kinase; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CDKN1A: cyclin dependent kinase inhibitor 1A; CDKN2A: cyclin dependent kinase inhibitor 2A; COPD: chronic obstructive pulmonary disease; CS: cigarette smoke; CSE: CS extract; CXCL1: C-X-C motif chemokine ligand 1; CXCL8: C-X-C motif chemokine ligand 8; HBEC: human bronchial epithelial cells; 4-HNE: 4-hydroxynonenal; IL: interleukin; KO: knockout; LF: lung fibroblasts; LPS: lipopolysaccharide; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; 8-OHdG: 8-hydroxy-2'-deoxyguanosine; OPTN: optineurin; PRKN: parkin RBR E3 ubiquitin protein ligase; PCD: programmed cell death; PFD: pirfenidone; PIK3C: phosphatidylinositol-4:5-bisphosphate 3-kinase catalytic subunit; PINK1: PTEN induced putative kinase 1; PTEN: phosphatase and tensin homolog; RA: rheumatoid arthritis; ROS: reactive oxygen species; SA-GLB1/β-Gal: senescence-associated-galactosidase, beta 1; SASP: senescence-associated secretory phenotype; SNP: single nucleotide polymorphism; TNF: tumor necrosis factor.
Collapse
Affiliation(s)
- Jun Araya
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Kazuya Tsubouchi
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan.,b Research Institute for Diseases of the Chest, Graduate School of Medical Sciences , Kyushu University , Fukuoka , Japan
| | - Nahoko Sato
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan.,c Department of Respiratory Medicine, Faculty of Life Science , Kumamoto University , Kumamoto , Japan
| | - Saburo Ito
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Shunsuke Minagawa
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Hiromichi Hara
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Yusuke Hosaka
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Akihiro Ichikawa
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Nayuta Saito
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Tsukasa Kadota
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Masahiro Yoshida
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Yu Fujita
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Hirofumi Utsumi
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Kenji Kobayashi
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Haruhiko Yanagisawa
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Mitsuo Hashimoto
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Hiroshi Wakui
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Takeo Ishikawa
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Takanori Numata
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Yumi Kaneko
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Hisatoshi Asano
- d Division of Chest Diseases, Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Makoto Yamashita
- d Division of Chest Diseases, Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Makoto Odaka
- d Division of Chest Diseases, Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Toshiaki Morikawa
- d Division of Chest Diseases, Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Stephen L Nishimura
- e Department of Pathology , University of California, San Francisco , San Francisco , CA , USA
| | - Katsutoshi Nakayama
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Kazuyoshi Kuwano
- a Division of Respiratory Diseases, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| |
Collapse
|
40
|
Walraven M, Hinz B. Therapeutic approaches to control tissue repair and fibrosis: Extracellular matrix as a game changer. Matrix Biol 2018; 71-72:205-224. [PMID: 29499355 DOI: 10.1016/j.matbio.2018.02.020] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 02/08/2023]
|
41
|
Cryo-EM structure of the αvβ8 integrin reveals a mechanism for stabilizing integrin extension. Nat Struct Mol Biol 2018; 25:698-704. [PMID: 30061598 PMCID: PMC6214843 DOI: 10.1038/s41594-018-0093-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/21/2018] [Indexed: 01/06/2023]
Abstract
Integrins are conformationally flexible cell surface receptors that survey the extracellular environment for their cognate ligands. Interactions with ligands are thought to be linked to global structural rearrangements involving transitions between bent, extended-closed and -open forms. Thus far, structural details are lacking for integrins in the extended conformation due to extensive flexibility between the headpiece and legs within this conformation. Here we present single-particle electron cryo-microscopy structures of human αvβ8 integrin in the extended-closed conformation, which has been considered to be a low-affinity intermediate. Our structures show the headpiece rotating about a flexible αv-knee, suggesting a ligand surveillance mechanism for integrins in their extended-closed form. Our model predicts that the extended conformation is mainly stabilized by an interface formed between flexible loops in the upper and lower domains of the αv-leg. Confirming these findings with the αvβ3 integrin suggests that our model of stabilizing the extended-closed conformation is generalizable to other integrins.
Collapse
|
42
|
Hu P, Luo B. The interface between the EGF1 and EGF2 domains is critical in integrin affinity regulation. J Cell Biochem 2018; 119:7264-7273. [DOI: 10.1002/jcb.26921] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 04/04/2018] [Indexed: 02/02/2023]
Affiliation(s)
- Ping Hu
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisiana
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginia
| | - Bing‐Hao Luo
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisiana
| |
Collapse
|
43
|
Miyazaki N, Iwasaki K, Takagi J. A systematic survey of conformational states in β1 and β4 integrins using negative-stain electron microscopy. J Cell Sci 2018; 131:jcs.216754. [PMID: 29700202 DOI: 10.1242/jcs.216754] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/19/2018] [Indexed: 01/23/2023] Open
Abstract
Structural analyses of β2 and β3 integrins have revealed that they generally assume a compact bent conformation in the resting state and undergo a global conformational transition involving extension during upregulation of ligand affinity, collectively called the 'switchblade model'. This hypothesis, however, has not been extensively tested for other classes of integrins. We prepared a set of recombinant integrin ectodomain fragments including αvβ3, α2β1, α3β1, α5β1, α6β1 and α6β4, and used negative-stain electron microscopy to examine their structures under various conditions. In contrast to αvβ3 integrin, which exhibited a severely bent conformation in low-affinity 5 mM Ca2+ conditions, all β1 integrin heterodimers displayed a mixed population of half-bent to fully extended conformations. Moreover, they did not undergo significant conformational change upon activation by Mn2+ Integrin α6β4 was even more resistant to conformational regulation, showing a completely extended structure regardless of the buffer conditions. These results suggest that the mechanisms of conformational regulation of integrins are more diverse and complex than previously thought, requiring more experimental scrutiny for each integrin subfamily member.
Collapse
Affiliation(s)
- Naoyuki Miyazaki
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kenji Iwasaki
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Junichi Takagi
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
44
|
Schuppan D, Ashfaq-Khan M, Yang AT, Kim YO. Liver fibrosis: Direct antifibrotic agents and targeted therapies. Matrix Biol 2018; 68-69:435-451. [PMID: 29656147 DOI: 10.1016/j.matbio.2018.04.006] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022]
Abstract
Liver fibrosis and in particular cirrhosis are the major causes of morbidity and mortality of patients with chronic liver disease. Their prevention or reversal have become major endpoints in clinical trials with novel liver specific drugs. Remarkable progress has been made with therapies that efficiently address the cause of the underlying liver disease, as in chronic hepatitis B and C. Highly effective antiviral therapy can prevent progression or even induce reversal in the majority of patients, but such treatment remains elusive for the majority of liver patients with advanced alcoholic or nonalcoholic steatohepatitis, genetic or autoimmune liver diseases. Moreover, drugs that would speed up fibrosis reversal are needed for patients with cirrhosis, since even with effective causal therapy reversal is slow or the disease may further progress. Therefore, highly efficient and specific antifibrotic agents are needed that can address advanced fibrosis, i.e., the detrimental downstream result of all chronic liver diseases. This review discusses targeted antifibrotic therapies that address molecules and mechanisms that are central to fibrogenesis or fibrolysis, including strategies that allow targeting of activated hepatic stellate cells and myofibroblasts and other fibrogenic effector cells. Focus is on collagen synthesis, integrins and cells and mechanisms specific including specific downregulation of TGFbeta signaling, major extracellular matrix (ECM) components, ECM-crosslinking, and ECM-receptors such as integrins and discoidin domain receptors, ECM-crosslinking and methods for targeted delivery of small interfering RNA, antisense oligonucleotides and small molecules to increase potency and reduce side effects. With an increased understanding of the biology of the ECM and liver fibrosis and an improved preclinical validation, the translation of these approaches to the clinic is currently ongoing. Application to patients with liver fibrosis and a personalized treatment is tightly linked to the development of noninvasive biomarkers of fibrosis, fibrogenesis and fibrolysis.
Collapse
Affiliation(s)
- Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| | - Muhammad Ashfaq-Khan
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| | - Ai Ting Yang
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| | - Yong Ook Kim
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| |
Collapse
|
45
|
Gotts JE, Abbott J, Fang X, Yanagisawa H, Takasaka N, Nishimura SL, Calfee CS, Matthay MA. Cigarette Smoke Exposure Worsens Endotoxin-Induced Lung Injury and Pulmonary Edema in Mice. Nicotine Tob Res 2018; 19:1033-1039. [PMID: 28340238 DOI: 10.1093/ntr/ntx062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/07/2017] [Indexed: 01/19/2023]
Abstract
Introduction Cigarette smoking (CS) remains a major public health concern and has recently been associated with an increased risk of developing acute respiratory distress syndrome (ARDS). Bronchoalveolar lavage (BAL) experiments in human volunteers have demonstrated that active smokers develop increased alveolar-epithelial barrier permeability to protein after inhaling lipopolysaccharide (LPS). Here we tested the hypothesis that short-term whole-body CS exposure would increase LPS-induced lung edema in mice. Methods Adult mice were exposed in a Teague TE-10 machine to CS from 3R4F cigarettes at 100 mg/m3 total suspended particulates for 12 days, then given LPS or saline intratracheally. Control mice were housed in the same room without CS exposure. Post-mortem measurements included gravimetric lung water and BAL protein, cell counts, and lung histology. Cytokines were measured in lung homogenate by ELISA and in plasma by Luminex and ELISA. Results In CS-exposed mice, intratracheal LPS caused greater increases in pulmonary edema by gravimetric measurement and histologic scoring. CS-exposed mice also had an increase in BAL neutrophilia, lung IL-6, and plasma CXCL9, a T-cell chemoattractant. Intratracheal LPS concentrated blood hemoglobin to a greater degree in CS-exposed mice, consistent with an increase in systemic vascular permeability. Conclusions These results demonstrate that CS exposure in endotoxin injured mice increases the severity of acute lung injury. The increased lung IL-6 in CS-exposed LPS-injured mice indicates that this potent cytokine, previously shown to predict mortality in patients with ARDS, may play a role in exacerbating lung injury in smokers and may have utility as a biomarker of tobacco-related lung injury. Implications Our results suggest that short-term CS exposure at levels that cause no overt lung injury may still prime the lung for acute inflammatory damage from a "second hit", a finding that mirrors the increased risk of developing ARDS in patients who smoke. This model may be useful for evaluating the acute pulmonary toxicity of existing and/or novel tobacco products and identifying biomarkers of tobacco-related lung injury.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Jason Abbott
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Xiaohui Fang
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Haru Yanagisawa
- Department of Pathology, University of California, San Francisco, CA
| | - Naoki Takasaka
- Department of Pathology, University of California, San Francisco, CA
| | | | - Carolyn S Calfee
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| |
Collapse
|
46
|
Abstract
Activation of TGF-β1 initiates a program of temporary collagen accumulation important to wound repair in many organs. However, the outcome of temporary extracellular matrix strengthening all too frequently morphs into progressive fibrosis, contributing to morbidity and mortality worldwide. To avoid this maladaptive outcome, TGF-β1 signaling is regulated at numerous levels and intimately connected to feedback signals that limit accumulation. Here, we examine the current understanding of the core functions of TGF-β1 in promoting collagen accumulation, parallel pathways that promote physiological repair, and pathological triggers that tip the balance toward progressive fibrosis. Implicit in better understanding of these processes is the identification of therapeutic opportunities that will need to be further advanced to limit or reverse organ fibrosis.
Collapse
Affiliation(s)
- Kevin K Kim
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan 48109
| | - Dean Sheppard
- Department of Medicine, Cardiovascular Research Institute, and Lung Biology Center, University of California, San Francisco, San Francisco, California 94143
| | - Harold A Chapman
- Department of Medicine, Cardiovascular Research Institute, and Lung Biology Center, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
47
|
Gotts JE, Chun L, Abbott J, Fang X, Takasaka N, Nishimura SL, Springer ML, Schick SF, Calfee CS, Matthay MA. Cigarette smoke exposure worsens acute lung injury in antibiotic-treated bacterial pneumonia in mice. Am J Physiol Lung Cell Mol Physiol 2018. [PMID: 29543040 DOI: 10.1152/ajplung.00405.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Evidence is accumulating that exposure to cigarette smoke (CS) increases the risk of developing acute respiratory distress syndrome (ARDS). Streptococcus pneumoniae is the most common cause of bacterial pneumonia, which in turn is the leading cause of ARDS. Chronic smokers have increased rates of pneumococcal colonization and develop more severe pneumococcal pneumonia than nonsmokers; yet mechanistic connections between CS exposure, bacterial pneumonia, and ARDS pathogenesis remain relatively unexplored. We exposed mice to 3 wk of moderate whole body CS or air, followed by intranasal inoculation with an invasive serotype of S. pneumoniae. CS exposure alone caused no detectable lung injury or bronchoalveolar lavage (BAL) inflammation. During pneumococcal infection, CS-exposed mice had greater survival than air-exposed mice, in association with reduced systemic spread of bacteria from the lungs. However, when mice were treated with antibiotics after infection to improve clinical relevance, the survival benefit was lost, and CS-exposed mice had more pulmonary edema, increased numbers of BAL monocytes, and elevated monocyte and lymphocyte chemokines. CS-exposed antibiotic-treated mice also had higher serum surfactant protein D and angiopoietin-2, consistent with more severe lung epithelial and endothelial injury. The results indicate that acute CS exposure enhances the recruitment of immune cells to the lung during bacterial pneumonia, an effect that may provide microbiological benefit but simultaneously exposes the mice to more severe inflammatory lung injury. The inclusion of antibiotic treatment in preclinical studies of acute lung injury in bacterial pneumonia may enhance clinical relevance, particularly for future studies of current or emerging tobacco products.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Lauren Chun
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Jason Abbott
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Xiaohui Fang
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Naoki Takasaka
- Department of Pathology, University of California , San Francisco, California
| | - Stephen L Nishimura
- Department of Pathology, University of California , San Francisco, California
| | - Matthew L Springer
- Department of Medicine, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Suzaynn F Schick
- Department of Medicine, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Carolyn S Calfee
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| |
Collapse
|
48
|
Hatley RJD, Macdonald SJF, Slack RJ, Le J, Ludbrook SB, Lukey PT. An αv-RGD Integrin Inhibitor Toolbox: Drug Discovery Insight, Challenges and Opportunities. Angew Chem Int Ed Engl 2018; 57:3298-3321. [DOI: 10.1002/anie.201707948] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Richard J. D. Hatley
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Simon J. F. Macdonald
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Robert J. Slack
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Joelle Le
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Steven B. Ludbrook
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Pauline T. Lukey
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| |
Collapse
|
49
|
Hatley RJD, Macdonald SJF, Slack RJ, Le J, Ludbrook SB, Lukey PT. Ein Instrumentarium von αv-RGD-Integrin-Inhibitoren: Wirkstoffsuche, Herausforderungen und Möglichkeiten. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201707948] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Richard J. D. Hatley
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Simon J. F. Macdonald
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Robert J. Slack
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Joelle Le
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Steven B. Ludbrook
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Pauline T. Lukey
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| |
Collapse
|
50
|
High integrin α Vβ 6 affinity reached by hybrid domain deletion slows ligand-binding on-rate. Proc Natl Acad Sci U S A 2018; 115:E1429-E1436. [PMID: 29378937 DOI: 10.1073/pnas.1718662115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The role of the hybrid domain in integrin affinity regulation is unknown, as is whether the kinetics of ligand binding is modulated by integrin affinity state. Here, we compare cell surface and soluble integrin αVβ6 truncation mutants for ligand-binding affinity, kinetics, and thermodynamics. Removal of the integrin transmembrane/cytoplasmic domains or lower legs has little effect on αVβ6 affinity, in contrast to β1 integrins. In integrin opening, rearrangement at the interface between the βI and hybrid domains is linked to remodeling at the ligand-binding site at the opposite end of the βI domain, which greatly increases in affinity in the open conformation. The larger size of the βI-hybrid interface in the closed state suggests that the hybrid domain stabilizes closing. In agreement, deletion of the hybrid domain raised affinity by 50-fold. Surface plasmon resonance and isothermal titration calorimetry gave similar results and the latter revealed tradeoffs between enthalpy and entropy not apparent from affinity. At extremely high affinity reached in Mn2+ with hybrid domain truncation, αVβ6 on-rate for both pro-TGF-β1 and fibronectin declined. The results suggest that the open conformation of αVβ6 has lower on-rate than the closed conformation, correlate with constriction of the ligand-binding pocket in open αVβ6 structures, and suggest that the extended-closed conformation is kinetically selected for ligand binding. Subsequent transition to the extended-open conformation is stabilized by its much higher affinity for ligand and would also be stabilized by force exerted across ligand-bound integrins by the actin cytoskeleton.
Collapse
|