101
|
Meijer EM, van Dijk CGM, Kramann R, Verhaar MC, Cheng C. Implementation of Pericytes in Vascular Regeneration Strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1-21. [PMID: 33231500 DOI: 10.1089/ten.teb.2020.0229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For the survival and integration of complex large-sized tissue-engineered (TE) organ constructs that exceed the maximal nutrients and oxygen diffusion distance required for cell survival, graft (pre)vascularization to ensure medium or blood supply is crucial. To achieve this, the morphology and functionality of the microcapillary bed should be mimicked by incorporating vascular cell populations, including endothelium and mural cells. Pericytes play a crucial role in microvascular function, blood vessel stability, angiogenesis, and blood pressure regulation. In addition, tissue-specific pericytes are important in maintaining specific functions in different organs, including vitamin A storage in the liver, renin production in the kidneys and maintenance of the blood-brain-barrier. Together with their multipotential differentiation capacity, this makes pericytes the preferred cell type for application in TE grafts. The use of a tissue-specific pericyte cell population that matches the TE organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)-vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts. Impact statement The use of a tissue-specific pericyte cell population that matches the tissue-engineered (TE) organ may benefit organ function. In this review, we provide an overview of the literature for graft (pre)vascularization strategies and highlight the possible advantages of using tissue-specific pericytes for specific TE organ grafts.
Collapse
Affiliation(s)
- Elana M Meijer
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christian G M van Dijk
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rafael Kramann
- Division of Nephrology and Institute of Experimental Medicine and Systems Biology, University Hospital RWTH Aachen, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Experimental Cardiology, Department of Cardiology, Thorax Center Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
102
|
Pastor L, Vera E, Marin JM, Sanz-Rubio D. Extracellular Vesicles from Airway Secretions: New Insights in Lung Diseases. Int J Mol Sci 2021; 22:E583. [PMID: 33430153 PMCID: PMC7827453 DOI: 10.3390/ijms22020583] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Lung diseases (LD) are one of the most common causes of death worldwide. Although it is known that chronic airway inflammation and excessive tissue repair are processes associated with LD such as asthma, chronic obstructive pulmonary disease (COPD) or idiopathic pulmonary fibrosis (IPF), their specific pathways remain unclear. Extracellular vesicles (EVs) are heterogeneous nanoscale membrane vesicles with an important role in cell-to-cell communication. EVs are present in general biofluids as plasma or urine but also in secretions of the airway as bronchoalveolar lavage fluid (BALF), induced sputum (IS), nasal lavage (NL) or pharyngeal lavage. Alterations of airway EV cargo could be crucial for understanding LD. Airway EVs have shown a role in the pathogenesis of some LD such as eosinophil increase in asthma, the promotion of lung cancer in vitro models in COPD and as biomarkers to distinguishing IPF in patients with diffuse lung diseases. In addition, they also have a promising future as therapeutics for LD. In this review, we focus on the importance of airway secretions in LD, the pivotal role of EVs from those secretions on their pathophysiology and their potential for biomarker discovery.
Collapse
Affiliation(s)
- Laura Pastor
- Translational Research Unit, Instituto de Investigación Sanitaria de Aragón (IISAragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain; (L.P.); (E.V.); (J.M.M.)
| | - Elisabeth Vera
- Translational Research Unit, Instituto de Investigación Sanitaria de Aragón (IISAragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain; (L.P.); (E.V.); (J.M.M.)
- Respiratory Service, Hospital Universitario Miguel Servet, University of Zaragoza, 50009 Zaragoza, Spain
| | - Jose M. Marin
- Translational Research Unit, Instituto de Investigación Sanitaria de Aragón (IISAragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain; (L.P.); (E.V.); (J.M.M.)
- Respiratory Service, Hospital Universitario Miguel Servet, University of Zaragoza, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERes), 28029 Madrid, Spain
| | - David Sanz-Rubio
- Translational Research Unit, Instituto de Investigación Sanitaria de Aragón (IISAragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain; (L.P.); (E.V.); (J.M.M.)
| |
Collapse
|
103
|
Chow RD, Majety M, Chen S. The aging transcriptome and cellular landscape of the human lung in relation to SARS-CoV-2. Nat Commun 2021; 12:4. [PMID: 33397975 PMCID: PMC7782551 DOI: 10.1038/s41467-020-20323-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 11/24/2020] [Indexed: 01/29/2023] Open
Abstract
Age is a major risk factor for severe coronavirus disease-2019 (COVID-19). Here, we interrogate the transcriptional features and cellular landscape of the aging human lung. By intersecting these age-associated changes with experimental data on SARS-CoV-2, we identify several factors that may contribute to the heightened severity of COVID-19 in older populations. The aging lung is transcriptionally characterized by increased cell adhesion and stress responses, with reduced mitochondria and cellular replication. Deconvolution analysis reveals that the proportions of alveolar type 2 cells, proliferating basal cells, goblet cells, and proliferating natural killer/T cells decrease with age, whereas alveolar fibroblasts, pericytes, airway smooth muscle cells, endothelial cells and IGSF21+ dendritic cells increase with age. Several age-associated genes directly interact with the SARS-CoV-2 proteome. Age-associated genes are also dysregulated by SARS-CoV-2 infection in vitro and in patients with severe COVID-19. These analyses illuminate avenues for further studies on the relationship between age and COVID-19.
Collapse
Affiliation(s)
- Ryan D Chow
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Systems Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- M.D.-Ph.D. Program, Yale University, New Haven, CT, USA
| | - Medha Majety
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Systems Biology Institute, Yale University, West Haven, CT, USA
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
- The College, Yale University, New Haven, CT, 06520, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- Systems Biology Institute, Yale University, West Haven, CT, USA.
- Center for Cancer Systems Biology, Yale University, West Haven, CT, USA.
- M.D.-Ph.D. Program, Yale University, New Haven, CT, USA.
- Immunobiology Program, Yale University, New Haven, CT, USA.
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA.
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA.
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Liver Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
104
|
Summers ME, Richmond BW, Kropski JA, Majka SA, Bastarache JA, Hatzopoulos AK, Bylund J, Ghosh M, Petrache I, Foronjy RF, Geraghty P, Majka SM. Balanced Wnt/Dickkopf-1 signaling by mesenchymal vascular progenitor cells in the microvascular niche maintains distal lung structure and function. Am J Physiol Cell Physiol 2021; 320:C119-C131. [PMID: 33085496 PMCID: PMC7846975 DOI: 10.1152/ajpcell.00277.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 02/08/2023]
Abstract
The well-described Wnt inhibitor Dickkopf-1 (DKK1) plays a role in angiogenesis as well as in regulation of growth factor signaling cascades in pulmonary remodeling associated with chronic lung diseases (CLDs) including emphysema and fibrosis. However, the specific mechanisms by which DKK1 influences mesenchymal vascular progenitor cells (MVPCs), microvascular endothelial cells (MVECs), and smooth muscle cells (SMCs) within the microvascular niche have not been elucidated. In this study, we show that knockdown of DKK1 in Abcg2pos lung mouse adult tissue resident MVPCs alters lung stiffness, parenchymal collagen deposition, microvessel muscularization and density as well as loss of tissue structure in response to hypoxia exposure. To complement the in vivo mouse modeling, we also identified cell- or disease-specific responses to DKK1, in primary lung chronic obstructive pulmonary disease (COPD) MVPCs, COPD MVECs, and SMCs, supporting a paradoxical disease-specific response of cells to well-characterized factors. Cell responses to DKK1 were dose dependent and correlated with varying expressions of the DKK1 receptor, CKAP4. These data demonstrate that DKK1 expression is necessary to maintain the microvascular niche whereas its effects are context specific. They also highlight DKK1 as a regulatory candidate to understand the role of Wnt and DKK1 signaling between cells of the microvascular niche during tissue homeostasis and during the development of chronic lung diseases.
Collapse
Affiliation(s)
- Megan E Summers
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Bradley W Richmond
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Sarah A Majka
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Julie A Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Antonis K Hatzopoulos
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jeffery Bylund
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Moumita Ghosh
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Irina Petrache
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Robert F Foronjy
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York
| | - Patrick Geraghty
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York
| | - Susan M Majka
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
- Department of Medicine, Pulmonary & Critical Care Medicine, Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| |
Collapse
|
105
|
Skibba M, Drelich A, Poellmann M, Hong S, Brasier AR. Nanoapproaches to Modifying Epigenetics of Epithelial Mesenchymal Transition for Treatment of Pulmonary Fibrosis. Front Pharmacol 2020; 11:607689. [PMID: 33384604 PMCID: PMC7770469 DOI: 10.3389/fphar.2020.607689] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronically progressive interstitial lung that affects over 3 M people worldwide and rising in incidence. With a median survival of 2-3 years, IPF is consequently associated with high morbidity, mortality, and healthcare burden. Although two antifibrotic therapies, pirfenidone and nintedanib, are approved for human use, these agents reduce the rate of decline of pulmonary function but are not curative and do not reverse established fibrosis. In this review, we discuss the prevailing epithelial injury hypothesis, wherein pathogenic airway epithelial cell-state changes known as Epithelial Mesenchymal Transition (EMT) promotes the expansion of myofibroblast populations. Myofibroblasts are principal components of extracellular matrix production that result in airspace loss and mortality. We review the epigenetic transition driving EMT, a process produced by changes in histone acetylation regulating mesenchymal gene expression programs. This mechanistic work has focused on the central role of bromodomain-containing protein 4 in mediating EMT and myofibroblast transition and initial preclinical work has provided evidence of efficacy. As nanomedicine presents a promising approach to enhancing the efficacy of such anti-IPF agents, we then focus on the state of nanomedicine formulations for inhalable delivery in the treatment of pulmonary diseases, including liposomes, polymeric nanoparticles (NPs), inorganic NPs, and exosomes. These nanoscale agents potentially provide unique properties to existing pulmonary therapeutics, including controlled release, reduced systemic toxicity, and combination delivery. NP-based approaches for pulmonary delivery thus offer substantial promise to modify epigenetic regulators of EMT and advance treatments for IPF.
Collapse
Affiliation(s)
- Melissa Skibba
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Adam Drelich
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
| | - Michael Poellmann
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
| | - Seungpyo Hong
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Yonsei Frontier Lab, Department of Pharmacy, Yonsei University, Seoul, South Korea
| | - Allan R. Brasier
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
106
|
Szepes M, Melchert A, Dahlmann J, Hegermann J, Werlein C, Jonigk D, Haverich A, Martin U, Olmer R, Gruh I. Dual Function of iPSC-Derived Pericyte-Like Cells in Vascularization and Fibrosis-Related Cardiac Tissue Remodeling In Vitro. Int J Mol Sci 2020; 21:E8947. [PMID: 33255686 PMCID: PMC7728071 DOI: 10.3390/ijms21238947] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Myocardial interstitial fibrosis (MIF) is characterized by excessive extracellular matrix (ECM) deposition, increased myocardial stiffness, functional weakening, and compensatory cardiomyocyte (CM) hypertrophy. Fibroblasts (Fbs) are considered the principal source of ECM, but the contribution of perivascular cells, including pericytes (PCs), has gained attention, since MIF develops primarily around small vessels. The pathogenesis of MIF is difficult to study in humans because of the pleiotropy of mutually influencing pathomechanisms, unpredictable side effects, and the lack of available patient samples. Human pluripotent stem cells (hPSCs) offer the unique opportunity for the de novo formation of bioartificial cardiac tissue (BCT) using a variety of different cardiovascular cell types to model aspects of MIF pathogenesis in vitro. Here, we have optimized a protocol for the derivation of hPSC-derived PC-like cells (iPSC-PCs) and present a BCT in vitro model of MIF that shows their central influence on interstitial collagen deposition and myocardial tissue stiffening. This model was used to study the interplay of different cell types-i.e., hPSC-derived CMs, endothelial cells (ECs), and iPSC-PCs or primary Fbs, respectively. While iPSC-PCs improved the sarcomere structure and supported vascularization in a PC-like fashion, the functional and histological parameters of BCTs revealed EC- and PC-mediated effects on fibrosis-related cardiac tissue remodeling.
Collapse
Affiliation(s)
- Monika Szepes
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.M.); (J.D.); (A.H.); (U.M.); (R.O.)
- REBIRTH—Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany;
| | - Anna Melchert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.M.); (J.D.); (A.H.); (U.M.); (R.O.)
- REBIRTH—Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany;
| | - Julia Dahlmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.M.); (J.D.); (A.H.); (U.M.); (R.O.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, 30625 Hannover, Germany;
| | - Jan Hegermann
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, 30625 Hannover, Germany;
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, 30625 Hannover, Germany
| | | | - Danny Jonigk
- REBIRTH—Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany;
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, 30625 Hannover, Germany;
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany;
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.M.); (J.D.); (A.H.); (U.M.); (R.O.)
- REBIRTH—Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany;
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, 30625 Hannover, Germany;
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.M.); (J.D.); (A.H.); (U.M.); (R.O.)
- REBIRTH—Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany;
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, 30625 Hannover, Germany;
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.M.); (J.D.); (A.H.); (U.M.); (R.O.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, 30625 Hannover, Germany;
| | - Ina Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (M.S.); (A.M.); (J.D.); (A.H.); (U.M.); (R.O.)
- REBIRTH—Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
107
|
Sono T, Hsu CY, Negri S, Miller S, Wang Y, Xu J, Meyers CA, Peault B, James AW. Platelet-derived growth factor receptor-β (PDGFRβ) lineage tracing highlights perivascular cell to myofibroblast transdifferentiation during post-traumatic osteoarthritis. J Orthop Res 2020; 38:2484-2494. [PMID: 32134140 PMCID: PMC7483913 DOI: 10.1002/jor.24648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 02/04/2023]
Abstract
Pericytes ubiquitously surround capillaries and microvessels within vascularized tissues and have diverse functions after tissue injury. In addition to regulation of angiogenesis and tissue regeneration after injury, pericytes also contribute to organ fibrosis. Destabilization of the medial meniscus (DMM) phenocopies post-traumatic osteoarthritis, yet little is known regarding the impact of DMM surgery on knee joint-associated pericytes and their cellular descendants. Here, inducible platelet-derived growth factor receptor-β (PDGFRβ)-CreERT2 reporter mice were subjected to DMM surgery, and lineage tracing studies performed over an 8-week period. Results showed that at baseline PDGFRβ reporter activity highlights abluminal perivascular cells within synovial and infrapatellar fat pad (IFP) tissues. DMM induces a temporospatially patterned increase in vascular density within synovial and subsynovial tissues. Marked vasculogenesis within IFP was accompanied by expansion of PDGFRβ reporter+ perivascular cell numbers, detachment of mGFP+ descendants from vessel walls, and aberrant adoption of myofibroblastic markers among mGFP+ cells including α-SMA, ED-A, and TGF-β1. At later timepoints, fibrotic changes and vascular maturation occurred within subsynovial tissues, with the redistribution of PDGFRβ+ cellular descendants back to their perivascular niche. In sum, PDGFRβ lineage tracing allows for tracing of perivascular cell fate within the diarthrodial joint. Further, destabilization of the joint induces vascular and fibrogenic changes of the IFP accompanied by perivascular to myofibroblast transdifferentiation.
Collapse
Affiliation(s)
- Takashi Sono
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States.,Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606–8507, Japan
| | - Ching-Yun Hsu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Sarah Miller
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095, University of Edinburgh, Edinburgh, United Kingdom,Center For Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
108
|
Abstract
Abstract
Purpose of Review
In this brief review, we will highlight important observational and experimental data in the literature that address the origin of scar-forming cells in lung fibrosis.
Recent Findings
Several cellular sources of activated scar-forming cells (myofibroblasts) have been postulated including alveolar epithelial cells; circulating fibrocytes; and lung stromal cell subpopulations including resident fibroblasts, pericytes, and resident mesenchymal stem cells. Recent advances in lineage-tracing models, however, fail to provide experimental evidence for epithelial and fibrocyte origins of lung myofibroblasts. Resident mesenchymal cells of the lung, which include various cell types including resident fibroblasts, pericytes, and resident mesenchymal stem cells, appear to be important sources of myofibroblasts in murine models of lung injury and fibrosis.
Summary
Lung myofibroblasts likely originate from multiple sources of lung-resident mesenchymal cells. Their relative contributions may vary depending on the type of injury. Although lineage-tracing experiments have failed to show significant contribution from epithelial cells or fibrocytes, they may play important functional roles in myofibroblast activation through paracrine signaling.
Collapse
|
109
|
Abstract
INTRODUCTION Myofibroblasts are the primary executor and influencer in lung fibrosis. Latest studies on lung myofibroblast pathobiology have significantly advanced the understanding of the pathogenesis of lung fibrosis and shed new light on strategies targeting these cells to treat this disease. AREAS COVERED This article reviewed the most recent progresses, mainly within the last 5 years, on the definition, origin, activity regulation, and targeting of lung myofibroblasts in lung fibrosis. We did a literature search on PubMed using the keywords below from the dates 2010 to 2020. EXPERT OPINION With the improved cell lineage characterization and the advent of scRNA-seq, the field is having much better picture of the lung myofibroblast origin and mesenchymal heterogeneity. Additionally, cellular metabolism has emerged as a key regulation of lung myofibroblast pathogenic phenotype and is a promising therapeutic target for treating a variety of lung fibrotic disorders.
Collapse
Affiliation(s)
- Dingyuan Jiang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital; National Clinical Research Center for Respiratory Diseases , Beijing, China
| | - Tapan Dey
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama, USA
| |
Collapse
|
110
|
Rosa I, Romano E, Fioretto BS, Manetti M. The contribution of mesenchymal transitions to the pathogenesis of systemic sclerosis. Eur J Rheumatol 2020; 7:S157-S164. [PMID: 31922472 PMCID: PMC7647682 DOI: 10.5152/eurjrheum.2019.19081] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/25/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic sclerosis (SSc) is a multifaceted connective tissue disease characterized by widespread vasculopathy and autoimmune reactions that evolve into progressive interstitial, perivascular, and vessel wall fibrosis that affects the skin and multiple internal organs. Such an uncontrolled fibrotic process gradually disrupts the physiologic architecture of the affected tissues and frequently leads to significant organ dysfunction, thus representing a major cause of death in SSc patients. The main fibrosis orchestrators in SSc are represented by chronically activated myofibroblasts, a peculiar population of mesenchymal cells combining the extracellular matrix-synthesizing features of fibroblasts with cytoskeletal characteristics of contractile smooth muscle cells. Multiple lines of evidence support the notion that profibrotic myofibroblasts may derive not only from the activation of tissue resident fibroblasts but also from a variety of additional cell types, including pericytes, epithelial cells, vascular endothelial cells and preadipocytes/adipocytes. Here we overview an emerging picture that espouses that several cell transitional processes may be novel essential contributors to the pool of profibrotic myofibroblasts in SSc, potentially representing new suitable targets for therapeutic purposes. An in-depth dissection of the multiple origins of myofibroblasts and the underlying molecular mechanisms may be crucial in the process of deciphering the cellular bases of fibrosis persistence and refractoriness to the treatment and, therefore, may help in developing more effective and personalized therapeutic opportunities for SSc patients.
Collapse
Affiliation(s)
- Irene Rosa
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Mirko Manetti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
111
|
Ray HC, Corliss BA, Bruce AC, Kesting S, Dey P, Mansour J, Seaman SA, Smolko CM, Mathews C, Dey BK, Owens GK, Peirce SM, Yates PA. Myh11+ microvascular mural cells and derived mesenchymal stem cells promote retinal fibrosis. Sci Rep 2020; 10:15808. [PMID: 32978500 PMCID: PMC7519078 DOI: 10.1038/s41598-020-72875-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/10/2020] [Indexed: 12/29/2022] Open
Abstract
Retinal diseases are frequently characterized by the accumulation of excessive scar tissue found throughout the neural retina. However, the pathophysiology of retinal fibrosis remains poorly understood, and the cell types that contribute to the fibrotic response are incompletely defined. Here, we show that myofibroblast differentiation of mural cells contributes directly to retinal fibrosis. Using lineage tracing technology, we demonstrate that after chemical ocular injury, Myh11+ mural cells detach from the retinal microvasculature and differentiate into myofibroblasts to form an epiretinal membrane. Inhibition of TGFβR attenuates Myh11+ retinal mural cell myofibroblast differentiation, and diminishes the subsequent formation of scar tissue on the surface of the retina. We demonstrate retinal fibrosis within a murine model of oxygen-induced retinopathy resulting from the intravitreal injection of adipose Myh11-derived mesenchymal stem cells, with ensuing myofibroblast differentiation. In this model, inhibiting TGFβR signaling does not significantly alter myofibroblast differentiation and collagen secretion within the retina. This work shows the complexity of retinal fibrosis, where scar formation is regulated both by TGFβR and non-TGFβR dependent processes involving mural cells and derived mesenchymal stem cells. It also offers a cautionary note on the potential deleterious, pro-fibrotic effects of exogenous MSCs once intravitreally injected into clinical patients.
Collapse
Affiliation(s)
- H Clifton Ray
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Bruce A Corliss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Sam Kesting
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Paromita Dey
- The RNA Institute, University at Albany, State University of New York, Albany, NY, USA
| | - Jennifer Mansour
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Scott A Seaman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Christian M Smolko
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Corbin Mathews
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Bijan K Dey
- The RNA Institute, University at Albany, State University of New York, Albany, NY, USA
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Paul A Yates
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
- Department of Ophthalmology, University of Virginia, PO Box 800715, Charlottesville, VA, 22908, USA.
| |
Collapse
|
112
|
Stephenson SE, Wilson CL, Bond NG, Kaur A, Alvarez X, Midkiff CC, Schnapp LM. Pericytes as novel targets for HIV/SIV infection in the lung. Am J Physiol Lung Cell Mol Physiol 2020; 319:L848-L853. [PMID: 32901522 DOI: 10.1152/ajplung.00296.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antiretroviral therapy in HIV patients has lengthened lifespan but led to an increased risk for secondary comorbidities, such as pulmonary complications characterized by vascular dysfunction. In the lung, PDGFRβ+ mesenchymal cells known as pericytes intimately associate with endothelial cells and are key for their survival both structurally and through the secretion of prosurvival factors. We hypothesize that in HIV infection there are functional changes in pericytes that may lead to destabilization of the microvasculature and ultimately to pulmonary abnormalities. Our objective in this study was to determine whether lung pericytes could be directly infected with HIV. We leveraged lung samples from macaque lungs with or without SIV infection and normal human lung for in vitro experiments. Pericytes were isolated based on the marker platelet-derived growth factor receptor-β (PDGFRβ). We determined that lung PDGFRβ-positive (PDGFRβ+) pericytes from both macaques and humans express CD4, the primary receptor for SIV/HIV, as well as the major coreceptors CXCR4 and CCR5. We found cells positive for both PDGFRβ and SIV in lungs from infected macaques. Lung pericytes isolated from these animals also harbored detectable SIV. To confirm relevance to human disease, we demonstrated that human lung pericytes are capable of being productively infected by HIV in vitro, with the time course of infection suggesting development of viral latency. In summary, we show for the first time that SIV/HIV directly infects lung pericytes, implicating these cells as a novel target and potential reservoir for the virus in vivo.
Collapse
Affiliation(s)
- Sarah E Stephenson
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Carole L Wilson
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Nell G Bond
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana
| | - Xavier Alvarez
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana
| | - Cecily C Midkiff
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana
| | - Lynn M Schnapp
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
113
|
Riccetti M, Gokey JJ, Aronow B, Perl AKT. The elephant in the lung: Integrating lineage-tracing, molecular markers, and single cell sequencing data to identify distinct fibroblast populations during lung development and regeneration. Matrix Biol 2020; 91-92:51-74. [PMID: 32442602 PMCID: PMC7434667 DOI: 10.1016/j.matbio.2020.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022]
Abstract
During lung development, the mesenchyme and epithelium are dependent on each other for instructive morphogenic cues that direct proliferation, cellular differentiation and organogenesis. Specification of epithelial and mesenchymal cell lineages occurs in parallel, forming cellular subtypes that guide the formation of both transitional developmental structures and the permanent architecture of the adult lung. While epithelial cell types and lineages have been relatively well-defined in recent years, the definition of mesenchymal cell types and lineage relationships has been more challenging. Transgenic mouse lines with permanent and inducible lineage tracers have been instrumental in identifying lineage relationships among epithelial progenitor cells and their differentiation into distinct airway and alveolar epithelial cells. Lineage tracing experiments with reporter mice used to identify fibroblast progenitors and their lineage trajectories have been limited by the number of cell specific genes and the developmental timepoint when the lineage trace was activated. In this review, we discuss major developmental mesenchymal lineages, focusing on time of origin, major cell type, and other lineage derivatives, as well as the transgenic tools used to find and define them. We describe lung fibroblasts using function, location, and molecular markers in order to compare and contrast cells with similar functions. The temporal and cell-type specific expression of fourteen "fibroblast lineage" genes were identified in single-cell RNA-sequencing data from LungMAP in the LGEA database. Using these lineage signature genes as guides, we clustered murine lung fibroblast populations from embryonic day 16.5 to postnatal day 28 (E16.5-PN28) and generated heatmaps to illustrate expression of transcription factors, signaling receptors and ligands in a temporal and population specific manner.
Collapse
Affiliation(s)
- Matthew Riccetti
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Jason J Gokey
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Bruce Aronow
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States
| | - Anne-Karina T Perl
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
114
|
Zhang X, Chen Q, Song H, Jiang W, Xie S, Huang J, Kang G. MicroRNA‑375 prevents TGF‑β‑dependent transdifferentiation of lung fibroblasts via the MAP2K6/P38 pathway. Mol Med Rep 2020; 22:1803-1810. [PMID: 32582987 PMCID: PMC7411355 DOI: 10.3892/mmr.2020.11261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
Transdifferentiation of lung fibroblasts to myofibroblasts is a crucial pathophysiological process in pulmonary fibrosis. MicroRNA‑375 (miR‑375) was initially identified as a tumor‑suppressive factor, and its expression was negatively associated with the severity of lung cancer; however, its role and potential mechanism in myofibroblast transdifferentiation and pulmonary fibrosis remain unclear. In the present study, human lung fibroblasts were stimulated with transforming growth factor‑β (TGF‑β) to induce myofibroblast transdifferentiation. A mimic and inhibitor of miR‑375, and their negative controls, were used to overexpress or suppress miR‑375 in lung fibroblasts, respectively. The mRNA expression levels of fibrotic markers, and protein expression of α‑smooth muscle actin and periostin, were subsequently detected by reverse transcription‑quantitative PCR and western blotting, to assess myofibroblast transdifferentiation. miR‑375 was markedly upregulated in human lung fibroblasts after TGF‑β stimulation. The miR‑375 mimic alleviated, whereas the miR‑375 inhibitor aggravated TGF‑β‑dependent transdifferentiation of lung fibroblasts. Mechanistically, miR‑375 prevented myofibroblast transdifferentiation and collagen synthesis by blocking the P38 mitogen‑activated protein kinases (P38) pathway, and P38 suppression abrogated the deleterious effect of the miR‑375 inhibitor on myofibroblast transdifferentiation. Furthermore, the present study revealed that mitogen‑activated protein kinase kinase 6 was involved in P38 inactivation by miR‑375. In conclusion, miR‑375 was implicated in modulating TGF‑β‑dependent transdifferentiation of lung fibroblasts, and targeting miR‑375 expression may help to develop therapeutic approaches for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Xinghua Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Chen
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hengya Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Huang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ganjun Kang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
115
|
Koliaraki V, Prados A, Armaka M, Kollias G. The mesenchymal context in inflammation, immunity and cancer. Nat Immunol 2020; 21:974-982. [PMID: 32747813 DOI: 10.1038/s41590-020-0741-2] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022]
Abstract
Mesenchymal cells are mesoderm-derived stromal cells that are best known for providing structural support to organs, synthesizing and remodeling the extracellular matrix (ECM) and regulating development, homeostasis and repair of tissues. Recent detailed mechanistic insights into the biology of fibroblastic mesenchymal cells have revealed they are also significantly involved in immune regulation, stem cell maintenance and blood vessel function. It is now becoming evident that these functions, when defective, drive the development of complex diseases, such as various immunopathologies, chronic inflammatory disease, tissue fibrosis and cancer. Here, we provide a concise overview of the contextual contribution of fibroblastic mesenchymal cells in physiology and disease and bring into focus emerging evidence for both their heterogeneity at the single-cell level and their tissue-specific, spatiotemporal functional diversity.
Collapse
Affiliation(s)
- Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Marietta Armaka
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
116
|
Crosstalk between pleural mesothelial cell and lung fibroblast contributes to pulmonary fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118806. [PMID: 32739525 DOI: 10.1016/j.bbamcr.2020.118806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a specific form of chronic, progressive and fibrosing interstitial pneumonia of unknown cause. The main feature of IPF is a heterogeneous appearance with areas of sub-pleural fibrosis. However, the mechanism of sub-pleural fibrosis was poorly understood. In this study, our in vivo study revealed that pleural mesothelial cells (PMCs) migrated into lung parenchyma and localized alongside lung fibroblasts in sub-pleural area in mouse pulmonary fibrosis. Our in vitro study displayed that cultured-PMCs-medium induced lung fibroblasts transforming into myofibroblast, cultured-fibroblasts-medium promoted mesothelial-mesenchymal transition of PMCs. Furthermore, these changes in lung fibroblasts and PMCs were prevented by blocking TGF-β1/Smad2/3 signaling with SB431542. TGF-β1 neutralized antibody attenuated bleomycin-induced pulmonary fibrosis. Similar to TGF-β1/Smad2/3 signaling, wnt/β-catenin signaling was also activated in the process of PMCs crosstalk with lung fibroblasts. Moreover, inhibition of CD147 attenuated cultured-PMCs-medium induced collagen-I synthesis in lung fibroblasts. Blocking CD147 signaling also prevented bleomycin-induced pulmonary fibrosis. Our data indicated that crosstalk between PMC and lung fibroblast contributed to sub-pleural pulmonary fibrosis. TGF-β1, Wnt/β-catenin and CD147 signaling was involved in the underling mechanism.
Collapse
|
117
|
Santos DM, Pantano L, Pronzati G, Grasberger P, Probst CK, Black KE, Spinney JJ, Hariri LP, Nichols R, Lin Y, Bieler M, Seither P, Nicklin P, Wyatt D, Tager AM, Medoff BD. Screening for YAP Inhibitors Identifies Statins as Modulators of Fibrosis. Am J Respir Cell Mol Biol 2020; 62:479-492. [PMID: 31944822 DOI: 10.1165/rcmb.2019-0296oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a lung disease with limited therapeutic options that is characterized by pathological fibroblast activation and aberrant lung remodeling with scar formation. YAP (Yes-associated protein) is a transcriptional coactivator that mediates mechanical and biochemical signals controlling fibroblast activation. In this study, we developed a high-throughput small-molecule screen for YAP inhibitors in primary human lung fibroblasts. Multiple HMG-CoA (hydroxymethylglutaryl-coenzyme A) reductase inhibitors (statins) were found to inhibit YAP nuclear localization via induction of YAP phosphorylation, cytoplasmic retention, and degradation. We further show that the mevalonate pathway regulates YAP activation, and that simvastatin treatment reduces fibrosis markers in activated human lung fibroblasts and in the bleomycin mouse model of pulmonary fibrosis. Finally, we show that simvastatin modulates YAP in vivo in mouse lung fibroblasts. Our results highlight the potential of small-molecule screens for YAP inhibitors and provide a mechanism for the antifibrotic activity of statins in idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
| | - Lorena Pantano
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Gina Pronzati
- Division of Pulmonary and Critical Care Medicine, and
| | | | | | | | | | - Lida P Hariri
- Division of Pulmonary and Critical Care Medicine, and.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Yufei Lin
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | | | | | | | - David Wyatt
- Biotherapeutics Discovery, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | | | | |
Collapse
|
118
|
Deng Z, Fear MW, Suk Choi Y, Wood FM, Allahham A, Mutsaers SE, Prêle CM. The extracellular matrix and mechanotransduction in pulmonary fibrosis. Int J Biochem Cell Biol 2020; 126:105802. [PMID: 32668329 DOI: 10.1016/j.biocel.2020.105802] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary fibrosis is characterised by excessive scarring in the lung which leads to compromised lung function, serious breathing problems and in some diseases, death. It includes several lung disorders with idiopathic pulmonary fibrosis (IPF) the most common and most severe. Pulmonary fibrosis is considered to be perpetuated by aberrant wound healing which leads to fibroblast accumulation, differentiation and activation, and deposition of excessive amounts of extracellular matrix (ECM) components, in particular, collagen. Recent studies have identified the importance of changes in the composition and structure of lung ECM during the development of pulmonary fibrosis and the interaction between ECM and lung cells. There is strong evidence that increased matrix stiffness induces changes in cell function including proliferation, migration, differentiation and activation. Understanding how changes in the ECM microenvironment influence cell behaviour during fibrogenesis, and the mechanisms regulating these changes, will provide insight for developing new treatments.
Collapse
Affiliation(s)
- Zhenjun Deng
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Institute for Respiratory Health, Nedlands, WA, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Burns Service of Western Australia, Perth Children's Hospital, Nedlands, WA, Australia; Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Amira Allahham
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Steven E Mutsaers
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
119
|
Abstract
Influenza A virus (IAV) causes annual epidemics and sporadic pandemics of respiratory disease. Secondary bacterial coinfection by organisms such as Staphylococcus aureus is the most common complication of primary IAV infection and is associated with high levels of morbidity and mortality. Here, we report the first identified S. aureus factor (lipase 1) that enhances IAV replication during infection via positive modulation of virus budding. The effect is observed in vivo in embryonated hen’s eggs and greatly enhances the yield of a vaccine strain, a finding that could be applied to address global shortages of influenza vaccines. Influenza A virus (IAV) causes annual epidemics of respiratory disease in humans, often complicated by secondary coinfection with bacterial pathogens such as Staphylococcus aureus. Here, we report that the S. aureus secreted protein lipase 1 enhances IAV replication in vitro in primary cells, including human lung fibroblasts. The proviral activity of lipase 1 is dependent on its enzymatic function, acts late in the viral life cycle, and results in increased infectivity through positive modulation of virus budding. Furthermore, the proviral effect of lipase 1 on IAV is exhibited during in vivo infection of embryonated hen’s eggs and, importantly, increases the yield of a vaccine strain of IAV by approximately 5-fold. Thus, we have identified the first S. aureus protein to enhance IAV replication, suggesting a potential role in coinfection. Importantly, this activity may be harnessed to address global shortages of influenza vaccines.
Collapse
|
120
|
Pericyte-myofibroblast transition in the human lung. Biochem Biophys Res Commun 2020; 528:269-275. [DOI: 10.1016/j.bbrc.2020.05.091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022]
|
121
|
Yamasaki Y, Kuwana M. Nintedanib for the treatment of systemic sclerosis-associated interstitial lung disease. Expert Rev Clin Immunol 2020; 16:547-560. [DOI: 10.1080/1744666x.2020.1777857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Yoshioki Yamasaki
- Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
122
|
Perivascular Fibro-Adipogenic Progenitor Tracing during Post-Traumatic Osteoarthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1909-1920. [PMID: 32533926 DOI: 10.1016/j.ajpath.2020.05.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/03/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
Perivascular mural cells surround capillaries and microvessels and have diverse regenerative or fibrotic functions after tissue injury. Subsynovial fibrosis is a well-known pathologic feature of osteoarthritis, yet transgenic animals for use in visualizing perivascular cell contribution to fibrosis during arthritic changes have not been developed. Here, inducible Pdgfra-CreERT2 reporter mice were subjected to joint-destabilization surgery to induce arthritic changes, and cell lineage was traced over an 8-week period with a focus on the joint-associated fat pad. Results showed that, at baseline, inducible Pdgfra reporter activity highlighted adventitial and, to a lesser extent, pericytic cells within the infrapatellar fat pad. Joint-destabilization surgery was associated with marked fibrosis of the infrapatellar fat pad, accompanied by an expansion of perivascular Pdgfra-expressing cellular descendants, many of which adopted α-smooth muscle actin expression. Gene expression analysis of microdissected infrapatellar fat pad confirmed enrichment in membrane-bound green fluorescent protein/Pdgfra-expressing cells, along with a gene signature that corresponded with injury-associated fibro-adipogenic progenitors. Our results highlight dynamic changes in joint-associated perivascular fibro-adipogenic progenitors during osteoarthritis.
Collapse
|
123
|
Ulrich H, Pillat MM. CD147 as a Target for COVID-19 Treatment: Suggested Effects of Azithromycin and Stem Cell Engagement. Stem Cell Rev Rep 2020; 16:434-440. [PMID: 32307653 PMCID: PMC7167302 DOI: 10.1007/s12015-020-09976-7] [Citation(s) in RCA: 288] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The expressive number of deaths and confirmed cases of SARS-CoV-2 call for an urgent demand of effective and available drugs for COVID-19 treatment. CD147, a receptor on host cells, is a novel route for SARS-CoV-2 invasion. Thus, drugs that interfere in the spike protein/CD147 interaction or CD147 expression may inhibit viral invasion and dissemination among other cells, including in progenitor/stem cells. Studies suggest beneficial effects of azithromycin in reducing viral load of hospitalized patients, possibly interfering with ligand/CD147 receptor interactions; however, its possible effects on SARS-CoV-2 invasion has not yet been evaluated. In addition to the possible effect in invasion, azithromycin decreases the expression of some metalloproteinases (downstream to CD147), induces anti-viral responses in primary human bronchial epithelial infected with rhinovirus, decreasing viral replication and release. Moreover, resident lung progenitor/stem are extensively differentiated into myofibroblasts during pulmonary fibrosis, a complication observed in COVID-19 patients. This process, and the possible direct viral invasion of progenitor/stem cells via CD147 or ACE2, could result in the decline of these cellular stocks and failing lung repair. Clinical tests with allogeneic MSCs from healthy individuals are underway to enhance endogenous lung repair and suppress inflammation.
Collapse
Affiliation(s)
- Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, São Paulo, Brazil
| | - Micheli M Pillat
- Department of Microbiology and Parasitology, Health Sciences Center, Federal University of Santa Maria-RS, Avenue Roraima n° 1000, Santa Maria, RS, 97105900, Brazil.
| |
Collapse
|
124
|
Cellular Senescence in the Lung: The Central Role of Senescent Epithelial Cells. Int J Mol Sci 2020; 21:ijms21093279. [PMID: 32384619 PMCID: PMC7247355 DOI: 10.3390/ijms21093279] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Cellular senescence is a key process in physiological dysfunction developing upon aging or following diverse stressors including ionizing radiation. It describes the state of a permanent cell cycle arrest, in which proliferating cells become resistant to growth-stimulating factors. Senescent cells differ from quiescent cells, which can re-enter the cell cycle and from finally differentiated cells: morphological and metabolic changes, restructuring of chromatin, changes in gene expressions and the appropriation of an inflammation-promoting phenotype, called the senescence-associated secretory phenotype (SASP), characterize cellular senescence. The biological role of senescence is complex, since both protective and harmful effects have been described for senescent cells. While initially described as a mechanism to avoid malignant transformation of damaged cells, senescence can even contribute to many age-related diseases, including cancer, tissue degeneration, and inflammatory diseases, particularly when senescent cells persist in damaged tissues. Due to overwhelming evidence about the important contribution of cellular senescence to the pathogenesis of different lung diseases, specific targeting of senescent cells or of pathology-promoting SASP factors has been suggested as a potential therapeutic approach. In this review, we summarize recent advances regarding the role of cellular (fibroblastic, endothelial, and epithelial) senescence in lung pathologies, with a focus on radiation-induced senescence. Among the different cells here, a central role of epithelial senescence is suggested.
Collapse
|
125
|
Wang Z, Qu S, Zhu J, Chen F, Ma L. Comprehensive analysis of lncRNA-associated competing endogenous RNA network and immune infiltration in idiopathic pulmonary fibrosis. J Thorac Dis 2020; 12:1856-1865. [PMID: 32642089 PMCID: PMC7330328 DOI: 10.21037/jtd-19-2842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a life-threatening lung disorder with an unknown aetiology. The roles of long non-coding RNAs (lncRNAs) and its related competing endogenous RNAs (ceRNA) network in IPF remains poorly understood. In this study, we aimed to build a lncRNA-miRNA-mRNA network and explore the pathogenesis of IPF. Methods We screened differentially expressed lncRNAs (DElncRNAs) and mRNAs (DEmRNAs) between IPF and control lung tissues from two datasets. The ceRNA network was built according to the interactions between DElncRNA, miRNA, and DEmRNA. Functional enrichment analysis of DemRNAs was performed using Metascape. CIBERSORT (Cell type Identification by Estimating Relative Subsets Of known RNA Transcripts) was applied to estimate the fraction of 22 immune cells in IPF and controls lung tissue samples. Then we investigated the correlation between immune cells and clinical traits. Results We constructed a lncRNA-miRNA-mRNA network, which was composed of two DElncRNAs, 18 miRNAs, 66 DemRNAs. Functional enrichment analysis showed that the DEmRNAs mainly participated in MicroRNAs in cancer. By applying CIBERSORT, we found that IPF tissue samples had a higher proportion of plasma cells, resting mast cells and a lower proportion of resting NK cells, monocytes, neutrophils compared with control tissue samples. Also, our results indicated that immune cells were associated with the severity of IPF. Conclusions In summary, this is the first study to build lncRNA-miRNA-mRNA ceRNA network of IPF, which may improve our understanding of IPF pathogenesis. Our study indicates that immune cells in lung tissues may predict disease severity and participate in the development of IPF. Future prospective studies are required to confirm the findings of the current study.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Infectious Diseases, Shandong University Qilu Hospital, Jinan 250012, China
| | - Shuoying Qu
- Department of Clinical Laboratory, Shandong University Qilu Hospital, Jinan 250012, China
| | - Jie Zhu
- Department of Infectious Diseases, Shandong University Qilu Hospital, Jinan 250012, China
| | - Fengzhe Chen
- Department of Infectious Diseases, Shandong University Qilu Hospital, Jinan 250012, China
| | - Lixian Ma
- Department of Infectious Diseases, Shandong University Qilu Hospital, Jinan 250012, China
| |
Collapse
|
126
|
Falke LL, He N, Chuva de Sousa Lopes SM, Broekhuizen R, Lyons K, Nguyen TQ, Goldschmeding R. Correction to: FoxD1-driven CCN2 deletion causes axial skeletal deformities, pulmonary hypoplasia, and neonatal asphyctic death. J Cell Commun Signal 2020; 14:47-52. [PMID: 32291573 DOI: 10.1007/s12079-020-00559-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pulmonary fibrosis is a severely disabling disease often leading to death. CCN2 (Cellular Communication Network factor 2, also known as CTGF) is a known mediator of fibrosis and clinical trials studying anti-CCN2 efficacy in pulmonary fibrosis are currently underway. Fork head box D1 (FoxD1) transcription factor is transiently expressed in several mesenchymal cell types, including those of fetal lungs. Differentiation of FoxD1-progenitor derived pericytes into myofibroblasts involves CCN2 expression and contributes importantly to maladaptive tissue remodeling in for example kidney and lung fibrosis models. To generate a model for studying the contribution of CCN2 expression in FoxD1-progenitor derived cells to development of fibrotic tissue remodeling, we set out to establish a FoxD1Cre - CCN2flox/flox mouse colony. However, all double-transgenic mice died soon after birth due to asphyxia. Histopathological examination revealed a reduction in alveolar space and lung weight, and subtle axial (thoracic and cervical) skeletal deformities. Together with the previously reported association of a FoxD1 containing locus with human adolescent idiopathic scoliosis, our data suggest that the fatal pulmonary hypoplasia resulting from selective deletion of CCN2 from FoxD1-progenitor derived mesenchymal cells developed secondary to impaired breathing movements due to aberrant axial skeletogenesis.
Collapse
Affiliation(s)
- Lucas L Falke
- Department. Pathology, University Medical Centre Utrecht, Heidelberglaan, 100 3584 CX, Utrecht, The Netherlands.,Department Internal medicine, Diakonessenhuis, Utrecht, the Netherlands
| | - Nannan He
- Department Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Roel Broekhuizen
- Department. Pathology, University Medical Centre Utrecht, Heidelberglaan, 100 3584 CX, Utrecht, The Netherlands
| | - Karen Lyons
- Department Orthopedic Surgery, University of California, Oakland, CA, USA.,Department Molecular Cell and Developmental Biology, University of California, Oakland, CA, USA
| | - Tri Q Nguyen
- Department. Pathology, University Medical Centre Utrecht, Heidelberglaan, 100 3584 CX, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Department. Pathology, University Medical Centre Utrecht, Heidelberglaan, 100 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
127
|
The Vascular Involvement in Soft Tissue Fibrosis-Lessons Learned from Pathological Scarring. Int J Mol Sci 2020; 21:ijms21072542. [PMID: 32268503 PMCID: PMC7177855 DOI: 10.3390/ijms21072542] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Soft tissue fibrosis in important organs such as the heart, liver, lung, and kidney is a serious pathological process that is characterized by excessive connective tissue deposition. It is the result of chronic but progressive accumulation of fibroblasts and their production of extracellular matrix components such as collagens. Research on pathological scars, namely, hypertrophic scars and keloids, may provide important clues about the mechanisms that drive soft tissue fibrosis, in particular the vascular involvement. This is because these dermal fibrotic lesions bear all of the fibrotic characteristics seen in soft tissue fibrosis. Moreover, their location on the skin surface means they are readily observable and directly treatable and therefore more accessible to research. We will focus here on the roles that blood vessel-associated cells play in cutaneous scar pathology and assess from the literature whether these cells also contribute to other soft tissue fibroses. These cells include endothelial cells, which not only exhibit aberrant functions but also differentiate into mesenchymal cells in pathological scars. They also include pericytes, hepatic stellate cells, fibrocytes, and myofibroblasts. This article will review with broad strokes the roles that these cells play in the pathophysiology of different soft tissue fibroses. We hope that this brief but wide-ranging overview of the vascular involvement in fibrosis pathophysiology will aid research into the mechanisms underlying fibrosis and that this will eventually lead to the development of interventions that can prevent, reduce, or even reverse fibrosis formation and/or progression.
Collapse
|
128
|
Steen EH, Wang X, Balaji S, Butte MJ, Bollyky PL, Keswani SG. The Role of the Anti-Inflammatory Cytokine Interleukin-10 in Tissue Fibrosis. Adv Wound Care (New Rochelle) 2020; 9:184-198. [PMID: 32117582 PMCID: PMC7047112 DOI: 10.1089/wound.2019.1032] [Citation(s) in RCA: 250] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023] Open
Abstract
Significance: Fibrosis is the endpoint of chronic disease in multiple organs, including the skin, heart, lungs, intestine, liver, and kidneys. Pathologic accumulation of fibrotic tissue results in a loss of structural integrity and function, with resultant increases in morbidity and mortality. Understanding the pathways governing fibrosis and identifying therapeutic targets within those pathways is necessary to develop novel antifibrotic therapies for fibrotic disease. Recent Advances: Given the connection between inflammation and fibrogenesis, Interleukin-10 (IL-10) has been a focus of potential antifibrotic therapies because of its well-known role as an anti-inflammatory mediator. Despite the apparent dissimilarity of diseases associated with fibrotic progression, pathways involving IL-10 appear to be a conserved molecular theme. More recently, many groups have worked to develop novel delivery tools for recombinant IL-10, such as hydrogels, and cell-based therapies, such as ex vivo activated macrophages, to directly or indirectly modulate IL-10 signaling. Critical Issues: Some efforts in this area, however, have been stymied by IL-10's pleiotropic and sometimes conflicting effects. A deeper, contextual understanding of IL-10 signaling and its interaction with effector cells, particularly immune cells, will be critical to future studies in the field. Future Directions: IL-10 is clearly a gatekeeper of fibrotic/antifibrotic signaling. The development of novel therapeutics and cell-based therapies that capitalize on targets within the IL-10 signaling pathway could have far-reaching implications for patients suffering from the consequences of organ fibrosis.
Collapse
Affiliation(s)
- Emily H. Steen
- Department of Surgery, Baylor College of Medicine, Houston, Texas
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, Texas
| | - Xinyi Wang
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, Texas
| | - Swathi Balaji
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, Texas
| | - Manish J. Butte
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, California
| | - Paul L. Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Sundeep G. Keswani
- Department of Surgery, Baylor College of Medicine, Houston, Texas
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, Texas
- Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital, Houston, Texas
| |
Collapse
|
129
|
Kopf KW, Harral JW, Staker EA, Summers ME, Petrache I, Kheyfets V, Irwin DC, Majka SM. Optimization of combined measures of airway physiology and cardiovascular hemodynamics in mice. Pulm Circ 2020; 10:2045894020912937. [PMID: 32206308 PMCID: PMC7074541 DOI: 10.1177/2045894020912937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
Pulmonary hypertension may arise as a complication of chronic lung disease typically associated with tissue hypoxia, as well as infectious agents or injury eliciting a type 2 immune response. The onset of pulmonary hypertension in this setting (classified as Group 3) often complicates treatment and worsens prognosis of chronic lung disease. Chronic lung diseases such as chronic obstructive lung disease (COPD), emphysema, and interstitial lung fibrosis impair airflow and alter lung elastance in addition to affecting pulmonary vascular hemodynamics that may culminate in right ventricle dysfunction. To date, functional endpoints in murine models of chronic lung disease have typically been limited to separately measuring airway and lung parenchyma physiology. These approaches may be lengthy and require a large number of animals per experiment. Here, we provide a detailed protocol for combined assessment of airway physiology with cardiovascular hemodynamics in mice. Ultimately, a comprehensive overview of pulmonary function in murine models of injury and disease will facilitate the integration of studies of the airway and vascular biology necessary to understand underlying pathophysiology of Group 3 pulmonary hypertension.
Collapse
Affiliation(s)
- Katrina W Kopf
- Biological Resource Center, National Jewish Health, Denver, USA
| | - Julie W Harral
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, USA
| | - Emily A Staker
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, USA
| | - Megan E Summers
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, USA
| | - Vitaly Kheyfets
- Department of Bioengineering, Anschutz Medical Campus University of Colorado, Aurora, USA
| | - David C Irwin
- Department of Medicine, Division of Cardiology, Anschutz Medical Campus University of Colorado, Aurora, USA
| | - Susan M Majka
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, USA.,Department of Biomedical Research, National Jewish Health, Denver, USA.,Gates Center for Regenerative Medicine and Stem Cell Biology and Cardiology University of Colorado Medical Center, Aurora, USA
| |
Collapse
|
130
|
Nwadozi E, Rudnicki M, Haas TL. Metabolic Coordination of Pericyte Phenotypes: Therapeutic Implications. Front Cell Dev Biol 2020; 8:77. [PMID: 32117997 PMCID: PMC7033550 DOI: 10.3389/fcell.2020.00077] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Pericytes are mural vascular cells found predominantly on the abluminal wall of capillaries, where they contribute to the maintenance of capillary structural integrity and vascular permeability. Generally quiescent cells in the adult, pericyte activation and proliferation occur during both physiological and pathological vascular and tissue remodeling. A considerable body of research indicates that pericytes possess attributes of a multipotent adult stem cell, as they are capable of self-renewal as well as commitment and differentiation into multiple lineages. However, pericytes also display phenotypic heterogeneity and recent studies indicate that lineage potential differs between pericyte subpopulations. While numerous microenvironmental cues and cell signaling pathways are known to regulate pericyte functions, the roles that metabolic pathways play in pericyte quiescence, self-renewal or differentiation have been given limited consideration to date. This review will summarize existing data regarding pericyte metabolism and will discuss the coupling of signal pathways to shifts in metabolic pathway preferences that ultimately regulate pericyte quiescence, self-renewal and trans-differentiation. The association between dysregulated metabolic processes and development of pericyte pathologies will be highlighted. Despite ongoing debate regarding pericyte classification and their functional capacity for trans-differentiation in vivo, pericytes are increasingly exploited as a cell therapy tool to promote tissue healing and regeneration. Ultimately, the efficacy of therapeutic approaches hinges on the capacity to effectively control/optimize the fate of the implanted pericytes. Thus, we will identify knowledge gaps that need to be addressed to more effectively harness the opportunity for therapeutic manipulation of pericytes to control pathological outcomes in tissue remodeling.
Collapse
Affiliation(s)
| | | | - Tara L. Haas
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
131
|
Falke LL, He N, Chuva de Sousa Lopes SM, Broekhuizen R, Lyons K, Nguyen TQ, Goldschmeding R. FoxD1-driven CCN2 deletion causes axial skeletal deformities, pulmonary hypoplasia, and neonatal asphyctic death. J Cell Commun Signal 2020; 13:573-577. [PMID: 32020419 DOI: 10.1007/s12079-020-00549-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
Pulmonary fibrosis is a severely disabling disease often leading to death. CCN2 (Cellular Communication Network factor 2, also known as CTGF) is a known mediator of fibrosis and clinical trials studying anti-CCN2 efficacy in pulmonary fibrosis are currently underway. Fork head box D1 (FoxD1) transcription factor is transiently expressed in several mesenchymal cell types, including those of fetal lungs. Differentiation of FoxD1-progenitor derived pericytes into myofibroblasts involves CCN2 expression and contributes importantly to maladaptive tissue remodeling in e.g. kidney and lung fibrosis models. To generate a model for studying the contribution of CCN2 expression in FoxD1-progenitor derived cells to development of fibrotic tissue remodeling, we set out to establish a FoxD1Cre - CCN2flox/flox mouse colony. However, all double-transgenic mice died soon after birth due to asphyxia. Histopathological examination revealed a reduction in alveolar space and lung weight, and subtle axial (thoracic and cervical) skeletal deformities. Together with the previously reported association of a FoxD1 containing locus with human adolescent idiopathic scoliosis, our data suggest that the development of fatal pulmonary hypoplasia caused by selective deletion of CCN2 from FoxD1-progenitor derived mesenchymal cells was secondary to aberrant axial skeletogenesis.
Collapse
Affiliation(s)
- Lucas L Falke
- Deparment Pathology, University Medical Centre Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands.,Department Internal medicine, Diakonessenhuis, Utrecht, the Netherlands
| | - Nannan He
- Department Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Roel Broekhuizen
- Deparment Pathology, University Medical Centre Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Karen Lyons
- Department Orthopedic Surgery, University of California, Berkeley, CA, USA.,Department Molecular Cell and Developmental Biology, University of California, Berkeley, CA, USA
| | - Tri Q Nguyen
- Deparment Pathology, University Medical Centre Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Deparment Pathology, University Medical Centre Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands.
| |
Collapse
|
132
|
Wu H, Yu Y, Huang H, Hu Y, Fu S, Wang Z, Shi M, Zhao X, Yuan J, Li J, Yang X, Bin E, Wei D, Zhang H, Zhang J, Yang C, Cai T, Dai H, Chen J, Tang N. Progressive Pulmonary Fibrosis Is Caused by Elevated Mechanical Tension on Alveolar Stem Cells. Cell 2020; 180:107-121.e17. [DOI: 10.1016/j.cell.2019.11.027] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/26/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023]
|
133
|
Ma Z, Yu R, Zhu Q, Sun L, Jian L, Wang X, Zhao J, Li C, Liu X. CXCL16/CXCR6 axis promotes bleomycin-induced fibrotic process in MRC-5 cells via the PI3K/AKT/FOXO3a pathway. Int Immunopharmacol 2019; 81:106035. [PMID: 31753588 DOI: 10.1016/j.intimp.2019.106035] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/01/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Interstitial lung disease (ILD) is a progressive and irreversible lung disease with very limited therapeutic options. Previous studies have found that chemokine ligands CXCL16 and CXCR6 play critical roles in organ fibrosis. However, whether CXCL16 and CXCR6 are also involved in the pathogenesis of ILD, as well as their regulatory role in pulmonary fibrosis, has not been reported. METHODS In this study, we detected CXCL16 levels in patients with rheumatoid arthritis-associated ILD (RA-ILD) and examined the critical role of the CXCL16/CXCR6 axis in the proliferation and collagen production of human pulmonary fibroblasts (MRC-5 cells). The effect of anti-CXCL16 antibody on the bleomycin-induced fibrogenesis in cultured MRC-5 cells was also evaluated. RESULTS Our results indicated that serum soluble CXCL16 was significantly higher in RA-ILD patients and also associated with the severity of lung fibrosis. CXCL16 facilitates fibrosis by enhancing proliferation, migration, and collagen production of MRC-5 cells. Furthermore, a synergistic fibrogenic effect of CXCL16 and bleomycin has been found. CXCL16 stimulated the activation of PI3K/AKT/FOXO3a signaling pathway in MRC-5 cells, and the inhibition by specific inhibitors Wortmannin and LY294002, or knockdown of CXCR6 by siRNA also suppressed the biological functions of MRC-5 cells mediated by CXCL16. Similarly, down-regulation of CXCR6 also partly blocked BLM-induced fibrogenesis in MRC-5 cells. CONCLUSIONS CXCL16/CXCR6 axis promotes proliferation and collagen production of MRC-5 cells by the PI3K/AKT/FOXO3a signaling pathway, and inhibition of the CXCL16/CXCR6 axis may provide a new therapeutic strategy targeting pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhenzhen Ma
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China
| | - Ruohan Yu
- Department of Rheumatology and Immunology, Beijing Tsinghua Changgung Hospital, Beijing 102218, China
| | - Qiao Zhu
- Department of Radiology, Peking University Third Hospital, Beijing 100191, China
| | - Lin Sun
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China
| | - Leilei Jian
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China
| | - Xinyu Wang
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China
| | - Jinxia Zhao
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China
| | - Changhong Li
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China.
| | - Xiangyuan Liu
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
134
|
Xie T, Wang Y, Deng N, Huang G, Taghavifar F, Geng Y, Liu N, Kulur V, Yao C, Chen P, Liu Z, Stripp B, Tang J, Liang J, Noble PW, Jiang D. Single-Cell Deconvolution of Fibroblast Heterogeneity in Mouse Pulmonary Fibrosis. Cell Rep 2019; 22:3625-3640. [PMID: 29590628 PMCID: PMC5908225 DOI: 10.1016/j.celrep.2018.03.010] [Citation(s) in RCA: 368] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/23/2018] [Accepted: 03/01/2018] [Indexed: 01/15/2023] Open
Abstract
Fibroblast heterogeneity has long been recognized in mouse and human lungs, homeostasis, and disease states. However, there is no common consensus on fibroblast subtypes, lineages, biological properties, signaling, and plasticity, which severely hampers our understanding of the mechanisms of fibrosis. To comprehensively classify fibroblast populations in the lung using an unbiased approach, single-cell RNA sequencing was performed with mesenchymal preparations from either uninjured or bleomycin-treated mouse lungs. Single-cell transcriptome analyses classified and defined six mesenchymal cell types in normal lung and seven in fibrotic lung. Furthermore, delineation of their differentiation trajectory was achieved by a machine learning method. This collection of single-cell transcriptomes and the distinct classification of fibroblast subsets provide a new resource for understanding the fibroblast landscape and the roles of fibroblasts in fibrotic diseases.
Collapse
Affiliation(s)
- Ting Xie
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Yizhou Wang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nan Deng
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Guanling Huang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Forough Taghavifar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yan Geng
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ningshan Liu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vrishika Kulur
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Changfu Yao
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Peter Chen
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zhengqiu Liu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Barry Stripp
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jie Tang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jiurong Liang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W Noble
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Dianhua Jiang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
135
|
Penke LR, Peters-Golden M. Molecular determinants of mesenchymal cell activation in fibroproliferative diseases. Cell Mol Life Sci 2019; 76:4179-4201. [PMID: 31563998 PMCID: PMC6858579 DOI: 10.1007/s00018-019-03212-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/01/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Uncontrolled scarring, or fibrosis, can interfere with the normal function of virtually all tissues of the body, ultimately leading to organ failure and death. Fibrotic diseases represent a major cause of death in industrialized countries. Unfortunately, no curative treatments for these conditions are yet available, highlighting the critical need for a better fundamental understanding of molecular mechanisms that may be therapeutically tractable. The ultimate indispensable effector cells responsible for deposition of extracellular matrix proteins that comprise scars are mesenchymal cells, namely fibroblasts and myofibroblasts. In this review, we focus on the biology of these cells and the molecular mechanisms that regulate their pertinent functions. We discuss key pro-fibrotic mediators, signaling pathways, and transcription factors that dictate their activation and persistence. Because of their possible clinical and therapeutic relevance, we also consider potential brakes on mesenchymal cell activation and cellular processes that may facilitate myofibroblast clearance from fibrotic tissue-topics that have in general been understudied.
Collapse
Affiliation(s)
- Loka R Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA.
| |
Collapse
|
136
|
Valenzi E, Bulik M, Tabib T, Morse C, Sembrat J, Trejo Bittar H, Rojas M, Lafyatis R. Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann Rheum Dis 2019; 78:1379-1387. [PMID: 31405848 PMCID: PMC7255436 DOI: 10.1136/annrheumdis-2018-214865] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/28/2019] [Accepted: 07/12/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Myofibroblasts are key effector cells in the extracellular matrix remodelling of systemic sclerosis-associated interstitial lung disease (SSc-ILD); however, the diversity of fibroblast populations present in the healthy and SSc-ILD lung is unknown and has prevented the specific study of the myofibroblast transcriptome. We sought to identify and define the transcriptomes of myofibroblasts and other mesenchymal cell populations in human healthy and SSc-ILD lungs to understand how alterations in fibroblast phenotypes lead to SSc-ILD fibrosis. METHODS We performed droplet-based, single-cell RNA-sequencing with integrated canonical correlation analysis of 13 explanted lung tissue specimens (56 196 cells) from four healthy control and four patients with SSc-ILD, with findings confirmed by cellular indexing of transcriptomes and epitopes by sequencing in additional samples. RESULTS Examination of gene expression in mesenchymal cells identified two major, SPINT2hi and MFAP5hi, and one minor, WIF1hi, fibroblast populations in the healthy control lung. Combined analysis of control and SSc-ILD mesenchymal cells identified SPINT2hi, MFAP5hi, few WIF1hi fibroblasts and a new large myofibroblast population with evidence of actively proliferating myofibroblasts. We compared differential gene expression between all SSc-ILD and control mesenchymal cell populations, as well as among the fibroblast subpopulations, showing that myofibroblasts undergo the greatest phenotypic changes in SSc-ILD and strongly upregulate expression of collagens and other profibrotic genes. CONCLUSIONS Our results demonstrate previously unrecognised fibroblast heterogeneity in SSc-ILD and healthy lungs, and define multimodal transcriptome-phenotypes associated with these populations. Our data indicate that myofibroblast differentiation and proliferation are key pathological mechanisms driving fibrosis in SSc-ILD.
Collapse
Affiliation(s)
- Eleanor Valenzi
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melissa Bulik
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tracy Tabib
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christina Morse
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John Sembrat
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Mauricio Rojas
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
137
|
Lemos DR, Duffield JS. Tissue-resident mesenchymal stromal cells: Implications for tissue-specific antifibrotic therapies. Sci Transl Med 2019; 10:10/426/eaan5174. [PMID: 29386358 DOI: 10.1126/scitranslmed.aan5174] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 01/08/2018] [Indexed: 11/02/2022]
Abstract
Recent scientific findings support the notion that fibrosis is driven by tissue-specific cellular and molecular mechanisms. Analysis of seemingly equivalent mesenchymal stromal cell (MSC) populations residing in different organs revealed unique properties and lineage capabilities that vary from one anatomical location to another. We review recently characterized tissue-resident MSC populations with a prominent role in fibrosis and highlight therapeutically relevant molecular pathways regulating their activity in chronic disease.
Collapse
Affiliation(s)
- Dario R Lemos
- Renal Division, Brigham and Women's Hospital, Boston, MA 02115, USA. .,Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy S Duffield
- Department of Medicine, University of Washington, Seattle, WA 98195, USA. .,Research and Development, Vertex Pharmaceuticals, Boston, MA 02210, USA
| |
Collapse
|
138
|
Majka SM, Rojas M, Petrache I, Foronjy RF. Mesenchymal Regulation of the Microvascular Niche in Chronic Lung Diseases. Compr Physiol 2019; 9:1431-1441. [PMID: 31688970 DOI: 10.1002/cphy.c180043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The adult lung is comprised of diverse vascular, epithelial, and mesenchymal progenitor cell populations that reside in distinct niches. Mesenchymal progenitor cells (MPCs) are intimately associated with both the epithelium and the vasculature, and new evidence is emerging to describe their functional roles in these niches. Also emerging, following lineage analysis and single cell sequencing, is a new understanding of the diversity of mesenchymal cell subpopulations in the lung. However, several gaps in knowledge remain, including how newly defined MPC lineages interact with cells in the vascular niche and the role of adult lung MPCs during lung repair and regeneration following injury, especially in chronic lung diseases. Here we summarize how the current evidence on MPC regulation of the microvasculature during tissue homeostasis and injury may inform studies on understanding their role in chronic lung disease pathogenesis or repair. © 2019 American Physiological Society. Compr Physiol 9:1431-1441, 2019.
Collapse
Affiliation(s)
- Susan M Majka
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Mauricio Rojas
- McGowan Institute for Regenerative Medicine, Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Robert F Foronjy
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York, USA
| |
Collapse
|
139
|
FOXF1 Inhibits Pulmonary Fibrosis by Preventing CDH2-CDH11 Cadherin Switch in Myofibroblasts. Cell Rep 2019; 23:442-458. [PMID: 29642003 PMCID: PMC5947867 DOI: 10.1016/j.celrep.2018.03.067] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/06/2018] [Accepted: 03/15/2018] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant accumulation of collagen-secreting myofibroblasts. Development of effective therapies is limited due to incomplete understanding of molecular mechanisms regulating myofibroblast expansion. FOXF1 transcription factor is expressed in resident lung fibroblasts, but its role in lung fibrosis remains unknown due to the lack of genetic mouse models. Through comprehensive analysis of human IPF genomics data, lung biopsies, and transgenic mice with fibroblast-specific inactivation of FOXF1, we show that FOXF1 inhibits pulmonary fibrosis. FOXF1 deletion increases myofibroblast invasion and collagen secretion and promotes a switch from N-cadherin (CDH2) to Cadherin-11 (CDH11), which is a critical step in the acquisition of the pro-fibrotic phenotype. FOXF1 directly binds to Cdh2 and Cdh11 promoters and differentially regulates transcription of these genes. Re-expression of CDH2 or inhibition of CDH11 in FOXF1-deficient cells reduces myofibroblast invasion in vitro. FOXF1 inhibits pulmonary fibrosis by regulating a switch from CDH2 to CDH11 in lung myofibroblasts.
Collapse
|
140
|
Wollin L, Distler JHW, Redente EF, Riches DWH, Stowasser S, Schlenker-Herceg R, Maher TM, Kolb M. Potential of nintedanib in treatment of progressive fibrosing interstitial lung diseases. Eur Respir J 2019; 54:1900161. [PMID: 31285305 PMCID: PMC6751387 DOI: 10.1183/13993003.00161-2019] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022]
Abstract
A proportion of patients with fibrosing interstitial lung diseases (ILDs) develop a progressive phenotype characterised by decline in lung function, worsening quality of life and early mortality. Other than idiopathic pulmonary fibrosis (IPF), there are no approved drugs for fibrosing ILDs and a poor evidence base to support current treatments. Fibrosing ILDs with a progressive phenotype show commonalities in clinical behaviour and in the pathogenic mechanisms that drive disease worsening. Nintedanib is an intracellular inhibitor of tyrosine kinases that has been approved for treatment of IPF and has recently been shown to reduce the rate of lung function decline in patients with ILD associated with systemic sclerosis (SSc-ILD). In vitro data demonstrate that nintedanib inhibits several steps in the initiation and progression of lung fibrosis, including the release of pro-inflammatory and pro-fibrotic mediators, migration and differentiation of fibrocytes and fibroblasts, and deposition of extracellular matrix. Nintedanib also inhibits the proliferation of vascular cells. Studies in animal models with features of fibrosing ILDs such as IPF, SSc-ILD, rheumatoid arthritis-ILD, hypersensitivity pneumonitis and silicosis demonstrate that nintedanib has anti-fibrotic activity irrespective of the trigger for the lung pathology. This suggests that nintedanib inhibits fundamental processes in the pathogenesis of fibrosis. A trial of nintedanib in patients with progressive fibrosing ILDs other than IPF (INBUILD) will report results in 2019.
Collapse
Affiliation(s)
- Lutz Wollin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Jörg H W Distler
- Dept of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Elizabeth F Redente
- Program in Cell Biology, Dept of Pediatrics, National Jewish Health, Denver, CO, USA
| | - David W H Riches
- Program in Cell Biology, Dept of Pediatrics, National Jewish Health, Denver, CO, USA
- University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, UK
- National Institute for Health Research Clinical Research Facility, Royal Brompton Hospital, London, UK
| | - Martin Kolb
- Dept of Respiratory Medicine, Pathology and Molecular Medicine, McMaster University and St Joseph's Healthcare, Hamilton, ON, Canada
| |
Collapse
|
141
|
Shammout B, Johnson JR. Pericytes in Chronic Lung Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:299-317. [PMID: 31147884 DOI: 10.1007/978-3-030-16908-4_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pericytes are supportive mesenchymal cells located on the abluminal surface of the microvasculature, with key roles in regulating microvascular homeostasis, leukocyte extravasation, and angiogenesis. A subpopulation of pericytes with progenitor cell function has recently been identified, with evidence demonstrating the capacity of tissue-resident pericytes to differentiate into the classic MSC triad, i.e., osteocytes, chondrocytes, and adipocytes. Beyond the regenerative capacity of these cells, studies have shown that pericytes play crucial roles in various pathologies in the lung, both acute (acute respiratory distress syndrome and sepsis-related pulmonary edema) and chronic (pulmonary hypertension, lung tumors, idiopathic pulmonary fibrosis, asthma, and chronic obstructive pulmonary disease). Taken together, this body of evidence suggests that, in the presence of acute and chronic pulmonary inflammation, pericytes are not associated with tissue regeneration and repair, but rather transform into scar-forming myofibroblasts, with devastating outcomes regarding lung structure and function. It is hoped that further studies into the mechanisms of pericyte-to-myofibroblast transition and migration to fibrotic foci will clarify the roles of pericytes in chronic lung disease and open up new avenues in the search for novel treatments for human pulmonary pathologies.
Collapse
Affiliation(s)
- Bushra Shammout
- Biosciences Department, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Jill R Johnson
- Biosciences Department, School of Life and Health Sciences, Aston University, Birmingham, UK.
| |
Collapse
|
142
|
Hung CF, Wilson CL, Schnapp LM. Pericytes in the Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:41-58. [PMID: 30937862 DOI: 10.1007/978-3-030-11093-2_3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The lung has numerous roles, including gas exchange, immune surveillance, and barrier function. Being a highly vascularized organ, the lung receives dual blood supply from both the pulmonary and bronchial circulation. Therefore, pericytes likely play a prominent role in lung physiology given their localization in the perivascular niche. New genetic approaches have increased our understanding of the origin and the diverse functions of lung pericytes. Lung pericytes are myofibroblast progenitors, contributing to development of fibrosis in mouse models. Lung pericytes are also capable of responding to danger signals and amplify the inflammatory response through elaboration of cytokines and adhesion molecules. In this chapter, we describe the molecular, anatomical, and phenotypical characterization of lung pericytes. We further highlight their potential roles in the pathogenesis of lung diseases including pulmonary fibrosis, asthma, and pulmonary hypertension. Finally, current gaps in knowledge and areas of ongoing investigation in lung pericyte biology are also discussed.
Collapse
Affiliation(s)
- Chi F Hung
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Carole L Wilson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Lynn M Schnapp
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
143
|
Xiao Y, Liu C, Chen Z, Blatchley MR, Kim D, Zhou J, Xu M, Gerecht S, Fan R. Senescent Cells with Augmented Cytokine Production for Microvascular Bioengineering and Tissue Repairs. ADVANCED BIOSYSTEMS 2019; 3:1900089. [PMID: 32270028 PMCID: PMC7141414 DOI: 10.1002/adbi.201900089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Indexed: 12/19/2022]
Abstract
Controlled delivery of cytokines and growth factors has been an area of intense research interest for molecular and cellular bioengineering, immunotherapy, and regenerative medicine. In this study, we show that primary human lung fibroblasts chemically induced to senescence (cell cycle arrest) can act as a living source to transiently produce factors essential for promoting vasculogenesis or angiogenesis, such as VEGF, HGF, and IL-8. Co-culture of senescent fibroblasts with HUVECs in a fibrin gel demonstrated accelerated formation and maturation of microvessel networks in as early as three days. Unlike the usage of non-senescent fibroblasts as the angiogenesis-promoting cells, this approach eliminates drawbacks related to the overproliferation of fibroblasts and the subsequent disruption of tissue architecture, integrity, or function. Co-culture of pancreatic islets with senescent fibroblasts and endothelial cells in a gel matrix maintains the viability and function of islets ex vivo for up to five days. Applying senescent fibroblasts to wound repair in vivo led to increased blood flow in a diabetic mouse model. Together, this work points to a new direction for engineering the delivery of cytokines and growth factors that promote microvascular tissue engineering and tissue repairs.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, U.S.A
| | - Chang Liu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, U.S.A
| | - Zhuo Chen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, U.S.A
| | - Michael R. Blatchley
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 20218, U.S.A
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University Baltimore, MD 20218, U.S.A
| | - Dongjoo Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, U.S.A
| | - Jing Zhou
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, U.S.A
- Department of Anesthesiology, Yale University, New Haven, CT 06520, U.S.A
| | - Ming Xu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, U.S.A
| | - Sharon Gerecht
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 20218, U.S.A
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University Baltimore, MD 20218, U.S.A
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 20218, U.S.A
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 20218, U.S.A
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, U.S.A
- Yale Comprehensive Cancer Center, New Haven, CT 06520, U.S.A
| |
Collapse
|
144
|
Kool HM, Bürgisser PE, Edel GG, de Kleer I, Boerema-de Munck A, de Laat I, Chrifi I, Cheng C, van Cappellen WA, Kremers GJ, Tibboel D, Rottier RJ. Inhibition of retinoic acid signaling induces aberrant pericyte coverage and differentiation resulting in vascular defects in congenital diaphragmatic hernia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L317-L331. [PMID: 31268349 DOI: 10.1152/ajplung.00104.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The mortality and morbidity of patients with congenital diaphragmatic hernia (CDH) is primarily caused by treatment-resistant, persistent pulmonary hypertension. Structural vascular changes, exemplified by extensive muscularization, are already present early in gestation, but the origin of these abnormalities is unknown. Understanding the origin of the vascular defects is important to improve treatment modalities. Here, we show that the distribution of pericytes is different and may thereby potentially initiate the vascular pathology in CDH. Transient inhibition of retinoic acid (RA) signaling early during pregnancy, the basis of the CDH mouse model, led to an increase in the number of pericytes, thereby affecting the angiogenic potential of pericytes in the fetuses. Pericytes of CDH lungs showed reduced proliferation and an increased ACTA2 expression, which indicates that these pericytes are more contractile than in control lung pericytes. This resulted in increased pericyte coverage of pulmonary vessels and reduced expansion of the capillary bed, the earliest pathological sign of the structural changes in CDH. Furthermore, the pericytes had reduced and altered collagen IV deposition in CDH, pointing to a loss of basal membrane integrity between pericytes and endothelial cells. Inhibition of RA signaling in vitro resulted in reduced migration of pericytes, reduced angiogenesis, and loss of collagen IV expression. Importantly, we confirmed our findings in lungs of human CDH patient samples. In summary, inhibition of RA signaling affects the lung pericyte population, leading to increased contractility, reduced pulmonary angiogenesis, and aberrant lung development, as observed in CDH.
Collapse
Affiliation(s)
- Heleen M Kool
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Petra E Bürgisser
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Gabriëla G Edel
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ismé de Kleer
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anne Boerema-de Munck
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Inge de Laat
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ihsan Chrifi
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline Cheng
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Gert-Jan Kremers
- Optical Imaging Centre, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dick Tibboel
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
145
|
Zhao Y, Tian B, Sun H, Zhang J, Zhang Y, Ivannikov M, Motamedi M, Liu Z, Zhou J, Kaphalia L, Calhoun WJ, Maroto R, Brasier AR. Pharmacoproteomics reveal novel protective activity of bromodomain containing 4 inhibitors on vascular homeostasis in TLR3-mediated airway remodeling. J Proteomics 2019; 205:103415. [PMID: 31195152 DOI: 10.1016/j.jprot.2019.103415] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/23/2019] [Accepted: 06/09/2019] [Indexed: 12/19/2022]
Abstract
Small molecule inhibitors of the epigenetic regulator bromodomain-containing protein 4 (BRD4) are potential therapeutics for viral and allergen-induced airway remodeling. A limitation of their preclinical advancement is the lack of detailed understanding of mechanisms of action and biomarkers of effect. We report a systems-level pharmacoproteomics in a standardized murine model of toll-like receptor TLR3-NFκB/RelA innate inflammation in the absence or presence of a highly selective BRD4 inhibitor (ZL0454) or nonselective bromodomain and extraterminal domain inhibitor (JQ1). Proteomics of bronchoalveolar lavage fluid (BALF) secretome and exosomal proteins from this murine model revealed increased, selective, capillary leak associated with pericyte-myofibroblast transition, a phenomenon blocked by BRD4 inhibitors. BALF proteomics also suggested that ZL0454 better reduced the vascular leakage and extracellular matrix deposition than JQ1. A significant subset of inflammation-mediated remodeling factors was also identified in a mouse model of idiopathic pulmonary fibrosis produced by bleomycin. BALF exosome analysis indicated that BRD4 inhibitors reduced the induction of exosomes enriched in coagulation factors whose presence correlated with interstitial fibrin deposition. Finally, BALF samples from humans with severe asthma demonstrated similar upregulations of ORM2, APCS, SPARCL1, FGA, and FN1, suggesting their potential as biomarkers for early detection of airway remodeling and/or monitoring of therapy response. SIGNIFICANCE: Repetitive and chronic viral upper respiratory tract infections trigger toll-like receptor (TLR)3-NFκB/RelA mediated airway remodeling which is linked to a progressive decline in pulmonary function in patients with asthma and chronic obstructive pulmonary disease. Small molecule inhibitors of the epigenetic regulator bromodomain-containing protein 4 (BRD4) are potential therapeutics for viral and allergen-induced airway remodeling. A limitation of their preclinical advancement is the lack of detailed understanding of mechanisms of action and biomarkers of effect. Our study revealed that the activation of (TLR)3-NFκB/RelA pathway in the lung induced an elevation in coagulation, complement, and platelet factors, indicating the increased vascular leak during airway remodeling. The mechanism of vascular leakage was chronic inflammation-induced pericyte-myofibroblast transition, which was blocked by BRD4 inhibitors. Finally, proteomics analysis of the bronchoalveolar lavage fluid samples from humans with severe asthma demonstrated similar findings that we observed in the animal model.
Collapse
Affiliation(s)
- Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA; Institute for Translational Sciences, UTMB, Galveston, TX, USA; Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA.
| | - Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA; Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA
| | - Hong Sun
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Jing Zhang
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Yueqing Zhang
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | | | | | - Zhiqing Liu
- Department of Pharmacology and Toxicology, UTMB, Galveston, TX, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, UTMB, Galveston, TX, USA
| | - Lata Kaphalia
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - William J Calhoun
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA; Institute for Translational Sciences, UTMB, Galveston, TX, USA; Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA
| | - Rosario Maroto
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Allan R Brasier
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
146
|
Rubio K, Singh I, Dobersch S, Sarvari P, Günther S, Cordero J, Mehta A, Wujak L, Cabrera-Fuentes H, Chao CM, Braubach P, Bellusci S, Seeger W, Günther A, Preissner KT, Wygrecka M, Savai R, Papy-Garcia D, Dobreva G, Heikenwalder M, Savai-Pullamsetti S, Braun T, Barreto G. Inactivation of nuclear histone deacetylases by EP300 disrupts the MiCEE complex in idiopathic pulmonary fibrosis. Nat Commun 2019; 10:2229. [PMID: 31110176 PMCID: PMC6527704 DOI: 10.1038/s41467-019-10066-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/12/2019] [Indexed: 01/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and highly lethal lung disease with unknown etiology and poor prognosis. IPF patients die within 2 years after diagnosis mostly due to respiratory failure. Current treatments against IPF aim to ameliorate patient symptoms and to delay disease progression. Unfortunately, therapies targeting the causes of or reverting IPF have not yet been developed. Here we show that reduced levels of miRNA lethal 7d (MIRLET7D) in IPF compromise epigenetic gene silencing mediated by the ribonucleoprotein complex MiCEE. In addition, we find that hyperactive EP300 reduces nuclear HDAC activity and interferes with MiCEE function in IPF. Remarkably, EP300 inhibition reduces fibrotic hallmarks of in vitro (patient-derived primary fibroblast), in vivo (bleomycin mouse model), and ex vivo (precision-cut lung slices, PCLS) IPF models. Our work provides the molecular basis for therapies against IPF using EP300 inhibition. Idiopathic pulmonary fibrosis (IPF) is a lethal disease with insufficient treatment strategies. Here the authors show that reduction of the microRNA MIRLET7D and hyperactivation of EP300 contribute to impaired epigenetic silencing by the MiCEE complex in pulmonary fibroblasts of IPF patients, and demonstrate the benefit of inhibiting EP300 for the treatment of IPF.
Collapse
Affiliation(s)
- Karla Rubio
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Indrabahadur Singh
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany. .,Division Chronic Inflammation and Cancer (F180), German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany.
| | - Stephanie Dobersch
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Pouya Sarvari
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Stefan Günther
- Department of Cardiac Development, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Julio Cordero
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Anatomy and Developmental Biology, CBTM, Heidelberg University, Mannheim, 68167, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Aditi Mehta
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, 81377, Germany
| | - Lukasz Wujak
- Faculty of Medicine, Biochemistry Institute, Justus Liebig University, Giessen, 35392, Germany
| | - Hector Cabrera-Fuentes
- Faculty of Medicine, Biochemistry Institute, Justus Liebig University, Giessen, 35392, Germany.,National Heart Research Institute, National Heart Centre Singapore, Singapore, 169609, Singapore.,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russian Federation.,Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, 64849, NL, Mexico.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, 169609, Singapore
| | - Cho-Ming Chao
- Chair for Lung Matrix Remodeling, Excellence Cluster Cardio Pulmonary System, Justus Liebig University, Giessen, 35392, Germany.,International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University and Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, 325035, China.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany.,German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany.,Department of General Pediatrics and Neonatology, University Children's Hospital Giessen, Justus Liebig University, Giessen, 35392, Germany
| | - Peter Braubach
- German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany.,Institute for Pathology, Hanover Medical School, Hanover, 30625, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH) Research Network, Hanover, 30625, Germany
| | - Saverio Bellusci
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russian Federation.,Chair for Lung Matrix Remodeling, Excellence Cluster Cardio Pulmonary System, Justus Liebig University, Giessen, 35392, Germany.,International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University and Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, 325035, China.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany.,German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany
| | - Werner Seeger
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany.,Department of General Pediatrics and Neonatology, University Children's Hospital Giessen, Justus Liebig University, Giessen, 35392, Germany.,Pulmonary and Critical Care Medicine, Department of Internal Medicine, Justus Liebig University, Giessen, 35392, Germany
| | - Andreas Günther
- Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany.,German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany.,Pulmonary and Critical Care Medicine, Department of Internal Medicine, Justus Liebig University, Giessen, 35392, Germany.,Agaplesion Lung Clinic Waldhof Elgershausen, Greifenstein, 35753, Germany
| | - Klaus T Preissner
- Faculty of Medicine, Biochemistry Institute, Justus Liebig University, Giessen, 35392, Germany.,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russian Federation.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany
| | - Malgorzata Wygrecka
- Faculty of Medicine, Biochemistry Institute, Justus Liebig University, Giessen, 35392, Germany.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany.,German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany.,German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany
| | - Dulce Papy-Garcia
- Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), CNRS ERL 9215, Université Paris Est Créteil, Université Paris Est, Créteil, F-94000, France
| | - Gergana Dobreva
- Anatomy and Developmental Biology, CBTM, Heidelberg University, Mannheim, 68167, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Mathias Heikenwalder
- Division Chronic Inflammation and Cancer (F180), German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
| | - Soni Savai-Pullamsetti
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany.,German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany
| | - Thomas Braun
- Department of Cardiac Development, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany
| | - Guillermo Barreto
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany. .,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, 420008, Russian Federation. .,Member of the Excellence Cluster Cardio Pulmonary System (ECCPS), The Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, 35392, Germany. .,German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), UGMLC, Giessen, 35392, Germany. .,Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), CNRS ERL 9215, Université Paris Est Créteil, Université Paris Est, Créteil, F-94000, France.
| |
Collapse
|
147
|
Role and Molecular Mechanisms of Pericytes in Regulation of Leukocyte Diapedesis in Inflamed Tissues. Mediators Inflamm 2019; 2019:4123605. [PMID: 31205449 PMCID: PMC6530229 DOI: 10.1155/2019/4123605] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/15/2019] [Accepted: 04/18/2019] [Indexed: 12/20/2022] Open
Abstract
Leukocyte recruitment is a hallmark of the inflammatory response. Migrating leukocytes breach the endothelium along with the vascular basement membrane and associated pericytes. While much is known about leukocyte-endothelial cell interactions, the mechanisms and role of pericytes in extravasation are poorly understood and the classical paradigm of leukocyte recruitment in the microvasculature seldom adequately discusses the involvement of pericytes. Emerging evidence shows that pericytes are essential players in the regulation of leukocyte extravasation in addition to their functions in blood vessel formation and blood-brain barrier maintenance. Junctions between venular endothelial cells are closely aligned with extracellular matrix protein low expression regions (LERs) in the basement membrane, which in turn are aligned with gaps between pericytes. This forms preferential paths for leukocyte extravasation. Breaching of the layer formed by pericytes and the basement membrane entails remodelling of LERs, leukocyte-pericyte adhesion, crawling of leukocytes on pericyte processes, and enlargement of gaps between pericytes to form channels for migrating leukocytes. Furthermore, inflamed arteriolar and capillary pericytes induce chemotactic migration of leukocytes that exit postcapillary venules, and through direct pericyte-leukocyte contact, they induce efficient interstitial migration to enhance the immunosurveillance capacity of leukocytes. Given their role as regulators of leukocyte extravasation, proper pericyte function is imperative in inflammatory disease contexts such as diabetic retinopathy and sepsis. This review summarizes research on the molecular mechanisms by which pericytes mediate leukocyte diapedesis in inflamed tissues.
Collapse
|
148
|
Kolb M, Vašáková M. The natural history of progressive fibrosing interstitial lung diseases. Respir Res 2019; 20:57. [PMID: 30871560 PMCID: PMC6417262 DOI: 10.1186/s12931-019-1022-1] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/04/2019] [Indexed: 12/11/2022] Open
Abstract
A proportion of patients with certain types of interstitial lung disease (ILD), including chronic hypersensitivity pneumonitis and ILDs associated with autoimmune diseases, develop a progressive fibrosing phenotype that shows similarities in clinical course to idiopathic pulmonary fibrosis. Irrespective of the clinical diagnosis, these progressive fibrosing ILDs show commonalities in the underlying pathogenetic mechanisms that drive a self-sustaining process of pulmonary fibrosis. The natural history of progressive fibrosing ILDs is characterized by decline in lung function, worsening of symptoms and health-related quality of life, and early mortality. Greater impairment in forced vital capacity or diffusion capacity of the lungs for carbon monoxide, and a greater extent of fibrotic changes on a computed tomography scan, are predictors of mortality in patients with fibrosing ILDs. However, the course of these diseases is heterogenous and cannot accurately be predicted for an individual patient. Data from ongoing clinical trials and patient registries will provide a better understanding of the clinical course and impact of progressive fibrosing ILDs.
Collapse
Affiliation(s)
- Martin Kolb
- McMaster University and St. Joseph’s Healthcare, 50 Charlton Avenue East, Hamilton, Ontario L8N 4A6 Canada
| | - Martina Vašáková
- Department of Respiratory Medicine, Thomayer Hospital, Videnska 800, 14059 Prague, Czech Republic
| |
Collapse
|
149
|
Parker GA, Li N, Takayama K, Farese AM, MacVittie TJ. Lung and Heart Injury in a Nonhuman Primate Model of Partial-body Irradiation with Minimal Bone Marrow Sparing: Histopathological Evidence of Lung and Heart Injury. HEALTH PHYSICS 2019; 116:383-400. [PMID: 30688698 PMCID: PMC6381599 DOI: 10.1097/hp.0000000000000936] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Male rhesus macaques were subjected to partial-body irradiation at 10, 11, or 12 Gy with 5% bone marrow protection. Animals were euthanized when dictated by prospectively determined clinical parameters or at approximately 180 d following irradiation. Histological sections of lung and heart were stained with hematoxylin and eosin as well as a battery of histochemical and immunohistochemical stains. Histopathological alterations in the lung were centered on fibrosis, inflammation, and reactive/proliferative changes in pneumocytes. These changes were noted in animals necropsied after approximately 85-100 d postirradiation and extending through the observation period. Interstitial and pleural fibrosis demonstrated by Masson's trichrome staining were associated with increased alpha smooth muscle actin and collagen 1 immunohistochemical staining. Areas of interstitial fibrosis had reduced microvascular density with CD31 immunohistochemical staining. Accumulations of CD163- and CD206-positive alveolar macrophages were present in areas of interstitial fibrosis. Unidentified cells termed "myxoid" cells in alveolar walls had histochemical and immunohistochemical staining characteristics of epithelial-, endothelial-, or pericyte-mesenchymal transition states that were developing myofibroblast features. Distinctive focal or multifocal alveolar-bronchiolar hyperplasia had microscopic features of preneoplastic proliferation. Delayed radiation-associated changes in the heart consisted primarily of myocardial fibrosis, with rare histological evidence of myofiber degeneration.
Collapse
Affiliation(s)
- George A. Parker
- Charles River Laboratories/Pathology Associates, Durham, NC, USA
| | - Na Li
- Charles River Laboratories/Pathology Associates, Durham, NC, USA
| | - Kyle Takayama
- Charles River Laboratories/Pathology Associates, Durham, NC, USA
| | - Ann M. Farese
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD, USA
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD, USA
| |
Collapse
|
150
|
Weng T, Ko J, Masamha CP, Xia Z, Xiang Y, Chen NY, Molina JG, Collum S, Mertens TC, Luo F, Philip K, Davies J, Huang J, Wilson C, Thandavarayan RA, Bruckner BA, Jyothula SS, Volcik KA, Li L, Han L, Li W, Assassi S, Karmouty-Quintana H, Wagner EJ, Blackburn MR. Cleavage factor 25 deregulation contributes to pulmonary fibrosis through alternative polyadenylation. J Clin Invest 2019; 129:1984-1999. [PMID: 30830875 DOI: 10.1172/jci122106] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and deadly disease with a poor prognosis and few treatment options. Pathological remodeling of the extracellular matrix (ECM) by myofibroblasts is a key factor that drives disease pathogenesis, although the underlying mechanisms remain unknown. Alternative polyadenylation (APA) has recently been shown to play a major role in cellular responses to stress by driving the expression of fibrotic factors and ECMs through altering microRNA sensitivity, but a connection to IPF has not been established. Here, we demonstrate that CFIm25, a global regulator of APA, is down-regulated in the lungs of patients with IPF and mice with pulmonary fibrosis, with its expression selectively reduced in alpha-smooth muscle actin (α-SMA) positive fibroblasts. Following the knockdown of CFIm25 in normal human lung fibroblasts, we identified 808 genes with shortened 3'UTRs, including those involved in the transforming growth factor-β signaling pathway, the Wnt signaling pathway, and cancer pathways. The expression of key pro-fibrotic factors can be suppressed by CFIm25 overexpression in IPF fibroblasts. Finally, we demonstrate that deletion of CFIm25 in fibroblasts or myofibroblast precursors using either the Col1a1 or the Foxd1 promoter enhances pulmonary fibrosis after bleomycin exposure in mice. Taken together, our results identified CFIm25 down-regulation as a novel mechanism to elevate pro-fibrotic gene expression in pulmonary fibrosis.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Junsuk Ko
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Chioniso P Masamha
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Zheng Xia
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, Texas, USA
| | - Yu Xiang
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Ning-Yuan Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Jose G Molina
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Scott Collum
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Tinne C Mertens
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Fayong Luo
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Kemly Philip
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Jonathan Davies
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Jingjing Huang
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cory Wilson
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | | | - Brian A Bruckner
- Houston Methodist DeBakey Transplant Center, Houston Methodist Hospital, Houston, Texas, USA
| | - Soma Sk Jyothula
- Department of Internal Medicine, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Kelly A Volcik
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Lei Li
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, Texas, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Wei Li
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, Texas, USA
| | - Shervin Assassi
- Department of Internal Medicine, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Eric J Wagner
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Michael R Blackburn
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, Texas, USA
| |
Collapse
|