1
|
Li X, Liu Y, Tang Y, Xia Z. Transformation of macrophages into myofibroblasts in fibrosis-related diseases: emerging biological concepts and potential mechanism. Front Immunol 2024; 15:1474688. [PMID: 39386212 PMCID: PMC11461261 DOI: 10.3389/fimmu.2024.1474688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Macrophage-myofibroblast transformation (MMT) transforms macrophages into myofibroblasts in a specific inflammation or injury microenvironment. MMT is an essential biological process in fibrosis-related diseases involving the lung, heart, kidney, liver, skeletal muscle, and other organs and tissues. This process consists of interacting with various cells and molecules and activating different signal transduction pathways. This review deeply discussed the molecular mechanism of MMT, clarified crucial signal pathways, multiple cytokines, and growth factors, and formed a complex regulatory network. Significantly, the critical role of transforming growth factor-β (TGF-β) and its downstream signaling pathways in this process were clarified. Furthermore, we discussed the significance of MMT in physiological and pathological conditions, such as pulmonary fibrosis and cardiac fibrosis. This review provides a new perspective for understanding the interaction between macrophages and myofibroblasts and new strategies and targets for the prevention and treatment of MMT in fibrotic diseases.
Collapse
Affiliation(s)
- Xiujun Li
- Health Science Center, Chifeng University, Chifeng, China
| | - Yuyan Liu
- Rehabilitation Medicine College, Shandong Second Medical University, Jinan, China
| | - Yongjun Tang
- Department of Emergency, Affiliated Hospital of Chifeng University, Chifeng, China
| | - Zhaoyi Xia
- Department of Library, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Library, Jinan Children’s Hospital, Jinan, China
| |
Collapse
|
2
|
Dahdah N, Tercero-Alcázar C, Malagón MM, Garcia-Roves PM, Guzmán-Ruiz R. Interrelation of adipose tissue macrophages and fibrosis in obesity. Biochem Pharmacol 2024; 225:116324. [PMID: 38815633 DOI: 10.1016/j.bcp.2024.116324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Obesity is characterized by adipose tissue expansion, extracellular matrix remodelling and unresolved inflammation that contribute to insulin resistance and fibrosis. Adipose tissue macrophages represent the most abundant class of immune cells in adipose tissue inflammation and could be key mediators of adipocyte dysfunction and fibrosis in obesity. Although macrophage activation states are classically defined by the M1/M2 polarization nomenclature, novel studies have revealed a more complex range of macrophage phenotypes in response to external condition or the surrounding microenvironment. Here, we discuss the plasticity of adipose tissue macrophages (ATMs) in response to their microenvironment in obesity, with special focus on macrophage infiltration and polarization, and their contribution to adipose tissue fibrosis. A better understanding of the role of ATMs as regulators of adipose tissue remodelling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Norma Dahdah
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carmen Tercero-Alcázar
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María M Malagón
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Pablo Miguel Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Rocío Guzmán-Ruiz
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain.
| |
Collapse
|
3
|
Tylek T, Wong J, Vaughan AE, Spiller KL. Biomaterial-mediated intracellular control of macrophages for cell therapy in pro-inflammatory and pro-fibrotic conditions. Biomaterials 2024; 308:122545. [PMID: 38547831 PMCID: PMC11264195 DOI: 10.1016/j.biomaterials.2024.122545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 05/03/2024]
Abstract
Macrophages are key modulators of all inflammatory diseases and essential for their resolution, making macrophage cell therapy a promising strategy for regenerative medicine. However, since macrophages change rapidly in response to microenvironmental cues, their phenotype must be controlled post-administration. We present a tunable biomaterial-based strategy to control macrophages intracellularly via small molecule-releasing microparticles. Poly(lactic-co-glycolic acid) microparticles encapsulating the anti-inflammatory and anti-fibrotic drug dexamethasone were administered to macrophages in vitro, with uptake rates controlled by different loading regimes. Microparticle dose and dexamethasone content directly affected macrophage phenotype and phagocytic capacity, independent of particle content per cell, leading to an overall pro-reparative, anti-inflammatory, anti-fibrotic phenotype with increased phagocytic and ECM degrading functionality. Intracellularly controlled macrophages partially maintained this phenotype in vivo in a murine pulmonary fibrosis model, with more prominent effects in a pro-fibrotic environment compared to pro-inflammatory. These results suggest that intracellular control using biomaterials has the potential to control macrophage phenotype post-administration, which is essential for successful macrophage cell therapy.
Collapse
Affiliation(s)
- Tina Tylek
- Drexel University, School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA 19104, USA
| | - Joanna Wong
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Kara L Spiller
- Drexel University, School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA 19104, USA.
| |
Collapse
|
4
|
Yang B, Qiao Y, Yan D, Meng Q. Targeting Interactions between Fibroblasts and Macrophages to Treat Cardiac Fibrosis. Cells 2024; 13:764. [PMID: 38727300 PMCID: PMC11082988 DOI: 10.3390/cells13090764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 05/13/2024] Open
Abstract
Excessive extracellular matrix (ECM) deposition is a defining feature of cardiac fibrosis. Most notably, it is characterized by a significant change in the concentration and volume fraction of collagen I, a disproportionate deposition of collagen subtypes, and a disturbed ECM network arrangement, which directly affect the systolic and diastolic functions of the heart. Immune cells that reside within or infiltrate the myocardium, including macrophages, play important roles in fibroblast activation and consequent ECM remodeling. Through both direct and indirect connections to fibroblasts, monocyte-derived macrophages and resident cardiac macrophages play complex, bidirectional, regulatory roles in cardiac fibrosis. In this review, we discuss emerging interactions between fibroblasts and macrophages in physiology and pathologic conditions, providing insights for future research aimed at targeting macrophages to combat cardiac fibrosis.
Collapse
Affiliation(s)
- Bo Yang
- Center for Organoid and Regeneration Medicine, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou 511466, China;
| | - Yan Qiao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China;
| | - Dong Yan
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China;
| | - Qinghang Meng
- Center for Organoid and Regeneration Medicine, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou 511466, China;
| |
Collapse
|
5
|
Harding J, Vintersten-Nagy K, Yang H, Tang JK, Shutova M, Jong ED, Lee JH, Massumi M, Oussenko T, Izadifar Z, Zhang P, Rogers IM, Wheeler MB, Lye SJ, Sung HK, Li C, Izadifar M, Nagy A. Immune-privileged tissues formed from immunologically cloaked mouse embryonic stem cells survive long term in allogeneic hosts. Nat Biomed Eng 2024; 8:427-442. [PMID: 37996616 PMCID: PMC11087263 DOI: 10.1038/s41551-023-01133-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/30/2023] [Indexed: 11/25/2023]
Abstract
The immunogenicity of transplanted allogeneic cells and tissues is a major hurdle to the advancement of cell therapies. Here we show that the overexpression of eight immunomodulatory transgenes (Pdl1, Cd200, Cd47, H2-M3, Fasl, Serpinb9, Ccl21 and Mfge8) in mouse embryonic stem cells (mESCs) is sufficient to immunologically 'cloak' the cells as well as tissues derived from them, allowing their survival for months in outbred and allogeneic inbred recipients. Overexpression of the human orthologues of these genes in human ESCs abolished the activation of allogeneic human peripheral blood mononuclear cells and their inflammatory responses. Moreover, by using the previously reported FailSafe transgene system, which transcriptionally links a gene essential for cell division with an inducible and cell-proliferation-dependent kill switch, we generated cloaked tissues from mESCs that served as immune-privileged subcutaneous sites that protected uncloaked allogeneic and xenogeneic cells from rejection in immune-competent hosts. The combination of cloaking and FailSafe technologies may allow for the generation of safe and allogeneically accepted cell lines and off-the-shelf cell products.
Collapse
Affiliation(s)
- Jeffrey Harding
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Kristina Vintersten-Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Huijuan Yang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jean Kit Tang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Maria Shutova
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Eric D Jong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Ju Hee Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad Massumi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Tatiana Oussenko
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Zohreh Izadifar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Puzheng Zhang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Ian M Rogers
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Michael B Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Stephen J Lye
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - ChengJin Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Mohammad Izadifar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
6
|
Mizote Y, Inoue T, Akazawa T, Kunimasa K, Tamiya M, Kumamoto Y, Tsuda A, Yoshida S, Tatsumi K, Ekawa T, Honma K, Nishino K, Tahara H. Potent CTLs can be induced against tumor cells in an environment of lower levels of systemic MFG-E8. Cancer Sci 2024; 115:1114-1128. [PMID: 38332689 PMCID: PMC11007000 DOI: 10.1111/cas.16099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
The direction and magnitude of immune responses are critically affected when dead cells are disposed of. Milk fat globule-epidermal growth factor-factor 8 (MFG-E8) promotes the engulfment of apoptotic normal and cancerous cells without inducing inflammation. We have previously reported that a certain proportion of the cancer cells express abundant MFG-E8, and that such expression is associated with the shorter survival of patients with esophageal cancer who had received chemotherapy before surgery. However, the influence of tumor-derived and systemically existing MFG-E8 on antitumor immune responses has not yet been fully investigated. Herein, we showed that CTL-dependent antitumor immune responses were observed in mice with no or decreased levels of systemic MFG-E8, and that such responses were enhanced further with the administration of anti-PD-1 antibody. In mice with decreased levels of systemic MFG-E8, the dominance of regulatory T cells in tumor-infiltrating lymphocytes was inverted to CD8+ T cell dominance. MFG-E8 expression by tumor cells appears to affect antitumor immune responses only when the level of systemic MFG-E8 is lower than the physiological status. We have also demonstrated in the clinical setting that lower levels of plasma MFG-E8, but not MFG-E8 expression in tumor cells, before the treatment was associated with objective responses to anti-PD-1 therapy in patients with non-small cell lung cancer. These results suggest that systemic MFG-E8 plays a critical role during the immunological initiation process of antigen-presenting cells to increase tumor-specific CTLs. Regulation of the systemic level of MFG-E8 might induce efficient antitumor immune responses and enhance the potency of anti-PD-1 therapy.
Collapse
Affiliation(s)
- Yu Mizote
- Department of Cancer Drug Discovery and Development, Research CenterOsaka International Cancer InstituteOsakaJapan
| | - Takako Inoue
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Takashi Akazawa
- Department of Cancer Drug Discovery and Development, Research CenterOsaka International Cancer InstituteOsakaJapan
| | - Kei Kunimasa
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Motohiro Tamiya
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Yachiyo Kumamoto
- Department of Cancer Drug Discovery and Development, Research CenterOsaka International Cancer InstituteOsakaJapan
| | - Arisa Tsuda
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Satomi Yoshida
- Department of Cancer Drug Discovery and Development, Research CenterOsaka International Cancer InstituteOsakaJapan
| | - Kumiko Tatsumi
- Department of Cancer Drug Discovery and Development, Research CenterOsaka International Cancer InstituteOsakaJapan
| | - Tomoya Ekawa
- Department of Cancer Drug Discovery and Development, Research CenterOsaka International Cancer InstituteOsakaJapan
| | - Keiichiro Honma
- Department of Diagnostic Pathology and CytologyOsaka International Cancer InstituteOsakaJapan
| | - Kazumi Nishino
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Hideaki Tahara
- Department of Cancer Drug Discovery and Development, Research CenterOsaka International Cancer InstituteOsakaJapan
- Project Division of Cancer Biomolecular Therapy, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| |
Collapse
|
7
|
Meizlish ML, Kimura Y, Pope SD, Matta R, Kim C, Philip NH, Meyaard L, Gonzalez A, Medzhitov R. Mechanosensing regulates tissue repair program in macrophages. SCIENCE ADVANCES 2024; 10:eadk6906. [PMID: 38478620 PMCID: PMC10936955 DOI: 10.1126/sciadv.adk6906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/29/2024] [Indexed: 03/17/2024]
Abstract
Tissue-resident macrophages play important roles in tissue homeostasis and repair. However, how macrophages monitor and maintain tissue integrity is not well understood. The extracellular matrix (ECM) is a key structural and organizational component of all tissues. Here, we find that macrophages sense the mechanical properties of the ECM to regulate a specific tissue repair program. We show that macrophage mechanosensing is mediated by cytoskeletal remodeling and can be performed in three-dimensional environments through a noncanonical, integrin-independent mechanism analogous to amoeboid migration. We find that these cytoskeletal dynamics also integrate biochemical signaling by colony-stimulating factor 1 and ultimately regulate chromatin accessibility to control the mechanosensitive gene expression program. This study identifies an "amoeboid" mode of ECM mechanosensing through which macrophages may regulate tissue repair and fibrosis.
Collapse
Affiliation(s)
- Matthew L. Meizlish
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yoshitaka Kimura
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Scott D. Pope
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rita Matta
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Catherine Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Naomi H. Philip
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Anjelica Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
8
|
Podolsky MJ, Kheyfets B, Pandey M, Beigh AH, Yang CD, Lizama CO, Datta R, Lin LL, Wang Z, Wolters PJ, McManus MT, Qi L, Atabai K. Genome-wide screens identify SEL1L as an intracellular rheostat controlling collagen turnover. Nat Commun 2024; 15:1531. [PMID: 38378719 PMCID: PMC10879544 DOI: 10.1038/s41467-024-45817-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024] Open
Abstract
Accumulating evidence has implicated impaired extracellular matrix (ECM) clearance as a key factor in fibrotic disease. Despite decades of research elucidating the effectors of ECM clearance, relatively little is understood regarding the upstream regulation of this process. Collagen is the most abundant constituent of normal and fibrotic ECM in mammalian tissues. Its catabolism occurs through extracellular proteolysis and cell-mediated uptake of collagen fragments for intracellular degradation. Given the paucity of information regarding the regulation of this latter process, here we execute unbiased genome-wide screens to understand the molecular underpinnings of cell-mediated collagen clearance. Using this approach, we discover a mechanism through which collagen biosynthesis is sensed by cells internally and directly regulates clearance of extracellular collagen. The sensing mechanism appears to be dependent on endoplasmic reticulum-resident protein SEL1L and occurs via a noncanonical function of this protein. This pathway functions as a homeostatic negative feedback loop that limits collagen accumulation in tissues. In human fibrotic lung disease, the induction of this collagen clearance pathway by collagen synthesis is impaired, thereby contributing to the pathological accumulation of collagen in lung tissue. Thus, we describe cell-autonomous, rheostatic collagen clearance as an important pathway of tissue homeostasis.
Collapse
Affiliation(s)
- Michael J Podolsky
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Benjamin Kheyfets
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Monika Pandey
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Afaq H Beigh
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Christopher D Yang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Carlos O Lizama
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Ritwik Datta
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Liangguang L Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Zhihong Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Michael T McManus
- Department of Microbiology and Immunology and UCSF Diabetes Center, University of California, San Francisco, CA, USA
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, CA, USA.
- Lung Biology Center, University of California, San Francisco, CA, USA.
| |
Collapse
|
9
|
Vujičić M, Broderick I, Salmantabar P, Perian C, Nilsson J, Sihlbom Wallem C, Wernstedt Asterholm I. A macrophage-collagen fragment axis mediates subcutaneous adipose tissue remodeling in mice. Proc Natl Acad Sci U S A 2024; 121:e2313185121. [PMID: 38300872 PMCID: PMC10861897 DOI: 10.1073/pnas.2313185121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/19/2023] [Indexed: 02/03/2024] Open
Abstract
Efficient removal of fibrillar collagen is essential for adaptive subcutaneous adipose tissue (SAT) expansion that protects against ectopic lipid deposition during weight gain. Here, we used mice to further define the mechanism for this collagenolytic process. We show that loss of collagen type-1 (CT1) and increased CT1-fragment levels in expanding SAT are associated with proliferation of resident M2-like macrophages that display increased CD206-mediated engagement in collagen endocytosis compared to chow-fed controls. Blockage of CD206 during acute high-fat diet-induced weight gain leads to SAT CT1-fragment accumulation associated with elevated inflammation and fibrosis markers. Moreover, these SAT macrophages' engagement in collagen endocytosis is diminished in obesity associated with elevated levels collagen fragments that are too short to assemble into triple helices. We show that such short fragments provoke M2-macrophage proliferation and fibroinflammatory changes in fibroblasts. In conclusion, our data delineate the importance of a macrophage-collagen fragment axis in physiological SAT expansion. Therapeutic targeting of this process may be a means to prevent pathological adipose tissue remodeling, which in turn may reduce the risk for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Milica Vujičić
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Isabella Broderick
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Pegah Salmantabar
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Charlène Perian
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Jonas Nilsson
- Proteomics Core Facility, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Carina Sihlbom Wallem
- Proteomics Core Facility, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| |
Collapse
|
10
|
Datta R, Mukherjee D, Podolsky MJ, Yang CD, Alba DL, Singh S, Arnold TD, Koliwad S, Lizama CO, Atabai K. PTP1B mediates the inhibitory effect of MFGE8 on insulin signaling through the β5 integrin. J Biol Chem 2024; 300:105631. [PMID: 38199575 PMCID: PMC10850974 DOI: 10.1016/j.jbc.2024.105631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/14/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Integrins are cell adhesion receptors that dimerize to mediate cell-cell interactions and regulate processes, including proliferation, inflammation, and tissue repair. The role of integrins in regulating insulin signaling is incompletely understood. We have previously shown that binding of the integrin ligand milk fat globule epidermal growth factor like 8 (MFGE8) to the αvβ5 integrin promotes termination of insulin receptor signaling in mice. Upon ligation of MFGE8, integrin β5 complexes with the insulin receptor beta (IRβ) in skeletal muscle, resulting in dephosphorylation of IRβ and reduction of insulin-stimulated glucose uptake. Here, we investigate the mechanism by which the interaction between β5 and IRβ impacts IRβ phosphorylation status. We show in in vitro and in vivo in skeletal muscle in mice that antibody-mediated blockade of the β5 integrin inhibits and recombinant MFGE8 promotes PTP1B binding to and dephosphorylation of IRβ resulting in increased or reduced insulin-stimulated glucose uptake, respectively. The β5-PTP1B complex is recruited by MFGE8 to IRβ leading to termination of canonical insulin signaling. β5 blockade enhances insulin-stimulated glucose uptake in wildtype but not Ptp1b KO mice indicating that PTP1B functions downstream of MFGE8 in modulating insulin receptor signaling. Furthermore, in a human cohort, we report serum MFGE8 levels correlate with indices of insulin resistance. These data provide mechanistic insights into the role of MFGE8 and β5 in regulating insulin signaling.
Collapse
Affiliation(s)
- Ritwik Datta
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Dibyanti Mukherjee
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Michael J Podolsky
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Christopher D Yang
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Diana L Alba
- Diabetes Center, University of California, San Francisco, California, USA; Division of Endocrinology, Department of Medicine, University of California, San Francisco, California, USA
| | - Sukhmani Singh
- Division of Endocrinology, Department of Medicine, University of California, San Francisco, California, USA
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Suneil Koliwad
- Diabetes Center, University of California, San Francisco, California, USA; Division of Endocrinology, Department of Medicine, University of California, San Francisco, California, USA
| | - Carlos O Lizama
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, California, USA; Diabetes Center, University of California, San Francisco, California, USA; Lung Biology Center, University of California, San Francisco, California, USA.
| |
Collapse
|
11
|
Zhu L, Gou W, Ou L, Liu B, Liu M, Feng H. Role and new insights of microfibrillar-associated protein 4 in fibrotic diseases. APMIS 2024; 132:55-67. [PMID: 37957836 DOI: 10.1111/apm.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023]
Abstract
Fibrosis is one of the most worrisome complications of chronic inflammatory diseases, leading to tissue damage, organ failure, and ultimately, death. The most notable pathological characteristic of fibrosis is the excessive accumulation of extracellular matrix (ECM) components such as collagen and fibronectin adjacent to foci of inflammation or damage. The human microfibrillar-associated protein 4 (MFAP4), an important member of the superfamily of fibrinogen-related proteins, is considered to have an extremely important role in ECM transformation of fibrogenesis. This review summarizes the structure, characteristics, and physiological functions of MFAP4 and the importance of MFAP4 in various fibrotic diseases. Meanwhile, we elaborated the underlying actions and mechanisms of MFAP4 in the development of fibrosis, suggesting that a better understand of MFAP4 broadens novel perspective for early screening, diagnosis, prognostic risk assessment, and treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Long Zhu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Wenqun Gou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
- Changsha Stomatological Hospital, Changsha, China
| | - Lijia Ou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Binjie Liu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Manyi Liu
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Hui Feng
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
12
|
Nofi CP, Tan C, Ma G, Kobritz M, Prince JM, Wang H, Aziz M, Wang P. A novel opsonic eCIRP inhibitor for lethal sepsis. J Leukoc Biol 2024; 115:385-400. [PMID: 37774691 PMCID: PMC10799304 DOI: 10.1093/jleuko/qiad119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
Sepsis is a life-threatening inflammatory condition partly orchestrated by the release of various damage-associated molecular patterns such as extracellular cold-inducible RNA-binding protein (eCIRP). Despite advances in understanding the pathogenic role of eCIRP in inflammatory diseases, novel therapeutic strategies to prevent its excessive inflammatory response are lacking. Milk fat globule-epidermal growth factor-VIII (MFG-E8) is critical for the opsonic clearance of apoptotic cells, but its potential involvement in the removal of eCIRP was previously unknown. Here, we report that MFG-E8 can strongly bind eCIRP to facilitate αvβ3-integrin-dependent internalization and lysosome-dependent degradation of MFG-E8/eCIRP complexes, thereby attenuating excessive inflammation. Genetic disruption of MFG-E8 expression exaggerated sepsis-induced systemic accumulation of eCIRP and other cytokines, and consequently exacerbated sepsis-associated acute lung injury. In contrast, MFG-E8-derived oligopeptide recapitulated its eCIRP binding properties, and significantly attenuated eCIRP-induced inflammation to confer protection against sepsis. Our findings suggest a novel therapeutic approach to attenuate eCIRP-induced inflammation to improve outcomes of lethal sepsis.
Collapse
Affiliation(s)
- Colleen P Nofi
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Chuyi Tan
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Gaifeng Ma
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Molly Kobritz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Jose M Prince
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Haichao Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Ping Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| |
Collapse
|
13
|
Kadono M, Nakashima A, Ishiuchi N, Sasaki K, Miura Y, Maeda S, Fujita A, Sasaki A, Nagamatsu S, Masaki T. Adipose-derived mesenchymal stem cells cultured in serum-free medium attenuate acute contrast-induced nephropathy by exerting anti-apoptotic effects. Stem Cell Res Ther 2023; 14:337. [PMID: 37993965 PMCID: PMC10664307 DOI: 10.1186/s13287-023-03553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Contrast-induced nephropathy (CIN) is a major clinical problem associated with acute kidney injury during hospitalization. However, effective treatments for CIN are currently lacking. Mesenchymal stem cells (MSCs) have protective effects against kidney injury by suppressing inflammation and fibrosis. We previously showed that MSCs cultured in serum-free medium (SF-MSCs) enhance their anti-inflammatory and anti-fibrotic effects. However, whether SF-MSCs potentiate their anti-apoptotic effects is unknown. Here, we investigated the effects of SF-MSCs on a CIN mouse model. METHODS To create CIN model mice, we removed right kidney at first. One week later, the left renal artery was clamped for 30 min to cause ischemia-reperfusion injury, and mice were injected with iohexol. Then the kidney received 10 Gy of irradiation, and MSCs or SF-MSCs were injected immediately. At 24 h post-injection, mice were sacrificed, and their blood and kidneys were collected to evaluate renal function, DNA damage, and apoptosis. In addition, apoptosis was induced in HEK-293 cells by irradiation and cells were treated with conditioned medium from MSCs or SF-MSCs. RESULTS Treatment of CIN model mice with SF-MSCs markedly improved renal function compared with MSCs treatment. Cleaved caspase-3 levels and TUNEL-positive cell numbers were strongly suppressed in CIN model mice treated with SF-MSCs compared with the findings in those treated with MSCs. γH2AX levels, a chromosome damage marker, were reduced by MSCs and further reduced by SF-MSCs. In addition, cleaved caspase-3 in irradiated HEK-293 cells was more strongly suppressed by conditioned medium from SF-MSCs than by that from MSCs. Secretion of epidermal growth factor (EGF) was enhanced by culturing MSCs in serum-free medium. Knockdown of EGF by siRNA attenuated the inhibitory effects of SF-MSCs on CIN-induced renal dysfunction and tubular apoptosis. CONCLUSIONS These findings strongly suggest that SF-MSCs improve CIN in model mice by exerting anti-apoptotic effects in a paracrine manner. Thus, SF-MSCs represent a potential novel therapy for CIN.
Collapse
Affiliation(s)
- Mitsuki Kadono
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yoshie Miura
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- TWOCELLS Company, Limited, 16-35 Hijiyama-Honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Satoshi Maeda
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- TWOCELLS Company, Limited, 16-35 Hijiyama-Honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Asuka Fujita
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayano Sasaki
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shogo Nagamatsu
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
14
|
Liu B, Zhang B, Qi J, Zhou H, Tan L, Huang J, Huang J, Fang X, Gong L, Luo J, Liu S, Fu L, Ling F, Ma S, Lai-wan Kwong D, Wang X, Guan XY. Targeting MFGE8 secreted by cancer-associated fibroblasts blocks angiogenesis and metastasis in esophageal squamous cell carcinoma. Proc Natl Acad Sci U S A 2023; 120:e2307914120. [PMID: 37816055 PMCID: PMC10589644 DOI: 10.1073/pnas.2307914120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/09/2023] [Indexed: 10/12/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) play vital roles in establishing a suitable tumor microenvironment. In this study, RNA sequencing data revealed that CAFs could promote cell proliferation, angiogenesis, and ECM reconstitution by binding to integrin families and activating PI3K/AKT pathways in esophageal squamous cell carcinoma (ESCC). The secretions of CAFs play an important role in regulating these biological activities. Among these secretions, we found that MFGE8 is specifically secreted by CAFs in ESCC. Additionally, the secreted MFGE8 protein is essential in CAF-regulated vascularization, tumor proliferation, drug resistance, and metastasis. By binding to Integrin αVβ3/αVβ5 receptors, MFGE8 promotes tumor progression by activating both the PI3K/AKT and ERK/AKT pathways. Interestingly, the biological function of MFGE8 secreted by CAFs fully demonstrated the major role of CAFs in ESCC and its mode of mechanism, showing that MFGE8 could be a driver factor of CAFs in remodeling the tumor environment. In vivo treatment targeting CAFs-secreting MFGE8 or its receptor produced significant inhibitory effects on ESCC growth and metastasis, which provides an approach for the treatment of ESCC.
Collapse
Affiliation(s)
- Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518053, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Baifeng Zhang
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518053, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Jiali Qi
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Hongyu Zhou
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Licheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Jinlin Huang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Jiao Huang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Xiaona Fang
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518053, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Lanqi Gong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518053, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Jie Luo
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Shan Liu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Li Fu
- Department of Pharmacology and International Cancer Center, Shenzhen University Health Science Center, Shenzhen518060, China
| | - Fei Ling
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou510006, China
| | - Stephianie Ma
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518053, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong852, China
| | - Dora Lai-wan Kwong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518053, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
| | - Xin Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Hong Kong510060, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518053, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong852, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou528200, China
- Ministry of Education Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou510275, China
| |
Collapse
|
15
|
Weckerle J, Mayr CH, Fundel-Clemens K, Lämmle B, Boryn L, Thomas MJ, Bretschneider T, Luippold AH, Huber HJ, Viollet C, Rist W, Veyel D, Ramirez F, Klee S, Kästle M. Transcriptomic and Proteomic Changes Driving Pulmonary Fibrosis Resolution in Young and Old Mice. Am J Respir Cell Mol Biol 2023; 69:422-440. [PMID: 37411041 DOI: 10.1165/rcmb.2023-0012oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/06/2023] [Indexed: 07/08/2023] Open
Abstract
Bleomycin-induced pulmonary fibrosis in mice mimics major hallmarks of idiopathic pulmonary fibrosis. Yet in this model, it spontaneously resolves over time. We studied molecular mechanisms of fibrosis resolution and lung repair, focusing on transcriptional and proteomic signatures and the effect of aging. Old mice showed incomplete and delayed lung function recovery 8 weeks after bleomycin instillation. This shift in structural and functional repair in old bleomycin-treated mice was reflected in a temporal shift in gene and protein expression. We reveal gene signatures and signaling pathways that underpin the lung repair process. Importantly, the downregulation of WNT, BMP, and TGFβ antagonists Frzb, Sfrp1, Dkk2, Grem1, Fst, Fstl1, and Inhba correlated with lung function improvement. Those genes constitute a network with functions in stem cell pathways, wound, and pulmonary healing. We suggest that insufficient and delayed downregulation of those antagonists during fibrosis resolution in old mice explains the impaired regenerative outcome. Together, we identified signaling pathway molecules with relevance to lung regeneration that should be tested in-depth experimentally as potential therapeutic targets for pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | - Bärbel Lämmle
- Global Computational Biology and Digital Sciences, and
| | | | | | - Tom Bretschneider
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Andreas H Luippold
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | | | | | - Wolfgang Rist
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Daniel Veyel
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Fidel Ramirez
- Global Computational Biology and Digital Sciences, and
| | - Stephan Klee
- Department of Immunology and Respiratory Disease Research
| | - Marc Kästle
- Department of Immunology and Respiratory Disease Research
| |
Collapse
|
16
|
Podolsky MJ, Kheyfets B, Pandey M, Beigh AH, Yang CD, Lizama Valenzuela C, Datta R, Wolters PJ, McManus M, Qi L, Atabai K. Genome-wide screens identify SEL1L as an intracellular rheostat controlling collagen turnover. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523943. [PMID: 36711851 PMCID: PMC9882208 DOI: 10.1101/2023.01.13.523943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Accumulating evidence has implicated impaired extracellular matrix (ECM) clearance as a key factor in fibrotic disease. Despite decades of research elucidating the effectors of ECM clearance, relatively little is understood regarding the upstream regulation of this process. Collagen is the most abundant constituent of normal and fibrotic ECM in mammalian tissues. Its catabolism occurs through extracellular proteolysis and cell-mediated uptake of collagen fragments for intracellular degradation. Given the paucity of information regarding the regulation of this latter process, we executed unbiased genome-wide screens to understand the molecular underpinnings of cell-mediated collagen clearance. Using this approach, we discovered a previously unappreciated mechanism through which collagen biosynthesis is sensed by cells internally and directly regulates clearance of extracellular collagen. The sensing mechanism is dependent on endoplasmic reticulum-resident protein SEL1L and occurs via a noncanonical function of SEL1L. This pathway functions as a homeostatic negative feedback loop that limits collagen accumulation in tissues. In human fibrotic lung disease, the induction of this collagen clearance pathway by collagen synthesis is impaired, thereby contributing to the pathological accumulation of collagen in lung tissue. Thus cell-autonomous, rheostatic collagen clearance is a previously unidentified pathway of tissue homeostasis.
Collapse
|
17
|
Bhattacharya M, Ramachandran P. Immunology of human fibrosis. Nat Immunol 2023; 24:1423-1433. [PMID: 37474654 DOI: 10.1038/s41590-023-01551-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/08/2023] [Indexed: 07/22/2023]
Abstract
Fibrosis, defined by the excess deposition of structural and matricellular proteins in the extracellular space, underlies tissue dysfunction in multiple chronic diseases. Approved antifibrotics have proven modest in efficacy, and the immune compartment remains, for the most part, an untapped therapeutic opportunity. Recent single-cell analyses have interrogated human fibrotic tissues, including immune cells. These studies have revealed a conserved profile of scar-associated macrophages, which localize to the fibrotic niche and interact with mesenchymal cells that produce pathological extracellular matrix. Here we review recent advances in the understanding of the fibrotic microenvironment in human diseases, with a focus on immune cell profiles and functional immune-stromal interactions. We also discuss the key role of the immune system in mediating fibrosis regression and highlight avenues for future study to elucidate potential approaches to targeting inflammatory cells in fibrotic disorders.
Collapse
Affiliation(s)
- Mallar Bhattacharya
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Prakash Ramachandran
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, Edinburgh, UK.
| |
Collapse
|
18
|
Salvador AFM, Dykstra T, Rustenhoven J, Gao W, Blackburn SM, Bhasiin K, Dong MQ, Guimarães RM, Gonuguntla S, Smirnov I, Kipnis J, Herz J. Age-dependent immune and lymphatic responses after spinal cord injury. Neuron 2023; 111:2155-2169.e9. [PMID: 37148871 PMCID: PMC10523880 DOI: 10.1016/j.neuron.2023.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 02/13/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
Spinal cord injury (SCI) causes lifelong debilitating conditions. Previous works demonstrated the essential role of the immune system in recovery after SCI. Here, we explored the temporal changes of the response after SCI in young and aged mice in order to characterize multiple immune populations within the mammalian spinal cord. We revealed substantial infiltration of myeloid cells to the spinal cord in young animals, accompanied by changes in the activation state of microglia. In contrast, both processes were blunted in aged mice. Interestingly, we discovered the formation of meningeal lymphatic structures above the lesion site, and their role has not been examined after contusive injury. Our transcriptomic data predicted lymphangiogenic signaling between myeloid cells in the spinal cord and lymphatic endothelial cells (LECs) in the meninges after SCI. Together, our findings delineate how aging affects the immune response following SCI and highlight the participation of the spinal cord meninges in supporting vascular repair.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Taitea Dykstra
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Justin Rustenhoven
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland 1023, New Zealand
| | - Wenqing Gao
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Susan M Blackburn
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kesshni Bhasiin
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michael Q Dong
- Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Rafaela Mano Guimarães
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Sriharsha Gonuguntla
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Igor Smirnov
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Jasmin Herz
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
19
|
Hoffman ET, Uhl FE, Asarian L, Deng B, Becker C, Uriarte JJ, Downs I, Young B, Weiss DJ. Regional and disease specific human lung extracellular matrix composition. Biomaterials 2023; 293:121960. [PMID: 36580718 PMCID: PMC9868084 DOI: 10.1016/j.biomaterials.2022.121960] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/25/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), are characterized by regional extracellular matrix (ECM) remodeling which contributes to disease progression. Previous proteomic studies on whole decellularized lungs have provided detailed characterization on the impact of COPD and IPF on total lung ECM composition. However, such studies are unable to determine the differences in ECM composition between individual anatomical regions of the lung. Here, we employ a post-decellularization dissection method to compare the ECM composition of whole decellularized lungs (wECM) and specific anatomical lung regions, including alveolar-enriched ECM (aECM), airway ECM (airECM), and vasculature ECM (vECM), between non-diseased (ND), COPD, and IPF human lungs. We demonstrate, using mass spectrometry, that individual regions possess a unique ECM signature characterized primarily by differences in collagen composition and basement-membrane associated proteins, including ECM glycoproteins. We further demonstrate that both COPD and IPF lead to alterations in lung ECM composition in a region-specific manner, including enrichment of type-III collagen and fibulin in IPF aECM. Taken together, this study provides methodology for future studies, including isolation of region-specific lung biomaterials, as well as a dataset that may be applied for the identification of novel ECM targets for therapeutics.
Collapse
Affiliation(s)
- Evan T. Hoffman
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Franziska E. Uhl
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Loredana Asarian
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Bin Deng
- Department of Biology, University of Vermont, Burlington, VT, 05405, USA
| | - Chloe Becker
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Juan J. Uriarte
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Isaac Downs
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Brad Young
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Daniel J. Weiss
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| |
Collapse
|
20
|
Yao H, Tang G. Macrophages in intestinal fibrosis and regression. Cell Immunol 2022; 381:104614. [PMID: 36182587 DOI: 10.1016/j.cellimm.2022.104614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
Intestinal macrophages are heterogenous cell populations with different developmental ontogeny and tissue anatomy. The concerted actions of intestinal macrophage subsets are critical to maintaining tissue homeostasis. However, the dysregulation of macrophages following tissue injury or chronic inflammation could also lead to intestinal fibrosis, with few treatment options in the clinic. In this review, we will characterize the features of intestinal macrophages in light of the latest advances in lineage tracing and single-cell sequencing technology. The roles of macrophages in distinct stages of intestinal fibrosis would be also elaborated. Finally, based on the reciprocal interaction between macrophages and intestinal fibrosis, we will propose the potential macrophage targeting anti-intestinal fibrosis therapies.
Collapse
Affiliation(s)
- Hui Yao
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China; National Center for Stomatology, Shanghai 200011, China; National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Guoyao Tang
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China; National Center for Stomatology, Shanghai 200011, China; National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.
| |
Collapse
|
21
|
Sari E, He C, Margaroli C. Plasticity towards Rigidity: A Macrophage Conundrum in Pulmonary Fibrosis. Int J Mol Sci 2022; 23:11443. [PMID: 36232756 PMCID: PMC9570276 DOI: 10.3390/ijms231911443] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, chronic, and ultimately fatal diffuse parenchymal lung disease. The molecular mechanisms of fibrosis in IPF patients are not fully understood and there is a lack of effective treatments. For decades, different types of drugs such as immunosuppressants and antioxidants have been tested, usually with unsuccessful results. Although two antifibrotic drugs (Nintedanib and Pirfenidone) are approved and used for the treatment of IPF, side effects are common, and they only slow down disease progression without improving patients' survival. Macrophages are central to lung homeostasis, wound healing, and injury. Depending on the stimulus in the microenvironment, macrophages may contribute to fibrosis, but also, they may play a role in the amelioration of fibrosis. In this review, we explore the role of macrophages in IPF in relation to the fibrotic processes, epithelial-mesenchymal transition (EMT), and their crosstalk with resident and recruited cells and we emphasized the importance of macrophages in finding new treatments.
Collapse
Affiliation(s)
- Ezgi Sari
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chao He
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Camilla Margaroli
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
22
|
Gu Y, Lawrence T, Mohamed R, Liang Y, Yahaya BH. The emerging roles of interstitial macrophages in pulmonary fibrosis: A perspective from scRNA-seq analyses. Front Immunol 2022; 13:923235. [PMID: 36211428 PMCID: PMC9536737 DOI: 10.3389/fimmu.2022.923235] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is an irreversible and progressive disease affecting the lungs, and the etiology remains poorly understood. This disease can be lethal and currently has no specific clinical therapeutic regimen. Macrophages, the most common type of immune cell in the lungs, have been reported to play a key role in the pathogenesis of fibrotic disease. The lung macrophage population is mostly composed of alveolar macrophages and interstitial macrophages, both of which have not been thoroughly studied in the pathogenesis of lung fibrosis. Interstitial macrophages have recently been recognised for their participation in lung fibrosis due to new technology arising from a combination of bioinformatics and single-cell RNA sequencing analysis. This paper reviews recent developments regarding lung macrophage classification and summarizes the origin and replenishment of interstitial macrophages and their function in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yanrong Gu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, Kepala Batas, Malaysia
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Toby Lawrence
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Centre for Inflammation Biology and Cancer Immunology, Cancer Research UK King’s Health Partners Centre, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Rafeezul Mohamed
- Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, Kepala Batas, Malaysia
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Yinming Liang, ; Badrul Hisham Yahaya,
| | - Badrul Hisham Yahaya
- Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, Kepala Batas, Malaysia
- *Correspondence: Yinming Liang, ; Badrul Hisham Yahaya,
| |
Collapse
|
23
|
Dooling LJ, Saini K, Anlaş AA, Discher DE. Tissue mechanics coevolves with fibrillar matrisomes in healthy and fibrotic tissues. Matrix Biol 2022; 111:153-188. [PMID: 35764212 PMCID: PMC9990088 DOI: 10.1016/j.matbio.2022.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022]
Abstract
Fibrillar proteins are principal components of extracellular matrix (ECM) that confer mechanical properties to tissues. Fibrosis can result from wound repair in nearly every tissue in adults, and it associates with increased ECM density and crosslinking as well as increased tissue stiffness. Such fibrotic tissues are a major biomedical challenge, and an emerging view posits that the altered mechanical environment supports both synthetic and contractile myofibroblasts in a state of persistent activation. Here, we review the matrisome in several fibrotic diseases, as well as normal tissues, with a focus on physicochemical properties. Stiffness generally increases with the abundance of fibrillar collagens, the major constituent of ECM, with similar mathematical trends for fibrosis as well as adult tissues from soft brain to stiff bone and heart development. Changes in expression of other core matrisome and matrisome-associated proteins or proteoglycans contribute to tissue stiffening in fibrosis by organizing collagen, crosslinking ECM, and facilitating adhesion of myofibroblasts. Understanding how ECM composition and mechanics coevolve during fibrosis can lead to better models and help with antifibrotic therapies.
Collapse
Affiliation(s)
- Lawrence J Dooling
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Karanvir Saini
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Alişya A Anlaş
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Zhao X, Chen J, Sun H, Zhang Y, Zou D. New insights into fibrosis from the ECM degradation perspective: the macrophage-MMP-ECM interaction. Cell Biosci 2022; 12:117. [PMID: 35897082 PMCID: PMC9327238 DOI: 10.1186/s13578-022-00856-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 07/16/2022] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is a pathological feature of a variety of chronic inflammatory diseases that can affect almost all organs, which can cause severe consequences and even lead to death. Fibrosis is characterized by the excessive accumulation of extracellular matrix (ECM) due to disruption of the balance between ECM production and degradation. Although overabundance of ECM proteins has long been the focus of studies on fibrosis, another facet of the problem-impaired degradation of the ECM-is gaining increasing attention. Matrix metalloproteinase (MMP) and the tissue inhibitor of metalloproteinase (TIMP) system is the main molecular system contributing to ECM degradation, and macrophages are the major regulators of ECM. However, the relationship among macrophages, the MMP/TIMP system and the ECM is not fully understood in the context of fibrosis. Here, we discuss in detail the role played by the ECM in the development of fibrosis and highlight the macrophage-MMP-ECM interaction that is involved in fibrogenesis and may be a potential therapeutic target for fibrosis.
Collapse
Affiliation(s)
- Xiangyu Zhao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayin Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxiang Sun
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Chakarov S, Blériot C, Ginhoux F. Role of adipose tissue macrophages in obesity-related disorders. J Exp Med 2022; 219:213212. [PMID: 35543703 PMCID: PMC9098652 DOI: 10.1084/jem.20211948] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/04/2022] Open
Abstract
The obesity epidemic has led researchers and clinicians to reconsider the etiology of this disease and precisely decipher its molecular mechanisms. The excessive accumulation of fat by cells, most notably adipocytes, which play a key role in this process, has many repercussions in tissue physiology. Herein, we focus on how macrophages, immune cells well known for their tissue gatekeeping functions, assume fundamental, yet ill-defined, roles in the genesis and development of obesity-related metabolic disorders. We first discuss the determinants of the biology of these cells before introducing the specifics of the adipose tissue environment, while highlighting its heterogeneity. Finally, we detail how obesity transforms both adipose tissue and local macrophage populations. Understanding macrophage diversity and their cross talk with the diverse cell types constituting the adipose tissue environment will allow us to frame the therapeutic potential of adipose tissue macrophages in obesity.
Collapse
Affiliation(s)
- Svetoslav Chakarov
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Camille Blériot
- Institut Gustave Roussy, Batiment de Médecine Moléculaire, Villejuif, France
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Institut Gustave Roussy, Batiment de Médecine Moléculaire, Villejuif, France.,Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| |
Collapse
|
26
|
MFG-E8 Knockout Aggravated Nonalcoholic Steatohepatitis by Promoting the Activation of TLR4/NF- κB Signaling in Mice. Mediators Inflamm 2022; 2022:5791915. [PMID: 35769208 PMCID: PMC9236848 DOI: 10.1155/2022/5791915] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/23/2022] [Accepted: 06/04/2022] [Indexed: 12/30/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the common liver disease characterized by hepatic steatosis, inflammation, and fibrosis; there are no approved drugs to treat this disease because of incomplete understanding of pathophysiological mechanisms of NASH. Milk fat globule-epidermal growth factor-factor 8 (MFG-E8), a multifunctional glycoprotein, has shown anti-inflammation and antifibrosis. Here, MFG-E8 was shown to play a key role in NASH progression. Using methionine and choline deficient (MCD) diet-fed mice, we found MFG-E8 knockout exacerbated hepatic damage and steatosis as indicated by increased plasma transaminases activities and hepatic histopathologic change, higher hepatic triglycerides (TGs), and lipid accumulation. Moreover, liver fibrosis and inflammation elicited by MCD were aggravated in MFG-E8 knockout mice. Mechanistically, MFG-E8 knockout facilitated activation of hepatic toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway in MCD-fed mice. In vitro experiment, the TLR4 specific antagonist TAK-242 rescued palmitic acid- (PA-) primed lipid formation and inflammation in MFG-E8 knockout primary murine hepatocytes. These findings indicated that MFG-E8 is involved in the progression of NASH and the possible mechanism by which MFG-E8 knockout exacerbated NASH in mice is associated with activation of the TLR4/NF-κB signaling pathway.
Collapse
|
27
|
Overexpression of V-ATPase B2 attenuates lung injury/fibrosis by stabilizing lysosomal membrane permeabilization and increasing collagen degradation. Exp Mol Med 2022; 54:662-672. [PMID: 35624153 PMCID: PMC9166714 DOI: 10.1038/s12276-022-00776-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 11/29/2022] Open
Abstract
Excessive oxidative stress causes lysosomal membrane permeabilization (LMP), which leads to cell death. Vacuolar ATPase (V-ATPase) is the enzyme responsible for pumping H+ into the cytosol and thus maintaining intracellular pH. Previously, we reported that V-ATPase B2 subunit expression is upregulated in the TiO2-exposed lung epithelium. We investigated the role of the lysosomal V-ATPase B2 subunit in oxidative stress-induced alveolar epithelial cell death and in an experimental lung injury/fibrosis model. Overexpression of V-ATPase B2 increased lysosomal pH and lysosomal activities in the cells. In the presence of H2O2, overexpression of V-ATPase B2 increased survival, and silencing of V-ATPase B2 dramatically increased cell death. Overexpression of V-ATPase B2 diminished H2O2-triggered LMP, as evidenced by a reduction in acridine orange staining and leakage of cathepsin D from the lysosome to the cytoplasm. In addition, V-ATPase B2-overexpressing macrophages exhibited significantly enhanced uptake and degradation of collagen. V-ATPase B2-overexpressing transgenic mice showed significant inhibition of the bleomycin-induced increases in lung inflammation and fibrosis. We conclude that V-ATPase B2 is critical for maintaining lysosomal activities against excessive oxidative stress by stabilizing LMP. Our findings reveal a previously unknown role of this V-ATPase subunit in a lung injury and fibrosis model. An enzyme involved in maintaining the correct pH inside the lysosome, an organelle involved in disposal of cellular waste, also plays a critical role in preventing lung injury. Using human lung cells and mouse models of lung fibrosis, a team led by Sung Woo Park from Soonchunhyang University Bucheon Hospital, South Korea, showed that a overexpression of subunit of the vacuolar ATPase enzyme B2, which pumps protons into cellular compartments to create more acidic environments, helps to boost lysosomal activities that lead to prevent oxidative stress-induced cell death and alleviate experimental lung injury/fibrosis. In addition, V-ATPase B2 overexpressed macrophages increased collagen uptake and degradation activities. The findings point to the vacuolar ATPase, and its B2 subunit in particular, as a promising drug target for future treatments of pulmonary fibrosis.
Collapse
|
28
|
Bain CC, MacDonald AS. The impact of the lung environment on macrophage development, activation and function: diversity in the face of adversity. Mucosal Immunol 2022; 15:223-234. [PMID: 35017701 PMCID: PMC8749355 DOI: 10.1038/s41385-021-00480-w] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/04/2021] [Accepted: 12/18/2021] [Indexed: 02/04/2023]
Abstract
The last decade has been somewhat of a renaissance period for the field of macrophage biology. This renewed interest, combined with the advent of new technologies and development of novel model systems to assess different facets of macrophage biology, has led to major advances in our understanding of the diverse roles macrophages play in health, inflammation, infection and repair, and the dominance of tissue environments in influencing all of these areas. Here, we discuss recent developments in our understanding of lung macrophage heterogeneity, ontogeny, metabolism and function in the context of health and disease, and highlight core conceptual advances and key unanswered questions that we believe should be focus of work in the coming years.
Collapse
Affiliation(s)
- Calum C Bain
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh Bioquarter, Edinburgh, EH16 4TJ, UK.
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9NT, UK.
| |
Collapse
|
29
|
Qiu J, Li W, Mu R, Wang L, Guo L, Ma L. MFGE8 decreased neuronal apoptosis and neuroinflammation to ameliorate early brain injury induced by subarachnoid hemorrhage through the inhibition of HMGB1. Hum Exp Toxicol 2022; 41:9603271221093635. [PMID: 35503264 DOI: 10.1177/09603271221093635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AIM Both MFGE8 and HMGB1 were vital players for aneurysmal subarachnoid hemorrhage. However, whether HMGB1 was served as the downstream target of MFGE8 was unknown. To test this new mechanism, we performed the SAH model in rats. METHOD All treatments were injected intraventricularly into the right lateral ventricles. SAH grade, brain water content, and neurological function scores were evaluated. HMGB1 expression was studied by double immunofluorescence staining. HE and Nissl's staining were performed to observe the pathological change. Inflammatory factors were measured by ELISA method. RESULTS High expression of MFGE8 could improve neurological function and reduce the brain edema and pro-inflammatory factors. Injection of rhMFGE8 inhibited HMGB1. To further verify the regulation of MFGE8 in HMGB1, we used rhHMGB1 and glycyrrhizin, and the results indicated MFGE8 produced excellent effect on SAH rats via inhibiting HMGB1. CONCLUSION In a word, MFGE8 improved EBI caused by SAH, depending on HMGB1 that was the potential mechanism.
Collapse
Affiliation(s)
- Jiaoxue Qiu
- Department of Neurology, 519688Yantaishan Hospital, Yantai, China
| | - Wenna Li
- Department of Neurology, 519688Yantaishan Hospital, Yantai, China
| | - Rutao Mu
- Department of Interventional Medicine, 519688Yantaishan Hospital, Yantai, China
| | - Lingling Wang
- Department of Neurology, 519688Yantaishan Hospital, Yantai, China
| | - Lei Guo
- Department of Neurology, 519688Yantaishan Hospital, Yantai, China
| | - Lili Ma
- Department of Neurology, 519688Yantaishan Hospital, Yantai, China
| |
Collapse
|
30
|
Tao L, Xu H, He Q. Potential influences of expression levels of MFGE8 and HMGB1 on the intestinal mucosal barrier function and inflammatory response after blunt abdominal injury in rats. Acta Cir Bras 2022; 37:e370303. [PMID: 35674581 PMCID: PMC9161623 DOI: 10.1590/acb370303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/25/2022] [Indexed: 08/30/2023] Open
Abstract
Purpose: To explore the influence of milk fat globule-EGF factor 8 protein (MFGE8) on blunt abdominal injury in Sprague Dawley (SD) rats through the RhoA/ROCK signaling pathway. Methods: The blunt abdominal injury model was generated in SD rats. A total of 44 rats was randomly assigned into three groups. Rat blunt abdominal injury was assessed by the abbreviated injury scale (AIS). The rats were sacrificed for observing the morphology of the abdominal cavity and intestines. Hematoxylin and eosin staining was performed to visualize the pathological changes of rat intestines. Positive expressions of MFGE8 and high mobility group box 1 (HMGB1) in rat intestines were examined by immunohistochemical staining. Protein levels were determined by Western blot. Serum levels of tumor necrosis factor α (TNF-α), IL-1β, IL-6 and malondialdehyde (MDA) were measured by enzyme linked immunosorbent assay (ELISA). Results: Blunt abdominal injury resulted in inflammatory response of intestinal tissues, increased serum levels of TNF-α, IL-1β, IL-6 and MDA, upregulation of HMGB1, RhoA and ROCK2, and downregulation of MFGE8 in rats, which were significantly alleviated by intervention of rhMFGE8. Conclusions: MFGE8 protects the intestinal mucosal barrier function after blunt abdominal injury in rats by downregulating HMGB1. Moreover, it alleviates inflammatory response and oxidative stress caused by blunt abdominal injury in rats through downregulating RhoA and ROCK.
Collapse
Affiliation(s)
- Lijun Tao
- Hospital of Wenzhou Medical University, China
| | - Hongbo Xu
- Hospital of Wenzhou Medical University, China
| | - Qianggui He
- Hospital of Wenzhou Medical University, China
| |
Collapse
|
31
|
Soliman E, Bhalla S, Elhassanny AEM, Malur A, Ogburn D, Leffler N, Malur AG, Thomassen MJ. Myeloid ABCG1 Deficiency Enhances Apoptosis and Initiates Efferocytosis in Bronchoalveolar Lavage Cells of Murine Multi-Walled Carbon Nanotube-Induced Granuloma Model. Int J Mol Sci 2021; 23:ijms23010047. [PMID: 35008476 PMCID: PMC8744594 DOI: 10.3390/ijms23010047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/12/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
The use of carbon nanotubes has increased in the past few decades. Carbon nanotubes are implicated in the pathogenesis of pulmonary sarcoidosis, a chronic granulomatous inflammatory condition. We developed a murine model of chronic granulomatous inflammation using multiwall carbon nanotubes (MWCNT) to investigate mechanisms of granuloma formation. Using this model, we demonstrated that myeloid deficiency of ATP-binding cassette (ABC) cholesterol transporter (ABCG1) promotes granuloma formation and fibrosis with MWCNT instillation; however, the mechanism remains unclear. Our previous studies showed that MWCNT induced apoptosis in bronchoalveolar lavage (BAL) cells of wild-type (C57BL/6) mice. Given that continual apoptosis causes persistent severe lung inflammation, we hypothesized that ABCG1 deficiency would increase MWCNT-induced apoptosis thereby promoting granulomatous inflammation and fibrosis. To test our hypothesis, we utilized myeloid-specific ABCG1 knockout (ABCG1 KO) mice. Our results demonstrate that MWCNT instillation enhances pulmonary fibrosis in ABCG1 KO mice compared to wild-type controls. Enhanced fibrosis is indicated by increased trichrome staining and transforming growth factor-beta (TGF-β) expression in lungs, together with an increased expression of TGF-β related signaling molecules, interleukin-13 (IL-13) and Smad-3. MWCNT induced more apoptosis in BAL cells of ABCG1 KO mice. Initiation of apoptosis is most likely mediated by the extrinsic pathway since caspase 8 activity and Fas expression are significantly higher in MWCNT instilled ABCG1 KO mice compared to the wild type. In addition, TUNEL staining shows that ABCG1 KO mice instilled with MWCNT have a higher percentage of TUNEL positive BAL cells and more efferocytosis than the WT control. Furthermore, BAL cells of ABCG1 KO mice instilled with MWCNT exhibit an increase in efferocytosis markers, milk fat globule-EGF factor 8 (MFG-E8) and integrin β3. Therefore, our observations suggest that ABCG1 deficiency promotes pulmonary fibrosis by MWCNT, and this effect may be due to an increase in apoptosis and efferocytosis in BAL cells.
Collapse
Affiliation(s)
- Eman Soliman
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (E.S.); (S.B.); (A.M.); (D.O.); (N.L.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Sophia Bhalla
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (E.S.); (S.B.); (A.M.); (D.O.); (N.L.)
| | - Ahmed E. M. Elhassanny
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA;
| | - Anagha Malur
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (E.S.); (S.B.); (A.M.); (D.O.); (N.L.)
| | - David Ogburn
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (E.S.); (S.B.); (A.M.); (D.O.); (N.L.)
| | - Nancy Leffler
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (E.S.); (S.B.); (A.M.); (D.O.); (N.L.)
| | - Achut G. Malur
- Department of Microbiology & Immunology, St. George’s University, St. George 999166, Grenada;
| | - Mary Jane Thomassen
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (E.S.); (S.B.); (A.M.); (D.O.); (N.L.)
- Correspondence:
| |
Collapse
|
32
|
Staab-Weijnitz CA. Fighting the Fiber: Targeting Collagen in Lung Fibrosis. Am J Respir Cell Mol Biol 2021; 66:363-381. [PMID: 34861139 DOI: 10.1165/rcmb.2021-0342tr] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Organ fibrosis is characterized by epithelial injury and aberrant tissue repair, where activated effector cells, mostly fibroblasts and myofibroblasts, excessively deposit collagen into the extracellular matrix. Fibrosis frequently results in organ failure and has been estimated to contribute to at least one third of all global deaths. Also lung fibrosis, in particular idiopathic pulmonary fibrosis (IPF), is a fatal disease with rising incidence worldwide. As current treatment options targeting fibrogenesis are insufficient, there is an urgent need for novel therapeutic strategies. During the last decade, several studies have proposed to target intra- and extracellular components of the collagen biosynthesis, maturation, and degradation machinery. This includes intra- and extracellular targets directly acting on collagen gene products, but also such that anabolize essential building blocks of collagen, in particular glycine and proline biosynthetic enzymes. Collagen, however, is a ubiquitous molecule in the body and fulfils essential functions as a macromolecular scaffold, growth factor reservoir, and receptor binding site in virtually every tissue. This review summarizes recent advances and future directions in this field. Evidence for the proposed therapeutic targets and where they currently stand in terms of clinical drug development for treatment of fibrotic disease is provided. The drug targets are furthermore discussed in light of (1) specificity for collagen biosynthesis, maturation and degradation, and (2) specificity for disease-associated collagen. As therapeutic success and safety of these drugs may largely depend on targeted delivery, different strategies for specific delivery to the main effector cells and to the extracellular matrix are discussed.
Collapse
Affiliation(s)
- Claudia A Staab-Weijnitz
- Helmholtz Zentrum Munchen Deutsches Forschungszentrum fur Gesundheit und Umwelt, 9150, Comprehensive Pneumology Center/Institute of Lung Biology and Disease, Member of the German Center of Lung Research (DZL), München, Germany;
| |
Collapse
|
33
|
Henn D, Chen K, Fehlmann T, Trotsyuk AA, Sivaraj D, Maan ZN, Bonham CA, Barrera JA, Mays CJ, Greco AH, Moortgat Illouz SE, Lin JQ, Steele SR, Foster DS, Padmanabhan J, Momeni A, Nguyen D, Wan DC, Kneser U, Januszyk M, Keller A, Longaker MT, Gurtner GC. Xenogeneic skin transplantation promotes angiogenesis and tissue regeneration through activated Trem2 + macrophages. SCIENCE ADVANCES 2021; 7:eabi4528. [PMID: 34851663 PMCID: PMC8635426 DOI: 10.1126/sciadv.abi4528] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 10/09/2021] [Indexed: 05/07/2023]
Abstract
Skin allo- and xenotransplantation are the standard treatment for major burns when donor sites for autografts are not available. The relationship between the immune response to foreign grafts and their impact on wound healing has not been fully elucidated. Here, we investigated changes in collagen architecture after xenogeneic implantation of human biologic scaffolds. We show that collagen deposition in response to the implantation of human split-thickness skin grafts (hSTSGs) containing live cells recapitulates normal skin architecture, whereas human acellular dermal matrix (ADM) grafts led to a fibrotic collagen deposition. We show that macrophage differentiation in response to hSTSG implantation is driven toward regenerative Trem2+ subpopulations and found that hydrogel delivery of these cells significantly accelerated wound closure. Our study identifies the preclinical therapeutic potential of Trem2+ macrophages to mitigate fibrosis and promote wound healing, providing a novel effective strategy to develop advanced cell therapies for complex wounds.
Collapse
Affiliation(s)
- Dominic Henn
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Kellen Chen
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Artem A. Trotsyuk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Dharshan Sivaraj
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Zeshaan N. Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Clark A. Bonham
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Janos A. Barrera
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Chyna J. Mays
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Autumn H. Greco
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Sylvia E. Moortgat Illouz
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - John Qian Lin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Sydney R. Steele
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Deshka S. Foster
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Jagannath Padmanabhan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Arash Momeni
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Dung Nguyen
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Derrick C. Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Ulrich Kneser
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Geoffrey C. Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
34
|
Abstract
Obesity is a chronic and progressive process affecting whole-body energy balance and is associated with comorbidities development. In addition to increased fat mass, obesity induces white adipose tissue (WAT) inflammation and fibrosis, leading to local and systemic metabolic dysfunctions, such as insulin resistance (IR). Accordingly, limiting inflammation or fibrosis deposition may improve IR and glucose homeostasis. Although no targeted therapy yet exists to slow or reverse adipose tissue fibrosis, a number of findings have clarified the underlying cellular and molecular mechanisms. In this review, we highlight adipose tissue remodeling events shown to be associated with fibrosis deposition, with a focus on adipose progenitors involved in obesity-induced healthy as well as unhealthy WAT expansion. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Geneviève Marcelin
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; ,
| | | | - Karine Clément
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; , .,Nutrition Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
35
|
Wilkinson AL, John AE, Barrett JW, Gower E, Morrison VS, Man Y, Pun KT, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Fisher AJ, Procopiou PA, Hatley RJD, Barrett TN, Marshall RP, Macdonald SJF, Jenkins RG, Slack RJ. Pharmacological characterisation of GSK3335103, an oral αvβ6 integrin small molecule RGD-mimetic inhibitor for the treatment of fibrotic disease. Eur J Pharmacol 2021; 913:174618. [PMID: 34762934 DOI: 10.1016/j.ejphar.2021.174618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 11/03/2021] [Indexed: 11/18/2022]
Abstract
Fibrosis is the formation of scar tissue due to injury or long-term inflammation and is a leading cause of morbidity and mortality. Activation of the pro-fibrotic cytokine transforming growth factor-β (TGFβ) via the alpha-V beta-6 (αvβ6) integrin has been identified as playing a key role in the development of fibrosis. Therefore, a drug discovery programme to identify an orally bioavailable small molecule αvβ6 arginyl-glycinyl-aspartic acid (RGD)-mimetic was initiated. As part of a medicinal chemistry programme GSK3335103 was identified and profiled in a range of pre-clinical in vitro and in vivo systems. GSK3335103 was shown to bind to the αvβ6 with high affinity and demonstrated fast binding kinetics. In primary human lung epithelial cells, GSK3335103-induced concentration- and time-dependent internalisation of αvβ6 with a rapid return of integrin to the cell surface observed after washout. Following sustained engagement of the αvβ6 integrin in vitro, lysosomal degradation was induced by GSK3335103. GSK3335103 was shown to engage with the αvβ6 integrin and inhibit the activation of TGFβ in both ex vivo IPF tissue and in a murine model of bleomycin-induced lung fibrosis, as measured by αvβ6 engagement, TGFβ signalling and collagen deposition, with a prolonged duration of action observed in vivo. In summary, GSK3335103 is a potent αvβ6 inhibitor that attenuates TGFβ signalling in vitro and in vivo with a well-defined pharmacokinetic/pharmacodynamic relationship. This translates to a significant reduction of collagen deposition in vivo and therefore GSK3335103 represents a potential novel oral therapy for fibrotic disorders.
Collapse
Affiliation(s)
- Alex L Wilkinson
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Alison E John
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, UK
| | - John W Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - E Gower
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Valerie S Morrison
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Yim Man
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - K Tao Pun
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - James A Roper
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Jeni C Luckett
- Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, UK
| | - Lee A Borthwick
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Ben S Barksby
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Rachel A Burgoyne
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Rory Barnes
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Andrew J Fisher
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK; Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS, Foundation Trust, Newcastle Upon Tyne, UK
| | | | - Richard J D Hatley
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Tim N Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Richard P Marshall
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Simon J F Macdonald
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - R Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, UK
| | - Robert J Slack
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK.
| |
Collapse
|
36
|
An GH, Lee J, Jin X, Chung J, Kim JC, Park JH, Kim M, Han C, Kim JH, Woo DH. Truncated Milk Fat Globule-EGF-like Factor 8 Ameliorates Liver Fibrosis via Inhibition of Integrin-TGFβ Receptor Interaction. Biomedicines 2021; 9:biomedicines9111529. [PMID: 34829758 PMCID: PMC8615163 DOI: 10.3390/biomedicines9111529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/27/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
Milk fat globule-EGF factor 8 (MFG-E8) protein is known as an immunomodulator in various diseases, and we previously demonstrated the anti-fibrotic role of MFG-E8 in liver disease. Here, we present a truncated form of MFG-E8 that provides an advanced therapeutic benefit in treating liver fibrosis. The enhanced therapeutic potential of the modified MFG-E8 was demonstrated in various liver fibrosis animal models, and the efficacy was further confirmed in human hepatic stellate cells and a liver spheroid model. In the subsequent analysis, we found that the modified MFG-E8 more efficiently suppressed transforming growth factor β (TGF-β) signaling than the original form of MFG-E8, and it deactivated the proliferation of hepatic stellate cells in the liver disease environment through interfering with the interactions between integrins (αvβ3 & αvβ5) and TGF-βRI. Furthermore, the protein preferentially delivered in the liver after administration, and the safety profiles of the protein were demonstrated in male and female rat models. Therefore, in conclusion, this modified MFG-E8 provides a promising new therapeutic strategy for treating fibrotic diseases.
Collapse
Affiliation(s)
- Geun Ho An
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Science Campus, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Jaehun Lee
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
| | - Xiong Jin
- School of Pharmacy, Henan University, Jin Ming Ave, Kaifeng 475004, China;
| | - Jinwoo Chung
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
| | - Joon-Chul Kim
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
| | - Jung-Hyuck Park
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
| | - Minkyung Kim
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
| | - Choongseong Han
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Science Campus, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
- Correspondence: (J.-H.K.); (D.-H.W.)
| | - Dong-Hun Woo
- Department of New Drug Development, NEXEL Co., Ltd., 8th Floor, 55 Magokdong-ro, Gangseo-gu, Seoul 07802, Korea; (G.H.A.); (J.L.); (J.C.); (J.-C.K.); (J.-H.P.); (M.K.); (C.H.)
- Correspondence: (J.-H.K.); (D.-H.W.)
| |
Collapse
|
37
|
Decaris ML, Schaub JR, Chen C, Cha J, Lee GG, Rexhepaj M, Ho SS, Rao V, Marlow MM, Kotak P, Budi EH, Hooi L, Wu J, Fridlib M, Martin SP, Huang S, Chen M, Muñoz M, Hom TF, Wolters PJ, Desai TJ, Rock F, Leftheris K, Morgans DJ, Lepist EI, Andre P, Lefebvre EA, Turner SM. Dual inhibition of α vβ 6 and α vβ 1 reduces fibrogenesis in lung tissue explants from patients with IPF. Respir Res 2021; 22:265. [PMID: 34666752 PMCID: PMC8524858 DOI: 10.1186/s12931-021-01863-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022] Open
Abstract
RATIONALE αv integrins, key regulators of transforming growth factor-β activation and fibrogenesis in in vivo models of pulmonary fibrosis, are expressed on abnormal epithelial cells (αvβ6) and fibroblasts (αvβ1) in fibrotic lungs. OBJECTIVES We evaluated multiple αv integrin inhibition strategies to assess which most effectively reduced fibrogenesis in explanted lung tissue from patients with idiopathic pulmonary fibrosis. METHODS Selective αvβ6 and αvβ1, dual αvβ6/αvβ1, and multi-αv integrin inhibitors were characterized for potency, selectivity, and functional activity by ligand binding, cell adhesion, and transforming growth factor-β cell activation assays. Precision-cut lung slices generated from lung explants from patients with idiopathic pulmonary fibrosis or bleomycin-challenged mouse lungs were treated with integrin inhibitors or standard-of-care drugs (nintedanib or pirfenidone) and analyzed for changes in fibrotic gene expression or TGF-β signaling. Bleomycin-challenged mice treated with dual αvβ6/αvβ1 integrin inhibitor, PLN-74809, were assessed for changes in pulmonary collagen deposition and Smad3 phosphorylation. MEASUREMENTS AND MAIN RESULTS Inhibition of integrins αvβ6 and αvβ1 was additive in reducing type I collagen gene expression in explanted lung tissue slices from patients with idiopathic pulmonary fibrosis. These data were replicated in fibrotic mouse lung tissue, with no added benefit observed from inhibition of additional αv integrins. Antifibrotic efficacy of dual αvβ6/αvβ1 integrin inhibitor PLN-74809 was confirmed in vivo, where dose-dependent inhibition of pulmonary Smad3 phosphorylation and collagen deposition was observed. PLN-74809 also, more potently, reduced collagen gene expression in fibrotic human and mouse lung slices than clinically relevant concentrations of nintedanib or pirfenidone. CONCLUSIONS In the fibrotic lung, dual inhibition of integrins αvβ6 and αvβ1 offers the optimal approach for blocking fibrogenesis resulting from integrin-mediated activation of transforming growth factor-β.
Collapse
Affiliation(s)
| | | | - Chun Chen
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Jacob Cha
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Gail G Lee
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Steve S Ho
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Vikram Rao
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Prerna Kotak
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Erine H Budi
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Lisa Hooi
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Jianfeng Wu
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | | | - Shaoyi Huang
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Ming Chen
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Manuel Muñoz
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Tushar J Desai
- Department of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - David J Morgans
- Pliant Therapeutics, South San Francisco, CA, USA
- Maze Therapeutics, South San Francisco, CA, USA
| | | | - Patrick Andre
- Pliant Therapeutics, South San Francisco, CA, USA
- Acceleron Pharma, Cambridge, MA, USA
| | | | | |
Collapse
|
38
|
Song S, Fu Z, Guan R, Zhao J, Yang P, Li Y, Yin H, Lai Y, Gong G, Zhao S, Yu J, Peng X, He Y, Luo Y, Zhong N, Su J. Intracellular hydroxyproline imprinting following resolution of bleomycin-induced pulmonary fibrosis. Eur Respir J 2021; 59:13993003.00864-2021. [PMID: 34561295 PMCID: PMC9068975 DOI: 10.1183/13993003.00864-2021] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/14/2021] [Indexed: 11/05/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with few treatment options. The poor success in developing anti-IPF strategies have impelled researchers to reconsider the importance of choice for animal model and assessment methodologies. Currently, it is still not settled whether the bleomycin-induced lung fibrosis mouse model finally returns to resolution.This study aimed to follow the dynamic fibrotic features of BLM (Bleomycin)-treated mouse lungs with extended durations through a combination of the latest technologies (micro-CT imaging and histological detection of degraded collagens) with traditional methods. In addition, we also applied immunohistochemistry to explore the distribution of all hydroxyproline-containing molecules.As determined by classical biochemical method, total lung hydroxyproline contents reached peak at 4-week after bleomycin injury and maintained a steady high level thereafter until the end of the experiments (16-week). This result seemed to partially contradict with the changes of other fibrosis evaluation parameters, which indicated a gradual degradation of collagens and a recovery of lung aeration post the fibrosis peak. This inconsistency was well reconciled by our data from immunostaining against hydroxyproline and a fluorescent peptide staining against degraded collagen, together showing large amounts of hydroxyproline-rich degraded collagen fragments detained and enriched within the intracellular regions at 10- or 16-week, rather than at 4-week post the BLM-treatment. Hence, our present data not only offer respiratory researchers a new perspective towards the resolution nature of mouse lung fibrosis, but also remind them to be cautious while using hydroxyproline content assay to evaluate the severity of fibrosis.
Collapse
Affiliation(s)
- Shengren Song
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China.,State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,These authors contributed equally to this work
| | - Zhenli Fu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,These authors contributed equally to this work
| | - Ruijuan Guan
- Shenzhen International Institute for Biomedical Research, Shenzhen, Guangdong, China.,These authors contributed equally to this work
| | - Jie Zhao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,These authors contributed equally to this work
| | - Penghui Yang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,These authors contributed equally to this work
| | - Yang Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hang Yin
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunxin Lai
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gencheng Gong
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Simin Zhao
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiangtian Yu
- Shenzhen International Institute for Biomedical Research, Shenzhen, Guangdong, China
| | - Xiaomin Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ying He
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yumei Luo
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Nanshan Zhong
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China .,State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jin Su
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China .,Shenzhen International Institute for Biomedical Research, Shenzhen, Guangdong, China
| |
Collapse
|
39
|
Disrupting biological sensors of force promotes tissue regeneration in large organisms. Nat Commun 2021; 12:5256. [PMID: 34489407 PMCID: PMC8421385 DOI: 10.1038/s41467-021-25410-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 08/06/2021] [Indexed: 12/31/2022] Open
Abstract
Tissue repair and healing remain among the most complicated processes that occur during postnatal life. Humans and other large organisms heal by forming fibrotic scar tissue with diminished function, while smaller organisms respond with scarless tissue regeneration and functional restoration. Well-established scaling principles reveal that organism size exponentially correlates with peak tissue forces during movement, and evolutionary responses have compensated by strengthening organ-level mechanical properties. How these adaptations may affect tissue injury has not been previously examined in large animals and humans. Here, we show that blocking mechanotransduction signaling through the focal adhesion kinase pathway in large animals significantly accelerates wound healing and enhances regeneration of skin with secondary structures such as hair follicles. In human cells, we demonstrate that mechanical forces shift fibroblasts toward pro-fibrotic phenotypes driven by ERK-YAP activation, leading to myofibroblast differentiation and excessive collagen production. Disruption of mechanical signaling specifically abrogates these responses and instead promotes regenerative fibroblast clusters characterized by AKT-EGR1. Humans and other large mammals heal wounds by forming fibrotic scar tissue with diminished function. Here, the authors show that disrupting mechanotransduction through the focal adhesion kinase pathway in large animals accelerates healing, prevents fibrosis, and enhances skin regeneration.
Collapse
|
40
|
Tang Z, Gao J, Wu J, Zeng G, Liao Y, Song Z, Liang X, Hu J, Hu Y, Liu M, Li N. Human umbilical cord mesenchymal stromal cells attenuate pulmonary fibrosis via regulatory T cell through interaction with macrophage. Stem Cell Res Ther 2021; 12:397. [PMID: 34256845 PMCID: PMC8278716 DOI: 10.1186/s13287-021-02469-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/21/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a growing clinical problem with limited therapeutic options. Human umbilical cord mesenchymal stromal cell (hucMSC) therapy is being investigated in clinical trials for the treatment of PF patients. However, little is known about the underlying molecular and cellular mechanisms of hucMSC therapy on PF. In this study, the molecular and cellular behavior of hucMSC was investigated in a bleomycin-induced mouse PF model. METHODS The effect of hucMSCs on mouse lung regeneration was determined by detecting Ki67 expression and EdU incorporation in alveolar type 2 (AT2) and lung fibroblast cells. hucMSCs were transfected to express the membrane localized GFP before transplant into the mouse lung. The cellular behavior of hucMSCs in mouse lung was tracked by GFP staining. Single cell RNA sequencing was performed to investigate the effects of hucMSCs on gene expression profiles of macrophages after bleomycin treatment. RESULTS hucMSCs could alleviate collagen accumulation in lung and decrease the mortality of mouse induced by bleomycin. hucMSC transplantation promoted AT2 cell proliferation and inhibited lung fibroblast cell proliferation. By using single cell RNA sequencing, a subcluster of interferon-sensitive macrophages (IFNSMs) were identified after hucMSC infusion. These IFNSMs elevate the secretion of CXCL9 and CXCL10 following hucMSC infusion and recruit more Treg cells to the injured lung. CONCLUSIONS Our study establishes a link between hucMSCs, macrophage, Treg, and PF. It provides new insights into how hucMSCs interact with macrophage during the repair process of bleomycin-induced PF and play its immunoregulation function.
Collapse
Affiliation(s)
- Zan Tang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, Guangdong, China
| | - Junxiao Gao
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jie Wu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, Guangdong, China
| | - Guifang Zeng
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, Guangdong, China
| | - Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, Guangdong, China
| | - Zhenkun Song
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, Guangdong, China
| | - Xiao Liang
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, Guangdong, China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, Guangdong, China
| | - Yong Hu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Muyun Liu
- National-Local Associated Engineering Laboratory for Personalized Cell Therapy, Shenzhen, Guangdong, China.
| | - Nan Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
41
|
Merkt W, Zhou Y, Han H, Lagares D. Myofibroblast fate plasticity in tissue repair and fibrosis: Deactivation, apoptosis, senescence and reprogramming. Wound Repair Regen 2021; 29:678-691. [PMID: 34117675 DOI: 10.1111/wrr.12952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
In response to tissue injury, fibroblasts differentiate into professional repair cells called myofibroblasts, which orchestrate many aspects of the normal tissue repair programme including synthesis, deposition and contraction of extracellular matrix proteins, leading to wound closure. Successful tissue repair responses involve termination of myofibroblast activities in order to prevent pathologic fibrotic scarring. Here, we discuss the cellular and molecular mechanisms limiting myofibroblast activities during physiological tissue repair, including myofibroblast deactivation, apoptosis, reprogramming and immune clearance of senescent myofibroblasts. In addition, we summarize pathological mechanisms leading to myofibroblast persistence and survival, a hallmark of fibrotic diseases. Finally, we discuss emerging anti-fibrotic therapies aimed at targeting myofibroblast fate such as senolytics, gene therapy, cellular immunotherapy and CAR-T cells.
Collapse
Affiliation(s)
- Wolfgang Merkt
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Yan Zhou
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Physiology, Xiangya Medical School, Central South University, Changsha, China
| | - Hongwei Han
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David Lagares
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Onursal C, Dick E, Angelidis I, Schiller HB, Staab-Weijnitz CA. Collagen Biosynthesis, Processing, and Maturation in Lung Ageing. Front Med (Lausanne) 2021; 8:593874. [PMID: 34095157 PMCID: PMC8172798 DOI: 10.3389/fmed.2021.593874] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 03/24/2021] [Indexed: 12/15/2022] Open
Abstract
In addition to providing a macromolecular scaffold, the extracellular matrix (ECM) is a critical regulator of cell function by virtue of specific physical, biochemical, and mechanical properties. Collagen is the main ECM component and hence plays an essential role in the pathogenesis and progression of chronic lung disease. It is well-established that many chronic lung diseases, e.g., chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) primarily manifest in the elderly, suggesting increased susceptibility of the aged lung or accumulated alterations in lung structure over time that favour disease. Here, we review the main steps of collagen biosynthesis, processing, and turnover and summarise what is currently known about alterations upon lung ageing, including changes in collagen composition, modification, and crosslinking. Recent proteomic data on mouse lung ageing indicates that, while the ER-resident machinery of collagen biosynthesis, modification and triple helix formation appears largely unchanged, there are specific changes in levels of type IV and type VI as well as the two fibril-associated collagens with interrupted triple helices (FACIT), namely type XIV and type XVI collagens. In addition, levels of the extracellular collagen crosslinking enzyme lysyl oxidase are decreased, indicating less enzymatically mediated collagen crosslinking upon ageing. The latter contrasts with the ageing-associated increase in collagen crosslinking by advanced glycation endproducts (AGEs), a result of spontaneous reactions of protein amino groups with reactive carbonyls, e.g., from monosaccharides or reactive dicarbonyls like methylglyoxal. Given the slow turnover of extracellular collagen such modifications accumulate even more in ageing tissues. In summary, the collective evidence points mainly toward age-induced alterations in collagen composition and drastic changes in the molecular nature of collagen crosslinks. Future work addressing the consequences of these changes may provide important clues for prevention of lung disease and for lung bioengineering and ultimately pave the way to novel targeted approaches in lung regenerative medicine.
Collapse
Affiliation(s)
- Ceylan Onursal
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Elisabeth Dick
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Ilias Angelidis
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Herbert B Schiller
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Claudia A Staab-Weijnitz
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
43
|
Arai H, Sato Y, Yanagita M. Fibroblast heterogeneity and tertiary lymphoid tissues in the kidney. Immunol Rev 2021; 302:196-210. [PMID: 33951198 PMCID: PMC8360208 DOI: 10.1111/imr.12969] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
Fibroblasts reside in various organs and support tissue structure and homeostasis under physiological conditions. Phenotypic alterations of fibroblasts underlie the development of diverse pathological conditions, including organ fibrosis. Recent advances in single‐cell biology have revealed that fibroblasts comprise heterogeneous subpopulations with distinct phenotypes, which exert both beneficial and detrimental effects on the host organs in a context‐dependent manner. In the kidney, phenotypic alterations of resident fibroblasts provoke common pathological conditions of chronic kidney disease (CKD), such as renal anemia and peritubular capillary loss. Additionally, in aged injured kidneys, fibroblasts provide functional and structural supports for tertiary lymphoid tissues (TLTs), which serve as the ectopic site of acquired immune reactions in various clinical contexts. TLTs are closely associated with aging and CKD progression, and the developmental stages of TLTs reflect the severity of renal injury. In this review, we describe the current understanding of fibroblast heterogeneity both under physiological and pathological conditions, with special emphasis on fibroblast contribution to TLT formation in the kidney. Dissecting the heterogeneous characteristics of fibroblasts will provide a promising therapeutic option for fibroblast‐related pathological conditions, including TLT formation.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Sato
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Medical Innovation Center, TMK Project, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
44
|
Datta R, Lizama CO, Soltani AK, Mckleroy W, Podolsky MJ, Yang CD, Huynh TL, Cautivo KM, Wang B, Koliwad SK, Abumrad NA, Atabai K. Autoregulation of insulin receptor signaling through MFGE8 and the αvβ5 integrin. Proc Natl Acad Sci U S A 2021; 118:e2102171118. [PMID: 33903257 PMCID: PMC8106306 DOI: 10.1073/pnas.2102171118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The role of integrins, in particular αv integrins, in regulating insulin resistance is incompletely understood. We have previously shown that the αvβ5 integrin ligand milk fat globule epidermal growth factor like 8 (MFGE8) regulates cellular uptake of fatty acids. In this work, we evaluated the impact of MFGE8 on glucose homeostasis. We show that acute blockade of the MFGE8/β5 pathway enhances while acute augmentation dampens insulin-stimulated glucose uptake. Moreover, we find that insulin itself induces cell-surface enrichment of MFGE8 in skeletal muscle, which then promotes interaction between the αvβ5 integrin and the insulin receptor leading to dampening of skeletal-muscle insulin receptor signaling. Blockade of the MFGE8/β5 pathway also enhances hepatic insulin sensitivity. Our work identifies an autoregulatory mechanism by which insulin-stimulated signaling through its cognate receptor is terminated through up-regulation of MFGE8 and its consequent interaction with the αvβ5 integrin, thereby establishing a pathway that can potentially be targeted to improve insulin sensitivity.
Collapse
Affiliation(s)
- Ritwik Datta
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
| | - Carlos O Lizama
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
| | - Amin K Soltani
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Lung Biology Center, University of California, San Francisco, CA 94158
| | - William Mckleroy
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Lung Biology Center, University of California, San Francisco, CA 94158
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
| | - Michael J Podolsky
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
| | - Christopher D Yang
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
| | - Tony L Huynh
- Department of Radiology and Biomedical imaging, University of California, San Francisco, CA 94107
| | - Kelly M Cautivo
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Biao Wang
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Department of Physiology, University of California, San Francisco, CA 94158
| | - Suneil K Koliwad
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
- Diabetes Center, University of California, San Francisco, CA 94143
| | - Nada A Abumrad
- Diabetes Research Center, Department of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO 63110
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158;
- Lung Biology Center, University of California, San Francisco, CA 94158
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
- Department of Physiology, University of California, San Francisco, CA 94158
| |
Collapse
|
45
|
Qu J, Yang SZ, Zhu Y, Guo T, Thannickal VJ, Zhou Y. Targeting mechanosensitive MDM4 promotes lung fibrosis resolution in aged mice. J Exp Med 2021; 218:e20202033. [PMID: 33688918 PMCID: PMC7953267 DOI: 10.1084/jem.20202033] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/18/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
Aging is a strong risk factor and an independent prognostic factor for progressive human idiopathic pulmonary fibrosis (IPF). Aged mice develop nonresolving pulmonary fibrosis following lung injury. In this study, we found that mouse double minute 4 homolog (MDM4) is highly expressed in the fibrotic lesions of human IPF and experimental pulmonary fibrosis in aged mice. We identified MDM4 as a matrix stiffness-regulated endogenous inhibitor of p53. Reducing matrix stiffness down-regulates MDM4 expression, resulting in p53 activation in primary lung myofibroblasts isolated from IPF patients. Gain of p53 function activates a gene program that sensitizes lung myofibroblasts to apoptosis and promotes the clearance of apoptotic myofibroblasts by macrophages. Destiffening of the fibrotic lung matrix by targeting nonenzymatic cross-linking or genetic ablation of Mdm4 in lung (myo)fibroblasts activates the Mdm4-p53 pathway and promotes lung fibrosis resolution in aged mice. These findings suggest that mechanosensitive MDM4 is a molecular target with promising therapeutic potential against persistent lung fibrosis associated with aging.
Collapse
Affiliation(s)
- Jing Qu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shan-Zhong Yang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Yi Zhu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ting Guo
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
- The Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Victor J. Thannickal
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Yong Zhou
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
46
|
Buechler MB, Fu W, Turley SJ. Fibroblast-macrophage reciprocal interactions in health, fibrosis, and cancer. Immunity 2021; 54:903-915. [PMID: 33979587 DOI: 10.1016/j.immuni.2021.04.021] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts and macrophages are present in all tissues, and mounting evidence supports that these cells engage in direct communication to influence the overall tissue microenvironment and affect disease outcomes. Here, we review the current understanding of the molecular mechanisms that underlie fibroblast-macrophage interactions in health, fibrosis, and cancer. We present an integrated view of fibroblast-macrophage interactions that is centered on the CSF1-CSF1R axis and discuss how additional molecular programs linking these cell types can underpin disease onset, progression, and resolution. These programs may be tissue and context dependent, affected also by macrophage and fibroblast origin and state, as seen most clearly in cancer. Continued efforts to understand these cells and the means by which they interact may provide therapeutic approaches for the treatment of fibrosis and cancer.
Collapse
Affiliation(s)
- Matthew B Buechler
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| | - Wenxian Fu
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
47
|
Human pluripotent stem-cell-derived alveolar organoids for modeling pulmonary fibrosis and drug testing. Cell Death Discov 2021; 7:48. [PMID: 33723255 PMCID: PMC7961057 DOI: 10.1038/s41420-021-00439-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/19/2021] [Accepted: 02/13/2021] [Indexed: 02/06/2023] Open
Abstract
Detailed understanding of the pathogenesis and development of effective therapies for pulmonary fibrosis (PF) have been hampered by lack of in vitro human models that recapitulate disease pathophysiology. In this study, we generated alveolar organoids (AOs) derived from human pluripotent stem cells (hPSCs) for use as an PF model and for drug efficacy evaluation. Stepwise direct differentiation of hPSCs into alveolar epithelial cells by mimicking developmental cues in a temporally controlled manner was used to generate multicellular AOs. Derived AOs contained the expected spectrum of differentiated cells, including alveolar progenitors, type 1 and 2 alveolar epithelial cells and mesenchymal cells. Treatment with transforming growth factor (TGF-β1) induced fibrotic changes in AOs, offering a PF model for therapeutic evaluation of a structurally truncated form (NP-011) of milk fat globule-EGF factor 8 (MFG-E8) protein. The significant fibrogenic responses and collagen accumulation that were induced by treatment with TGF-β1 in these AOs were effectively ameliorated by treatment with NP-011 via suppression of extracellular signal-regulated kinase (ERK) signaling. Furthermore, administration of NP-011 reversed bleomycin-induced lung fibrosis in mice also via ERK signaling suppression and collagen reduction. This anti-fibrotic effect mirrored that following Pirfenidone and Nintedanib administration. Furthermore, NP-011 interacted with macrophages, which accelerated the collagen uptake for eliminating accumulated collagen in fibrotic lung tissues. This study provides a robust in vitro human organoid system for modeling PF and assessing anti-fibrotic mechanisms of potential drugs and suggests that modified MGF-E8 protein has therapeutic potential for treating PF.
Collapse
|
48
|
Atabai K, Yang CD, Podolsky MJ. You Say You Want a Resolution (of Fibrosis). Am J Respir Cell Mol Biol 2020; 63:424-435. [PMID: 32640171 DOI: 10.1165/rcmb.2020-0182tr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In pathological fibrosis, aberrant tissue remodeling with excess extracellular matrix leads to organ dysfunction and eventual morbidity. Diseases of fibrosis create significant global health and economic burdens and are often deadly. Although fibrosis has traditionally been thought of as an irreversible process, a growing body of evidence demonstrates that organ fibrosis can reverse in certain circumstances, especially if an underlying cause of injury can be removed. This body of evidence has uncovered more and more contributors to persistent and nonresolving tissue fibrosis. Here, we review the present knowledge on resolution of organ fibrosis and restoration of near-normal tissue architecture. We emphasize three critical areas of tissue homeostasis that are necessary for fibrosis resolution, namely, the elimination of matrix-producing cells, the clearance of excess matrix, and the regeneration of normal tissue constituents. In so doing, we also highlight how profibrotic pathways interact with one another and where there may be therapeutic opportunities to intervene and remediate pathological persistent fibrosis.
Collapse
Affiliation(s)
- Kamran Atabai
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | - Michael J Podolsky
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
49
|
Shi T, Denney L, An H, Ho LP, Zheng Y. Alveolar and lung interstitial macrophages: Definitions, functions, and roles in lung fibrosis. J Leukoc Biol 2020; 110:107-114. [PMID: 33155728 DOI: 10.1002/jlb.3ru0720-418r] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Mϕs are the main innate immune cells in the lung at homeostasis, with important roles in host defence and immune modulation. Alveolar Mϕs (AMs) and interstitial Mϕs (IMs) are the two lung Mϕ subsets, so called according to the sites they reside in. These subsets are also defined by their origins and immunological microenvironment, which endow these cells with distinct features and plasticity. This review summarizes the latest definitions and functions of lung Mϕs during homeostasis and provides exemplar of their divergent roles in lung fibrosis.
Collapse
Affiliation(s)
- Ting Shi
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Laura Denney
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Huazhang An
- Clinical Cancer Institute, Center of Translational Medicine, Second Military Medical University, Shanghai, China
| | - Ling-Pei Ho
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Yuejuan Zheng
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Key Laboratory of Health Identification and Assessment, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
50
|
Ogger PP, Albers GJ, Hewitt RJ, O'Sullivan BJ, Powell JE, Calamita E, Ghai P, Walker SA, McErlean P, Saunders P, Kingston S, Molyneaux PL, Halket JM, Gray R, Chambers DC, Maher TM, Lloyd CM, Byrne AJ. Itaconate controls the severity of pulmonary fibrosis. Sci Immunol 2020; 5:5/52/eabc1884. [PMID: 33097591 DOI: 10.1126/sciimmunol.abc1884] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/29/2020] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease in which airway macrophages (AMs) play a key role. Itaconate has emerged as a mediator of macrophage function, but its role during fibrosis is unknown. Here, we reveal that itaconate is an endogenous antifibrotic factor in the lung. Itaconate levels are reduced in bronchoalveolar lavage, and itaconate-synthesizing cis-aconitate decarboxylase expression (ACOD1) is reduced in AMs from patients with IPF compared with controls. In the murine bleomycin model of pulmonary fibrosis, Acod1-/- mice develop persistent fibrosis, unlike wild-type (WT) littermates. Profibrotic gene expression is increased in Acod1-/- tissue-resident AMs compared with WT, and adoptive transfer of WT monocyte-recruited AMs rescued mice from disease phenotype. Culture of lung fibroblasts with itaconate decreased proliferation and wound healing capacity, and inhaled itaconate was protective in mice in vivo. Collectively, these data identify itaconate as critical for controlling the severity of lung fibrosis, and targeting this pathway may be a viable therapeutic strategy.
Collapse
Affiliation(s)
- Patricia P Ogger
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Gesa J Albers
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Richard J Hewitt
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Brendan J O'Sullivan
- Queensland Lung Transplant Service, Prince Charles Hospital, Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Joseph E Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.,Cellular Genomics Futures Institute, University of New South Wales, Kensington, Sydney, Australia
| | - Emily Calamita
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Poonam Ghai
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Simone A Walker
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Peter McErlean
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Peter Saunders
- Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Shaun Kingston
- Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - John M Halket
- Mass Spectrometry Facility King's College London, London SE1 9NH, UK
| | - Robert Gray
- Mass Spectrometry Facility King's College London, London SE1 9NH, UK
| | - Daniel C Chambers
- Queensland Lung Transplant Service, Prince Charles Hospital, Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK.,Hastings Centre for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK. .,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| |
Collapse
|