1
|
Dong X, Xiang H, Li J, Hao A, Wang H, Gou Y, Li A, Rahaman S, Qiu Y, Li J, Mei O, Zhong J, You W, Shen G, Wu X, Li J, Shu Y, Shi LL, Zhu Y, Reid RR, He TC, Fan J. Dermal fibroblast-derived extracellular matrix (ECM) synergizes with keratinocytes in promoting re-epithelization and scarless healing of skin wounds: Towards optimized skin tissue engineering. Bioact Mater 2025; 47:1-17. [PMID: 39872210 PMCID: PMC11762682 DOI: 10.1016/j.bioactmat.2024.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025] Open
Abstract
Skin serves as the first-order protective barrier against the environment and any significant disruptions in skin integrity must be promptly restored. Despite significant advances in therapeutic strategies, effective management of large chronic skin wounds remains a clinical challenge. Dermal fibroblasts are the primary cell type responsible for remodeling the extracellular matrix (ECM) in wound healing. Here, we investigated whether ECM derived from exogenous fibroblasts, in combination with keratinocytes, promoted scarless cutaneous wound healing. To overcome the limited lifespan of primary dermal fibroblasts, we established reversibly immortalized mouse dermal fibroblasts (imDFs), which were non-tumorigenic, expressed dermal fibroblast markers, and were responsive to TGF-β1 stimulation. The decellularized ECM prepared from both imDFs and primary dermal fibroblasts shared similar expression profiles of extracellular matrix proteins and promoted the proliferation of keratinocyte (iKera) cells. The imDFs-derived ECM solicited no local immune response. While the ECM and to a lesser extent imDFs enhanced skin wound healing with excessive fibrosis, a combination of imDFs-derived ECM and iKera cells effectively promoted the re-epithelization and scarless healing of full-thickness skin wounds. These findings strongly suggest that dermal fibroblast-derived ECM, not fibroblasts themselves, may synergize with keratinocytes in regulating scarless healing and re-epithelialization of skin wounds. Given its low immunogenic nature, imDFs-derived ECM should be a valuable resource of skin-specific biomaterial for wound healing and skin tissue engineering.
Collapse
Affiliation(s)
- Xiangyu Dong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Han Xiang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiajia Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ailing Hao
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hao Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Aohua Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Saidur Rahaman
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yiheng Qiu
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiahao Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ou Mei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopedic Surgery, Jiangxi Hospital of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Wulin You
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedic Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China
| | - Guowei Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedic Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210019, China
| | - Xingye Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jingjing Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Oncology, The Affiliated Hospital of Shandong Second Medical University, Weifang, 261053, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Stem Cell Biology and Therapy Laboratory of the Pediatric Research Institute, the National Clinical Research Center for Child Health and Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, the Children's Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Western Institute of Digital-Intelligent Medicine, Chongqing, 401329, China
| |
Collapse
|
2
|
Jimenez SA, Mendoza FA, Piera-Velazquez S. A review of recent studies on the pathogenesis of Systemic Sclerosis: focus on fibrosis pathways. Front Immunol 2025; 16:1551911. [PMID: 40308583 PMCID: PMC12040652 DOI: 10.3389/fimmu.2025.1551911] [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: 12/26/2024] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology characterized by the development of frequently progressive cutaneous and internal organ fibrosis accompanied by severe vascular alterations. The pathogenesis of SSc is highly complex and, despite extensive investigation, has not been fully elucidated. Numerous studies have suggested that unknown etiologic factors cause multiple alterations in genetically receptive hosts, leading to SSc development and progression. These events may be functionally and pathologically interconnected and include: 1) Structural and functional microvascular and endothelial cell abnormalities; 2) Severe oxidative stress and high reactive oxygen species (3); Frequently progressive cutaneous and visceral fibrosis; 4) Transdifferentiation of various cell types into activated myofibroblasts, the cells ultimately responsible for the fibrotic process; 5) Establishment of a chronic inflammatory process in various affected tissues; 6) Release of cytokines, chemokines, and growth factors from the inflammatory cells; 7) Abnormalities in humoral and cellular immunity with the production of specific autoantibodies; and 8) Epigenetic alterations including changes in multiple non-coding RNAs. These events manifest with different levels of intensity in the affected organs and display remarkable individual variability, resulting in a wide heterogeneity in the extent and severity of clinical manifestations. Here, we will review some of the recent studies related to SSc pathogenesis.
Collapse
Affiliation(s)
- Sergio A. Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Fabian A. Mendoza
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
- Division of Rheumatology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
3
|
Sun J, Liu Y, Sun J, Ding J, Chen X. Biomaterials‐Involved Construction of Extracellular Matrices for Tumor Blockade Therapy. EXPLORATION 2025. [DOI: 10.1002/exp.20240229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/28/2025] [Indexed: 05/14/2025]
Abstract
ABSTRACTExtracellular matrices (ECMs) play a crucial role in the onset and progression of tumors by providing structural support and promoting the proliferation and metastases of tumor cells. Current therapeutic approaches targeting tumor ECMs focus on two main strategies: Inhibiting matrix degradation to prevent metastases and facilitating matrix degradation to enhance the penetration of drugs and immune cells. However, these strategies may lead to unintended consequences, such as tumor growth promotion, drug resistance, and side effects like fibrotic changes in healthy tissues. Biomaterials have made significant progress in fabricating artificial ECMs for tumor therapy by inducing biomineralization, fibrogenesis, or gelation. This perspective explores the fundamental concepts, benefits, and challenges of each technique. Additionally, future improvements and research directions in artificial ECMs are discussed, highlighting their potential to advance tumor therapy.
Collapse
Affiliation(s)
- Jinfeng Sun
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun P. R. China
- College of Chemistry Jilin University Changchun P. R. China
| | - Yang Liu
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun P. R. China
- School of Applied Chemistry and Engineering University of Science and Technology of China Hefei P. R. China
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Jingshan Sun
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun P. R. China
- School of Applied Chemistry and Engineering University of Science and Technology of China Hefei P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun P. R. China
- School of Applied Chemistry and Engineering University of Science and Technology of China Hefei P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun P. R. China
- College of Chemistry Jilin University Changchun P. R. China
- School of Applied Chemistry and Engineering University of Science and Technology of China Hefei P. R. China
| |
Collapse
|
4
|
Abraham DJ, Black CM, Denton CP, Distler JHW, Domsic R, Feghali-Bostwick C, Gourh P, Hinchcliff M, Kolling F, Kuwana M, Lafyatis R, Landegren U, Mahoney JM, Martin J, Matucci-Cerinic M, McMahan ZH, Mora AL, Mouthon L, Rabinovitch M, Rojas M, Rubin K, Trojanowska M, Varga J, Whitfield ML, Gabrielli A, Krieg T. An international perspective on the future of systemic sclerosis research. Nat Rev Rheumatol 2025; 21:174-187. [PMID: 39953141 DOI: 10.1038/s41584-024-01217-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2024] [Indexed: 02/17/2025]
Abstract
Systemic sclerosis (SSc) remains a challenging and enigmatic systemic autoimmune disease, owing to its complex pathogenesis, clinical and molecular heterogeneity, and the lack of effective disease-modifying treatments. Despite a century of research in SSc, the interconnections among microvascular dysfunction, autoimmune phenomena and tissue fibrosis in SSc remain unclear. The absence of validated biomarkers and reliable animal models complicates diagnosis and treatment, contributing to high morbidity and mortality. Advances in the past 5 years, such as single-cell RNA sequencing, next-generation sequencing, spatial biology, transcriptomics, genomics, proteomics, metabolomics, microbiome profiling and artificial intelligence, offer new avenues for identifying the early pathogenetic events that, once treated, could change the clinical history of SSc. Collaborative global efforts to integrate these approaches are crucial to developing a comprehensive, mechanistic understanding and enabling personalized therapies. Challenges include disease classification, clinical heterogeneity and the establishment of robust biomarkers for disease activity and progression. Innovative clinical trial designs and patient-centred approaches are essential for developing effective treatments. Emerging therapies, including cell-based and fibroblast-targeting treatments, show promise. Global cooperation, standardized protocols and interdisciplinary research are vital for advancing SSc research and improving patient outcomes. The integration of advanced research techniques holds the potential for important breakthroughs in the diagnosis, treatment and care of individuals with SSc.
Collapse
Affiliation(s)
- David J Abraham
- Department of Inflammation and Rare Diseases, UCL Centre for Rheumatology, UCL Division of Medicine, Royal Free Hospital Campus, London, UK.
| | - Carol M Black
- Department of Inflammation and Rare Diseases, UCL Centre for Rheumatology, UCL Division of Medicine, Royal Free Hospital Campus, London, UK
| | - Christopher P Denton
- Department of Inflammation and Rare Diseases, UCL Centre for Rheumatology, UCL Division of Medicine, Royal Free Hospital Campus, London, UK
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Robyn Domsic
- Division of Rheumatology, Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carol Feghali-Bostwick
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Pravitt Gourh
- Scleroderma Genomics and Health Disparities Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Monique Hinchcliff
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Fred Kolling
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Masataka Kuwana
- Department of Allergy and Rheumatology. Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology. University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ulf Landegren
- Department of Immunology, Genetics and Pathology, Research programme: Molecular Tools and Functional Genomics, Uppsala University, Uppsala, Sweden
| | | | - Javier Martin
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Marco Matucci-Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases and Inflammation, fibrosis and aging Initiative, IRCCS Ospedle San Raffaele and Vita Salute University San Raffaele, Milan, Italy
| | - Zsuzsanna H McMahan
- Department of Internal Medicine, Division of Rheumatology, UTHealth Houston, Houston, TX, USA
| | - Ana L Mora
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung research Institute, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Luc Mouthon
- Department of Internal Medicine, Reference Center for Rare Systemic Autoimmune and Auto-Inflammatory diseases in Île-de-France, East and West, Cochin Hospital, Public Assistance-Hospitals of Paris, Paris-Centre, Paris Cité University, Paris, France
| | - Marlene Rabinovitch
- Department of Paediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Basic Science and Engineering (BASE) Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung research Institute, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristofer Rubin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Maria Trojanowska
- Boston University, Department of Medicine, Arthritis & Autoimmune Diseases Research Center, Boston, MA, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, USA
| | - Michael L Whitfield
- Department of Biomedical Data Science, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Armando Gabrielli
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany.
- Foundation of Molecular Medicine and Cellular Therapy Polytechnic University of Marche, Via Tronto, Ancona, Italy.
| | - Thomas Krieg
- Translational Matrix Biology, Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC) University of Cologne, Cologne, Germany.
| |
Collapse
|
5
|
Wang W, Li Y, Zhang C, Zhou H, Li C, Cheng R, Chen X, Pu Y, Chen Y. Small Extracellular Vesicles from Young Healthy Human Plasma Inhibit Cardiac Fibrosis After Myocardial Infarction via miR-664a-3p Targeting SMAD4. Int J Nanomedicine 2025; 20:557-579. [PMID: 39830157 PMCID: PMC11740580 DOI: 10.2147/ijn.s488368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025] Open
Abstract
Purpose Cardiac fibrosis, a key contributor to ventricular pathologic remodeling and heart failure, currently lacks effective therapeutic approaches. Patients and Methods Small extracellular vesicles from young healthy human plasma (Young-sEVs) were characterized via protein marker, transmission electron microscopy, and nanoparticle tracking analysis, then applied in cellular models and mouse models of cardiac fibrosis. Western blotting and qRT-PCR were used to identify protective signaling pathways in cardiac fibroblasts (CFs). Results Young-sEVs significantly inhibited cardiac fibrosis and subsequent cardiac dysfunction post-myocardial infarction (MI) in mice. The main findings included that echocardiographic assessments four weeks post-MI indicated that Young-sEVs improved left ventricular ejection fraction (LVEF) and fractional shortening (LVFS), and reduced left ventricular internal diameter in diastole (LVIDd) and systole (LVIDs). Treatment with Young-sEVs also decreased Masson-positive fibroblast areas and collagen synthesis in cardiac tissue. However, sEVs from the old control group did not achieve the above effect. Consistent with in vivo results, Young-sEVs could also inhibit the proliferation, migration, and collagen synthesis of CFs in the TGF-β1-induced cellular fibrosis model. High-throughput microRNA (miRNA) sequencing and qRT-PCR analysis revealed that miR-664a-3p was abundant in Young-sEVs. The high expression of miR-664a-3p significantly inhibited the proliferation, migration, and collagen synthesis of TGF-β1-induced CFs. However, suppressing the expression of miR-664a-3p in Young-sEVs eliminated their therapeutic effect on cardiac fibrosis in mice. Further studies confirmed SMAD4 as a direct downstream target of miR-664a-3p, whose overexpression could reverse the anti-fibrotic effects of miR-664a-3p. Conclusion In summary, these findings firstly revealed that Young-sEVs could directly bind to the 3'-untranslated region of SMAD4 mRNA through miR-664a-3p, thereby inhibiting the TGF-β/SMAD4 signaling pathway to protect heart from fibrosis and improve cardiac function. Considering the ease of obtaining plasma-derived sEVs, our study offers a promising therapeutic strategy for heart failure, with the potential for rapid clinical translation in the near future.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Ying Li
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Cheng Zhang
- Long Jiang Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Haoyang Zhou
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Chunyu Li
- Long Jiang Intensive Care Unit, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Rong Cheng
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Xufeng Chen
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Yanan Pu
- Department of Clinical Laboratory, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, People’s Republic of China
| | - Yan Chen
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People’s Republic of China
- Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, People’s Republic of China
- Department of Emergency Management, School of Health Policy & Management, Nanjing Medical University, Nanjing, 211166, People’s Republic of China
| |
Collapse
|
6
|
Bahrami M, Abbaszadeh HA, Norouzian M, Abdollahifar MA, Roozbahany NA, Saber M, Azimi M, Ehsani E, Bakhtiyari M, Serra AL, Moghadasali R. Enriched human embryonic stem cells-derived CD133 +, CD24 + renal progenitors engraft and restore function in a gentamicin-induced kidney injury in mice. Regen Ther 2024; 27:506-518. [PMID: 38745839 PMCID: PMC11091464 DOI: 10.1016/j.reth.2024.04.015] [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: 02/21/2024] [Revised: 03/30/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction Acute kidney injury (AKI) is a common health problem that leads to high morbidity and potential mortality. The failure of conventional treatments to improve forms of this condition highlights the need for innovative and effective treatment approaches. Regenerative therapies with Renal Progenitor Cells (RPCs) have been proposed as a promising new strategy. A growing body of evidence suggests that progenitor cells differentiated from different sources, including human embryonic stem cells (hESCs), can effectively treat AKI. Methods Here, we describe a method for generating RPCs and directed human Embryoid Bodies (EBs) towards CD133+CD24+ renal progenitor cells and evaluate their functional activity in alleviating AKI. Results The obtained results show that hESCs-derived CD133+CD24+ RPCs can engraft into damaged renal tubules and restore renal function and structure in mice with gentamicin-induced kidney injury, and significantly decrease blood urea nitrogen levels, suppress oxidative stress and inflammation, and attenuate histopathological disturbances, including tubular necrosis, tubular dilation, urinary casts, and interstitial fibrosis. Conclusion The results suggest that RPCs have a promising regenerative potential in improving renal disease and can lay the foundation for future cell therapy and disease modeling.
Collapse
Affiliation(s)
- Maryam Bahrami
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Norouzian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Ahmady Roozbahany
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Private Practice, Bradford ON, Canada
| | - Maryam Saber
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masoumeh Azimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ehsan Ehsani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Mohsen Bakhtiyari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Andreas L. Serra
- Department of Internal Medicine and Nephrology, Klinik Hirslanden, Zurich, Switzerland
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
7
|
Yao Q, Tan W, Bai F. Gut microbiome and metabolomics in systemic sclerosis: feature, link and mechanisms. Front Immunol 2024; 15:1475528. [PMID: 39559369 PMCID: PMC11570262 DOI: 10.3389/fimmu.2024.1475528] [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: 08/04/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Systemic sclerosis (SSc) is a rare and highly heterogeneous chronic autoimmune disease characterized by multi-organ and tissue fibrosis, often accompanied by a poor prognosis and high mortality rates. The primary pathogenic mechanisms of SSc are considered to involve tissue fibrosis, autoimmune dysfunction, and microvascular abnormalities. Recent studies have shed light on the gut microbiota (GM) and metabolites in SSc patients, revealing their association with gastrointestinal symptoms and disease phenotypes. However, further elucidation is needed on the specific mechanisms underlying the interactions between GM, metabolites, and the immune system and their roles in the pathogenesis of SSc. This review outlines the characteristics of GM and metabolites in SSc patients, exploring their interrelationships and analyzing their correlations with the clinical phenotypes of SSc. The findings indicate that while the α-diversity of GM in SSc patients resembles that of healthy individuals, notable differences exist in the β-diversity and the abundance of specific bacterial genera, which are closely linked to gastrointestinal symptoms. Moreover, alterations in the levels of amino acids and lipid metabolites in SSc patients are prominently observed and significantly associated with clinical phenotypes. Furthermore, this review delves into the potential immunopathological mechanisms of GM and metabolites in SSc, emphasizing the critical role of interactions between GM, metabolites, and the immune system in comprehending the immunopathological processes of SSc. These insights may offer new scientific evidence for the development of future treatment strategies.
Collapse
Affiliation(s)
- Qicen Yao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Nanjing Medical University, Nanjing, China
| | - Wenfeng Tan
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
8
|
Li W, Liu J, Jiao R, Liu Z, Zhang T, Chai D, Meng L, Yang Z, Liu Y, Gu X, Li X, Yang C. Baricitinib alleviates cardiac fibrosis and inflammation induced by chronic sympathetic activation. Int Immunopharmacol 2024; 140:112894. [PMID: 39126736 DOI: 10.1016/j.intimp.2024.112894] [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: 06/27/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Cardiac fibrosis is characterized by the over-proliferation, over-transdifferentiation and over-deposition of extracellular matrix (ECM) of cardiac fibroblasts (CFs). Cardiac sympathetic activation is one of the leading causes of myocardial fibrosis. Meanwhile, cardiac fibrosis is often together with cardiac inflammation, which accelerates fibrosis by mediating inflammatory cytokines secretion. Recently, the Janus kinase/signal transducer and activator of transcription (JAK/STAT3) signaling pathway has been confirmed by its vital role during the progression of cardiac fibrosis. Thus, JAK/STAT3 signaling pathway is thought to be a potential therapeutic target for cardiac fibrosis. Baricitinib (BR), a novel JAK1/2 inhibitor, has been reported excellent effects of anti-fibrosis in multiple fibrotic diseases. However, little is known about whether and how BR ameliorates cardiac fibrosis caused by chronic sympathetic activation. Isoproterenol (ISO), a β-Adrenergic receptor (β-AR) nonselective agonist, was used to modulate chronic sympathetic activation in mice. As expected, our results proved that BR ameliorated ISO-induced cardiac dysfunction. Meanwhile, BR attenuated ISO-induced cardiac fibrosis and cardiac inflammation in mice. Moreover, BR also inhibited ISO-induced cardiac fibroblasts activation and macrophages pro-inflammatory secretion. As for mechanism studies, BR reduced ISO-induced cardiac fibroblasts by JAK2/STAT3 and PI3K/Akt signaling, while reduced ISO-induced macrophages pro-inflammatory secretion by JAK1/STAT3 and NF-κB signaling. In summary, BR alleviates cardiac fibrosis and inflammation caused by chronic sympathetic activation. The underlying mechanism involves BR-mediated suppression of JAK1/2/STAT3, PI3K/Akt and NF-κB signaling.
Collapse
Affiliation(s)
- Wenqi Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Jing Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Ran Jiao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Zhigang Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Tiantian Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Dan Chai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Lingxin Meng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Zhongyi Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Yuming Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | - Xiaoting Gu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; Tianjin Key Laboratory of Molecular Drug Research, International Joint Academy of Biomedicine, Tianjin 300457, China.
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; Tianjin Key Laboratory of Molecular Drug Research, International Joint Academy of Biomedicine, Tianjin 300457, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; Tianjin Key Laboratory of Molecular Drug Research, International Joint Academy of Biomedicine, Tianjin 300457, China.
| |
Collapse
|
9
|
Gao L, Wang H, Fang F, Liu J, Zhao C, Niu J, Wang Z, Zhong Y, Wang X. The roles of orphan nuclear receptor 4 group A1 and A2 in fibrosis. Int Immunopharmacol 2024; 139:112705. [PMID: 39029235 DOI: 10.1016/j.intimp.2024.112705] [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: 05/16/2024] [Revised: 07/11/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024]
Abstract
Fibrosis is not a disease but rather an outcome of the pathological tissue repair response. Many myofibroblasts are activated which lead to the excessive accumulation of extracellular matrix components such as collagen and fibronectin with fibrosis. A variety of organs, including kidney, liver, lung, heart and skin, can undergo fibrosis under the stimulation of exogenous or endogenous pathogenic factors. The orphan nuclear receptor 4 group A1 (NR4A1) and nuclear receptor 4 group A2(NR4A2)are belong to the nuclear receptor subfamily and inhibit the occurrence and development of fibrosis. NR4A1 is an inhibitory factor of TGF-β signaling transduction. Overexpression of NR4A1 in fibroblasts can reduce TGF-β induced collagen deposition and fibrosis related gene expression. Here, we summarize the current research progress on the NR4A1/2 and fibrosis, providing reference for the treatment of fibrosis.
Collapse
Affiliation(s)
- Lanjun Gao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Hongshuang Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Fang Fang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Jiazhi Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Chenchen Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Jieqi Niu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Zheng Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yan Zhong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| | - Xiangting Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China.
| |
Collapse
|
10
|
Zhang J, Shimozaki K, Hattori S, Pastukh V, Maloney D, Hogan MV, Wang JHC. Metformin lotion promotes scarless skin tissue formation through AMPK activation, TGF-β1 inhibition, and reduced myofibroblast numbers. PLoS One 2024; 19:e0311147. [PMID: 39331598 PMCID: PMC11433050 DOI: 10.1371/journal.pone.0311147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024] Open
Abstract
Scar tissue formation following skin wound healing is a challenging public health problem. Skin regeneration and preventing the formation of scar tissue by currently available commercial products are largely ineffective. This study aimed to test the efficacy of a novel topical metformin lotion (ML) in inhibiting scar tissue formation during skin wound healing in rats and to determine the mechanisms of action involved. A 6% ML was prepared in our laboratory. A skin wound healing model in rats was used. The wounded rats were divided into two groups and treated daily for 10 days as follows: Group 1 received a daily application of 50 mg of control lotion, or 0% ML (totaling 100 mg of lotion per rat), and Group 2 received a daily application of 50 mg of 6% ML (totaling 100 mg of 6% ML per rat). Blood samples from the heart of each rat were analyzed for inflammatory markers, HMGB1 and IL-1β, using ELISA, and immunological and histological analyses were performed on skin tissue sections. ML decreased levels of inflammatory markers HMGB1 and IL-1β in the serum of rats and inhibited the release of HMGB1 from cell nuclei into the skin tissue matrix. Additionally, ML demonstrated anti-fibrotic properties by enhancing AMPK activity, decreasing the expression of TGF-β1, reducing the number of myofibroblasts, decreasing the production of collagen III, and increasing the expression of collagen I. ML promotes the regeneration of high-quality skin during wound healing by reducing scar tissue formation. This effect is mediated through the activation of AMPK, inhibition of TGF-β1, and a decrease in the number of myofibroblasts.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kengo Shimozaki
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Soichi Hattori
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Vasyl Pastukh
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Derek Maloney
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
11
|
Primerano A, De Domenico E, Cianfarani F, De Luca N, Floriddia G, Teson M, Cristofoletti C, Cardarelli S, Scaglione GL, Baldini E, Cangelosi D, Uva P, Reinoso Sánchez JF, Roubaty C, Dengjel J, Nyström A, Mastroeni S, Ulisse S, Castiglia D, Odorisio T. Histone deacetylase inhibition mitigates fibrosis-driven disease progression in recessive dystrophic epidermolysis bullosa. Br J Dermatol 2024; 191:568-579. [PMID: 38820176 DOI: 10.1093/bjd/ljae225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Recessive dystrophic epidermolysis bullosa (RDEB) is a blistering disease caused by mutations in the gene encoding type VII collagen (C7). RDEB is associated with fibrosis, which is responsible for severe complications. The phenotypic variability observed in siblings with RDEB suggests that epigenetic modifications contribute to disease severity. Identifying epigenetic changes may help to uncover molecular mechanisms underlying RDEB pathogenesis and new therapeutic targets. OBJECTIVES To investigate histone acetylation in RDEB skin and to explore histone deacetylase inhibitors (HDACi) as therapeutic molecules capable of counteracting fibrosis and disease progression in RDEB mice. METHODS Acetylated histone levels were detected in human skin by immunofluorescence and in RDEB fibroblasts by enzyme-linked immunosorbent assay (ELISA). The effects of givinostat and valproic acid (VPA) on RDEB fibroblast fibrotic behaviour were assessed by a collagen-gel contraction assay, Western blot and immunocytofluorescence for α-smooth muscle actin, and ELISA for released transforming growth factor (TGF)-β1. RNA sequencing was performed in HDACi- and vehicle-treated RDEB fibroblasts. VPA was systemically administered to RDEB mice and effects on overt phenotype were monitored. Fibrosis was investigated in the skin using histological and immunofluorescence analyses. Eye and tongue defects were examined microscopically. Mass spectrometry proteomics was performed on skin protein extracts from VPA-treated RDEB and control mice. RESULTS Histone acetylation decreases in RDEB skin and primary fibroblasts. RDEB fibroblasts treated with HDACi lowered fibrotic traits, including contractility, TGF-β1 release and proliferation. VPA administration to RDEB mice mitigated severe manifestations affecting the eyes and paws. These effects were associated with fibrosis inhibition. Proteomic analysis of mouse skin revealed that VPA almost normalized protein sets involved in protein synthesis and immune response, processes linked to the increased susceptibility to cancer and bacterial infections seen in people with RDEB. CONCLUSIONS Dysregulated histone acetylation contributes to RDEB pathogenesis by facilitating the progression of fibrosis. Repurposing of HDACi could be considered for disease-modifying treatments in RDEB.
Collapse
Affiliation(s)
| | | | | | - Naomi De Luca
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | | | - Massimo Teson
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | | | - Silvia Cardarelli
- Laboratory of Experimental Medicine, Department of Surgery, Sapienza University, Rome, Italy
| | | | - Enke Baldini
- Laboratory of Experimental Medicine, Department of Surgery, Sapienza University, Rome, Italy
| | - Davide Cangelosi
- Clinical Bioinformatics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paolo Uva
- Clinical Bioinformatics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Carole Roubaty
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Alexander Nyström
- Department of Dermatology, University of Freiburg, Freiburg, Germany
| | | | - Salvatore Ulisse
- Laboratory of Experimental Medicine, Department of Surgery, Sapienza University, Rome, Italy
| | | | - Teresa Odorisio
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| |
Collapse
|
12
|
Sanchez S, McDowell-Sanchez AK, Al-Meerani SB, Cala-Garcia JD, Waich Cohen AR, Ochsner SA, McKenna NJ, Celada LJ, Wu M, Assassi S, Rosas IO, Tsoyi K. PIK-III exerts anti-fibrotic effects in activated fibroblasts by regulating p38 activation. PLoS One 2024; 19:e0306624. [PMID: 39240940 PMCID: PMC11379285 DOI: 10.1371/journal.pone.0306624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 06/20/2024] [Indexed: 09/08/2024] Open
Abstract
Systemic sclerosis (SSc), also known as scleroderma, is an autoimmune-driven connective tissue disorder that results in fibrosis of the skin and internal organs such as the lung. Fibroblasts are known as the main effector cells involved in the progression of SSc through the induction of extracellular matrix (ECM) proteins and myofibroblast differentiation. Here, we demonstrate that 4'-(cyclopropylmethyl)-N2-4-pyridinyl-[4,5'-bipyrimidine]-2,2'-diamine (PIK-III), known as class III phosphatidylinositol 3-kinase (PIK3C3/VPS34) inhibitor, exerts potent antifibrotic effects in human dermal fibroblasts (HDFs) by attenuating transforming growth factor-beta 1 (TGF-β1)-induced ECM expression, cell contraction and myofibroblast differentiation. Unexpectedly, neither genetic silencing of PIK3C3 nor other PIK3C3 inhibitors (e.g., SAR405 and Autophinib) were able to mimic PIK-III-mediated antifibrotic effect in dermal fibroblasts, suggesting that PIK-III inhibits fibroblast activation through another signaling pathway. We identified that PIK-III effectively inhibits p38 activation in TGF-β1-stimulated dermal fibroblasts. Finally, PIK-III administration significantly attenuated dermal and lung fibrosis in bleomycin-injured mice.
Collapse
Affiliation(s)
- Santiago Sanchez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Aaron K McDowell-Sanchez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Sharaz B Al-Meerani
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Juan D Cala-Garcia
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Alan R Waich Cohen
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Lindsay J Celada
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Minghua Wu
- Division of Rheumatology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States of America
| | - Shervin Assassi
- Division of Rheumatology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States of America
| | - Ivan O Rosas
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Konstantin Tsoyi
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| |
Collapse
|
13
|
Albu MT, Matei AE, Distler JHW, Giesel FL, Mori Y. Fibroblast activation protein inhibitor PET/CT as an emerging diagnostic modality in interstitial lung disease and other fibrotic conditions. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:152-156. [PMID: 39439976 PMCID: PMC11492823 DOI: 10.2478/rir-2024-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/31/2024] [Indexed: 10/25/2024]
Abstract
Interstitial lung diseases (ILD) encompass a wide range of disorders characterized by alveolar inflammation and fibrotic tissue remodeling, marked by significant morbidity and mortality. Systemic sclerosis (SSc), among other connective tissue diseases, is a frequent cause of ILD. Assessment of pulmonary fibrosis is frequently constrained by the delayed manifestations of profibrotic activation of fibroblasts, which results in late macroscopic alterations detectable by standard imaging techniques such as computed tomography (CT) and magnetic resonance imaging (MRI) scans. 68Ga-labeled fibroblast activation protein inhibitors (68Ga-FAPI [fibroblast activation protein inhibitor]) are novel radionuclides used in the selective positron emission tomography/computed tomography (PET/CT) detection of profibrotic fibroblasts, a key player in fibrotic tissue remodeling. Application of 68Ga-FAPI in different target organs undergoing fibrosis, such as lung and heart, highlights its efficacy in detecting ongoing fibrotic processes, since FAPI tracer uptake has been correlated with clinical disease progression markers in SSc-ILD. This feature could enable physicians to detect subclinical fibrotic activity and tailor an individualised therapy plan on a case by case basis. The use of 68Ga-FAPI in ILD and other fibrotic conditions may emerge as a novel tool in future clinical practice for both activity monitoring and treatment optimisation. Other tracers tested in ILD of different etiologies have shown promising results and may in future also be considered for potential application in SSc-ILD.
Collapse
Affiliation(s)
- Mihai Tudor Albu
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
| | - Alexandru-Emil Matei
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
| | - Jörg H. W. Distler
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
| | - Frederik L. Giesel
- Department of Nuclear Medicine, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
- Institute for Radiation Sciences, Osaka University, Osaka, Japan
| | - Yuriko Mori
- Department of Nuclear Medicine, University Hospital Düsseldorf, Medical Faculty of Heinrich-Heine University; Düsseldorf, Germany
| |
Collapse
|
14
|
Liang M, Wang L, Tian X, Wang K, Zhu X, Huang L, Li Q, Ye W, Chen C, Yang H, Wu W, Chen X, Zhu X, Xue Y, Wan W, Wu Y, Lu L, Wang J, Zou H, Ying T, Zhou F. Identification and validation of anti-protein arginine methyltransferase 5 (PRMT5) antibody as a novel biomarker for systemic sclerosis (SSc). Ann Rheum Dis 2024; 83:1144-1155. [PMID: 38684324 PMCID: PMC11420721 DOI: 10.1136/ard-2024-225596] [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: 01/27/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVES In the complex panorama of autoimmune diseases, the characterisation of pivotal contributing autoantibodies that are involved in disease progression remains challenging. This study aimed to employ a global antibody profiling strategy to identify novel antibodies and investigate their association with systemic sclerosis (SSc). METHODS We implemented this strategy by conducting immunoprecipitation (IP) following on-bead digestion with the sera of patients with SSc or healthy donors, using antigen pools derived from cell lysates. The enriched antigen-antibody complex was proceeded with mass spectrometry (MS)-based quantitative proteomics and over-represented by bioinformatics analysis. The candidate antibodies were then orthogonally validated in two independent groups of patients with SSc. Mice were immunised with the target antigen, which was subsequently evaluated by histological examination and RNA sequencing. RESULTS The IP-MS analysis, followed by validation in patients with SSc, revealed a significant elevation in anti-PRMT5 antibodies among patients with SSc. These antibodies exhibited robust diagnostic accuracy in distinguishing SSc from healthy controls and other autoimmune conditions, including systemic lupus erythematosus and Sjögren's syndrome, with an area under the curve ranging from 0.900 to 0.988. The elevation of anti-PRMT5 antibodies was verified in a subsequent independent group with SSc using an additional method, microarray. Notably, 31.11% of patients with SSc exhibited seropositivity for anti-PRMT5 antibodies. Furthermore, the titres of anti-PRMT5 antibodies demonstrated a correlation with the progression or regression trajectory in SSc. PRMT5 immunisation displayed significant inflammation and fibrosis in both the skin and lungs of mice. This was concomitant with the upregulation of multiple proinflammatory and profibrotic pathways, thereby underscoring a potentially pivotal role of anti-PRMT5 antibodies in SSc. CONCLUSIONS This study has identified anti-PRMT5 antibodies as a novel biomarker for SSc.
Collapse
Affiliation(s)
- Minrui Liang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lingbiao Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaolong Tian
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kun Wang
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyi Zhu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Linlin Huang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Li
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haihua Yang
- Department of Respiratory and Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanqing Wu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiguo Wan
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liwei Lu
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Feng Zhou
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol 2024; 25:617-638. [PMID: 38589640 DOI: 10.1038/s41580-024-00716-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
The term 'fibroblast' often serves as a catch-all for a diverse array of mesenchymal cells, including perivascular cells, stromal progenitor cells and bona fide fibroblasts. Although phenotypically similar, these subpopulations are functionally distinct, maintaining tissue integrity and serving as local progenitor reservoirs. In response to tissue injury, these cells undergo a dynamic fibroblast-myofibroblast transition, marked by extracellular matrix secretion and contraction of actomyosin-based stress fibres. Importantly, whereas transient activation into myofibroblasts aids in tissue repair, persistent activation triggers pathological fibrosis. In this Review, we discuss the roles of mechanical cues, such as tissue stiffness and strain, alongside cell signalling pathways and extracellular matrix ligands in modulating myofibroblast activation and survival. We also highlight the role of epigenetic modifications and myofibroblast memory in physiological and pathological processes. Finally, we discuss potential strategies for therapeutically interfering with these factors and the associated signal transduction pathways to improve the outcome of dysregulated healing.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andrew E Miller
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Fabio M V Rossi
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada.
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
16
|
Rodrigues de Almeida A, Jaime Bezerra Mendonça Junior F, Tavares Dantas A, Eduarda de Oliveira Gonçalves M, Chêne C, Jeljeli M, Chouzenoux S, Thomas M, David de Azevedo Valadares L, Andreza Bezerra Correia M, Ângela da Silva Alves W, Carvalho Lira E, Doridot L, Jesus Barreto de Melo Rêgo M, Cristiny Pereira M, Luzia Branco Pinto Duarte A, Saes Parra Abdalla D, Nicco C, Batteux F, Galdino da Rocha Pitta M. IBPA a mutual prodrug of ibuprofen and acetaminophen alleviates inflammation, immune dysregulation and fibrosis in preclinical models of systemic sclerosis. Int Immunopharmacol 2024; 136:112344. [PMID: 38833846 DOI: 10.1016/j.intimp.2024.112344] [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: 04/03/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/06/2024]
Abstract
Systemic sclerosis (SSc) is a devastating autoimmune illness with a wide range of clinical symptoms, including vascular abnormalities, inflammation, and persistent and progressive fibrosis. The disease's complicated pathophysiology makes it difficult to develop effective therapies, necessitating research into novel therapeutic options. Molecular hybridization is a strategy that can be used to develop new drugs that act on two or multiple targets and represents an interesting option to be explored for the treatment of complex diseases. We aimed to evaluate the effects of a hybrid mutual prodrug of ibuprofen and acetaminophen (IBPA) in peripheral blood mononuclear cells (PBMC) isolated from SSc patients, and in an in vivo model of SSc induced in BALB/c mice by intradermal injections of hypochlorous acid (HOCl) for 6 weeks. The mice were treated at the same time with daily intraperitoneal injections of IBPA (40 mg/kg). Pulmonary and skin fibrosis as well as immune responses were evaluated. IBPA significantly decreased the release of cytokines in PBMC culture supernatants from SSc patients after stimulation with phytohemagglutinin-M (IL-2, IL-4, IL-6, IL-10, IL-13, IL-17A, TNF and IFN-γ).In HOCl-induced SSc, IBPA treatment prevented dermal and pulmonary fibrosis, in addition to reducing CD4 + T and B cells activation and reversing the M2 polarization of macrophages in spleen cells, and inhibiting IFN-γ secretion in splenocyte cultures. These results show the anti-inflammatory and antifibrotic effects of IBPA in SSc and highlight the therapeutic potential of this mutual prodrug, providing support for future studies.
Collapse
Affiliation(s)
- Anderson Rodrigues de Almeida
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, PE, Brazil; Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | - Francisco Jaime Bezerra Mendonça Junior
- Laboratório de Síntese e Vetorização de Moléculas, Departamento de Ciências Biológicas, Centro de Ciências Biológicas e Sociais Aplicadas, Universidade Estadual da Paraíba, João Pessoa, PB, Brazil
| | - Andréa Tavares Dantas
- Serviço de Reumatologia, Hospital das Clínicas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Maria Eduarda de Oliveira Gonçalves
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Charlotte Chêne
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | - Mohamed Jeljeli
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | | | - Marine Thomas
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | | | - Maria Andreza Bezerra Correia
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Eduardo Carvalho Lira
- Departamento de Fisiologia e Farmacologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Ludivine Doridot
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | - Moacyr Jesus Barreto de Melo Rêgo
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Michelly Cristiny Pereira
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, PE, Brazil; Departamento de Fisiologia e Farmacologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil.
| | | | - Dulcineia Saes Parra Abdalla
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Carole Nicco
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | - Frédéric Batteux
- Université Paris Cité, Institut Cochin, Inserm, CNRS, Paris, France
| | - Maira Galdino da Rocha Pitta
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, PE, Brazil
| |
Collapse
|
17
|
Drzyzga Ł, Śpiewak D, Dorecka M, Wyględowska-Promieńska D. Available Therapeutic Options for Corneal Neovascularization: A Review. Int J Mol Sci 2024; 25:5479. [PMID: 38791518 PMCID: PMC11121997 DOI: 10.3390/ijms25105479] [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: 03/25/2024] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Corneal neovascularization can impair vision and result in a poor quality of life. The pathogenesis involves a complex interplay of angiogenic factors, notably vascular endothelial growth factor (VEGF). This review provides a comprehensive overview of potential therapies for corneal neovascularization, covering tissue inhibitors of metalloproteinases (TIMPs), transforming growth factor beta (TGF-β) inhibitors, interleukin-1L receptor antagonist (IL-1 Ra), nitric oxide synthase (NOS) isoforms, galectin-3 inhibitors, retinal pigment epithelium-derived factor (PEDF), platelet-derived growth factor (PDGF) receptor inhibitors, and surgical treatments. Conventional treatments include anti-VEGF therapy and laser interventions, while emerging therapies such as immunosuppressive drugs (cyclosporine and rapamycin) have been explored. Losartan and decorin are potential antifibrotic agents that mitigate TGF-β-induced fibrosis. Ocular nanosystems are innovative drug-delivery platforms that facilitate the targeted release of therapeutic agents. Gene therapies, such as small interfering RNA and antisense oligonucleotides, are promising approaches for selectively inhibiting angiogenesis-related gene expression. Aganirsen is efficacious in reducing the corneal neovascularization area without significant adverse effects. These multifaceted approaches underscore the corneal neovascularization management complexity and highlight ideas for enhancing therapeutic outcomes. Furthermore, the importance of combination therapies and the need for further research to develop specific inhibitors while considering their therapeutic efficacy and potential adverse effects are discussed.
Collapse
Affiliation(s)
- Łukasz Drzyzga
- Department of Ophthalmology, Prof. K. Gibiński University Clinical Center, Medical University of Silesia, 40-055 Katowice, Poland
- Clinical Ophthalmology Center Okolux, 40-754 Katowice, Poland
| | - Dorota Śpiewak
- Department of Ophthalmology, Prof. K. Gibiński University Clinical Center, Medical University of Silesia, 40-055 Katowice, Poland
- Clinical Ophthalmology Center Okolux, 40-754 Katowice, Poland
| | - Mariola Dorecka
- Department of Ophthalmology, Prof. K. Gibiński University Clinical Center, Medical University of Silesia, 40-055 Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-514 Katowice, Poland
| | - Dorota Wyględowska-Promieńska
- Department of Ophthalmology, Prof. K. Gibiński University Clinical Center, Medical University of Silesia, 40-055 Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-514 Katowice, Poland
| |
Collapse
|
18
|
Zanini G, Bertani G, Di Tinco R, Pisciotta A, Bertoni L, Selleri V, Generali L, Marconi A, Mattioli AV, Pinti M, Carnevale G, Nasi M. Dental Pulp Stem Cells Modulate Inflammasome Pathway and Collagen Deposition of Dermal Fibroblasts. Cells 2024; 13:836. [PMID: 38786058 PMCID: PMC11120068 DOI: 10.3390/cells13100836] [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: 02/27/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Fibrosis is a pathological condition consisting of a delayed deposition and remodeling of the extracellular matrix (ECM) by fibroblasts. This deregulation is mostly triggered by a chronic stimulus mediated by pro-inflammatory cytokines, such as TNF-α and IL-1, which activate fibroblasts. Due to their anti-inflammatory and immunosuppressive potential, dental pulp stem cells (DPSCs) could affect fibrotic processes. This study aims to clarify if DPSCs can affect fibroblast activation and modulate collagen deposition. We set up a transwell co-culture system, where DPSCs were seeded above the monolayer of fibroblasts and stimulated with LPS or a combination of TNF-α and IL-1β and quantified a set of genes involved in inflammasome activation or ECM deposition. Cytokines-stimulated co-cultured fibroblasts, compared to unstimulated ones, showed a significant increase in the expression of IL-1β, IL-6, NAIP, AIM2, CASP1, FN1, and TGF-β genes. At the protein level, IL-1β and IL-6 release as well as FN1 were increased in stimulated, co-cultured fibroblasts. Moreover, we found a significant increase of MMP-9 production, suggesting a role of DPSCs in ECM remodeling. Our data seem to suggest a crosstalk between cultured fibroblasts and DPSCs, which seems to modulate genes involved in inflammasome activation, ECM deposition, wound healing, and fibrosis.
Collapse
Affiliation(s)
- Giada Zanini
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.Z.)
| | - Giulia Bertani
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| | - Rosanna Di Tinco
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| | - Alessandra Pisciotta
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| | - Laura Bertoni
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| | - Valentina Selleri
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.Z.)
- National Institute for Cardiovascular Research—INRC, 40126 Bologna, Italy;
| | - Luigi Generali
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| | - Alessandra Marconi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| | - Anna Vittoria Mattioli
- National Institute for Cardiovascular Research—INRC, 40126 Bologna, Italy;
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.Z.)
| | - Gianluca Carnevale
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| | - Milena Nasi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.B.); (R.D.T.); (A.P.); (L.B.); (L.G.); (A.M.); (G.C.); (M.N.)
| |
Collapse
|
19
|
Zou W, Lu J, Zhang L, Sun D. Tetrahedral framework nucleic acids for improving wound healing. J Nanobiotechnology 2024; 22:113. [PMID: 38491372 PMCID: PMC10943864 DOI: 10.1186/s12951-024-02365-z] [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: 11/08/2023] [Accepted: 02/21/2024] [Indexed: 03/18/2024] Open
Abstract
Wounds are one of the most common health issues, and the cost of wound care and healing has continued to increase over the past decade. In recent years, there has been growing interest in developing innovative strategies to enhance the efficacy of wound healing. Tetrahedral framework nucleic acids (tFNAs) have emerged as a promising tool for wound healing applications due to their unique structural and functional properties. Therefore, it is of great significance to summarize the applications of tFNAs for wound healing. This review article provides a comprehensive overview of the potential of tFNAs as a novel therapeutic approach for wound healing. In this review, we discuss the possible mechanisms of tFNAs in wound healing and highlight the role of tFNAs in modulating key processes involved in wound healing, such as cell proliferation and migration, angiogenesis, and tissue regeneration. The targeted delivery and controlled release capabilities of tFNAs offer advantages in terms of localized and sustained delivery of therapeutic agents to the wound site. In addition, the latest research progress on tFNAs in wound healing is systematically introduced. We also discuss the biocompatibility and biosafety of tFNAs, along with their potential applications and future directions for research. Finally, the current challenges and prospects of tFNAs are briefly discussed to promote wider applications.
Collapse
Affiliation(s)
- Wanqing Zou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, Guangdong, China
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China.
| | - Luyong Zhang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.
| | - Duanping Sun
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, Guangdong, China.
| |
Collapse
|
20
|
Janjetovic Z, Qayyum S, Reddy SB, Podgorska E, Scott SG, Szpotan J, Mobley AA, Li W, Boda VK, Ravichandran S, Tuckey RC, Jetten AM, Slominski AT. Novel Vitamin D3 Hydroxymetabolites Require Involvement of the Vitamin D Receptor or Retinoic Acid-Related Orphan Receptors for Their Antifibrogenic Activities in Human Fibroblasts. Cells 2024; 13:239. [PMID: 38334631 PMCID: PMC10854953 DOI: 10.3390/cells13030239] [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: 11/27/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
We investigated multiple signaling pathways activated by CYP11A1-derived vitamin D3 hydroxymetabolites in human skin fibroblasts by assessing the actions of these molecules on their cognate receptors and by investigating the role of CYP27B1 in their biological activities. The actions of 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3 and 1,20,23(OH)3D3 were compared to those of classical 1,25(OH)2D3. This was undertaken using wild type (WT) fibroblasts, as well as cells with VDR, RORs, or CYP27B1 genes knocked down with siRNA. Vitamin D3 hydroxymetabolites had an inhibitory effect on the proliferation of WT cells, but this effect was abrogated in cells with silenced VDR or RORs. The collagen expression by WT cells was reduced upon secosteroid treatment. This effect was reversed in cells where VDR or RORs were knocked down where the inhibition of collagen production and the expression of anti-fibrotic genes in response to the hydroxymetabolites was abrogated, along with ablation of their anti-inflammatory action. The knockdown of CYP27B1 did not change the effect of either 20(OH)D3 or 20,23(OH)2D3, indicating that their actions are independent of 1α-hydroxylation. In conclusion, the expression of the VDR and/or RORα/γ receptors in fibroblasts is necessary for the inhibition of both the proliferation and fibrogenic activity of hydroxymetabolites of vitamin D3, while CYP27B1 is not required.
Collapse
Affiliation(s)
- Zorica Janjetovic
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Shariq Qayyum
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
- Brigham’s Women’s Hospital, Harvard University, Boston, MA 02115, USA
| | - Sivani B. Reddy
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Ewa Podgorska
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - S. Gates Scott
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Justyna Szpotan
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Alisa A. Mobley
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Wei Li
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA; (W.L.); (V.K.B.)
| | - Vijay K. Boda
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA; (W.L.); (V.K.B.)
| | - Senthilkumar Ravichandran
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
| | - Robert C. Tuckey
- School of Molecular Science, The University of Western Australia, Perth 6009, Australia;
| | - Anton M. Jetten
- Cell Biology Section, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA;
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Z.J.); (S.Q.); (S.B.R.); (E.P.); (S.G.S.); (J.S.); (A.A.M.); (S.R.)
- Cancer Chemoprevention Program, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- VA Medical Center, Birmingham, AL 35294, USA
| |
Collapse
|
21
|
Kulebyakina M, Basalova N, Butuzova D, Arbatsky M, Chechekhin V, Kalinina N, Tyurin-Kuzmin P, Kulebyakin K, Klychnikov O, Efimenko A. Balance between Pro- and Antifibrotic Proteins in Mesenchymal Stromal Cell Secretome Fractions Revealed by Proteome and Cell Subpopulation Analysis. Int J Mol Sci 2023; 25:290. [PMID: 38203461 PMCID: PMC10779358 DOI: 10.3390/ijms25010290] [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: 10/27/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) regulate tissue repair through paracrine activity, with secreted proteins being significant contributors. Human tissue repair commonly results in fibrosis, where fibroblast differentiation into myofibroblasts is a major cellular mechanism. MSCs' paracrine activity can inhibit fibrosis development. We previously demonstrated that the separation of MSC secretome, represented by conditioned medium (CM), into subfractions enriched with extracellular vesicles (EV) or soluble factors (SF) boosts EV and SF antifibrotic effect. This effect is realized through the inhibition of fibroblast-to-myofibroblast differentiation in vitro. To unravel the mechanisms of MSC paracrine effects on fibroblast differentiation, we performed a comparative proteomic analysis of MSC secretome fractions. We found that CM was enriched in NF-κB activators and confirmed via qPCR that CM, but not EV or SF, upregulated NF-κB target genes (COX2, IL6, etc.) in human dermal fibroblasts. Furthermore, we revealed that EV and SF were enriched in TGF-β, Notch, IGF, and Wnt pathway regulators. According to scRNAseq, 11 out of 13 corresponding genes were upregulated in a minor MSC subpopulation disappearing in profibrotic conditions. Thus, protein enrichment of MSC secretome fractions and cellular subpopulation patterns shift the balance in fibroblast-to-myofibroblast differentiation, which should be considered in studies of MSC paracrine effects and the therapeutic use of MSC secretome.
Collapse
Affiliation(s)
- Maria Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
- Institute for Regenerative Medicine, Medical Research and Educational Center, Lomonosov Moscow State University, 27/10, Lomonosovskiy Av., 119192 Moscow, Russia
| | - Nataliya Basalova
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
- Institute for Regenerative Medicine, Medical Research and Educational Center, Lomonosov Moscow State University, 27/10, Lomonosovskiy Av., 119192 Moscow, Russia
| | - Daria Butuzova
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
| | - Mikhail Arbatsky
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
| | - Vadim Chechekhin
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
| | - Natalia Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
| | - Pyotr Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
| | - Konstantin Kulebyakin
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
- Institute for Regenerative Medicine, Medical Research and Educational Center, Lomonosov Moscow State University, 27/10, Lomonosovskiy Av., 119192 Moscow, Russia
| | - Oleg Klychnikov
- Faculty of Biology, Lomonosov Moscow State University, 1-12, Leninskie Gory, Lomonosovskiy Av., 119991 Moscow, Russia;
| | - Anastasia Efimenko
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovskiy Av., 119192 Moscow, Russia; (M.K.); (N.B.); (D.B.); (M.A.); (V.C.); (N.K.); (P.T.-K.); (K.K.)
- Institute for Regenerative Medicine, Medical Research and Educational Center, Lomonosov Moscow State University, 27/10, Lomonosovskiy Av., 119192 Moscow, Russia
| |
Collapse
|
22
|
Zhang J, Brown R, Hogan MV, Wang JHC. Mitigating Scar Tissue Formation in Tendon Injuries: Targeting HMGB1, AMPK Activation, and Myofibroblast Migration All at Once. Pharmaceuticals (Basel) 2023; 16:1739. [PMID: 38139865 PMCID: PMC10748062 DOI: 10.3390/ph16121739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Tendon injuries, while prevalent, present significant challenges regarding their structural and functional restoration. Utilizing alpha-smooth muscle actin (α-SMA)-Ai9-scleraxis (Scx)-green fluorescent protein (GFP) transgenic mice, which exhibit both Scx (a tendon cell marker) and α-SMA (a myofibroblast marker), we explored the effects of metformin (Met) on tendon healing, repair, and its mechanisms of action. Our findings revealed that intraperitoneal (IP) injections of Met, administered before or after injury, as well as both, effectively prevented the release of HMGB1 into the tendon matrix and reduced circulating levels of HMGB1. Additionally, Met treatment increased and activated AMPK and suppressed TGF-β1 levels within the healing tendon. Tendon healing was also improved by blocking the migration of α-SMA+ myofibroblasts, reducing the prevalence of disorganized collagen fibers and collagen type III. It also enhanced the presence of collagen type I. These outcomes highlight Met's anti-fibrotic properties in acutely injured tendons and suggest its potential for repurposing as a therapeutic agent to minimize scar tissue formation in tendon injuries, which could have profound implications in clinical practice.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
| | - Roshawn Brown
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
23
|
Higuchi T, Takagi K, Tochimoto A, Ichimura Y, Hirose H, Sawada T, Shibata N, Harigai M, Kawaguchi Y. Antifibrotic effect of apremilast in systemic sclerosis dermal fibroblasts and bleomycin-induced mouse model. Sci Rep 2023; 13:19378. [PMID: 37938601 PMCID: PMC10632419 DOI: 10.1038/s41598-023-46737-1] [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: 02/21/2023] [Accepted: 11/04/2023] [Indexed: 11/09/2023] Open
Abstract
Phosphodiesterase (PDE) 4 inhibitors have been reported to suppress the progression of dermal fibrosis in patients with systemic sclerosis (SSc); however, the precise mechanisms remain to be elucidated. Therefore, we conducted experiments focusing on the antifibrotic and anti-inflammatory effects of apremilast using dermal fibroblasts derived from patients with SSc and an SSc mouse model. Dermal fibroblasts derived from healthy controls and patients with SSc were incubated with apremilast in the presence or absence of 10 ng/ml transforming growth factor (TGF)-β1 for the measurement of intracellular cAMP levels and evaluation of mRNA and protein expression. A bleomycin-induced dermal fibrosis mouse model was used to evaluate the inhibitory effects of apremilast on the progression of dermal fibrosis. Intracellular cAMP levels were significantly reduced in dermal fibroblasts derived from patients with SSc compared with those derived from healthy controls. Apremilast reduced the mRNA expression of profibrotic markers and the protein expression of type I collagen and Cellular Communication Network Factor 2 (CCN2) in dermal fibroblasts. Additionally, apremilast inhibited the progression of dermal fibrosis in mice, partly by acting on T cells. These results suggest that apremilast may be a potential candidate for treating dermal fibrosis in SSc.
Collapse
Affiliation(s)
- Tomoaki Higuchi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan.
- Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Kae Takagi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Akiko Tochimoto
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Yuki Ichimura
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
- Department of Dermatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hikaru Hirose
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Tatsuo Sawada
- Department of Pathology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Noriyuki Shibata
- Department of Pathology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Yasushi Kawaguchi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Massemin A, Goehrig D, Flaman J, Jaber S, Griveau A, Djebali S, Marcos E, Payen L, Marvel J, Parent R, Adnot S, Bertolino P, Rieusset J, Tortereau A, Vindrieux D, Bernard D. Loss of Pla2r1 decreases cellular senescence and age-related alterations caused by aging and Western diets. Aging Cell 2023; 22:e13971. [PMID: 37667516 PMCID: PMC10652324 DOI: 10.1111/acel.13971] [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: 08/02/2022] [Revised: 06/21/2023] [Accepted: 08/04/2023] [Indexed: 09/06/2023] Open
Abstract
Cellular senescence is induced by many stresses including telomere shortening, DNA damage, oxidative, or metabolic stresses. Senescent cells are stably cell cycle arrested and they secrete many factors including cytokines and chemokines. Accumulation of senescent cells promotes many age-related alterations and diseases. In this study, we investigated the role of the pro-senescent phospholipase A2 receptor 1 (PLA2R1) in regulating some age-related alterations in old mice and in mice subjected to a Western diet, whereas aged wild-type mice displayed a decreased ability to regulate their glycemia during glucose and insulin tolerance tests, aged Pla2r1 knockout (KO) mice efficiently regulated their glycemia and displayed fewer signs of aging. Loss of Pla2r1 was also found protective against the deleterious effects of a Western diet. Moreover, these Pla2r1 KO mice were partially protected from diet-induced senescent cell accumulation, steatosis, and fibrosis. Together these results support that Pla2r1 drives several age-related alterations, especially in the liver, arising during aging or through a Western diet.
Collapse
Affiliation(s)
- Amélie Massemin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Delphine Goehrig
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Jean‐Michel Flaman
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Sara Jaber
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
| | - Audrey Griveau
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
| | - Sophia Djebali
- Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, École Normale Supérieure de LyonUniversité de Lyon, Université Claude Bernard Lyon 1LyonFrance
| | - Elisabeth Marcos
- INSERM U955, Département de Physiologie ‐ Explorations fonctionnelles, Hôpital Henri MondorAP‐HP, FHU SENECCréteilFrance
| | - Léa Payen
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Lyon SudHospices Civils de LyonPierre BéniteFrance
| | - Jacqueline Marvel
- Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, École Normale Supérieure de LyonUniversité de Lyon, Université Claude Bernard Lyon 1LyonFrance
| | - Romain Parent
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
| | - Serge Adnot
- INSERM U955, Département de Physiologie ‐ Explorations fonctionnelles, Hôpital Henri MondorAP‐HP, FHU SENECCréteilFrance
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - Jennifer Rieusset
- CarMeN Laboratory, UMR INSERM U1060/INRA U1397Lyon 1 UniversityPierre béniteFrance
| | - Antonin Tortereau
- VetAgro Sup, Interactions Cellules Environnement (ICE)Université de LyonMarcy l'EtoileFrance
| | - David Vindrieux
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon BérardUniversité de LyonLyonFrance
- Equipe Labellisée la Ligue Contre le CancerLyonFrance
| |
Collapse
|
25
|
Özgüden-Akkoc CG, Mutlu AM, Keskin A, Yumuşak E, Akkoc A. Phenotypic evaluation of mast cells in bovine mammary tissue and mastitis in the context of fibrosis. J DAIRY RES 2023; 90:387-392. [PMID: 38186214 DOI: 10.1017/s0022029923000651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
This research paper addresses the hypothesis that mast cells (MCs) contribute to the formation of mammary fibrosis. MCs are important immune regulatory and immune modulatory cells that play major roles in the inflammatory process. Since there is no detailed knowledge, this research study aimed to comparatively investigate the presence, localization, and immunophenotypes of MCs in healthy and mastitic mammary tissues. A total of 264 mammary samples were evaluated for the examination of mast cells and fibrosis. The mean mast cell number in both acute and chronic mastitis samples were very significantly higher than the control group P < 0.001). A 7.9-fold increase in the number of mast cells was found when the chronic mastitis group was compared with the control (healthy) group. Immunohistochemistry revealed presence of all three immune phenotypes in control and mastitic mammary samples (tryptase + (MCT), chymase + (MCC) and both chymase and tryptase + (MCTC). The mean MCT, MCC, and MCTC numbers in the chronic mastitis group were found to be significantly higher than the control (P < 0.001 for all three phenotypes) but did not differ significantly between control and acute mastitis samples. When the mean numbers of MCT, MCC, and MCTC in the control group and chronic mastitis group were compared, a 10.5, 7.8, and a 4.1-fold increase was observed, respectively. The amount of connective tissue was strongly increased in tissues with chronic mastitis and a 3.01-fold increase was detected compared to the control group. A statistically significant relation was also found between the amount of fibrosis and the increased number of total MCs (P < 0.001).
Collapse
Affiliation(s)
- Cansel Güzin Özgüden-Akkoc
- Department of Histology & Embryology, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Ayşe Meriç Mutlu
- Department of Pathology, Health Sciences Institute, Bursa Uludağ University, Bursa, Turkey
| | - Abdülkadir Keskin
- Department of Gynaecology & Obstetrics, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Ezgi Yumuşak
- Department of Pathology, Health Sciences Institute, Bursa Uludağ University, Bursa, Turkey
| | - Ahmet Akkoc
- Department of Pathology, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa, Turkey
| |
Collapse
|
26
|
Ishi S, Kanno E, Tanno H, Kurosaka S, Shoji M, Imai T, Yamaguchi K, Kotsugai K, Niiyama M, Kurachi H, Makabe F, Watanabe T, Sato K, Ishii K, Hara H, Imai Y, Kawakami K. Cutaneous wound healing promoted by topical administration of heat-killed Lactobacillus plantarum KB131 and possible contribution of CARD9-mediated signaling. Sci Rep 2023; 13:15917. [PMID: 37741861 PMCID: PMC10517988 DOI: 10.1038/s41598-023-42919-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/16/2023] [Indexed: 09/25/2023] Open
Abstract
Optimal conditions for wound healing require a smooth transition from the early stage of inflammation to proliferation, and during this time alternatively activated (M2) macrophages play a central role. Recently, heat-killed lactic acid bacteria (LAB), such as Lactobacillus plantarum (L. plantarum) have been reported as possible modulators affecting the immune responses in wound healing. However, how signaling molecules regulate this process after the administration of heat-killed LAB remains unclear. In this study, we examined the effect of heat-killed L. plantarum KB131 (KB131) administration on wound healing and the contribution of CARD9, which is an essential signaling adaptor molecule for NF-kB activation upon triggering through C-type lectin receptors, in the effects of this bacterium. We analyzed wound closure, histological findings, and inflammatory responses. We found that administration of KB131 accelerated wound closure, re-epithelialization, granulation area, CD31-positive vessels, and α-SMA-positive myofibroblast accumulated area, as well as the local infiltration of leukocytes. In particular, M2 macrophages were increased, in parallel with CCL5 synthesis. The acceleration of wound healing responses by KB131 was canceled in CARD9-knockout mice. These results indicate that the topical administration of KB131 accelerates wound healing, accompanying increased M2 macrophages, which suggests that CARD9 may be involved in these responses.
Collapse
Affiliation(s)
- Shinyo Ishi
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Emi Kanno
- Department of Translational Science for Nursing, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Hiromasa Tanno
- Department of Translational Science for Nursing, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Shiho Kurosaka
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
- Bio-Lab Co., Ltd, 2-1-3 Komagawa, Hidaka-shi, Japan
| | - Miki Shoji
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Toshiro Imai
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Kenji Yamaguchi
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Kanna Kotsugai
- Department of Translational Science for Nursing, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Momoko Niiyama
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Haruko Kurachi
- Department of Translational Science for Nursing, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Fuko Makabe
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | | | - Ko Sato
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
- Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
- Department of Clinical Microbiology and Infection, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Keiko Ishii
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yoshimichi Imai
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
- Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Japan
| |
Collapse
|
27
|
Xu M, Zhao C, Song H, Wang C, Li H, Qiu X, Jing H, Zhuang W. Inhibitory effects of Schisandrin C on collagen behavior in pulmonary fibrosis. Sci Rep 2023; 13:13475. [PMID: 37596361 PMCID: PMC10439186 DOI: 10.1038/s41598-023-40631-6] [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: 04/07/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Pulmonary fibrosis (PF) is a serious progressive fibrotic disease that is characterized by excessive accumulation of extracellular matrix (ECM), thus resulting in stiff lung tissues. Lysyl oxidase (LOX) is an enzyme involved in fibrosis by catalyzing collagen cross-linking. Studies found that the ingredients in schisandra ameliorated bleomycin (BLM)-induced PF, but it is unknown whether the anti-PF of schisandra is related to LOX. In this study, we established models of PF including a mouse model stimulated by BLM and a HFL1 cell model induced by transforming growth factor (TGF)-β1 to evaluate the inhibition effects of Schisandrin C (Sch C) on PF. We observed that Sch C treatment decreased pulmonary indexes compared to control group. Treatment of Sch C showed a significant reduction in the accumulation of ECM as evidenced by decreased expressions of α-SMA, FN, MMP2, MMP9, TIMP1 and collagen proteins such as Col 1A1, and Col 3A1. In addition, the expression of LOX in the lung tissue of mice after Sch C treatment was effectively decreased compared with the MOD group. The inhibition effects in vitro were consistent with those in vivo. Mechanistic studies revealed that Sch C significantly inhibited TGF-β1/Smad2/3 and TNF-α/JNK signaling pathways. In conclusion, our data demonstrated that Sch C significantly ameliorated PF in vivo and vitro, which may play an important role by reducing ECM deposition and inhibiting the production of LOX.
Collapse
Affiliation(s)
- Mingchen Xu
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China
| | - Chenghe Zhao
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China
| | - Haiming Song
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China
| | - Chunmei Wang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, China
| | - He Li
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, China
| | - Xudong Qiu
- Department of Hand Surgery, Affiliated Hospital, Beihua University, Jilin, China
| | - He Jing
- Department of Hand Surgery, Affiliated Hospital, Beihua University, Jilin, China
| | - Wenyue Zhuang
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China.
| |
Collapse
|
28
|
Kohon MY, Zaaroor Levy M, Hornik-Lurie T, Shalom A, Berl A, Drucker L, Levy Y, Tartakover Matalon S. αvβ3 Integrin as a Link between the Development of Fibrosis and Thyroid Hormones in Systemic Sclerosis. Int J Mol Sci 2023; 24:ijms24108927. [PMID: 37240272 DOI: 10.3390/ijms24108927] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of the skin and internal organs. Key players mediating fibrosis are myofibroblasts (MF) that, following transforming growth factor β (TGFβ) exposure, produce a collagen-rich extracellular matrix (ECM) that induces myofibroblast differentiation. Myofibroblasts express αvβ3 integrin (a membrane receptor for thyroid hormones) and miRNA-21 that promotes deiodinase-type-3 expression (D3), causing the degradation of triiodothyronine (T3) that attenuates fibrosis. We hypothesized that αvβ3 affects the fibrotic processes through its thyroid hormones (THs) binding site. To test this, dermal fibroblasts (DF) were cultured with/without TGFβ and removed with a base, leaving only normal/fibrotic ECMs in wells. Then, DF were cultured on the ECMs with/without tetrac (αvβ3 ligand, T4 antagonist), and evaluated for pro-fibrotic characteristics, αvβ3, miRNA-21, and D3 levels. Blood free-T3 (fT3), miRNA-21 levels, and the modified Rodnan skin score (MRSS) were evaluated in SSc patients. We found that the "fibrotic-ECM" significantly increased the pro-fibrotic characteristics of DF and the levels of miRNA-21, D3, and αvβ3, compared to the "normal-ECM." Tetrac significantly inhibited the effects of the "fibrotic-ECM" on the cells. In accordance with tetrac's effect on D3/miRNA-21, a negative correlation was found between the patients' fT3 to miRNA-21 levels, and to the development of pulmonary arterial hypertension (PAH). We conclude that occupying the THs binding site of αvβ3 may delay the development of fibrosis.
Collapse
Affiliation(s)
- Maia Yamila Kohon
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Autoimmune Research Laboratory, Meir Medical Center, Kfar Saba 4428164, Israel
| | - Mor Zaaroor Levy
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Autoimmune Research Laboratory, Meir Medical Center, Kfar Saba 4428164, Israel
| | - Tzipi Hornik-Lurie
- Data Research Department, Meir Medical Center, Kfar Saba 4428164, Israel
| | - Avshalom Shalom
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Plastic Surgery, Meir Medical Center, Kfar Saba 4428164, Israel
| | - Ariel Berl
- Department of Plastic Surgery, Meir Medical Center, Kfar Saba 4428164, Israel
| | - Liat Drucker
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Oncogenetics Laboratory, Meir Medical Center, Kfar Saba 4428164, Israel
| | - Yair Levy
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Autoimmune Research Laboratory, Meir Medical Center, Kfar Saba 4428164, Israel
- Department of Internal Medicine E, Meir Medical Center, Kfar Saba 4428164, Israel
| | - Shelly Tartakover Matalon
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Autoimmune Research Laboratory, Meir Medical Center, Kfar Saba 4428164, Israel
| |
Collapse
|
29
|
Li R, Kang H, Chen S. From Basic Research to Clinical Practice: Considerations for Treatment Drugs for Silicosis. Int J Mol Sci 2023; 24:ijms24098333. [PMID: 37176040 PMCID: PMC10179659 DOI: 10.3390/ijms24098333] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Silicosis, characterized by irreversible pulmonary fibrosis, remains a major global public health problem. Nowadays, cumulative studies are focusing on elucidating the pathogenesis of silicosis in order to identify preventive or therapeutic antifibrotic agents. However, the existing research on the mechanism of silica-dust-induced pulmonary fibrosis is only the tip of the iceberg and lags far behind clinical needs. Idiopathic pulmonary fibrosis (IPF), as a pulmonary fibrosis disease, also has the same problem. In this study, we examined the relationship between silicosis and IPF from the perspective of their pathogenesis and fibrotic characteristics, further discussing current drug research and limitations of clinical application in silicosis. Overall, this review provided novel insights for clinical treatment of silicosis with the hope of bridging the gap between research and practice in silicosis.
Collapse
Affiliation(s)
- Rou Li
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Huimin Kang
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Shi Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha 410013, China
| |
Collapse
|
30
|
Wei L, Liu L, Bai M, Ning X, Sun S. CircRNAs: versatile players and new targets in organ fibrosis. Cell Commun Signal 2023; 21:90. [PMID: 37131173 PMCID: PMC10152639 DOI: 10.1186/s12964-023-01051-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/15/2023] [Indexed: 05/04/2023] Open
Abstract
Organ fibrosis can occur in virtually all major organs with relentlessly progressive and irreversible progress, ultimately resulting in organ dysfunction and potentially death. Unfortunately, current clinical treatments cannot halt or reverse the progression of fibrosis to end-stage organ failure, and thus, advanced antifibrotic therapeutics are urgently needed. In recent years, a growing body of research has revealed that circular RNAs (circRNAs) play pivotal roles in the development and progression of organ fibrosis through highly diverse mechanisms of action. Thus, manipulating circRNAs has emerged as a promising strategy to mitigate fibrosis across different organ types. In this review, we systemically summarize the current state of knowledge about circRNA biological properties and the regulatory mechanisms of circRNAs. A comprehensive overview of major fibrotic signaling pathways and representative circRNAs that are known to modulate fibrotic signals are outlined. Then, we focus on the research progress of the versatile functional roles and underlying molecular mechanisms of circRNAs in various fibrotic diseases in different organs, including the heart, liver, lung, kidney and skin. Finally, we offer a glimpse into the prospects of circRNA-based interference and therapy, as well as their utilization as biomarkers in the diagnosis and prognosis of fibrotic diseases. Video abstract.
Collapse
Affiliation(s)
- Lei Wei
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Limin Liu
- School of Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710032, Shaanxi, China
| | - Ming Bai
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, No. 127 Changle West Road, Xi'an, Shaanxi, China
| | - Xiaoxuan Ning
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an, 710032, Shaanxi, China.
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, No. 127 Changle West Road, Xi'an, Shaanxi, China.
| |
Collapse
|
31
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
32
|
Wenglén C, Demirel I, Eremo AG, Grenegård M, Paramel GV. Targeting serotonin receptor 2B inhibits TGFβ induced differentiation of human vascular smooth muscle cells. Eur J Pharmacol 2023; 944:175570. [PMID: 36781042 DOI: 10.1016/j.ejphar.2023.175570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/13/2023]
Abstract
Vascular Smooth Muscle Cells (VSMCs) are known to be the key drivers of intimal thickening which contribute to early progression of atherosclerosis. VSMCs are the major producers of extracellular matrix within the vessel wall and in response to atherogenic stimuli they could modify the type of matrix proteins produced. Serotonin receptor 2B (5-HT2B receptor/HTR2B) has been implicated in several chronic fibrotic and vascular diseases. Although studies have successfully demonstrated the efficacy of HTR2B blockade in attenuating fibrotic disease, the role of 5-HT2B receptor in TGFβ mediated VSMC differentiation remain largely unknown. In the present study, we investigated the potential of targeting the 5-HT2B receptor to prevent TGFβ induced VSMCs differentiation. Our results showed that 5-HT2B receptors are expressed in human atherosclerotic lesion and HTR2B expression positively correlated to the VSMCs markers. We show that AM1125, a selective 5-HT2B receptor inhibitor, significantly inhibits TGFβ1 induced production of collagen and CTGF. The investigation of underlying mechanisms indicated that 5-HT2B receptor antagonism blocks phospho-Smad2 mediated downstream signaling of TGFβ1 in vascular smooth muscle cells. Collectively, the HTR2B/TGF-β1/Phospho-Smad2 pathway plays a critical role in the regulation of VSMCs differentiation. Our findings might serve 5-HT2B receptor as a therapeutic target to limit TGF-β1 induced VSMC differentiation.
Collapse
Affiliation(s)
| | - Isak Demirel
- Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Anna Göthlin Eremo
- Department of Clinical Research Laboratory, Faculty of Medicine, and Health, Örebro University, Örebro, Sweden
| | - Magnus Grenegård
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Geena V Paramel
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden.
| |
Collapse
|
33
|
Garufi A, Pistritto G, D’Orazi G. HIPK2 as a Novel Regulator of Fibrosis. Cancers (Basel) 2023; 15:1059. [PMID: 36831402 PMCID: PMC9954661 DOI: 10.3390/cancers15041059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Fibrosis is an unmet medical problem due to a lack of evident biomarkers to help develop efficient targeted therapies. Fibrosis can affect almost every organ and eventually induce organ failure. Homeodomain-interacting protein kinase 2 (HIPK2) is a protein kinase that controls several molecular pathways involved in cell death and development and it has been extensively studied, mainly in the cancer biology field. Recently, a role for HIPK2 has been highlighted in tissue fibrosis. Thus, HIPK2 regulates several pro-fibrotic pathways such as Wnt/β-catenin, TGF-β and Notch involved in renal, pulmonary, liver and cardiac fibrosis. These findings suggest a wider role for HIPK2 in tissue physiopathology and highlight HIPK2 as a promising target for therapeutic purposes in fibrosis. Here, we will summarize the recent studies showing the involvement of HIPK2 as a novel regulator of fibrosis.
Collapse
Affiliation(s)
- Alessia Garufi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Giuseppa Pistritto
- Centralized Procedures Office, Italian Medicines Agency (AIFA), 00187 Rome, Italy
| | - Gabriella D’Orazi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
| |
Collapse
|
34
|
Dolivo DM, Sun LS, Rodrigues AE, Galiano RD, Mustoe TA, Hong SJ. Epidermal Potentiation of Dermal Fibrosis: Lessons from Occlusion and Mucosal Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:510-519. [PMID: 36740181 DOI: 10.1016/j.ajpath.2023.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Fibrotic skin conditions, such as hypertrophic and keloid scars, frequently result from injury to the skin and as sequelae to surgical procedures. The development of skin fibrosis may lead to patient discomfort, limitation in range of motion, and cosmetic disfigurement. Despite the frequency of skin fibrosis, treatments that seek to address the root causes of fibrosis are lacking. Much research into fibrotic pathophysiology has focused on dermal pathology, but less research has been performed to understand aberrations in fibrotic epidermis, leading to an incomplete understanding of dermal fibrosis. The literature on occlusion, a treatment modality known to reduce dermal fibrosis, in part through accelerating wound healing and regulating aberrant epidermal inflammation that otherwise drives fibrosis in the dermis, is reviewed. There is a focus on epidermal-dermal crosstalk, which contributes to the development and maintenance of dermal fibrosis, an underemphasized interplay that may yield novel strategies for treatment if understood in more detail.
Collapse
Affiliation(s)
- David M Dolivo
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lauren S Sun
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Adrian E Rodrigues
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Robert D Galiano
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Thomas A Mustoe
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Seok Jong Hong
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
35
|
Dean LS, Chow DC, Ndhlovu LC, Boisvert WA, Chang SP, Shikuma CM, Park J. Characterization of Circulating Fibrocytes in People Living with HIV on Stable Antiretroviral Therapy. Immunohorizons 2022; 6:760-767. [PMID: 36445359 PMCID: PMC10402248 DOI: 10.4049/immunohorizons.2200085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 01/04/2023] Open
Abstract
Highly effective combination antiretroviral therapy has reduced HIV infection to a manageable chronic disease, shifting the clinical landscape toward management of noninfectious comorbidities in people living with HIV (PLWH). These comorbidities are diverse, generally associated with accelerated aging, and present within multiple organ systems. Mechanistically, immune dysregulation and chronic inflammation, both of which persist in PLWH with well-controlled virally suppressive HIV infection, are suggested to create and exacerbate noninfectious comorbidity development. Persistent inflammation often leads to fibrosis, which is the common end point pathologic feature associated with most comorbidities. Fibrocytes are bone marrow-derived fibroblast-like cells, which emerged as key effector cells in tissue repair and pathologic fibrotic diseases. Despite their relevance to fibrosis, the circulating fibrocyte concentration in PLWH remains poorly characterized, and an understanding of their functional role in chronic HIV is limited. In this study, utilizing PBMCs from a cross-sectional adult HIV cohort study with matched uninfected controls (HIV-), we aimed to identify and compare circulating fibrocytes in blood. Both the percentage and number of fibrocytes and α-smooth muscle actin+ fibrocytes in circulation did not differ between the HIV+ and HIV- groups. However, circulating fibrocyte levels were significantly associated with increasing age in both the HIV+ and HIV- groups (the percentage and number; r = 0.575, p ≤ 0.0001 and r = 0.558, p ≤ 0.0001, respectively). Our study demonstrates that circulating fibrocyte levels and their fibroblast-like phenotype defined as collagen I and α-smooth muscle actin+ expression are comparable between, and strongly associated with, age irrespective of HIV status.
Collapse
Affiliation(s)
- Logan S. Dean
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii at Manoa, Honolulu, HI
- Hawaii Center for AIDS, John A. Burns School Medicine, University of Hawaii at Manoa, Honolulu, HI
| | - Dominic C. Chow
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii at Manoa, Honolulu, HI
- Hawaii Center for AIDS, John A. Burns School Medicine, University of Hawaii at Manoa, Honolulu, HI
| | - Lishomwa C. Ndhlovu
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii at Manoa, Honolulu, HI
| | - William A. Boisvert
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI
| | - Sandra P. Chang
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii at Manoa, Honolulu, HI
| | - Cecilia M. Shikuma
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii at Manoa, Honolulu, HI
- Hawaii Center for AIDS, John A. Burns School Medicine, University of Hawaii at Manoa, Honolulu, HI
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, University of Hawaii at Manoa, Honolulu, HI
- Hawaii Center for AIDS, John A. Burns School Medicine, University of Hawaii at Manoa, Honolulu, HI
| |
Collapse
|
36
|
Kundu A, Gali S, Sharma S, Park JH, Kyung SY, Kacew S, Kim IS, Lee KY, Kim HS. Tenovin-1 Ameliorates Renal Fibrosis in High-Fat-Diet-Induced Diabetic Nephropathy via Antioxidant and Anti-Inflammatory Pathways. Antioxidants (Basel) 2022; 11:antiox11091812. [PMID: 36139886 PMCID: PMC9495519 DOI: 10.3390/antiox11091812] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
High-fat diet (HFD)-induced obesity has been involved in the development of diabetic nephropathy (DN). Tenovin-1, a potent selective SIRT1/2 inhibitor, regulates various target proteins. The present study evaluated the protective effect of Tenovin-1 against renal fibrosis in HFD-induced Zucker diabetic fatty (ZDF) rats. Rats were fed a normal chow diet or HFD. Tenovin-1 (45 mg/kg) administered to HFD-fed rats decreased inflammatory cytokine expression in the serum of the rats. The antioxidant status and oxidative damage to lipids or DNA were significantly restored by Tenovin-1. Additionally, Tenovin-1 reduced the levels of blood urea nitrogen (BUN), serum creatinine (sCr), microalbumin, and urinary protein-based biomarkers in the urine of HFD-fed rats. The abnormal architecture of the kidney and pancreas was restored by Tenovin-1 administration. Tenovin-1 also reduced apoptosis in the kidneys of the HFD-fed rats and HG-treated NRK-52E cells. It significantly lowered the levels of ECM proteins in the kidneys of HFD-fed rats and HG-treated NRK-52E cells. Additionally, Tenovin-1 markedly reduced claudin-1, SIRT1, and SIRT2, but increased SIRT3 and SIRT4 in HFD-fed rats and NRK-52E cells treated with HG. Furthermore, Tenovin-1 altered epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor-β (PDGFR-β), and signal transducer and activator of transcription 3 (STAT3) levels in the kidneys of HFD-fed rats. Conclusively, this study shows that Tenovin-1 can be a potential candidate drug for the treatment of HFD-induced renal fibrosis, in vivo and in vitro models.
Collapse
Affiliation(s)
- Amit Kundu
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 440-746, Korea
| | - Sreevarsha Gali
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 440-746, Korea
| | - Swati Sharma
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 440-746, Korea
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 440-746, Korea
| | - So Young Kyung
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 440-746, Korea
| | - Sam Kacew
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 440-746, Korea
| | - Kwang Youl Lee
- College of Pharmacy, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju 61186, Korea
- Correspondence: (K.Y.L.); (H.S.K.)
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 440-746, Korea
- Correspondence: (K.Y.L.); (H.S.K.)
| |
Collapse
|
37
|
Bell TJ, Nagel DJ, Woeller CF, Kottmann RM. Ogerin mediated inhibition of TGF-β(1) induced myofibroblast differentiation is potentiated by acidic pH. PLoS One 2022; 17:e0271608. [PMID: 35901086 PMCID: PMC9333254 DOI: 10.1371/journal.pone.0271608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 07/01/2022] [Indexed: 01/28/2023] Open
Abstract
Transforming growth factor beta (TGF-β) induced myofibroblast differentiation is central to the pathological scarring observed in Idiopathic Pulmonary Fibrosis (IPF) and other fibrotic diseases. Our lab has recently identified expression of GPR68 (Ovarian Cancer Gene Receptor 1, OGR1), a pH sensing G-protein coupled receptor, as a negative regulator of TGF-β induced profibrotic effects in primary human lung fibroblasts (PHLFs). We therefore hypothesized that small molecule activators of GPR68 would inhibit myofibroblast differentiation. Ogerin is a positive allosteric modulator (PAM) of GPR68, inducing a leftward shift of the dose response curve to proton induced signaling. Using PHLFs derived from patients with both non-fibrotic and IPF diagnoses, we show that Ogerin inhibits, and partially reverses TGF-β induced myofibroblast differentiation in a dose dependent manner. This occurs at the transcriptional level without inhibition of canonical TGF-β induced SMAD signaling. Ogerin induces PKA dependent CREB phosphorylation, a marker of Gαs pathway activation. The ability of Ogerin to inhibit both basal and TGF-β induced collagen gene transcription, and induction of Gαs signaling is enhanced at an acidic pH (pH 6.8). Similar findings were also found using fibroblasts derived from dermal, intestinal, and orbital tissue. The biological role of GPR68 in different tissues, cell types, and disease states is an evolving and emerging field. This work adds to the understanding of Gαs coupled GPCRs in fibrotic lung disease, the ability to harness the pH sensing properties of GPR68, and conserved mechanisms of fibrosis across different organ systems.
Collapse
Affiliation(s)
- Tyler J. Bell
- Department of Environmental Medicine Toxicology Training Program, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - David J. Nagel
- Department of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Collynn F. Woeller
- Department of Ophthalmology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - R. Mathew Kottmann
- Department of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
- * E-mail:
| |
Collapse
|
38
|
Ortiz-Zapater E, Signes-Costa J, Montero P, Roger I. Lung Fibrosis and Fibrosis in the Lungs: Is It All about Myofibroblasts? Biomedicines 2022; 10:biomedicines10061423. [PMID: 35740444 PMCID: PMC9220162 DOI: 10.3390/biomedicines10061423] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022] Open
Abstract
In the lungs, fibrosis is a growing clinical problem that results in shortness of breath and can end up in respiratory failure. Even though the main fibrotic disease affecting the lung is idiopathic pulmonary fibrosis (IPF), which affects the interstitial space, there are many fibrotic events that have high and dangerous consequences for the lungs. Asthma, chronic obstructive pulmonary disease (COPD), excessive allergies, clearance of infection or COVID-19, all are frequent diseases that show lung fibrosis. In this review, we describe the different kinds of fibrosis and analyse the main types of cells involved-myofibroblasts and other cells, like macrophages-and review the main fibrotic mechanisms. Finally, we analyse present treatments for fibrosis in the lungs and highlight potential targets for anti-fibrotic therapies.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Faculty of Medicine-IIS INCLIVA, University of Valencia, 46010 Valencia, Spain
- Correspondence:
| | | | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (P.M.); (I.R.)
| | - Inés Roger
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (P.M.); (I.R.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
| |
Collapse
|
39
|
Binatti E, Gerussi A, Barisani D, Invernizzi P. The Role of Macrophages in Liver Fibrosis: New Therapeutic Opportunities. Int J Mol Sci 2022; 23:6649. [PMID: 35743092 PMCID: PMC9224467 DOI: 10.3390/ijms23126649] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation is the hallmark of fibrotic disorders and is characterized by the activation of immune cells in the damaged tissues. Macrophages have emerged as central players in the fibrotic process since they initiate, sustain and amplify the inflammatory reaction. As regards the liver, distinct populations of phagocytic cells, like Kupffer cells and monocyte-derived macrophages, are indisputably key cells implicated in the pathogenesis of several chronic liver diseases. In this review, we summarize the current knowledge on the origin, role and functions of macrophages in fibrotic conditions, with a specific focus on liver fibrosis; then, we discuss some innovative therapeutic strategies targeting macrophages in fibrotic liver diseases.
Collapse
Affiliation(s)
- Eleonora Binatti
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, Università degli Studi di Milano Bicocca, 20900 Monza, Italy; (A.G.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| | - Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, Università degli Studi di Milano Bicocca, 20900 Monza, Italy; (A.G.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| | - Donatella Barisani
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, Università degli Studi di Milano Bicocca, 20900 Monza, Italy; (A.G.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| |
Collapse
|
40
|
Li Z, Zhu Z, Liu Y, Liu Y, Zhao H. Function and regulation of GPX4 in the development and progression of fibrotic disease. J Cell Physiol 2022; 237:2808-2824. [PMID: 35605092 DOI: 10.1002/jcp.30780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023]
Abstract
Fibrosis is a common feature of fibrotic diseases that poses a serious threat to global health due to high morbidity and mortality in developing countries. There exist some chemical compounds and biomolecules associated with the development of fibrosis, including cytokines, hormones, and enzymes. Among them, glutathione peroxidase 4 (GPX4), as a selenoprotein antioxidant enzyme, is widely found in the embryo, testis, brain, liver, heart, and photoreceptor cells. Moreover, it is shown that GPX4 elicits diverse biological functions by suppressing phospholipid hydroperoxide at the expense of decreased glutathione (GSH), including loss of neurons, autophagy, cell repair, inflammation, ferroptosis, apoptosis, and oxidative stress. Interestingly, these processes are intimately related to the occurrence of fibrotic disease. Recently, GPX4 has been reported to exhibit a decline in fibrotic disease and inhibit fibrosis, suggesting that alterations of GPX4 can change the course or dictate the outcome of fibrotic disease. In this review, we summarize the role and underlying mechanisms of GPX4 in fibrosis diseases such as lung fibrosis, liver fibrosis, kidney fibrosis, cardiac fibrosis, and myelofibrosis.
Collapse
Affiliation(s)
- Zhaobing Li
- Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunnan, China
| | - Zigui Zhu
- Department of Intensive Care Units, The Affiliated Nanhua Hospital, Hengyang Medical school, University of South China, Hengyang, Hunnan, China
| | - Yulu Liu
- Department of Intensive Care Units, The Affiliated Nanhua Hospital, Hengyang Medical school, University of South China, Hengyang, Hunnan, China
| | - Yannan Liu
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Hong Zhao
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
41
|
Mitchell S, Lee A, Stenquist R, Yatsonsky II D, Mooney ML, Shendge VB. Extensive adhesion formation in a total knee replacement in the setting of a gastrointestinal stromal tumor: A case report. World J Orthop 2022; 13:538-543. [PMID: 35633745 PMCID: PMC9124998 DOI: 10.5312/wjo.v13.i5.538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/04/2022] [Accepted: 04/24/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are rare primary neoplasms of the gastrointestinal tract, accounting for 1% to 2% of all gastrointestinal neoplasms worldwide. GISTs are frequently discovered incidentally during workup for other diagnosis or intestinal obstruction, as they can present with few or no symptoms. Simultaneously, GISTs confer a high degree of malignant transformation, with a progression in about 10% to 30% of cases.
CASE SUMMARY A 63-year-old healthy female presented to our institution with complaints of right knee pain and limited passive and active motion in the setting of a previous right total knee arthroplasty (TKA). One year after TKA, the patient was incidentally diagnosed with a GIST, which was successfully removed. After removal, the patient continued to have limited range of motion of the right knee and subsequently underwent revision TKA. Intraoperatively significant fibrotic adhesions were found encapsulating the femoral and tibial components. The patient’s pain improved postoperatively, however, she continued to have decreased range of motion with difficulty ambulating.
CONCLUSION We propose that this case may demonstrate a proinflammatory milieu arising from a GIST, which had a direct influence on the outcome of recent total knee arthroplasty. This proposed mechanism between neoplastic cytokinetic activity and adhesion formation could have implications on preoperative and postoperative orthopedic management of total knee arthroplasty.
Collapse
Affiliation(s)
- Steven Mitchell
- Department ofOrthopaedic Surgery, The University of Toledo Medical Center, Toledo, OH 43614, United States
| | - Anderson Lee
- Department ofOrthopaedic Surgery, The University of Toledo Medical Center, Toledo, OH 43614, United States
| | - Ryan Stenquist
- Department ofOrthopaedic Surgery, The University of Toledo Medical Center, Toledo, OH 43614, United States
| | - David Yatsonsky II
- Department ofOrthopaedic Surgery, The University of Toledo Medical Center, Toledo, OH 43614, United States
| | - Megan L Mooney
- Department ofOrthopaedic Surgery, The University of Toledo Medical Center, Toledo, OH 43614, United States
| | - Vithal B Shendge
- Department ofOrthopaedic Surgery, The University of Toledo Medical Center, Toledo, OH 43614, United States
| |
Collapse
|
42
|
Treatment of Chronic Kidney Disease with Extracellular Vesicles from Mesenchymal Stem Cells and CD133 + Expanded Cells: A Comparative Preclinical Analysis. Int J Mol Sci 2022; 23:ijms23052521. [PMID: 35269664 PMCID: PMC8910174 DOI: 10.3390/ijms23052521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic kidney disease (CKD) is characterized by structural abnormalities and the progressive loss of kidney function. Extracellular vesicles (EVs) from human umbilical cord tissue (hUCT)-derived mesenchymal stem cells (MSCs) and expanded human umbilical cord blood (hUCB)-derived CD133+ cells (eCD133+) maintain the characteristics of the parent cells, providing a new form of cell-free treatment. We evaluated the effects of EVs from hUCT-derived MSCs and hUCB-derived CD133+ cells on rats with CDK induced by an adenine-enriched diet. EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis (NTA) and electron microscopy. The animals were randomized and divided into the MSC-EV group, eEPC-EV group and control group. Infusions occurred on the seventh and 14th days after CKD induction. Evaluations of kidney function were carried out by biochemical and histological analyses. Intense labeling of the α-SMA protein was observed when comparing the control with MSC-EVs. In both groups treated with EVs, a significant increase in serum albumin was observed, and the increase in cystatin C was inhibited. The results indicated improvements in renal function in CKD, demonstrating the therapeutic potential of EVs derived from MSCs and eCD133+ cells and suggesting the possibility that in the future, more than one type of EV will be used concurrently.
Collapse
|
43
|
Coentro JQ, Di Nubila A, May U, Prince S, Zwaagstra J, Järvinen TAH, Zeugolis D. Dual drug delivery collagen vehicles for modulation of skin fibrosis in vitro. Biomed Mater 2022; 17. [PMID: 35176732 DOI: 10.1088/1748-605x/ac5673] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/17/2022] [Indexed: 11/11/2022]
Abstract
Single molecule drug delivery systems have failed to yield functional therapeutic outcomes, triggering investigations into multi-molecular drug delivery vehicles. In the context of skin fibrosis, although multi-drug systems have been assessed, no system has assessed molecular combinations that directly and specifically reduce cell proliferation, collagen synthesis and transforming growth factor β1 (TGFβ1) expression. Herein, a core-shell collagen type I hydrogel system was developed for the dual delivery of a TGFβ trap, a soluble recombinant protein that inhibits TGFβ signalling, and Trichostatin A (TSA), a small molecule inhibitor of histone deacetylases. The antifibrotic potential of the dual delivery system was assessed in an in vitro skin fibrosis model induced by macromolecular crowding (MMC) and TGFβ1. SDS-PAGE and HPLC analyses revealed that ~ 50 % of the TGFβ trap and ~ 30 % of the TSA were released from the core and shell compartments, respectively, of the hydrogel system after 10 days (longest time point assessed) in culture. As a direct consequence of this slow release, the core (TGFβ trap) / shell (TSA) hydrogel system induced significantly (p < 0.05) lower than the control group (MMC and TGFβ1) collagen type I deposition (assessed via SDS-PAGE and immunocytochemistry), α smooth muscle actin (αSMA) expression (assessed via immunocytochemistry) and cellular proliferation (assessed via DNA quantification) and viability (assessed via calcein AM and ethidium homodimer-I staining) after 10 days in culture. On the other hand, direct TSA-TGFβ supplementation induced the lowest (p < 0.05) collagen type I deposition, αSMA expression and cellular proliferation and viability after 10 days in culture. Our results illustrate the potential of core-shell collagen hydrogel systems for sustained delivery of antifibrotic molecules.
Collapse
Affiliation(s)
- João Q Coentro
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Biomedical Sciences Building, Galway, Galway, IRELAND
| | - Alessia Di Nubila
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Biomedical Sciences Building, Galway, Galway, IRELAND
| | - Ulrike May
- Faculty of Medicine & Health Technology, Tampere University, Kalevantie 4, Tampere, 33014, FINLAND
| | - Stuart Prince
- Faculty of Medicine & Health Technology, Tampere University, Kalevantie 4, Tampere, 33014, FINLAND
| | - John Zwaagstra
- Human Health Therapeutics Research Centre, National Research Council Canada, Human Health Therapeutics Research Centre, Montreal, Quebec, K1A 0R6, CANADA
| | - Tero A H Järvinen
- Faculty of Medicine & Health Technology, Tampere University, Faculty of Medicine & Health Technology, Tampere, 33014, FINLAND
| | - Dimitrios Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, University College Dublin, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, Dublin, 4, IRELAND
| |
Collapse
|
44
|
Liang M, Matteson EL, Abril A, Distler JH. The role of antifibrotics in the treatment of rheumatoid arthritis-associated interstitial lung disease. Ther Adv Musculoskelet Dis 2022; 14:1759720X221074457. [PMID: 35186127 PMCID: PMC8852164 DOI: 10.1177/1759720x221074457] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/03/2022] [Indexed: 12/21/2022] Open
Abstract
The major pulmonary complication of rheumatoid arthritis (RA) is interstitial lung disease (ILD), which causes significant morbidity and mortality and influences the natural course of disease. Recent advances in the management of arthritis have improved patient outcomes. However, exceptionally high medical needs still remain for effective therapies for the patients with ILD in RA. Better understanding of the shared and distinct pathophysiology of fibrotic diseases led to the development of novel antifibrotic agents such as nintedanib and pirfenidone. The further stratification analysis of the phase III INBUILD trial demonstrated beneficial effects of nintedanib in RA-ILD with a progressive phenotype by reducing the rate of decline in forced vital capacity (FVC) over 52 weeks by 60%. Pirfenidone is another antifibrotic agent currently under phase II clinical study (TRAIL1) aiming to evaluate its effects for RA-ILD. This review provides an overview of state-of-the-art pathogenesis and the current therapeutic options for RA-ILD, with a focus on antifibrotic strategies.
Collapse
Affiliation(s)
- Minrui Liang
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Eric L. Matteson
- Division of Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Andy Abril
- Division of Rheumatology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Jörg H.W. Distler
- Rheumatology and Clinical Immunology, Department of Internal Medicine 3, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
| |
Collapse
|
45
|
Gut Microbiome and Organ Fibrosis. Nutrients 2022; 14:nu14020352. [PMID: 35057530 PMCID: PMC8781069 DOI: 10.3390/nu14020352] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is a pathological process associated with most chronic inflammatory diseases. It is defined by an excessive deposition of extracellular matrix proteins and can affect nearly every tissue and organ system in the body. Fibroproliferative diseases, such as intestinal fibrosis, liver cirrhosis, progressive kidney disease and cardiovascular disease, often lead to severe organ damage and are a leading cause of morbidity and mortality worldwide, for which there are currently no effective therapies available. In the past decade, a growing body of evidence has highlighted the gut microbiome as a major player in the regulation of the innate and adaptive immune system, with severe implications in the pathogenesis of multiple immune-mediated disorders. Gut microbiota dysbiosis has been associated with the development and progression of fibrotic processes in various organs and is predicted to be a potential therapeutic target for fibrosis management. In this review we summarize the state of the art concerning the crosstalk between intestinal microbiota and organ fibrosis, address the relevance of diet in different fibrotic diseases and discuss gut microbiome-targeted therapeutic approaches that are current being explored.
Collapse
|
46
|
Höppner J, Bruni C, Distler O, Robson SC, Burmester GR, Siegert E, Distler JHW. Purinergic signaling in systemic sclerosis. Rheumatology (Oxford) 2021; 61:2770-2782. [PMID: 34849624 DOI: 10.1093/rheumatology/keab859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune rheumatic disease that involves numerous organs and presents major management challenges. The histopathologic hallmarks of SSc include vasculopathy, fibrosis and autoimmune phenomena involving both innate and adaptive immune systems. Purinergic signalling is a pathway that may be implicated in the pathophysiology of several of these disease manifestations. Extracellular purines are potent signalling mediators, which have been shown to be dysregulated in SSc. As examples, purines can exacerbate vasculopathy and provoke platelet dysfunction; as well as contributing to immune dysregulation. Elements of purinergic signalling further promote organ and tissue fibrosis in several disease models. Here, we provide an overview of extracellular purine metabolism in purinergic signalling and link disorders of these to the molecular pathology of SSc. We also discuss targeting the purinergic signalling and explore the translational applications for new therapeutic options in SSc.
Collapse
Affiliation(s)
- Jakob Höppner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Careggi University Hospital, University of Florence, Florence, Italy.,Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Simon C Robson
- Departments of Anesthesia and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elise Siegert
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
47
|
Huang CW, Lee SY, Wei TT, Kuo YH, Wu ST, Ku HC. A novel caffeic acid derivative prevents renal remodeling after ischemia/reperfusion injury. Biomed Pharmacother 2021; 142:112028. [PMID: 34399201 DOI: 10.1016/j.biopha.2021.112028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/19/2021] [Accepted: 08/07/2021] [Indexed: 11/29/2022] Open
Abstract
Acute kidney disease due to renal ischemia/reperfusion (I/R) is a major clinical problem without effective therapies. The injured tubular epithelial cells may undergo epithelial-mesenchymal transition (EMT). It will loss epithelial phenotypes and express the mesenchymal characteristics. The formation of scar tissue in the interstitial space during renal remodeling is caused by the excessive accumulation of extracellular matrix components and induced fibrosis. This study investigated the effect of caffeic acid ethanolamide (CAEA), a novel caffeic acid derivative, on renal remodeling after injury. The inhibitory role of CAEA on EMT was determined by western blotting, real-time PCR, and immunohistochemistry staining. Treating renal epithelial cells with CAEA in TGF-β exposed cell culture successfully maintained the content of E-cadherin and inhibited the expression of mesenchymal marker, indicating that CAEA prevented renal epithelial cells undergo EMT after TGF-β exposure. Unilateral renal I/R were performed in mice to induce renal remodeling models. CAEA can protect against I/R-induced renal remodeling by inhibiting inflammatory reactions and consecutively inhibiting TGF-β-induced EMT, characterized by the preserved E-cadherin expression and alleviated α-SMA and collagen expression, as well as the alleviated of renal fibrosis. We also revealed that CAEA may exhibits biological activity by targeting TGFBRI. CAEA may antagonize TGF-β signaling by interacting with TGFBR1, thereby blocking binding between TGF-β and TGFBR1 and reducing downstream signaling, such as Smad3 phosphorylation. Our data support the administration of CAEA after I/R as a viable method for preventing the progression of acute renal injury to renal fibrosis.
Collapse
Affiliation(s)
- Cheng-Wei Huang
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Shih-Yi Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, MacKay Memorial Hospital, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taitung MacKay Memorial Hospital, Taiwan
| | - Tzu-Tang Wei
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Shao-Tung Wu
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hui-Chun Ku
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
48
|
Böhm M. In search of the needle in a haystack: Finding a suitable serum biomarker for monitoring disease activity of systemic sclerosis. Exp Dermatol 2021; 30:880-886. [PMID: 34121239 DOI: 10.1111/exd.14403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Markus Böhm
- Department of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
49
|
Plikus MV, Krieg T. More than just bricks and mortar: Fibroblasts and ECM in skin health and disease. Exp Dermatol 2021; 30:4-9. [PMID: 33349992 PMCID: PMC9911308 DOI: 10.1111/exd.14257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Maksim V. Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA,Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA,NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA,Authors for correspondence: Maksim V. Plikus, Ph.D., Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA, and Thomas Krieg, M.D., FRCP, Translational Matrix Biology, University of Cologne, Jospeh-Stelzmann-Str. 52, D-50931 Cologne, Germany,
| | - Thomas Krieg
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany,Authors for correspondence: Maksim V. Plikus, Ph.D., Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA, and Thomas Krieg, M.D., FRCP, Translational Matrix Biology, University of Cologne, Jospeh-Stelzmann-Str. 52, D-50931 Cologne, Germany,
| |
Collapse
|
50
|
Petkovic M, Sørensen AE, Leal EC, Carvalho E, Dalgaard LT. Mechanistic Actions of microRNAs in Diabetic Wound Healing. Cells 2020; 9:E2228. [PMID: 33023156 PMCID: PMC7601058 DOI: 10.3390/cells9102228] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Wound healing is a complex biological process that is impaired under diabetes conditions. Chronic non-healing wounds in diabetes are some of the most expensive healthcare expenditures worldwide. Early diagnosis and efficacious treatment strategies are needed. microRNAs (miRNAs), a class of 18-25 nucleotide long RNAs, are important regulatory molecules involved in gene expression regulation and in the repression of translation, controlling protein expression in health and disease. Recently, miRNAs have emerged as critical players in impaired wound healing and could be targets for potential therapies for non-healing wounds. Here, we review and discuss the mechanistic background of miRNA actions in chronic wounds that can shed the light on their utilization as specific wound healing biomarkers.
Collapse
Affiliation(s)
- Marija Petkovic
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark; (A.E.S.); (L.T.D.)
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (E.C.L.); (E.C.)
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Anja Elaine Sørensen
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark; (A.E.S.); (L.T.D.)
| | - Ermelindo Carreira Leal
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (E.C.L.); (E.C.)
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Eugenia Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (E.C.L.); (E.C.)
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Department of Geriatrics, University of Arkansas for Medical Sciences, and Arkansas Children’s Research Institute, Little Rock, AR 72205, USA
| | - Louise Torp Dalgaard
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark; (A.E.S.); (L.T.D.)
| |
Collapse
|