1
|
Liao C, Wang P, Zeng Q, Yan G, Gao J, Liu J, Yan J, Zhang G, Liu Y, Wang X. Piezo1-Mediated Calcium Flux Transfers Mechanosignal to Yes-Associated Protein to Stimulate Matrix Production in Keloid. J Invest Dermatol 2025:S0022-202X(25)00415-4. [PMID: 40254148 DOI: 10.1016/j.jid.2025.03.039] [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: 11/14/2023] [Revised: 03/17/2025] [Accepted: 03/29/2025] [Indexed: 04/22/2025]
Abstract
Keloids are fibroproliferative diseases affecting millions of people worldwide, but curing keloids remains challenging. Mechanical force is a common initiator and driver of keloids, and blocking the proadhesive signaling pathways is expected to cure keloids. This study found higher levels of Piezo1 in human keloid fibroblasts than in normal skin fibroblasts. Single-cell transcriptome analysis revealed a correlation of Piezo1 with Yes-associated protein (YAP) in keloid fibroblasts. Knockdown of Piezo1/YAP in keloid fibroblasts versus fibroblasts decreased CCN2 and CCN1 expression and fibrosis-related cell behaviors, identifying Piezo1 and YAP as upstream signals of proadhesive signaling loop in keloids. Treatment of patient-derived keloid xenograft model with Piezo1 inhibitor GsMTx4 and YAP inhibitor verteporfin reduced keloid volume and decreased type I/III collagen ratio. Atomic force microscopy further confirmed the biomechanical improvements of keloids in elasticity, viscoelasticity, and roughness ex vivo. In addition, the calcium ion-sensitive fluorescent indicator Fluo-3/AM and double-labeling immunofluorescence stains showed that Piezo1 transferred mechanosignal to increase YAP nuclear translocation through calcium flux. Finally, transcriptomics revealed target genes of the Piezo1/YAP signaling pathway, such as TBX3, SESN2, SMAD7, FOSB, JARID2, and HAS2. Consequently, the Piezo1/calcium flux/YAP signaling axis contributes to the mechanically induced proadhesive signaling pathway, and thus, Piezo1 and YAP are promising targets for keloid treatment.
Collapse
Affiliation(s)
- Caihe Liao
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiru Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingyu Zeng
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guorong Yan
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiawen Gao
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jia Liu
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jia Yan
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guolong Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yeqiang Liu
- Department of Pathology at Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
2
|
Gallardo FS, Cruz-Soca M, Bock-Pereda A, Faundez-Contreras J, Gutiérrez-Rojas C, Gandin A, Torresan V, Casar JC, Ravasio A, Brandan E. Role of TGF-β/SMAD/YAP/TAZ signaling in skeletal muscle fibrosis. Am J Physiol Cell Physiol 2025; 328:C1015-C1028. [PMID: 39925133 DOI: 10.1152/ajpcell.00541.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/22/2024] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Skeletal muscle fibrosis is strongly associated with the differentiation of its resident multipotent fibro/adipogenic progenitors (FAPs) toward the myofibroblast phenotype. Although transforming growth factor type β (TGF-β) signaling is well-known for driving FAPs differentiation and fibrosis, due to its pleiotropic functions its complete inhibition is not suitable for treating fibrotic disorders such as muscular dystrophies. Here, we describe that TGF-β operates through the mechanosensitive transcriptional regulators Yes-associated protein (YAP)/ transcriptional coactivator with PDZ-binding motif (TAZ) to determine the myofibroblast fate of FAPs and skeletal muscle fibrosis. Spatial transcriptomics analyses of dystrophic and acute injured muscles showed that areas with active fibrosis and TGF-β signaling displayed high YAP/TAZ activity. Using a TGF-β-driven fibrotic mouse model, we found that activation of YAP/TAZ in activated FAPs is associated with the fibrotic process. Mechanistically, primary culture of FAPs reveals the remarkable ability of TGF-β1 to activate YAP/TAZ through its canonical SMAD3 pathway. Moreover, inhibition of YAP/TAZ, either by disrupting its activity (with Verteporfin) or cellular mechanotransduction (with the Rho inhibitor C3 or soft matrices), decreased TGF-β1-dependent FAPs differentiation into myofibroblasts. In vivo, administration of Verteporfin in mice limits the deposition of collagen and fibronectin, and the activation of FAPs during the development of fibrosis. Overall, our work provides robust evidence for considering YAP/TAZ as a potential target in muscular fibroproliferative disorders.NEW & NOTEWORTHY The understanding of the nuclear factors governing the differentiation of muscular fibro/adipogenic progenitors (FAPs) into myofibroblasts is in its infancy. Here, we comprehensively elucidate the status, regulation, and role of the mechanotransducers Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) in the muscular fibrotic process. Our findings reveal that inhibiting cellular mechanotransduction limits FAP differentiation and the extent of muscular fibrosis exerted by transforming growth factor type β (TGF-β). This research shed new lights on the molecular mechanisms dictating the cell fate of FAPs and the muscular fibrosis.
Collapse
Affiliation(s)
- Felipe S Gallardo
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Meilyn Cruz-Soca
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Alexia Bock-Pereda
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Jennifer Faundez-Contreras
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Cristian Gutiérrez-Rojas
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
- Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alessandro Gandin
- Department of Industrial Engineering, University of Padova and INSTM, Padova, Italy
| | - Veronica Torresan
- Department of Industrial Engineering, University of Padova and INSTM, Padova, Italy
| | - Juan Carlos Casar
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrea Ravasio
- Institute for Biological and Medical Engineering, School of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrique Brandan
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
3
|
Du H, Rose JP, Bons J, Guo L, Valentino TR, Wu F, Burton JB, Basisty N, Manwaring-Mueller M, Makhijani P, Chen N, Chang V, Winer S, Campisi J, Furman D, Nagy A, Schilling B, Winer DA. Substrate stiffness dictates unique doxorubicin-induced senescence-associated secretory phenotypes and transcriptomic signatures in human pulmonary fibroblasts. GeroScience 2025:10.1007/s11357-025-01507-x. [PMID: 39826027 DOI: 10.1007/s11357-025-01507-x] [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: 11/20/2024] [Accepted: 12/31/2024] [Indexed: 01/20/2025] Open
Abstract
Cells are subjected to dynamic mechanical environments which impart forces and induce cellular responses. In age-related conditions like pulmonary fibrosis, there is both an increase in tissue stiffness and an accumulation of senescent cells. While senescent cells produce a senescence-associated secretory phenotype (SASP), the impact of physical stimuli on both cellular senescence and the SASP is not well understood. Here, we show that mechanical tension, modeled using cell culture substrate rigidity, influences senescent cell markers like SA-β-gal and secretory phenotypes. Comparing human primary pulmonary fibroblasts (IMR-90) cultured on physiological (2 kPa), fibrotic (50 kPa), and plastic (approximately 3 GPa) substrates, followed by senescence induction using doxorubicin, we identified unique high-stiffness-driven secretory protein profiles using mass spectrometry and transcriptomic signatures, both showing an enrichment in collagen proteins. Consistently, clusters of p21 + cells are seen in fibrotic regions of bleomycin induced pulmonary fibrosis in mice. Computational meta-analysis of single-cell RNA sequencing datasets from human interstitial lung disease confirmed these stiffness SASP genes are highly expressed in disease fibroblasts and strongly correlate with mechanotransduction and senescence-related pathways. Thus, mechanical forces shape cell senescence and their secretory phenotypes.
Collapse
Affiliation(s)
- Huixun Du
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Jacob P Rose
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Joanna Bons
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Li Guo
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | | | - Fei Wu
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Jordan B Burton
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Nathan Basisty
- Longitudinal Studies Section, Translational Gerontology Branch, NIA, NIH, Baltimore, MA, USA
| | | | - Priya Makhijani
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Nan Chen
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Veronica Chang
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, , Canada
| | - Judith Campisi
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - David Furman
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
| | - Andras Nagy
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Birgit Schilling
- Buck Institute for Research On Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Daniel A Winer
- Buck Institute for Research On Aging, Novato, CA, 94945, USA.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
4
|
Du H, Rose JP, Bons J, Guo L, Valentino TR, Wu F, Burton JB, Basisty N, Manwaring-Mueller M, Makhijani P, Chen N, Chang V, Winer S, Campisi J, Furman D, Nagy A, Schilling B, Winer DA. Substrate Stiffness Dictates Unique Doxorubicin-induced Senescence-associated Secretory Phenotypes and Transcriptomic Signatures in Human Pulmonary Fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.623471. [PMID: 39605579 PMCID: PMC11601487 DOI: 10.1101/2024.11.18.623471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cells are subjected to dynamic mechanical environments which impart forces and induce cellular responses. In age-related conditions like pulmonary fibrosis, there is both an increase in tissue stiffness and an accumulation of senescent cells. While senescent cells produce a senescence-associated secretory phenotype (SASP), the impact of physical stimuli on both cellular senescence and the SASP is not well understood. Here, we show that mechanical tension, modeled using cell culture substrate rigidity, influences senescent cell markers like SA-β-gal and secretory phenotypes. Comparing human primary pulmonary fibroblasts (IMR-90) cultured on physiological (2 kPa), fibrotic (50 kPa), and plastic (approximately 3 GPa) substrates, followed by senescence induction using doxorubicin, we identified unique high-stiffness-driven secretory protein profiles using mass spectrometry and transcriptomic signatures, both showing an enrichment in collagen proteins. Consistently, clusters of p21+ cells are seen in fibrotic regions of bleomycin induced pulmonary fibrosis in mice. Computational meta-analysis of single-cell RNA sequencing datasets from human interstitial lung disease confirmed these stiffness SASP genes are highly expressed in disease fibroblasts and strongly correlate with mechanotransduction and senescence-related pathways. Thus, mechanical forces shape cell senescence and their secretory phenotypes.
Collapse
Affiliation(s)
- Huixun Du
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jacob P Rose
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Li Guo
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | | | - Fei Wu
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Nathan Basisty
- Longitudinal Studies Section, Translational Gerontology Branch, NIA, NIH, Baltimore, Maryland, USA
| | | | | | - Nan Chen
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| | - Veronica Chang
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, CA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Andras Nagy
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Birgit Schilling
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Daniel A Winer
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
5
|
Davan-Wetton CSA, Montero-Melendez T. An optimised protocol for the detection of lipofuscin, a versatile and quantifiable marker of cellular senescence. PLoS One 2024; 19:e0306275. [PMID: 39008441 PMCID: PMC11249248 DOI: 10.1371/journal.pone.0306275] [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: 05/14/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Lipofuscin is a yellow-brown pigment typically found in the lysosomes that contains a mixture of molecules including lipids, metals and misfolded proteins. The use of Sudan black B to detect lipofuscin accumulation, a well described marker of cellular senescence and ageing, was first described in 2013 by Georgakopoulou, et al. Here, we provide an optimisation of the original protocol. Firstly, we adjusted the staining methodology for increased ease of use on cultured cells. Secondly, we show that Sudan black B-stained lipofuscin emits strong fluorescence in the far-red channel making it suitable for fluorescence microscopy detection and quantification. Moreover, we also demonstrate that this optimised protocol can be utilised in conjunction with standard immunofluorescence staining techniques, making possible the simultaneous detection of lipofuscin and other cellular proteins of interest, like additional markers of senescence. This is a significant advantage over the most commonly used method for senescence detection, based on beta galactosidase enzymatic activity. We therefore believe that these findings and the provided optimised protocol will represent a useful tool for the scientific community in the field of cellular senescence.
Collapse
Affiliation(s)
- Camilla S. A. Davan-Wetton
- The William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Trinidad Montero-Melendez
- The William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
6
|
Gao J, Pi C, Pan J, Zhou W. Research progress on Hippo signaling pathway effector molecules in rheumatic immune system diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:376-381. [PMID: 38899353 PMCID: PMC11348685 DOI: 10.3724/zdxbyxb-2023-0567] [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: 12/04/2023] [Accepted: 04/10/2024] [Indexed: 06/21/2024]
Abstract
The core components of the Hippo signaling pathway encompass upstream regulatory molecules, core kinase cascade complexes, and downstream transcriptional regulation complexes. This pathway modulates cellular behaviors by influencing the effector molecules of its core components and plays a pivotal role in immune regulation. Effector molecules,such as Yes-associated protein (YAP), transcriptional coactivator with PDZ-binding motif (TAZ), transcriptional enhanced associate domain transcriptional factor (TEAD), monopolar spindle-one binder (MOB1), large tumor suppressor (LATS), can stimulate fibroblast-like synovial cell migration and invasion in rheumatoid arthritis, regulate osteoarthritis disease progression, promote pathological new bone formation in ankylosing spondylitis, sustain submandibular gland development while delaying Sjogren's syndrome progression, mediate alpha-smooth muscle actin in systemic sclerosis, and refine the regulation of target genes associated with pulmonary fibrosis. This article provides an overview of the regulatory mechanisms involving Hippo signaling pathway-related effector molecules in the pathogenesis and progression of rheumatic immune system diseases, to serve as a reference for exploring novel therapeutic targets of rheumatic immune system diseases.
Collapse
Affiliation(s)
- Jie Gao
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou 225003, Jiangsu Province, China.
- Graduate School of Dalian Medical University, Dalian 116044, Liaoning Province, China.
| | - Caihong Pi
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou 225003, Jiangsu Province, China
- Graduate School of Dalian Medical University, Dalian 116044, Liaoning Province, China
| | - Junmei Pan
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou 225003, Jiangsu Province, China
| | - Wei Zhou
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou 225003, Jiangsu Province, China.
- Graduate School of Dalian Medical University, Dalian 116044, Liaoning Province, China.
| |
Collapse
|
7
|
Chen X, Wu Y, Jia S, Zhao M. Fibroblast: A Novel Target for Autoimmune and Inflammatory Skin Diseases Therapeutics. Clin Rev Allergy Immunol 2024; 66:274-293. [PMID: 38940997 DOI: 10.1007/s12016-024-08997-1] [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] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Fibroblasts are crucial components of the skin structure. They were traditionally believed to maintain the skin's structure by producing extracellular matrix and other elements. Recent research illuminated that fibroblasts can respond to external stimuli and exhibit diverse functions, such as the secretion of pro-inflammatory factors, adipogenesis, and antigen presentation, exhibiting remarkable heterogeneity and plasticity. This revelation positions fibroblasts as active contributors to the pathogenesis of skin diseases, challenging the traditional perspective that views fibroblasts solely as structural entities. Based on their diverse functions, fibroblasts can be categorized into six subtypes: pro-inflammatory fibroblasts, myofibroblasts, adipogenic fibroblasts, angiogenic fibroblasts, mesenchymal fibroblasts, and antigen-presenting fibroblasts. Cytokines, metabolism, and epigenetics regulate functional abnormalities in fibroblasts. The dynamic changes fibroblasts exhibit in different diseases and disease states warrant a comprehensive discussion. We focus on dermal fibroblasts' aberrant manifestations and pivotal roles in inflammatory and autoimmune skin diseases, including psoriasis, vitiligo, lupus erythematosus, scleroderma, and atopic dermatitis, and propose targeting aberrantly activated fibroblasts as a potential therapeutic strategy for inflammatory and autoimmune skin diseases.
Collapse
Affiliation(s)
- Xiaoyun Chen
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yutong Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Sujie Jia
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
8
|
Rogovaya OS, Abolin DS, Cherkashina OL, Smyslov AD, Vorotelyak EA, Kalabusheva EP. In vitro and in vivo Evaluation of Antifibrotic Properties of Verteporfin in a Composition of a Collagen Scaffold. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:942-957. [PMID: 38880654 DOI: 10.1134/s0006297924050146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/05/2024] [Accepted: 03/31/2024] [Indexed: 06/18/2024]
Abstract
Extensive skin damage requires specialized therapy that stimulates regeneration processes without scarring. The possibility of using combination of a collagen gel application as a wound dressing and fibroblast attractant with verteporfin as an antifibrotic agent was examined in vivo and in vitro. In vitro effects of verteporfin on viability and myofibroblast markers expression were evaluated using fibroblasts isolated from human scar tissue. In vivo the collagen gel and verteporfin (individually and in combination) were applied into the wound to investigate scarring during skin regeneration: deviations in skin layer thickness, collagen synthesis, and extracellular matrix fibers were characterized. The results indicate that verteporfin reduces fibrotic phenotype by suppressing expression of the contractile protein Sm22α without inducing cell death. However, administration of verteporfin in combination with the collagen gel disrupts its ability to direct wound healing in a scarless manner, which may be related to incompatibility of the mechanisms by which collagen and verteporfin control regeneration.
Collapse
Affiliation(s)
- Olga S Rogovaya
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Danila S Abolin
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Olga L Cherkashina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Artem D Smyslov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Ekaterina A Vorotelyak
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Ekaterina P Kalabusheva
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
9
|
Naik A, Chitturi P, Nguyen J, Leask A. The yes-associated protein-1 (YAP1) inhibitor celastrol suppresses the ability of transforming growth factor β to activate human gingival fibroblasts. Arch Oral Biol 2024; 160:105910. [PMID: 38364717 DOI: 10.1016/j.archoralbio.2024.105910] [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: 07/24/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024]
Abstract
OBJECTIVE To determine whether celastrol, an inhibitor of the mechanosensitive transcriptional cofactor yes-associated protein-1 (YAP1), impairs the ability of TGFβ1 to stimulate fibrogenic activity in human gingival fibroblast cell line. DESIGN Human gingival fibroblasts were pre-treated with celastrol or DMSO followed by stimulation with or without TGFβ1 (4 ng/ml). We then utilized bulk RNA sequencing (RNAseq), real-time polymerase chain reaction (RT-PCR), Western blot, immunofluorescence, cell proliferation assays to determine if celastrol impaired TGFβ1-induced responses in a human gingival fibroblast cell line. RESULTS Celastrol impaired the ability of TGFβ1 to induce expression of the profibrotic marker and mediator CCN2. Bulk RNAseq analysis of gingival fibroblasts treated with TGFβ1, in the presence or absence of celastrol, revealed that celastrol impaired the ability of TGFβ1 to induce mRNA expression of genes within extracellular matrix, wound healing, focal adhesion and cytokine/Wnt signaling clusters. RT-PCR analysis of extracted RNAs confirmed that celastrol antagonized the ability of TGFβ1 to induce expression of genes anticipated to contribute to fibrotic responses. Celastrol also reduced gingival fibroblast proliferation, and YAP1 nuclear localization in response to TGFβ1. CONCLUSION YAP1 inhibitors such as celastrol could be used to impair pro-fibrotic responses to TGFβ1 in human gingival fibroblasts.
Collapse
Affiliation(s)
- Angha Naik
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - John Nguyen
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
10
|
Burgess JK, Weiss DJ, Westergren-Thorsson G, Wigen J, Dean CH, Mumby S, Bush A, Adcock IM. Extracellular Matrix as a Driver of Chronic Lung Diseases. Am J Respir Cell Mol Biol 2024; 70:239-246. [PMID: 38190723 DOI: 10.1165/rcmb.2023-0176ps] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
The extracellular matrix (ECM) is not just a three-dimensional scaffold that provides stable support for all cells in the lungs, but also an important component of chronic fibrotic airway, vascular, and interstitial diseases. It is a bioactive entity that is dynamically modulated during tissue homeostasis and disease, that controls structural and immune cell functions and drug responses, and that can release fragments that have biological activity and that can be used to monitor disease activity. There is a growing recognition of the importance of considering ECM changes in chronic airway, vascular, and interstitial diseases, including 1) compositional changes, 2) structural and organizational changes, and 3) mechanical changes and how these affect disease pathogenesis. As altered ECM biology is an important component of many lung diseases, disease models must incorporate this factor to fully recapitulate disease-driver pathways and to study potential novel therapeutic interventions. Although novel models are evolving that capture some or all of the elements of the altered ECM microenvironment in lung diseases, opportunities exist to more fully understand cell-ECM interactions that will help devise future therapeutic targets to restore function in chronic lung diseases. In this perspective article, we review evolving knowledge about the ECM's role in homeostasis and disease in the lung.
Collapse
Affiliation(s)
- Janette K Burgess
- Department of Pathology and Medical Biology
- Groningen Research Institute for Asthma and COPD, and
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont
| | | | - Jenny Wigen
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Andrew Bush
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
- Centre for Pediatrics and Child Health, Imperial College and Royal Brompton Hospital, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| |
Collapse
|
11
|
Leask A, Fadl A, Naik A. A modest proposal: targeting αv integrin-mediated activation of latent TGFbeta as a novel therapeutic approach to treat scleroderma fibrosis. Expert Opin Investig Drugs 2024; 33:279-285. [PMID: 38393748 DOI: 10.1080/13543784.2024.2323528] [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: 11/04/2023] [Accepted: 02/22/2024] [Indexed: 02/25/2024]
Abstract
INTRODUCTION The potent profibrotic cytokine transforming growth factor-β (TGF-β) has been associated with the onset and progression of the fibrosis seen in the autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc). AREA COVERED This review explores the data supporting the notion that TGF-β contributes to SSc fibrosis and examines why initiating clinical trials in SSc aimed at targeting integrin-mediated latent TGF-β activation is timely. EXPERT OPINION Targeting TGF-β directly has not been proven to be clinically effective in this disease. Conversely, targeting matrix stiffness, which perpetuates fibrosis, may have more promise. Intriguingly, targeting integrin-mediated activation of latent TGF-β, which bridges these concepts, may have therapeutic value.
Collapse
Affiliation(s)
- Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Asmaa Fadl
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Angha Naik
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
12
|
van Straalen KR, Ma F, Tsou PS, Plazyo O, Gharaee-Kermani M, Calbet M, Xing X, Sarkar MK, Uppala R, Harms PW, Wasikowski R, Nahlawi L, Nakamura M, Eshaq M, Wang C, Dobry C, Kozlow JH, Cherry-Bukowiec J, Brodie WD, Wolk K, Uluçkan Ö, Mattichak MN, Pellegrini M, Modlin RL, Maverakis E, Sabat R, Kahlenberg JM, Billi AC, Tsoi LC, Gudjonsson JE. Single-cell sequencing reveals Hippo signaling as a driver of fibrosis in hidradenitis suppurativa. J Clin Invest 2024; 134:e169225. [PMID: 38051587 PMCID: PMC10836805 DOI: 10.1172/jci169225] [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: 01/31/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic inflammatory disease characterized by abscesses, nodules, dissecting/draining tunnels, and extensive fibrosis. Here, we integrate single-cell RNA sequencing, spatial transcriptomics, and immunostaining to provide an unprecedented view of the pathogenesis of chronic HS, characterizing the main cellular players and defining their interactions. We found a striking layering of the chronic HS infiltrate and identified the contribution of 2 fibroblast subtypes (SFRP4+ and CXCL13+) in orchestrating this compartmentalized immune response. We further demonstrated the central role of the Hippo pathway in promoting extensive fibrosis in HS and provided preclinical evidence that the profibrotic fibroblast response in HS can be modulated through inhibition of this pathway. These data provide insights into key aspects of HS pathogenesis with broad therapeutic implications.
Collapse
Affiliation(s)
| | - Feiyang Ma
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Mehrnaz Gharaee-Kermani
- Department of Dermatology and
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Marta Calbet
- Almirall SA, R&D Center, Sant Feliu de Llobregat, Barcelona, Spain
| | | | | | | | - Paul W. Harms
- Department of Dermatology and
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | | | - Cong Wang
- Laboratory for Experimental Immunodermatology, Department of Dermatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | | | - Jill Cherry-Bukowiec
- Section of General Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - William D. Brodie
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kerstin Wolk
- Interdisciplinary group Molecular Immunopathology, Dermatology/Medical Immunology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Özge Uluçkan
- Almirall SA, R&D Center, Sant Feliu de Llobregat, Barcelona, Spain
| | - Megan N. Mattichak
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Emanual Maverakis
- Department of Dermatology, University of California, Sacramento, California, USA
| | - Robert Sabat
- Interdisciplinary group Molecular Immunopathology, Dermatology/Medical Immunology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - J. Michelle Kahlenberg
- Department of Dermatology and
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
13
|
Yang JT, Wu D, Li J, Zhao C, Zhu L, Xu C, Xu N. An Injectable Composite Hydrogel of Verteporfin-Bonded Carboxymethyl Chitosan and Oxidized Sodium Alginate Facilitates Scarless Full-Thickness Skin Regeneration. Macromol Biosci 2024; 24:e2300165. [PMID: 37681479 DOI: 10.1002/mabi.202300165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/22/2023] [Indexed: 09/09/2023]
Abstract
Full-thickness skin defect has always been a major challenge in clinics due to fibrous hyperplasia in the repair process. Hydrogel composite dressings loaded with anti-fibrotic drugs have been considered as a promising strategy for scarless skin regeneration. In this work, a hydrogel composite (VP-CMCS-OSA) of carboxymethyl chitosan (CMCS) and oxidized sodium alginate (OSA), with loading anti-fibrotic drug verteporfin (VP), is developed based on two-step chemical reactions. Verteporfin is bonded with carboxymethyl chitosan through EDC/NHS treatment to form VP-CMCS, and then VP-CMCS is crosslinked with oxidized sodium alginate by Schiff base reaction to form VP-CMCS-OSA hydrogel. The characterization by SEM, FTIR, and UV-Vis shows the microstructure and chemical bonding of VP-CMCS-OSA. VP-CMCS-OSA hydrogel demonstrates the properties of high tissue adhesion, strong self-healing, and tensile ability. In the full-thickness skin defect model, the VP-CMCS-OSA composite hydrogels hasten wound healing due to the synergistic effects of hydrogels and verteporfin administration. The histological examination reveals the regular collagen arrangement and more skin appendages after VP-CMCS-OSA composite hydrogel treatment, indicating the full-thickness skin regeneration without potential scar formation. The outcomes suggest that the verteporfin-loaded composite hydrogel could be a potential method for scarless skin regeneration.
Collapse
Affiliation(s)
- Jiang-Tao Yang
- College of Life Sciences and Health, Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Dingwei Wu
- College of Life Sciences and Health, Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Jianping Li
- The State Key Laboratory of Refractories and Metallurgy, Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Chenchen Zhao
- College of Life Sciences and Health, Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Lian Zhu
- College of Life Sciences and Health, Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Chengchen Xu
- College of Life Sciences and Health, Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Na Xu
- College of Life Sciences and Health, Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| |
Collapse
|
14
|
Ma F, Tsou PS, Gharaee-Kermani M, Plazyo O, Xing X, Kirma J, Wasikowski R, Hile GA, Harms PW, Jiang Y, Xing E, Nakamura M, Ochocki D, Brodie WD, Pillai S, Maverakis E, Pellegrini M, Modlin RL, Varga J, Tsoi LC, Lafyatis R, Kahlenberg JM, Billi AC, Khanna D, Gudjonsson JE. Systems-based identification of the Hippo pathway for promoting fibrotic mesenchymal differentiation in systemic sclerosis. Nat Commun 2024; 15:210. [PMID: 38172207 PMCID: PMC10764940 DOI: 10.1038/s41467-023-44645-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Systemic sclerosis (SSc) is a devastating autoimmune disease characterized by excessive production and accumulation of extracellular matrix, leading to fibrosis of skin and other internal organs. However, the main cellular participants in SSc skin fibrosis remain incompletely understood. Here using differentiation trajectories at a single cell level, we demonstrate a dual source of extracellular matrix deposition in SSc skin from both myofibroblasts and endothelial-to-mesenchymal-transitioning cells (EndoMT). We further define a central role of Hippo pathway effectors in differentiation and homeostasis of myofibroblast and EndoMT, respectively, and show that myofibroblasts and EndoMTs function as central communication hubs that drive key pro-fibrotic signaling pathways in SSc. Together, our data help characterize myofibroblast differentiation and EndoMT phenotypes in SSc skin, and hint that modulation of the Hippo pathway may contribute in reversing the pro-fibrotic phenotypes in myofibroblasts and EndoMTs.
Collapse
Affiliation(s)
- Feiyang Ma
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pei-Suen Tsou
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - Mehrnaz Gharaee-Kermani
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Olesya Plazyo
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xianying Xing
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Joseph Kirma
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rachael Wasikowski
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Grace A Hile
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Paul W Harms
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yanyun Jiang
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Enze Xing
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mio Nakamura
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Danielle Ochocki
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - William D Brodie
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shiv Pillai
- Ragon Institute, Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Boston, MA, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Matteo Pellegrini
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Robert L Modlin
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - John Varga
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Robert Lafyatis
- Division of Rheumatology, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Michelle Kahlenberg
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Allison C Billi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Dinesh Khanna
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA.
| | | |
Collapse
|
15
|
Leask A, Naik A, Stratton RJ. Back to the future: targeting the extracellular matrix to treat systemic sclerosis. Nat Rev Rheumatol 2023; 19:713-723. [PMID: 37789119 DOI: 10.1038/s41584-023-01032-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
Fibrosis is the excessive deposition of a stable extracellular matrix (ECM); fibrotic tissue is composed principally of highly crosslinked type I collagen and highly contractile myofibroblasts. Systemic sclerosis (SSc) is a multisystem autoimmune connective tissue disease characterized by skin and organ fibrosis. The fibrotic process has been recognized in SSc for >40 years, but drugs with demonstrable efficacy against SSc fibrosis in ameliorating the lung involvement have only recently been identified. Unfortunately, these treatments are ineffective at improving the skin score in patients with SSc. Previous clinical trials in SSc have largely focused on the cross-purposing of anti-inflammatory drugs and the use of immunosuppressive drugs from the transplantation field, which address inflammatory and/or autoimmune processes. Limited examination has taken place of specific anti-fibrotic agents developed through their ability to directly target the ECM in SSc by, for example, alleviating the persistent matrix stiffness and mechanotransduction that might be required for both the initiation and maintenance of fibrosis, including in SSc. However, because of the importance of the ECM in the SSc phenotype, attempts have now been made to identify drugs that specifically target the ECM, including some drugs that are currently under consideration for the treatment of cancer.
Collapse
Affiliation(s)
- Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Angha Naik
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Richard J Stratton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Division of Medicine, London, UK
| |
Collapse
|
16
|
Chitturi P, Xu S, Ahmed Abdi B, Nguyen J, Carter DE, Sinha S, Arora R, Biernaskie J, Stratton RJ, Leask A. Tripterygium wilfordii derivative celastrol, a YAP inhibitor, has antifibrotic effects in systemic sclerosis. Ann Rheum Dis 2023; 82:1191-1204. [PMID: 37328193 DOI: 10.1136/ard-2023-223859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 06/05/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVES Systemic sclerosis (SSc) is characterised by extensive tissue fibrosis maintained by mechanotranductive/proadhesive signalling. Drugs targeting this pathway are therefore of likely therapeutic benefit. The mechanosensitive transcriptional co-activator, yes activated protein-1 (YAP1), is activated in SSc fibroblasts. The terpenoid celastrol is a YAP1 inhibitor; however, if celastrol can alleviate SSc fibrosis is unknown. Moreover, the cell niches required for skin fibrosis are unknown. METHODS Human dermal fibroblasts from healthy individuals and patients with diffuse cutaneous SSc were treated with or without transforming growth factor β1 (TGFβ1), with or without celastrol. Mice were subjected to the bleomycin-induced model of skin SSc, in the presence or absence of celastrol. Fibrosis was assessed using RNA Sequencing, real-time PCR, spatial transcriptomic analyses, Western blot, ELISA and histological analyses. RESULTS In dermal fibroblasts, celastrol impaired the ability of TGFβ1 to induce an SSc-like pattern of gene expression, including that of cellular communication network factor 2, collagen I and TGFβ1. Celastrol alleviated the persistent fibrotic phenotype of dermal fibroblasts cultured from lesions of SSc patients. In the bleomycin-induced model of skin SSc, increased expression of genes associated with reticular fibroblast and hippo/YAP clusters was observed; conversely, celastrol inhibited these bleomycin-induced changes and blocked nuclear localisation of YAP. CONCLUSIONS Our data clarify niches within the skin activated in fibrosis and suggest that compounds, such as celastrol, that antagonise the YAP pathway may be potential treatments for SSc skin fibrosis.
Collapse
Affiliation(s)
- Pratyusha Chitturi
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Shiwen Xu
- Centre for Rheumatology and Connective Tissue Diseases, UCL Medical School, Royal Free Campus, London, UK
| | - Bahja Ahmed Abdi
- Centre for Rheumatology and Connective Tissue Diseases, UCL Medical School, Royal Free Campus, London, UK
| | - John Nguyen
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | - Sartak Sinha
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rohit Arora
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Richard J Stratton
- Centre for Rheumatology and Connective Tissue Diseases, UCL Medical School, Royal Free Campus, London, UK
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
17
|
Zhu L, Liu L, Wang A, Liu J, Huang X, Zan T. Positive feedback loops between fibroblasts and the mechanical environment contribute to dermal fibrosis. Matrix Biol 2023; 121:1-21. [PMID: 37164179 DOI: 10.1016/j.matbio.2023.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/06/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Dermal fibrosis is characterized by excessive deposition of extracellular matrix in the dermis and affects millions of people worldwide and causes limited movement, disfigurement and psychological distress in patients. Fibroblast dysfunction of plays a central role in the pathogenesis of dermal fibrosis and is controlled by distinct factors. Recent studies support the hypothesis that fibroblasts can drive matrix deposition and stiffening, which in turn can exacerbate the functional dysregulation of fibroblasts. Ultimately, through a positive feedback loop, uncontrolled pathological fibrosis develops. This review aims to summarize the phenomenon and mechanism of the positive feedback loop in dermal fibrosis, and discuss potential therapeutic targets to help further elucidate the pathogenesis of dermal fibrosis and develop therapeutic strategies. In this review, fibroblast-derived compositional and structural changes in the ECM that lead to altered mechanical properties are briefly discussed. We focus on the mechanisms by which mechanical cues participate in dermal fibrosis progression. The mechanosensors discussed in the review include integrins, DDRs, proteoglycans, and mechanosensitive ion channels. The FAK, ERK, Akt, and Rho pathways, as well as transcription factors, including MRTF and YAP/TAZ, are also discussed. In addition, we describe stiffness-induced biological changes in the ECM on fibroblasts that contribute to the formation of a positive feedback loop. Finally, we discuss therapeutic strategies to treat the vicious cycle and present important suggestions for researchers conducting in-depth research.
Collapse
Affiliation(s)
- Liang Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lechen Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Aoli Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jinwen Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Chen K, Liu Y, Liu X, Guo Y, Liu J, Ding J, Zhang Z, Ni X, Chen Y. Hyaluronic acid-modified and verteporfin-loaded polylactic acid nanogels promote scarless wound healing by accelerating wound re-epithelialization and controlling scar formation. J Nanobiotechnology 2023; 21:241. [PMID: 37496007 PMCID: PMC10369727 DOI: 10.1186/s12951-023-02014-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Wound healing is a common occurrence. However, delayed healing and aberrant scarring result in pathological wound healing. Accordingly, a scarless wound healing remains a significant clinical challenge. In this study, we constructed hyaluronic acid (HA)-modified and verteporfin (VP)-loaded polylactic acid (PLA) nanogels (HA/VP-PLA) to promote scarless wound healing by accelerating wound re-epithelialization and controlling scar formation. Owing to the unique structure of HA incorporating and coating in VP-loaded PLA nanoparticles, HA/VP-PLA could be topically applied on wound to achieve targeted delivery to fibroblasts. Then, HA/VP-PLA released HA and lactic acid (LA) to stimulate the proliferation and migration of fibroblasts, as well as VP to inhibit Yes-associated protein (YAP) expression and nuclear localization to suppress fibrosis. In vitro (skin fibroblasts) and in vivo (rat and rabbit models) experiments strongly suggested that HA/VP-PLA promoted scarless wound healing by accelerating wound re-epithelialization and controlling scar formation. Therefore, our work provides a feasible strategy for scarless wound healing, and the sophisticated HA/VP-PLA exhibit a great potential for clinical applications.
Collapse
Affiliation(s)
- Kun Chen
- Department of Burn and Plastic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yuanhu Liu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children' s Hospital, Capital Medical University, National Center for Children' s Health, Beijing, 100045, China
- Shunyi Maternal and Children's Hospital of Beijing Children's Hospital, Beijing, China
| | - Xiaohui Liu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children' s Hospital, Capital Medical University, National Center for Children' s Health, Beijing, 100045, China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jing Liu
- Department of Burn and Plastic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jiaojiao Ding
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children' s Hospital, Capital Medical University, National Center for Children' s Health, Beijing, 100045, China
| | - Zheng Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Xin Ni
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children' s Hospital, Capital Medical University, National Center for Children' s Health, Beijing, 100045, China.
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Yunsheng Chen
- Department of Burn, Shanghai Burn Institute, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
19
|
Golubev DA, Zemskaya NV, Gorbunova AA, Kukuman DV, Moskalev A, Shaposhnikov MV. Studying the Geroprotective Properties of YAP/TAZ Signaling Inhibitors on Drosophila melanogaster Model. Int J Mol Sci 2023; 24:ijms24066006. [PMID: 36983079 PMCID: PMC10058302 DOI: 10.3390/ijms24066006] [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: 12/01/2022] [Revised: 02/28/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
The transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are the main downstream effectors of the evolutionarily conserved Hippo signaling pathway. YAP/TAZ are implicated in the transcriptional regulation of target genes that are involved in a wide range of key biological processes affecting tissue homeostasis and play dual roles in the aging process, depending on the cellular and tissue context. The aim of the present study was to investigate whether pharmacological inhibitors of Yap/Taz increase the lifespan of Drosophila melanogaster. Real-time qRT-PCR was performed to measure the changes in the expression of Yki (Yorkie, the Drosophila homolog of YAP/TAZ) target genes. We have revealed a lifespan-increasing effect of YAP/TAZ inhibitors that was mostly associated with decreased expression levels of the wg and E2f1 genes. However, further analysis is required to understand the link between the YAP/TAZ pathway and aging.
Collapse
Affiliation(s)
- Denis A Golubev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Nadezhda V Zemskaya
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Anastasia A Gorbunova
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Daria V Kukuman
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Mikhail V Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| |
Collapse
|
20
|
Gallardo FS, Córdova-Casanova A, Bock-Pereda A, Rebolledo DL, Ravasio A, Casar JC, Brandan E. Denervation Drives YAP/TAZ Activation in Muscular Fibro/Adipogenic Progenitors. Int J Mol Sci 2023; 24:ijms24065585. [PMID: 36982659 PMCID: PMC10059792 DOI: 10.3390/ijms24065585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/17/2023] Open
Abstract
Loss of motoneuron innervation (denervation) is a hallmark of neurodegeneration and aging of the skeletal muscle. Denervation induces fibrosis, a response attributed to the activation and expansion of resident fibro/adipogenic progenitors (FAPs), i.e., multipotent stromal cells with myofibroblast potential. Using in vivo and in silico approaches, we revealed FAPs as a novel cell population that activates the transcriptional coregulators YAP/TAZ in response to skeletal muscle denervation. Here, we found that denervation induces the expression and transcriptional activity of YAP/TAZ in whole muscle lysates. Using the PdgfraH2B:EGFP/+ transgenic reporter mice to trace FAPs, we demonstrated that denervation leads to increased YAP expression that accumulates within FAPs nuclei. Consistently, re-analysis of published single-nucleus RNA sequencing (snRNA-seq) data indicates that FAPs from denervated muscles have a higher YAP/TAZ signature level than control FAPs. Thus, our work provides the foundations to address the functional role of YAP/TAZ in FAPs in a neurogenic pathological context, which could be applied to develop novel therapeutic approaches for the treatment of muscle disorders triggered by motoneuron degeneration.
Collapse
Affiliation(s)
- Felipe S. Gallardo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Adriana Córdova-Casanova
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Alexia Bock-Pereda
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Daniela L. Rebolledo
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| | - Andrea Ravasio
- Institute for Biological and Medical Engineering, School of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Juan Carlos Casar
- Departamento de Neurología, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510602, Chile
- Correspondence:
| |
Collapse
|
21
|
Jia X, He L, Yang Z. Recent advances in the role of Yes-associated protein in dermatosis. Skin Res Technol 2023; 29:e13285. [PMID: 36973973 PMCID: PMC10155855 DOI: 10.1111/srt.13285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 03/04/2023]
Abstract
BACKGROUND Dermatosis is a general term for diseases of the skin and skin appendages including scleroderma, psoriasis, bullous disease, atopic dermatitis, basal cell carcinoma, squamous cell carcinoma, and melanoma. These diseases affect millions of individuals globally and are a serious public health concern. However, the pathogenesis of skin diseases is not fully understood, and treatments are not optimal. Yes-associated protein (YAP) is a transcriptional coactivator that plays a role in the regulation of gene transcription and signal transduction. AIMS To study the role of Yes-associated protein in skin diseases. MATERIALS AND METHODS The present review summarizes recent advances in our understanding of the role of YAP in skin diseases, current treatments that target YAP, and potential avenues for novel therapies. RESULTS Abnormal YAP expression has been implicated in occurrence and development of dermatosis. YAP regulates the cell homeostasis, proliferation, differentiation, apoptosis, angiopoiesis, and epithelial-to-mesenchymal transition, among other processes. As well as, it serves as a potential target in many biological processes for treating dermatosis. CONCLUSIONS The effects of YAP on the skin are complex and require multidimensional investigational approaches. YAP functions as an oncoprotein that can promote the occurrence and development of cancer, but there is currently limited information on the therapeutic potential of YAP inhibition for cancer treatment. Additional studies are also needed to clarify the role of YAP in the development and maturation of dermal fibroblasts; skin barrier function, homeostasis, aging, and melanin production; and dermatosis.
Collapse
Affiliation(s)
- Xiaorong Jia
- Department of DermatologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Li He
- Department of DermatologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Zhi Yang
- Department of DermatologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| |
Collapse
|
22
|
Gharbia FZ, Abouhashem AS, Moqidem YA, Elbaz AA, Abdellatif A, Singh K, Sen CK, Azzazy HME. Adult skin fibroblast state change in murine wound healing. Sci Rep 2023; 13:886. [PMID: 36650180 PMCID: PMC9845335 DOI: 10.1038/s41598-022-27152-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023] Open
Abstract
Wound healing is a well-organized dynamic process involving coordinated consecutive phases: homeostasis, inflammation, proliferation and resolution. Fibroblasts play major roles in skin wound healing such as in wound contraction and release of growth factors which are of importance in angiogenesis and tissue remodeling. Abnormal fibroblast phenotypes have been identified in patients with chronic wounds. In this work, we analyzed scRNA-seq datasets of normal and wounded skin from mice at day 4 post-wound to investigate fibroblast heterogeneity during the proliferative phase of wound healing. Compositional analysis revealed a specific subset of fibroblast (cluster 3) that primarily increased in wounded skin (14%) compared to normal skin (3.9%). This subset was characterized by a gene signature marked by the plasma membrane proteins Sfrp2 + Sfrp4 + Sfrp1 + and the transcription factors Ebf1 + Prrx1 + Maged1 + . Differential gene expression and enrichment analysis identified epithelial to mesenchymal transition (EMT) and angiogenesis to be upregulated in the emerging subset of fibroblasts of the wounded skin. Using two other datasets for murine wounded skin confirmed the increase in cluster 3-like fibroblasts at days 2, 7 and 14 post-wounding with a peak at day 7. By performing a similarity check between the differential gene expression profile between wounded and normal skin for this emerging fibroblast subset with drug signature from the ConnectivityMap database, we identified drugs capable of mimicking the observed gene expression change in fibroblasts during wound healing. TTNPB, verteprofin and nicotinic acid were identified as candidate drugs capable of inducing fibroblast gene expression profile necessary for wound healing. On the other hand, methocarbamol, ifosfamide and penbutolol were recognized to antagonize the identified fibroblast differential expression profile during wound healing which might cause delay in wound healing. Taken together, analysis of murine transcriptomic skin wound healing datasets suggested a subset of fibroblasts capable of inducing EMT and further inferred drugs that might be tested as potential candidates to induce wound closure.
Collapse
Affiliation(s)
- Fatma Z Gharbia
- Graduate Nanotechnology Program, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Ahmed S Abouhashem
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- Sharkia Clinical Research Department, Ministry of Health & Population, Zagazig, 44511, Sharkia, Egypt
- CytoTalk LLC, Cheyenne, WY, 82001, USA
| | - Yomna A Moqidem
- Department of Biology, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
| | - Ahmed A Elbaz
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- CytoTalk LLC, Cheyenne, WY, 82001, USA
| | - Ahmed Abdellatif
- Department of Biology, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Hassan M E Azzazy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt.
- Department of Nanobiophotonics, Leibniz Institute for Photonic Technology, Albert Einstein Str. 9, 07745, Jena, Germany.
| |
Collapse
|
23
|
Peidl A, Nguyen J, Chitturi P, Riser BL, Leask A. Using the Bleomycin-Induced Model of Fibrosis to Study the Contribution of CCN Proteins to Scleroderma Fibrosis. Methods Mol Biol 2023; 2582:309-321. [PMID: 36370359 DOI: 10.1007/978-1-0716-2744-0_21] [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] [Indexed: 06/16/2023]
Abstract
Approximately 45% of the deaths in the developed world result from conditions with a fibrotic component. Although no specific, focused anti-fibrotic therapies have been approved for clinical use, a long-standing concept is that targeting CCN proteins may be useful to treat fibrosis. Herein, we summarize current data supporting the concept that targeting CCN2 may be a viable anti-fibrotic approach to treat scleroderma. Testing this hypothesis has been made possible by using a mouse model of inflammation-driven skin and lung fibrosis.
Collapse
Affiliation(s)
- Alexander Peidl
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - John Nguyen
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Bruce L Riser
- BLR Bio LLC, Kenosha, WI, USA
- Center for Cancer Cell Biology, Immunology and Infection, Department of Physiology and Biophysics, and Department of Medicine Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
24
|
Puerta Cavanzo N, Riesmeijer SA, Holt-Kedde IL, Werker PMN, Piersma B, Olinga P, Bank RA. Verteporfin ameliorates fibrotic aspects of Dupuytren's disease nodular fibroblasts irrespective the activation state of the cells. Sci Rep 2022; 12:13940. [PMID: 35977978 PMCID: PMC9386017 DOI: 10.1038/s41598-022-18116-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 08/05/2022] [Indexed: 11/28/2022] Open
Abstract
Dupuytren’s disease is a chronic, progressive fibroproliferative condition of the hand fascia which results in digital contraction. So far, treatments do not directly interfere with the (myo)fibroblasts that are responsible for the formation of the collagen-rich cords and its contraction. Here we investigated whether verteporfin (VP) is able to inhibit the activation and subsequent differentiation of DD nodular fibroblasts into myofibroblasts. Fibroblasts were isolated from nodules of 7 Dupuytren patients. Cells are treated (1) for 48 h with 5 ng/ml transforming growth factor β1 (TGF-β1) followed by 48 h with/without 250 nM VP in the absence of TGF-β1, or treated (2) for 48 h with TGF-β1 followed by 48 h with/without VP in the presence of TGF-β1. mRNA levels were measured by means of Real-Time PCR, and proteins were visualized by means of Western blotting and/or immunofluorescence. Quantitative data were statistically analyzed with GraphPad Prism using the paired t-test. We found that fibroblasts activated for 48 h with TGF-β1 show a decrease in mRNA levels of COL1A1, COL3A1, COL4A1, PLOD2, FN1EDA, CCN2 and SERPINE1 when exposed for another 48 h with VP, whereas no decrease is seen for ACTA2, YAP1, SMAD2 and SMAD3 mRNA levels. Cells exposed for an additional 48 h with TGF-β1, but now in the presence of VP, are not further activated anymore, whereas in the absence of VP the cells continue to differentiate into myofibroblasts. Collagen type I, fibronectin-extra domain A, α-smooth muscle actin, YAP1, Smad2 and Smad3 protein levels were attenuated by both VP treatments. We conclude that VP has strong anti-fibrotic properties: it is able to halt the differentiation of fibroblasts into myofibroblasts, and is also able to reverse the activation status of fibroblasts. The decreased protein levels of YAP1, Smad2 and Smad3 in the presence of VP explain in part the strong anti-fibrotic properties of VP. Verteporfin is clinically used as a photosensitizer for photodynamic therapy to eliminate abnormal blood vessels in the eye to attenuate macular degeneration. The antifibrotic properties of VP do not rely on photo-activation, as we used the molecule in its non-photoinduced state.
Collapse
Affiliation(s)
- Nataly Puerta Cavanzo
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,MATRIX Research Group, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Sophie A Riesmeijer
- Department of Plastic Surgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Iris L Holt-Kedde
- Department of Plastic Surgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Paul M N Werker
- Department of Plastic Surgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Bram Piersma
- MATRIX Research Group, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Ruud A Bank
- MATRIX Research Group, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
25
|
Martínez-Nieto GA, Teppo HR, Petrelius N, Izzi V, Devarajan R, Petäistö T, Liu H, Kim KS, Karppinen SM, Ruotsalainen H, Koivunen J, Mäki JM, Walker GC, Pihlajaniemi T, Gullberg D, Heljasvaara R. Upregulated integrin α11 in the stroma of cutaneous squamous cell carcinoma promotes skin carcinogenesis. Front Oncol 2022; 12:981009. [PMID: 36003785 PMCID: PMC9393502 DOI: 10.3389/fonc.2022.981009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Integrin α11β1 is a collagen-binding integrin that is needed to induce and maintain the myofibroblast phenotype in fibrotic tissues and during wound healing. The expression of the α11 is upregulated in cancer-associated fibroblasts (CAFs) in various human neoplasms. We investigated α11 expression in human cutaneous squamous cell carcinoma (cSCC) and in benign and premalignant human skin lesions and monitored its effects on cSCC development by subjecting α11-knockout (Itga11−/−) mice to the DMBA/TPA skin carcinogenesis protocol. α11-deficient mice showed significantly decreased tumor cell proliferation, leading to delayed tumor development and reduced tumor burden. Integrin α11 expression was significantly upregulated in the desmoplastic tumor stroma of human and mouse cSCCs, and the highest α11 expression was detected in high-grade tumors. Our results point to a reduced ability of α11-deficient stromal cells to differentiate into matrix-producing and tumor-promoting CAFs and suggest that this is one causative mechanism underlying the observed decreased tumor growth. An unexpected finding in our study was that, despite reduced CAF activation, the α11-deficient skin tumors were characterized by the presence of thick and regularly aligned collagen bundles. This finding was attributed to a higher expression of TGFβ1 and collagen crosslinking lysyl oxidases in the Itga11-/- tumor stroma. In summary, our data suggest that α11β1 operates in a complex interactive tumor environment to regulate ECM synthesis and collagen organization and thus foster cSCC growth. Further studies with advanced experimental models are still needed to define the exact roles and molecular mechanisms of stromal α11β1 in skin tumorigenesis.
Collapse
Affiliation(s)
- Guillermo A. Martínez-Nieto
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hanna-Riikka Teppo
- Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Pathology, Oulu University Hospital, Oulu, Finland
| | - Noora Petrelius
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Valerio Izzi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
- Finnish Cancer Institute, Helsinki, Finland
| | - Raman Devarajan
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Tiina Petäistö
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hengshuo Liu
- Matrix Biology Group, Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Kris S. Kim
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Sanna-Maria Karppinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Heli Ruotsalainen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Jarkko Koivunen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Joni M. Mäki
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | | | - Taina Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Donald Gullberg
- Matrix Biology Group, Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Matrix Biology Group, Department of Biomedicine, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- *Correspondence: Ritva Heljasvaara,
| |
Collapse
|
26
|
Canadian Contributions in Fibroblast Biology. Cells 2022; 11:cells11152272. [PMID: 35892569 PMCID: PMC9331635 DOI: 10.3390/cells11152272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 02/04/2023] Open
Abstract
Fibroblasts are stromal cells found in virtually every tissue and organ of the body. For many years, these cells were often considered to be secondary in functional importance to parenchymal cells. Over the past 2 decades, focused research into the roles of fibroblasts has revealed important roles for these cells in the homeostasis of healthy tissue, and has demonstrated that activation of fibroblasts to myofibroblasts is a key step in disease initiation and progression in many tissues, with fibrosis now recognized as not only an outcome of disease, but also a central contributor to tissue dysfunction, particularly in the heart and lungs. With a growing understanding of both fibroblast and myofibroblast heterogeneity, and the deciphering of the humoral and mechanical cues that impact the phenotype of these cells, fibroblast biology is rapidly becoming a major focus in biomedical research. In this review, we provide an overview of fibroblast and myofibroblast biology, particularly in the heart, and including a discussion of pathophysiological processes such as fibrosis and scarring. We then discuss the central role of Canadian researchers in moving this field forwards, particularly in cardiac fibrosis, and highlight some of the major contributions of these individuals to our understanding of fibroblast and myofibroblast biology in health and disease.
Collapse
|
27
|
Shutova MS, Boehncke WH. Mechanotransduction in Skin Inflammation. Cells 2022; 11:2026. [PMID: 35805110 PMCID: PMC9265324 DOI: 10.3390/cells11132026] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
In the process of mechanotransduction, the cells in the body perceive and interpret mechanical stimuli to maintain tissue homeostasis and respond to the environmental changes. Increasing evidence points towards dysregulated mechanotransduction as a pathologically relevant factor in human diseases, including inflammatory conditions. Skin is the organ that constantly undergoes considerable mechanical stresses, and the ability of mechanical factors to provoke inflammatory processes in the skin has long been known, with the Koebner phenomenon being an example. However, the molecular mechanisms and key factors linking mechanotransduction and cutaneous inflammation remain understudied. In this review, we outline the key players in the tissue's mechanical homeostasis, the available data, and the gaps in our current understanding of their aberrant regulation in chronic cutaneous inflammation. We mainly focus on psoriasis as one of the most studied skin inflammatory diseases; we also discuss mechanotransduction in the context of skin fibrosis as a result of chronic inflammation. Even though the role of mechanotransduction in inflammation of the simple epithelia of internal organs is being actively studied, we conclude that the mechanoregulation in the stratified epidermis of the skin requires more attention in future translational research.
Collapse
Affiliation(s)
- Maria S. Shutova
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| |
Collapse
|
28
|
Yi Z, Zeng J, Chen Z, Chen L, Lu HB, Zhang Q, Yang X, Qi Z. The Role of Verteporfin in Prevention of Periprosthetic Capsular Fibrosis: An Experimental Study. Aesthet Surg J 2022; 42:820-829. [PMID: 35420670 DOI: 10.1093/asj/sjac083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Capsular contracture (CC) characterized by excessive fibrosis is one of the most common complications after silicone implant surgery. Verteporfin (VP), an inhibitor of Yes-associated protein 1 (YAP1), has recently been found to reduce the fibrotic process. OBJECTIVES The aim of this study was to use an in vivo rabbit model to evaluate the efficacy of VP for the prevention of CC. METHODS Twenty-four New Zealand rabbits received 10-cc smooth saline silicone implants inserted in the dorsal skin and were randomly divided into 2 groups to receive 2 mL VP (1.5 mg/mL) or 2 mL phosphate-buffered saline solution instillation in the implant pocket. When the animals were killed on Day 60, capsule formation was observed both macroscopically and microscopically. Histologic evaluation included capsule thickness, fibrosis degree, and myofibroblast (α smooth muscle actin positive) content. In addition, the YAP1 expression level was examined by immunofluorescence staining. Transforming growth factor β1, collagen I, and connective tissue growth factor expression were measured by real-time quantitative polymerase chain reaction. RESULTS The VP-treated group exhibited thinner, more transparent capsules and less fibrosis than the control group at 60 days postsurgery (P < 0.05). Moreover, the VP treatment significantly reduced α smooth muscle actin, YAP1, transforming growth factor β1, collagen I, and connective tissue growth factor expression levels in the capsular tissues (P < 0.05). CONCLUSIONS VP reduced capsule formation after silicone implantation by inhibiting YAP1-mediated mechanical signaling, thereby attenuating excessive collagen deposition in the rabbit model. This preclinical study may provide a feasible strategy to prevent periprosthetic capsular fibrosis in clinical application.
Collapse
Affiliation(s)
- Zhen Yi
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Jinshi Zeng
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Zixiang Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Lulu Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Hai-bin Lu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xiaonan Yang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Zuoliang Qi
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| |
Collapse
|
29
|
Qin Z, He T, Guo C, Quan T. Age-related downregulation of CCN2 is regulated by cell size in a YAP/TAZ-dependent manner in human dermal fibroblasts: impact on dermal aging. JID INNOVATIONS 2022; 2:100111. [PMID: 35480397 PMCID: PMC9035808 DOI: 10.1016/j.xjidi.2022.100111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/17/2022] Open
Abstract
CCN2, a member of the CCN family of matricellular proteins, is a key mediator and biomarker of tissue fibrosis. We previously reported that CCN2 is significantly reduced in aged human dermis, which contributes to dermal aging through the downregulation of collagen production, the major structural protein in the skin. In this study, we investigated the underlying mechanisms of the age-related downregulation of CCN2 in human skin dermal fibroblasts. Dermal fibroblasts isolation and laser-capture microdissection‒coupled RT-PCR from human skin confirmed that age-related reduction of CCN2 expression is regulated by epigenetics. Mechanistic investigation revealed that age-related reduction of CCN2 is regulated by impaired dermal fibroblast spreading/cell size, which is a prominent feature of aged dermal fibroblasts in vivo. Gain-of-function and loss-of-function analysis confirmed that age-related downregulation of CCN2 is regulated by YAP/TAZ in response to reduced cell size. We further confirmed that restoration of dermal fibroblast size rapidly reversed the downregulation of CCN2 in a YAP/TAZ-dependent manner. Finally, we confirmed that reduced YAP/TAZ nuclear staining is accompanied by loss of CCN2 in aged human skin in vivo. Our data reveal a mechanism by which age-related reduction in fibroblast spreading/size drives YAP/TAZ-dependent downregulation of CCN2 expression, which in turn contributes to loss of collagen in aged human skin.
Collapse
Affiliation(s)
- Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tianyuan He
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Chunfang Guo
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Correspondence: Taihao Quan, Department of Dermatology, University of Michigan Medical School, 1301 Catherine, Medical Science I, Room 6447, Ann Arbor, Michigan 48109-0609, USA.
| |
Collapse
|
30
|
Parry D, Allison K. Is the future scarless? - Fibroblasts as targets for scarless wound healing: a narrative review. Scars Burn Heal 2022; 8:20595131221095348. [PMID: 36082315 PMCID: PMC9445533 DOI: 10.1177/20595131221095348] [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] [Indexed: 11/16/2022] Open
Abstract
Introduction: Scarless healing is the ideal outcome of wound healing and is exhibited in some species. This narrative review assembles the current understanding of fibroblast heterogenicity along with the latest fibroblast-related targets for scar reduction therapies. Human regenerative wound healing is deemed possible due to the wound regeneration already seen in the early gestation foetus. Methods: This literature narrative review was undertaken by searching PubMed and Web of Science databases and Google Scholar to find articles concerning the fibroblast involvement in wound healing. We evaluated and collated these articles to form a consensus of the current understanding of the field. Discussion: This article describes current understanding of fibroblast heterogenicity and involvement in wound healing, focusing on the role of fibroblasts during physiological scarring. We also present the current most promising targets involving fibroblasts in the reduction of scarring and how we can manipulate the behaviour of fibroblasts to mimic the wound regeneration models in the human foetus. These targets include the pro-fibrotic EN1 positive fibroblast lineage, TGFβ1 inhibition, and genetic therapies utilising miRNAs and siRNAs. Conclusion: No therapies are currently available to eradicate scarring; however, treatment options are available to reduce the appearance of scarring. Further research into the heterogenicity and interactions of fibroblasts in both the foetus and adult is needed, and this may lead to the development of novel treatments against scarring.
Collapse
Affiliation(s)
- Dylan Parry
- Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Keith Allison
- South Tees Hospitals NHS Foundation Trust, Middlesbrough, UK
| |
Collapse
|
31
|
Lescoat A, Varga J, Matucci-Cerinic M, Khanna D. New promising drugs for the treatment of systemic sclerosis: pathogenic considerations, enhanced classifications, and personalized medicine. Expert Opin Investig Drugs 2021; 30:635-652. [PMID: 33909517 PMCID: PMC8292968 DOI: 10.1080/13543784.2021.1923693] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
Introduction: Systemic sclerosis (SSc), also known as scleroderma, is a complex orphan disease characterized by early inflammatory features, vascular hyper-reactivity, and fibrosis of the skin and internal organs. Although substantial progress has been made in the understanding of the pathogenesis of SSc, there is still no disease-modifying drug that could significantly impact the natural history of the disease.Areas covered: This review discusses the rationale, preclinical evidence, first clinical eevidence,and pending issues concerning new promising therapeutic options that are under investigation in SSc. The search strategy was based on PubMed database and clinical trial.gov, highlighting recent key pathogenic aspects and phase I or II trials of investigational drugs in SSc.Expert opinion: The identification of new molecular entities that potentially impact inflammation and fibrosis may constitute promising options for a disease modifying-agent in SSc. The early combinations of antifibrotic drugs (such as pirfenidone) with immunomodulatory agents (such as mycophenolate mofetil) may also participate to achieve such a goal. A more refined stratification of patients, based on clinical features, molecular signatures, and identification of subpopulations with distinct clinical trajectories, may also improve management strategies in the future.
Collapse
Affiliation(s)
- Alain Lescoat
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Dinesh Khanna
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|