1
|
Tang H, Wang X, Qiu S, Wang Y, Zhang X, Zhang Y. Low-density electrospun fibrous network promotes mechanotransduction and matrix remodeling in fibroblasts. BIOMATERIALS ADVANCES 2025; 174:214316. [PMID: 40245813 DOI: 10.1016/j.bioadv.2025.214316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
The mechanical interactions between cells and fiber-dominated extracellular matrix (ECM) are crucial in regulating matrix-remodeling and cellular physiological processes. Electrospun fibers, as a type of biomimicking fibers, provide an ideal platform for engineering a variety of tissues in vitro. However, the mechanisms by which electrospun fibers promote cellular matrix-remodeling, particularly concerning the characteristic mechanical compliance in the fibers, remain inadequately understood due to the crossing and entanglement of electrospun ultrafine fibers in those densely packed fibrous mats. This study devised low-density fibrous network and mechanically sensitive fibroblasts to investigate how cells sense, respond to, and remodel the residing microenvironment at both cellular and molecular levels. The results showed that the fibroblasts cultured on the low-density fibrous network exhibited a contractile phenotype, as evidenced by the upregulated transcription and synthesis of ECM-related proteins as well as fiber recruitment capability, thereby displaying a greater capacity in matrix-remodeling. Analysis of mechanotransduction-related markers revealed that the RhoA-ROCK signaling pathway was activated in the low-density fibrous network-substrated fibroblasts. Additionally, enhanced cytoskeletal assembly, cell contractility, YAP nuclear translocation, and activation of Piezo1 were observed. Inhibition of ROCK disrupted mechanotransduction, consequently impairing the cell's matrix-remodeling capacity. These findings demonstrate that the low-density electrospun fibrous network promotes the cell-mediated matrix-remodeling by facilitating mechanotransduction signaling. This study establishes a theoretical framework for understanding how electrospun fibers regulate cellular function at the micro-mechanical level and may shed insights on the design of biomimetic fibrous scaffolds for promoting tissue regeneration.
Collapse
Affiliation(s)
- Han Tang
- Institute of Pharmacology & Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang Key Laboratory of Neuropsychopharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; International Institute for Science, Proya Cosmetics Co., Ltd., Hangzhou 310023, China; College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xiaoli Wang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Sha Qiu
- Department of Traditional Chinese Medicine, Qingdao Central Hospital University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, China
| | - Yuying Wang
- International Institute for Science, Proya Cosmetics Co., Ltd., Hangzhou 310023, China
| | - Xiangnan Zhang
- Institute of Pharmacology & Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang Key Laboratory of Neuropsychopharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Yanzhong Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
2
|
Lv R, Liu Z, Guo H, Zhang B, Wang X, Peng Y, Chang Y, Yang F, Xiong Y, Hao J, Gao X, Wang X, Xu Q, Shimosawa T, Qiang P. Fibroblast to macrophage-like cell transition in renal inflammatory injury through the MR/CSF1 pathway induced by aldosterone. Life Sci 2025; 372:123627. [PMID: 40216224 DOI: 10.1016/j.lfs.2025.123627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/25/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
AIMS Inflammatory injury promotes the chronic kidney disease (CKD) progression,with renal macrophage accumulation and proliferation of as typical manifestations of inflammatory injury. We aimed to verify fibroblast to macrophage-like cell transition as a new source of macrophages that participate in renal inflammatory injury. MATERIALS AND METHODS Wistar rats were divided into Sham, ALD (aldosterone infusion for 12 weeks), and ESA (aldosterone infusion and esaxerenone by diet for 12 weeks) groups. Rat kidney interstitial fibroblast (RKF) were cultured, induced with aldosterone or CSF1, and treated with antagonists in vitro. The proportions of FSP-1+ F4/80+ cells in the rat kidney and RKF, including M1 marker iNOS/CD86 and M2 marker CD206/CD163 were assessed by flow cytometry and immunofluorescence staining. Single-cell RNA sequencing was used to assess the origin of macrophages in the rat kidneys and related gene expression. Additionally, immunofluorescence was used to detect FSP-1+ F4/80+ cells in kidney biopsy samples from CKD patients. KEY FINDINGS Fibroblast to macrophage-like cell transition was observed in both the kidneys of aldosterone-infused rats and in vitro aldosterone-treated RKF, with a predominant differentiation into the M1 phenotype. This transformation was mediated through the MR/CSF1 signalling pathway, revealing a novel source of macrophages and providing significant insights into the mechanisms underlying organ fibrosis. SIGNIFICANCE Aldosterone induces fibroblast to macrophage-like cell transition through the MR/ CSF1 pathway.
Collapse
Affiliation(s)
- Ruyan Lv
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ziqian Liu
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Haixia Guo
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Boya Zhang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xuan Wang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yunsong Peng
- Shijiazhuang Hospital of Traditional Chinese Medicine, Shijiazhuang 050200, China
| | - Yi Chang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Fan Yang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yunzhao Xiong
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Juan Hao
- Shijiazhuang Hospital of Traditional Chinese Medicine, Shijiazhuang 050200, China
| | - Xiaomeng Gao
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xiangting Wang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Qingyou Xu
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, International University of Health and Welfare, Narita 286-8686, Japan.
| | - Panpan Qiang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
3
|
Yang Y, Li L, Dai F, Deng L, Yang K, He C, Chen Y, Yang X, Song L. Fibroblast-derived versican exacerbates periodontitis progression by regulating macrophage migration and inflammatory cytokine secretion. Cell Signal 2025; 131:111755. [PMID: 40112905 DOI: 10.1016/j.cellsig.2025.111755] [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: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Versican (VCAN), a prominent extracellular matrix component upregulated in inflammatory diseases, demonstrates context-specific regulatory mechanisms. Periodontitis, a chronic inflammatory disease leading to periodontal tissue destruction and tooth loss, the pathological role of it remains poorly defined. Our study aims to examine VCAN-mediated mechanisms in periodontitis. METHODS We conducted a comprehensive analysis of bulk RNA sequencing and single-cell RNA sequencing data to examine VCAN expression level and source in periodontitis. Functional and correlation analyses were used to explore its biological functions. We then validated VCAN expression using quantitative real-time polymerase chain reaction, immunohistochemical staining, and immunofluorescence staining in animal models and investigated its biological functions in inflammation through in vitro experiments. RESULTS Our findings reveal that VCAN is mainly generated by fibroblast in periodontitis, and its expression significantly upregulated at both mRNA and protein levels. Using VCAN-overexpressing L929 cells, we demonstrated enhanced proliferative capacity and inflammatory potential. Co-culture experiments with RAW264.7 cells showed promoted migration, adhesion, M1 polarization, and mitogen-activated protein kinase (MAPK) pathway activation. CONCLUSION VCAN enhances fibroblast proliferation and migration, and upregulates inflammatory cytokines expression. Furthermore, fibroblast-derived VCAN not only induces macrophage chemotaxis, migration, adhesion, and polarization toward the proinflammatory M1 phenotype, but also activates MAPK signaling of macrophage, which may amplify inflammatory cascades to exacerbate periodontal tissue destruction. Targeted regulation of VCAN expression may become a promising precision treatment strategy for periodontitis.
Collapse
Affiliation(s)
- Yuting Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Li Li
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Fang Dai
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China
| | - Libin Deng
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, China
| | - Kaiqiang Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Chenjiang He
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Yeke Chen
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Xinbo Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Li Song
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China.
| |
Collapse
|
4
|
Zhang Q, Song Q, Li Z, Wu X, Chen Y, Lin H. Targeting fibroblasts in pathological bone formation: mechanisms and treatments. Front Cell Dev Biol 2025; 13:1612950. [PMID: 40491950 PMCID: PMC12146285 DOI: 10.3389/fcell.2025.1612950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Accepted: 05/19/2025] [Indexed: 06/11/2025] Open
Abstract
Fibroblasts are integral to the pathological processes underlying abnormal bone formation, including heterotopic ossification (HO), ankylosing spondylitis (AS), and ossification of the posterior longitudinal ligament (OPLL). This review summarized the diverse roles of fibroblasts, from their transdifferentiation into osteoblast-like cells to their influence on inflammatory and mechanical signal transduction pathways, including those mediated by BMP, TGF-β, and Wnt/β-catenin. In particular, senescent fibroblasts can secrete Activin A to activate the BMP pathway to drive HO formation, and fibroblasts can also differentiate into osteoblasts via interactions among the TGF-β1, BMP-2, and FGF-2 pathways. In AS and OPLL, fibroblasts respond to inflammatory signals and mechanical stress, contributing to pathological bone formation through extracellular matrix remodeling and osteogenic gene expression. In rare cases, fibroblast-mediated abnormal ossification also occurs in diffuse idiopathic skeletal hyperostosis (DISH) and systemic sclerosis (SSc). Therapeutic strategies targeting fibroblast signaling pathways, inflammation, and senescence are highlighted as potential interventions to mitigate these conditions.
Collapse
Affiliation(s)
- Qianyu Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Qimin Song
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Zeyin Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Xinyi Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Yuxiong Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Hui Lin
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Yu M, Si C, Xinjue H, Pan Y, Dai Y, Jin C, Han T, Yu C, Zhang J. Biglycan deficiency alleviates intestinal fibrosis through BMP-7-mediated Smad1/5/8 signaling. J Crohns Colitis 2025; 19:jjaf065. [PMID: 40249230 DOI: 10.1093/ecco-jcc/jjaf065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Indexed: 04/19/2025]
Abstract
BACKGROUND Biglycan (BGN) is a small proteoglycan rich in leucine, which plays a crucial role in the excessive production of extracellular matrix (ECM) and its association with fibrosis across various organs. Nevertheless, the precise contribution of BGN to intestinal fibrosis remains undisclosed. This study aimed to investigate the role and mechanism of BGN in intestinal fibrosis. METHODS Human Crohn's disease (CD) tissue samples were obtained from patients with Crohn's disease who underwent surgical resection of the intestine and were categorized as stenotic/nonstenotic regions. A dextran sodium sulfate (DSS)-induced mouse model of intestinal fibrosis was established. Bgn-/0 (BGN KO) mice and primary human intestinal fibroblasts were applied for the study of experimental fibrosis. Coimmunoprecipitation, immunofluorescence staining, western blot, and qRT-PCR were conducted to identify the regulatory effects of BGN on bone morphogenetic protein-7 (BMP-7) expression and intesinal fibrosis. RESULTS In both human CD samples and the DSS-induced mouse model of intestinal fibrosis, we observed a significant upregulation of BGN in areas activated by fibrosis. The genetic deletion of BGN resulted in the alleviation of intestinal fibrosis in mice administered DSS. The knockdown of BGN through siRNA significantly attenuated TGF-β1-induced ECM deposition and fibroblastic activation in primary human intestinal fibroblasts. Mechanistically, BGN directly interacted with and negatively regulated the anti-fibrotic protein BMP-7. Rescue experiments demonstrated that BGN facilitated intestinal fibrosis by modulating Smad1/5/8 phosphorylation and activating ECM deposition. CONCLUSION Our data indicate that BGN deficiency inhibits intestinal fibrosis through activation of the BMP-7-Smad1/5/8 signaling pathway. BGN and BMP-7 may become new biomarkers of intestinal fibrosis and novel targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Mengli Yu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chenqin Si
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 200054, China
| | - He Xinjue
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yuanyuan Pan
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yiyang Dai
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chengfeng Jin
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Tiemei Han
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chaohui Yu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
6
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
7
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
8
|
Moses JC, Sapkota A, Wu Y, Martinez I, Handa H, Brisbois EJ. In Situ Nitric Oxide Generating Nano-Bioactive Glass-Based Coatings and Its Therapeutic Ion Release toward Attenuating Implant-Associated Fibrosis and Infection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411984. [PMID: 39989185 PMCID: PMC11962685 DOI: 10.1002/smll.202411984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/24/2025] [Indexed: 02/25/2025]
Abstract
Nitric oxide (NO) is a potent gasotransmitter that exhibits a pleiotropic effect in regulating homeostasis and pathophysiology. Though it is a versatile biomaterial, silicone-based devices are still challenged by implant-associated infections and fibrous capsule formation complications. Here, a NO-generating (NOgen) interface is developed from copper or strontium-doped mesoporous bioactive glass-based coating on silicone substrates to facilitate metal-ion catalysis of endogenous S-nitrosothiols. The copper or strontium-based interfaces can generate physiologically relevant NO levels, which have bactericidal and antithrombotic effects to combat implant-associated early onsite infection and thrombosis. The NO generated in tandem with the low therapeutic release of strontium ions from the NOgen interface regulates cellular fate pertaining to fibroblasts, macrophages, and endothelial cells. Strontium suppresses the collagen expression and migration of activated fibroblasts while favoring M2 phenotype bias in macrophages. Differential NO flux observed over time from NOgen interfaces helps switch macrophages from proinflammatory M1 phenotype to M2 anti-inflammatory phenotype. Moreover, the synergistic effect of leachate and NO generated by the silicone substrate demonstrates a proangiogenic effect by aiding endothelial network maturation in vitro. Thus, the multifunctional features of the developed strontium-doped bioactive glass-based coating hold promise in regulating local immune-micromilieu and attenuating implant-associated fibrosis of silicone-based implantable devices.
Collapse
Affiliation(s)
- Joseph Christakiran Moses
- School of ChemicalMaterials and Biomedical EngineeringCollege of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Aasma Sapkota
- School of ChemicalMaterials and Biomedical EngineeringCollege of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Yi Wu
- School of ChemicalMaterials and Biomedical EngineeringCollege of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Isabel Martinez
- School of ChemicalMaterials and Biomedical EngineeringCollege of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Hitesh Handa
- School of ChemicalMaterials and Biomedical EngineeringCollege of EngineeringUniversity of GeorgiaAthensGA30602USA
- Pharmaceutical and Biomedical Sciences DepartmentCollege of PharmacyUniversity of GeorgiaAthensGA30602USA
| | - Elizabeth J. Brisbois
- School of ChemicalMaterials and Biomedical EngineeringCollege of EngineeringUniversity of GeorgiaAthensGA30602USA
| |
Collapse
|
9
|
Hung YL, Sato A, Takino Y, Ishigami A, Machida S. Resistance training suppresses accumulation of senescent fibro-adipogenic progenitors and senescence-associated secretory phenotype in aging rat skeletal muscle. GeroScience 2025; 47:1669-1683. [PMID: 39298108 PMCID: PMC11979060 DOI: 10.1007/s11357-024-01338-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Accumulation of senescent cells in tissues contributes to multiple aging-related pathologies. Senescent fibro-adipogenic progenitors (FAPs) contribute to aging-related muscle atrophy. Resistance training can help to maintain skeletal muscle mass, improve mobility, and reduce certain health risks commonly associated with aging. We investigated, using rat model, the impact of resistance training on FAPs in aging skeletal muscle, which remains unclear. Twenty-two-month-old female rats were divided into sedentary and training groups. The training group rodents were trained to climb a ladder while bearing a load for 20 training sessions over 2 months, after which, the flexor hallucis longus muscles were collected and analyzed. Senescent cells were identified using a senescence-associated β-galactosidase stain and p21 immunohistochemistry (IHC), and FAPs were identified using platelet-derived growth factor receptor alpha IHC. The results indicate that resistance training in rats prevented aging-associated skeletal muscle atrophy and suppressed M2 polarization of macrophages. The number of senescent cells was significantly reduced in the 24-month-old training group, with most of them being FAPs. Conversely, the number of senescent FAPs increased significantly in the 24-month-old sedentary group compared with that in the 18-month-old sedentary group. The number of senescent FAPs in the 24-month-old training group decreased significantly. Resistance training also suppressed the senescence-associated secretory phenotype (SASP). The killer T cell-specific marker, CD8α, was elevated in the skeletal muscles of the aging rats following resistance training, indicating upregulation of recognition and elimination of senescent cells. Overall, resistance training suppressed the accumulation of senescent FAPs and acquisition of SASP in aging skeletal muscles.
Collapse
Affiliation(s)
- Yung-Li Hung
- Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-Ku, Tokyo, 102-0083, Japan
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| | - Ayami Sato
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Yuka Takino
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
- Institute of Health and Sports Science & Medicine, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| |
Collapse
|
10
|
Gupta R, Schärer P, Liao Y, Roy B, Benoit RM, Shivashankar GV. Regulation of p65 nuclear localization and chromatin states by compressive force. Mol Biol Cell 2025; 36:ar37. [PMID: 39908115 PMCID: PMC12005105 DOI: 10.1091/mbc.e23-11-0431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
The tumor microenvironment (TME) is a dynamic ecosystem, that evolves with the developing tumor to support its growth and metastasis. A key aspect of TME evolution is the recruitment of stromal fibroblasts, carried out via the release of various tumor signals including tumor necrosis factor (TNFα). These tumor signals in turn alter the mechanical properties of the TME as the tumor grows. Because of the important role of stromal cells in supporting tumor progression, new therapies aim to target stromal fibroblasts. However, these therapies have been unsuccessful in part due to the limited understanding of cross-talk between chemical and altered mechanical signaling within stromal fibroblasts. To investigate this, we designed a coculture assay with YFP-TNFα releasing spheroids embedded within collagen gels alongside fibroblasts to mimic the stromal response within the TME. This resulted in the nuclear translocation of p65 in the stromal fibroblasts which was further intensified by the addition of compressive stress. The combination of mechanical and chemical signals led to cytoskeletal disruption and induced a distinct chromatin state in the stromal fibroblasts. These results highlight the important cross-talk between cytokine signaling and mechanical forces on stromal cells within the TME and facilitate the development of a better spheroid model for therapeutic interventions.
Collapse
Affiliation(s)
- Rajshikhar Gupta
- Laboratory of Multiscale Bioimaging, Paul Scherrer Institut, Villigen, Aargau, Switzerland 5232
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland 8092
| | - Paulina Schärer
- Laboratory of Multiscale Bioimaging, Paul Scherrer Institut, Villigen, Aargau, Switzerland 5232
| | - Yawen Liao
- Laboratory of Multiscale Bioimaging, Paul Scherrer Institut, Villigen, Aargau, Switzerland 5232
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland 8092
| | - Bibhas Roy
- Laboratory of Multiscale Bioimaging, Paul Scherrer Institut, Villigen, Aargau, Switzerland 5232
- Department of Biological Science, BITS-Pilani Hyderabad Campus, Secunderabad, Telngana, India 500078
| | - Roger M. Benoit
- Laboratory of Multiscale Bioimaging, Paul Scherrer Institut, Villigen, Aargau, Switzerland 5232
| | - G. V. Shivashankar
- Laboratory of Multiscale Bioimaging, Paul Scherrer Institut, Villigen, Aargau, Switzerland 5232
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland 8092
| |
Collapse
|
11
|
Estrada AC, Humphrey JD. Multi-Scale Multi-Cell Computational Model of Inflammation-Mediated Aortic Remodeling in Hypertension. Ann Biomed Eng 2025; 53:1014-1023. [PMID: 39904866 PMCID: PMC12067544 DOI: 10.1007/s10439-025-03685-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Multiple cell types interact within the aortic wall to control development, homeostasis, and adaptation as well as to drive disease progression. Given the complexity of these interactions and their manifestations at the tissue level, there is a pressing need for a new class of computational models that integrate data across scales. METHODS We meld logic-based cell signaling models of vascular smooth muscle cells, adventitial fibroblasts, and macrophages and couple this multi-cell model with a tissue level-constrained mixture model of aortic growth and remodeling. The coupled multi-scale model is parameterized using data from the literature and then specialized for the case of angiotensin II-induced hypertensive remodeling of the descending thoracic aorta in wild-type mice. RESULTS We contrast important contributions of chemo- and mechano-stimulation of cell responses and identify critical roles of recruited macrophages in driving the non-homeostatic thickening of the adventitial layer that reduces biaxial wall stress below setpoint values. CONCLUSION We show the utility of a multi-scale, multi-cell model in delineating effects of different chemo-mechanical stimuli in aortic remodeling in hypertension.
Collapse
MESH Headings
- Animals
- Hypertension/pathology
- Hypertension/physiopathology
- Hypertension/metabolism
- Mice
- Models, Cardiovascular
- Vascular Remodeling
- Macrophages/pathology
- Macrophages/metabolism
- Angiotensin II
- Inflammation/pathology
- Inflammation/physiopathology
- Inflammation/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Muscle, Smooth, Vascular/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/metabolism
- Fibroblasts/pathology
- Fibroblasts/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Humans
- Computer Simulation
Collapse
Affiliation(s)
- Ana C Estrada
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Electrical and Biomedical Engineering, Fairfield University, Fairfield, CT, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
12
|
Khang A, Barmore A, Tseropoulos G, Bera K, Batan D, Anseth KS. Automated prediction of fibroblast phenotypes using mathematical descriptors of cellular features. Nat Commun 2025; 16:2841. [PMID: 40121192 PMCID: PMC11929917 DOI: 10.1038/s41467-025-58082-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
Fibrosis is caused by pathological activation of resident fibroblasts to myofibroblasts that leads to aberrant tissue stiffening and diminished function of affected organs with limited pharmacological interventions. Despite the prevalence of myofibroblasts in fibrotic tissue, existing methods to grade fibroblast phenotypes are typically subjective and qualitative, yet important for screening of new therapeutics. Here, we develop mathematical descriptors of cell morphology and intracellular structures to identify quantitative and interpretable cell features that capture the fibroblast-to-myofibroblast phenotypic transition in immunostained images. We train and validate models on features extracted from over 3000 primary heart valve interstitial cells and test their predictive performance on cells treated with the small molecule drugs 5-azacytidine and bisperoxovanadium (HOpic), which inhibited and promoted myofibroblast activation, respectively. Collectively, this work introduces an analytical framework that unveils key features associated with distinct fibroblast phenotypes via quantitative image analysis and is broadly applicable for high-throughput screening assays of candidate treatments for fibrotic diseases.
Collapse
Affiliation(s)
- Alex Khang
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Abigail Barmore
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Kaustav Bera
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Dilara Batan
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
13
|
Qu C, Koskinen Holm C. Impact of a Heterozygous C1R R301P/WT Mutation on Collagen Metabolism and Inflammatory Response in Human Gingival Fibroblasts. Cells 2025; 14:479. [PMID: 40214433 PMCID: PMC11987961 DOI: 10.3390/cells14070479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Periodontal Ehlers-Danlos syndrome arising from heterozygous pathogenic mutation in C1R and/or C1S genes is an autosomal-dominant disorder characterized by early-onset periodontitis. Due to the difficulties in obtaining and culturing the patient-derived gingival fibroblasts, we established a model system by introducing a heterozygous C1RR301P/WT mutation into human TERT-immortalized gingival fibroblasts (hGFBs) to investigate its specific effects on collagen metabolism and inflammatory responses. A heterozygous C1RR301P/WT mutation was introduced into hGFBs using engineered prime editing. The functional consequences of this mutation were assessed at cellular, molecular, and enzymatic levels using a variety of techniques, including cell growth analysis, collagen deposition quantification, immunocytochemistry, enzyme-linked immunosorbent assay, and quantitative real-time reverse transcription polymerase chain reaction. The C1RR301P/WT-mutated hGFBs (mhGFBs) exhibited normal morphology and growth rate compared to wild-type hGFBs. However, mhGFBs displayed upregulated procollagen α1(V), MMP-1, and IL-6 mRNA expression while simultaneously downregulating collagen deposition and C1r protein levels. A modest accumulation of unfolded collagens was observed in mhGFBs. The mhGFBs exhibited a heightened inflammatory response, with a more pronounced increase in MMP-1 and IL-6 mRNA expression compared to TNF-α/IL-1β-stimulated hGFBs. Unlike cytokine-stimulated hGFBs, cytokine-stimulated mhGFB did not increase C1R, C1S, procollagen α1(III), and procollagen α1(V) mRNA expression. Our results suggest that the C1RR301P/WT mutation specifically disrupts collagen metabolism and inflammatory pathways in hGFBs, highlighting the mutation's role in these processes. While other cellular functions appear largely unaffected, these findings underscore the potential of targeting collagen metabolism and inflammation for therapeutic interventions in pEDS.
Collapse
Affiliation(s)
- Chengjuan Qu
- Department of Odontology, Umeå University, 90185 Umeå, Sweden;
| | | |
Collapse
|
14
|
Liao Q, Chen J, Liu G. Low intensity pulsed ultrasound alleviates synovial fibrosis in osteoarthritis via the PI3K/AKT pathway. Sci Rep 2025; 15:9644. [PMID: 40113833 PMCID: PMC11926212 DOI: 10.1038/s41598-025-92413-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
Previous studies have reported that low-intensity pulsed ultrasound (LIPUS) can alleviate cartilage degradation in osteoarthritis (OA). However, the functions and mechanisms of LIPUS in synovial fibrosis with OA require further study. To investigate the role of the PI3K/AKT signaling pathway in synovial fibrosis and in LIPUS treatment in synovial fibrosis, a TGF-β stimulated rat FLS cell model and a rat OA animal model based on anterior cruciate ligament transection (ACLT) and partial medial meniscectomy (MMx) were used. The results revealed that LIPUS delayed the progression of OA. Masson staining revealed that LIPUS reduced the collagen deposition of synovial tissue in OA rats. Correspondingly, immunofluorescence demonstrated that LIPUS significantly downregulated the expression of α-SMA, Col1a1 and Col3a1 in OA rats. Moreover, TGF-β stimulation upregulated fibrosis markers at the mRNA and protein levels in FLS, as well as increased phosphorylation-dependent activation of the PI3K/Akt pathway. 740Y-P was found to promote the fibrotic change of FLS induced by TGF-β, but LY294002 reduced its expression. However, LIPUS inhibits the fibrotic change and activation of the PI3K/Akt pathway in FLS under stimulation of TGF-β. In conclusion, LIPUS alleviates synovial fibrosis by blocking the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qing Liao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Jun Chen
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, 510000, China
- Taihe Hospital, Hubei University of Medicine, Shiyan City, 442000, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
15
|
Rieder F, Nagy LE, Maher TM, Distler JHW, Kramann R, Hinz B, Prunotto M. Fibrosis: cross-organ biology and pathways to development of innovative drugs. Nat Rev Drug Discov 2025:10.1038/s41573-025-01158-9. [PMID: 40102636 DOI: 10.1038/s41573-025-01158-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/20/2025]
Abstract
Fibrosis is a pathophysiological mechanism involved in chronic and progressive diseases that results in excessive tissue scarring. Diseases associated with fibrosis include metabolic dysfunction-associated steatohepatitis (MASH), inflammatory bowel diseases (IBDs), chronic kidney disease (CKD), idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc), which are collectively responsible for substantial morbidity and mortality. Although a few drugs with direct antifibrotic activity are approved for pulmonary fibrosis and considerable progress has been made in the understanding of mechanisms of fibrosis, translation of this knowledge into effective therapies continues to be limited and challenging. With the aim of assisting developers of novel antifibrotic drugs, this Review integrates viewpoints of biologists and physician-scientists on core pathways involved in fibrosis across organs, as well as on specific characteristics and approaches to assess therapeutic interventions for fibrotic diseases of the lung, gut, kidney, skin and liver. This discussion is used as a basis to propose strategies to improve the translation of potential antifibrotic therapies.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA.
- Program for Global Translational Inflammatory Bowel Diseases (GRID), Chicago, IL, USA.
| | - Laura E Nagy
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Toby M Maher
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- National Heart and Lung Institute, Imperial College, London, UK
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen; Medical Faculty, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
16
|
Walker JT, Cooper TT, Dunmore-Buyze J, Serack FE, Brooks C, Grant A, Drangova M, Lajoie G, Dekaban GA, Flynn LE. Syngeneic adipose-derived stromal cells modulate the immune response but have limited persistence within decellularized adipose tissue implants in C57BL/6 mice. Acta Biomater 2025; 195:169-182. [PMID: 39922513 DOI: 10.1016/j.actbio.2025.02.015] [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: 10/18/2024] [Revised: 01/14/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
The delivery of adipose-derived stromal cells (ASCs) on cell-instructive decellularized adipose tissue (DAT) scaffolds is a promising strategy for stimulating host-derived soft tissue regeneration. However, a better understanding of the mechanisms through which ASCs modulate regeneration in vivo is needed to harness these cells more effectively. In this study, DAT scaffolds, both with and without seeded syngeneic DsRED+ mouse ASCs, were implanted into immunocompetent C57BL/6 mice. Downstream analyses focused on assessing donor ASC persistence and phenotype, as well as the effects of ASC seeding on host macrophage polarization and the perfused host vascular network. Notably, most donor ASCs were cleared from the scaffolds by 2 weeks. Mass spectrometry-based proteomics indicated that the transplanted ASCs maintained their pre-implantation phenotype up to 1 week in vivo, suggesting that the cells were not undergoing programmed cell death. A higher fraction of the infiltrating host macrophages expressed CD68 and Arginase-1 in the ASC-seeded implants up to 1-week post-implantation. Interestingly, a small population of phagocytic macrophages, identified by uptake of DsRED protein, was present in the DAT implants in the first 2 weeks and showed enhanced expression of CD68, Arginase-1, and CD163, along with reduced expression of iNOS. MicroCT angiography revealed a similar perfused vessel network in the seeded and unseeded groups at 4- and 8-weeks post-implantation. Overall, seeding with syngeneic ASCs modulated the host macrophage response to the DAT bioscaffolds at early timepoints, but did not impact long-term regenerative outcomes, potentially due to the rapid clearance of the donor cell population in this model. STATEMENT OF SIGNIFICANCE: Decellularized adipose tissue (DAT) is a promising biomaterial for treating soft tissue defects. Seeding with adipose-derived stromal cells (ASCs) can augment fat regeneration within DAT in pre-clinical models, but our understanding of how ASCs contribute to tissue regeneration in vivo remains limited. Furthermore, ASC clearance from implanted biomaterials is well described, but poorly understood. Here, ASC-seeded DAT was implanted subcutaneously in immunocompetent mice to assess how ASCs altered the host macrophage response, functional vascular regeneration, and long-term integration with the host tissues. Additionally, ASC phenotype and persistence were assessed to determine how these cells might be cleared from the implants. Such understanding is critical to design biomaterials that can better harness the therapeutic benefits of ASCs.
Collapse
Affiliation(s)
- John T Walker
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Tyler T Cooper
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Joy Dunmore-Buyze
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Fiona E Serack
- School of Biomedical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Aaron Grant
- Division of Plastic and Reconstructive Surgery, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, Ontario N6A 5C1, Canada
| | - Maria Drangova
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada; Department of Medical Biophysics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Gilles Lajoie
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Gregory A Dekaban
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada; Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Lauren E Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada; School of Biomedical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario N6A 3K7, Canada; Department of Chemical and Biochemical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada.
| |
Collapse
|
17
|
Froom ZSCS, Medd K, Wheeler BP, Osborne ND, Rempe CN, Woodworth KE, Charron C, Davenport Huyer L. Antifibrotic Function of Itaconate-Based Degradable Polyester Materials. ACS Biomater Sci Eng 2025; 11:1549-1561. [PMID: 39961606 DOI: 10.1021/acsbiomaterials.4c02444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2025]
Abstract
Pathological fibrosis is a chronic disease, characterized by excessive extracellular matrix deposition, that remains a significant global health challenge. Despite its prevalence, current antifibrotic therapies are limited due to the complex interplay and signaling of profibrotic macrophages and fibroblast cells that underlies fibrotic tissue microenvironments. This study investigates a novel approach to combat fibrosis, harnessing the antifibrotic properties of the endogenous metabolite itaconate (IA) to target the pathological activation of the macrophage-fibroblast axis in fibrotic disease. To achieve therapeutic delivery relevant to the chronic nature of fibrotic conditions, we incorporated IA into the backbone of biodegradable polyester polymers, poly(dodecyl itaconate) (poly[IA-DoD]), capable of long-term localized release of IA. Degradation characterization of poly(IA-DoD) revealed that IA, as well as water-soluble IA-containing oligomeric groups, is released in a sustained manner. Treatment of murine bone marrow-derived macrophages and human dermal fibroblasts demonstrated that the degradation products of poly(IA-DoD) effectively modulated profibrotic behavior. Macrophages exposed to the degradation products exhibited reduced profibrotic responses, while fibroblasts showed decreased proliferation and myofibroblast α-smooth muscle actin expression. These findings suggest that poly(IA-DoD) has the potential to disrupt the fibrotic cycle by targeting key cellular players. This polymer-based delivery system offers a promising strategy for the treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kyle Medd
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Brenden P Wheeler
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Natasha D Osborne
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Christian N Rempe
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kaitlyn E Woodworth
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Carlie Charron
- Department of Chemistry, Faculty of Science, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Nova Scotia Health, Halifax, NS B3S 0H6, Canada
| |
Collapse
|
18
|
Kim DH, Jeong CH, Han JH, Lim SJ, Kwon HC, Kim YJ, Keum DH, Lee KH, Han SG. Comparative toxicity study of hyaluronic acid fillers crosslinked with 1,4-butanediol diglycidyl ether or poly (ethylene glycol) diglycidyl ether. Int J Biol Macromol 2025; 296:139620. [PMID: 39788250 DOI: 10.1016/j.ijbiomac.2025.139620] [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: 02/27/2024] [Revised: 10/15/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Dermal fillers comprising injectable hyaluronic acid (HA) are widely used for soft tissue augmentation, often using crosslinking agents such as 1,4-butanediol diglycidyl ether (BDDE) or poly (ethylene glycol) diglycidyl ether (PEGDE). Here, we assessed the physical properties, toxicity, and inflammatory reactions of HA fillers crosslinked with either BDDE (HA-BDDE filler) or PEGDE (HA-PEGDE filler) in in vitro and in vivo investigations. The HA-PEGDE filler exhibited higher G', tan δ, G*, and complex viscosity values compared to the HA-BDDE filler, while maintaining similar cohesivity. The filler extracts were used to evaluate cytotoxicity. HA-PEGDE filler extracts exhibited reduced cytotoxicity, oxidative stress, and inflammation in human keratinocytes (HaCaTs) and fibroblasts (HDFs) than that of HA-BDDE filler extracts. In animal studies, SKH1-hairless mice were injected subcutaneously with varying volumes of fillers and euthanized at 1- and 4-weeks post-injection. Compared with the HA-BDDE filler, the HA-PEGDE filler displayed favorable biocompatibility, decreased tumor necrosis factor (TNF)-α and interleukin (IL)-1β expression and reduced inflammatory cell infiltration. Our results demonstrate that the HA-PEGDE filler has comparable rheological properties and lower toxicity than the HA-BDDE filler, suggesting its suitability as an alternative in applications where minimal inflammatory response is crucial.
Collapse
Affiliation(s)
- Do Hyun Kim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Chang Hee Jeong
- Technology Innovation Research Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Jong Hyeon Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Su Jin Lim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyuk Cheol Kwon
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Yea Ji Kim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Dong Hyun Keum
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Keyong Ho Lee
- R&D Department, Organoflora, Chuncheon 42237, Republic of Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
19
|
Li Z, Jiang J, Cai K, Qiao Y, Zhang X, Wang L, Kang Y, Wu X, Zhao B, Wang X, Zhang T, Lin Z, Wu J, Lu S, Gao H, Jin H, Xu C, Huangfu X, James Z, Chen Q, Zheng X, Liu NN, Zhao J. CCN2 mediates fibroblast-macrophage interaction in knee arthrofibrosis based on single-cell RNA-seq analysis. Bone Res 2025; 13:26. [PMID: 39994205 PMCID: PMC11850813 DOI: 10.1038/s41413-025-00400-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 12/02/2024] [Accepted: 12/31/2024] [Indexed: 02/26/2025] Open
Abstract
Knee arthrofibrosis, characterized by excessive matrix protein production and deposition, substantially impairs basic daily functions, causing considerable distress and financial burden. However, the underlying pathomechanisms remain unclear. Here, we characterized the heterogeneous cell populations and cellular pathways by combination of flow cytometry and single-cell RNA-seq analysis of synovial tissues from six patients with or without knee arthrofibrosis. Increased macrophages and fibroblasts were observed with decreased numbers of fibroblast-like synoviocytes, endothelial cells, vascular smooth muscle cells, and T cells in the arthrofibrosis group compared with negative controls. Notably, fibroblasts were discovered to interact with macrophages, and lead to fibrosis through TGF-β pathway induced CCN2 expression in fibroblasts. CCN2 was demonstrated to be required for fibroblast pro-fibrotic functions (activation, proliferation, and migration) through TGFBR/SMAD pathway. The expression of CCN2 was positively correlated with the collagen volume and TGF-β expression and negatively associated with patient-reported outcome measures in another cohort of patients with knee arthrofibrosis. Our study reveals the role of CCN2 in the fibroblast-macrophage interaction through TGF-β pathway which might help to shed light on CCN2 as a potential biomarker.
Collapse
Affiliation(s)
- Ziyun Li
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jia Jiang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Kangwen Cai
- Shanghai Normal University, Shanghai, 200233, China
| | - Yi Qiao
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xuancheng Zhang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liren Wang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuhao Kang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiulin Wu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Benpeng Zhao
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiuli Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tianyi Zhang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhiqi Lin
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jinlong Wu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Simin Lu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haihan Gao
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haocheng Jin
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Caiqi Xu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaoqiao Huangfu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhengzhi James
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qiuhua Chen
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaoqi Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jinzhong Zhao
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
20
|
Sheng K, Ran Y, Feng X, Wang Y, Zhou S, Guan Y, Tan P, Qian S, Zhao Z, Zhang B, Ji W, Niu C, Guo X. PTN secreted by cardiac fibroblasts promotes myocardial fibrosis and inflammation of pressure overload-induced hypertrophic cardiomyopathy through the PTN-SDC4 pathway. Life Sci 2025; 363:123356. [PMID: 39765325 DOI: 10.1016/j.lfs.2024.123356] [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: 10/20/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is characterized by unexplained left ventricular hypertrophy (LVH) with key pathologic processes including myocardial necrosis, fibrosis, inflammation, and hypertrophy, which are involved in heart failure (HF), stroke, and even sudden death. Our aim was to explore the communication network among various cells in the heart of transverse aortic constriction (TAC) surgery induced HCM mice. MATERIALS AND METHODS Single-cell RNA-seq data of GSE137167 was downloaded from the Gene Expression Omnibus (GEO) database. Seurat was used to perform the standard workflow. CellChat was utilized to compute the cell-cell interaction network and analyze the ligand-receptor pairs. Weighted gene co-expression network analysis (WGCNA) was conducted to identify gene co-expression modules. In vitro and in vivo studies were performed to verify bioinformatic analysis findings through real-time quantitative PCR (RT-qPCR), Edu staining, transwell assay, western blot, immunofluorescence assay, CCK-8, hematoxylin and eosin (H&E) staining, and echocardiography based on TAC mouse model. KEY FINDINGS Our results showed that after TAC surgery, the interaction between cardiac fibroblasts and macrophages was very common, and the increasing pleiotrophin (PTN) ligand secreted by cardiac fibroblasts could promote the self-proliferation or invasion for myocardial fibrosis as well as stimulate the inflammatory response of macrophages to contribute TAC surgery induced HCM through acting on Syndecan 4 (SDC4) receptor. SIGNIFICANCE Our study demonstrates that PTN derived from cardiac fibroblasts may play potential role in pressure overload-induced HCM through activating the PTN-SDC4 pathway in cardiac fibroblasts and macrophages, which may be a potential therapeutic target for pressure overload-induced HCM patients.
Collapse
Affiliation(s)
- Ke Sheng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqing Ran
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xing Feng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijia Wang
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Size Zhou
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Guan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pingping Tan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songwei Qian
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiguang Zhao
- Department of Pathology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bing Zhang
- Engineering Research Center of Techniques and Instruments for Diagnosis and Treatment of Congenital Heart Disease, , Institute of Developmental and Regenerative Medicine, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiping Ji
- Department of General Surgery, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China; Department of General Surgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chao Niu
- Pediatric Research Institute, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China.
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
21
|
Allen RS, Seifert AW. Spiny mice (Acomys) have evolved cellular features to support regenerative healing. Ann N Y Acad Sci 2025; 1544:5-26. [PMID: 39805008 PMCID: PMC11830558 DOI: 10.1111/nyas.15281] [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: 01/16/2025]
Abstract
Spiny mice (Acomys spp.) are warm-blooded (homeothermic) vertebrates whose ability to restore missing tissue through regenerative healing has coincided with the evolution of unique cellular and physiological adaptations across different tissue types. This review seeks to explore how these bizarre rodents deploy unique or altered injury response mechanisms to either enhance tissue repair or fully regenerate excised tissue compared to closely related, scar-forming mammals. First, we examine overall trends in healing Acomys tissues, including the cellular stress response, the ability to activate and maintain cell cycle progression, and the expression of certain features in reproductive adults that are normally associated with embryos. Second, we focus on specific cell types that exhibit precisely regulated proliferation to restore missing tissue. While Acomys utilize many of the same cell types involved in scar formation, these cells exhibit divergent activation profiles during regenerative healing. Considered together, current lines of evidence support sustained deployment of proregenerative pathways in conjunction with transient activation of fibrotic pathways to facilitate regeneration and improve tissue repair in Acomys.
Collapse
Affiliation(s)
- Robyn S. Allen
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Ashley W. Seifert
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
- The Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky, USA
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
22
|
Monaco CF, Jones CM, Sayles HR, Rudloff B, McFee R, Cupp AS, Davis JS. Luteal fibroblasts produce prostaglandins in response to IL1β in a MAPK-mediated manner. Mol Cell Endocrinol 2025; 596:112420. [PMID: 39577796 DOI: 10.1016/j.mce.2024.112420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/09/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
The corpus luteum is a temporary endocrine gland that is crucial for pregnancy, as it produces the progesterone needed to maintain optimal uterine conditions for implantation. In the absence of a conceptus, the corpus luteum becomes non-functional and undergoes rapid tissue remodeling to regress into a fibrotic corpus albicans. Early luteal regression is characterized by increased cytokine release. Because the role of fibroblasts in the bovine corpus luteum remains to be elucidated, the aim of this study was to elucidate the response of bovine luteal fibroblasts to inflammatory cytokines, tumor necrosis factor α (TNFα), and interleukin 1β (IL1β). Both cytokines induced canonical mitogen activated protein kinase (MAPK) signaling in luteal fibroblasts by phosphorylation of ERK1/2, p38 MAPK, and JNK. IL1β elevated expression and phosphorylation of cytosolic phospholipase A2 (cPLA2), an enzyme that mobilizes arachidonic acid for prostanoid synthesis. IL1β also elevated expression of prostaglandin-endoperoxide synthase 2 (PTGS2), another enzyme needed to synthesize prostanoids. IL1β increased PGF2α and PGE2 levels in the culture medium over 20-fold. Inhibition of MAPKs with small-molecule inhibitors abrogated the stimulatory effects of IL1β. IL1β also induced prostaglandin production in steroidogenic cells; however, there was no elevation in cPLA2. Therefore, actions of IL1β differ based on ovarian cell type. All together, we have identified luteal fibroblasts as potential inflammatory mediators during luteal regression.
Collapse
Affiliation(s)
- Corrine F Monaco
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Chloe M Jones
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Harlan R Sayles
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brooke Rudloff
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Renee McFee
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA; US Department of Veterans Affairs VA Medical Center, Omaha, NE, USA.
| |
Collapse
|
23
|
Steffan B, Grossmann T, Gerstenberger C, Gugatschka M, Hortobagyi D, Kirsch A, Grill M. Functional Characteristics of the Crosstalk Between Vocal Fold Fibroblasts and Macrophages-The Role of Vibration in Vocal Fold Inflammation. J Voice 2025:S0892-1997(24)00478-8. [PMID: 39799073 DOI: 10.1016/j.jvoice.2024.12.043] [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: 11/25/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025]
Abstract
OBJECTIVES This in vitro study investigated the interaction between human vocal fold fibroblasts (hVFF) and macrophages under the influence of cigarette smoke extract (CSE) and vibration as potential regulators of vocal fold (VF) inflammation. STUDY DESIGN Experimental in vitro pilot study. METHODS Immortalized hVFF were cultured in flexible-bottomed cell culture plates, treated with CSE, and subjected to static or dynamic conditions in a phonomimetic bioreactor. For coculture, unstimulated or lipopolysaccharide/IFNγ-stimulated THP-1 (human leukemia monocytic cell line) macrophages were added in inserts for a final 24 hours of vibration period. We measured messenger ribonucleic acid (mRNA) (quantitative polymerase chain reaction [qPCR]) and protein levels (Western Blot, ELISA, and LUMINEX®) of hVFF and analyzed the results using two- and three-way ANOVA with post hoc tests. RESULTS Under inflammatory stimulation, we observed a reduction of collagen (COL) type 1A1, 1A2, and 3A1, and increased gene expression of COL4A1, matrix metallopeptidase 2, and vascular endothelial growth factor A in hVFF. Additionally, the pro-inflammatory markers cyclooxygenase (COX) 1 and 2, interleukin (IL) 1β, IL-6, and IL-8 were upregulated. CSE increased COX1 and COX2 levels, whereas vibration reduced CSE-induced increases of COL4A1 and COX2 in pro-inflammatory stimulated hVFF. CONCLUSION This study indicates that vibration may mitigate CSE-induced inflammatory damage in the hVFF, thereby offering new insights into the cellular crosstalk that underlies the pathophysiology of VF inflammation in smoking-related voice disorders.
Collapse
Affiliation(s)
- Barbara Steffan
- Division of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Graz, Austria
| | - Tanja Grossmann
- Division of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Graz, Austria.
| | - Claus Gerstenberger
- Division of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Graz, Austria
| | - Markus Gugatschka
- Division of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Graz, Austria
| | - David Hortobagyi
- Division of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Graz, Austria
| | - Andrijana Kirsch
- Division of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Graz, Austria; Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Magdalena Grill
- Division of Phoniatrics, ENT University Hospital Graz, Medical University of Graz, Graz, Austria; Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| |
Collapse
|
24
|
Ramamoorthi L, Jeyabalan S, Sankar S, Begum MY, Duraipandian C, Sekar M, Wong LS, Subramaniyan V. Anti-arthritic potential of crude sulfated polysaccharide from marine macroalgae Sargassum ilicifolium (Turner) C. Agardh: Regulation of cytokine cascade. Biomol Concepts 2025; 16:bmc-2022-0050. [PMID: 39846213 DOI: 10.1515/bmc-2022-0050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/30/2024] [Indexed: 01/24/2025] Open
Abstract
Seaweeds have been utilized as food, fodder, fertilizer, and medicine since ancient times; nevertheless, they have received only a little attention. In the current work, we extracted the sulfated polysaccharide from a marine source and investigated its anti-arthritic potential in vivo. The isolated and freeze-dried polysaccharide was tested for acute oral toxicity based on OECD 423. This step was followed by investigations on clinical signs and gross pathological alterations seen. A complete Freund's adjuvant-induced arthritis was used to test the in vivo activity in female Sprague-Dawley rats, which were divided into five groups: (1) normal control, (2) arthritic control, (3) methotrexate treatment (0.1 mg/kg), (4) crude sulfated polysaccharide (CSP) (5 mg/kg), and (5) CSP (10 mg/kg). CSP was from the marine brown algae Sargassum ilicifolium from the Gulf of Mannar. The body weight, paw volume, and biochemical markers (alanine aminotransferase, aspartate aminotransferase, creatinine, urea, and C-reactive protein levels) were also measured for each group coupled with histopathological and immunohistochemistry studies. The acute toxicity investigation indicated that the lethal dose of 50% (LD50) of the polysaccharide was more than 2,000 mg/kg. In addition, animals from the methotrexate and CSP (5 mg/kg, p.o.) groups had a substantial reduction in paw volume compared to other treatment groups. Methotrexate and CSP treatment dramatically decreased the levels of the investigated marker enzymes. Histopathology revealed that low-dose CSP (5 mg/kg, p.o.) significantly reduced the severity of synovitis, panniculitis, liver necrosis, inflammatory cell infiltration, and cortical and paracortical necrotic foci in node, compared to the high dose (10 mg/kg, p.o.). Immunohistochemical studies revealed that CSP (5 mg/kg) significantly inhibited pro-inflammatory cytokines such as tumor necrosis factor-alpha, interleukin-2, and CD4 cells. Overall, it can be concluded that a low-dose CSP (5 mg/kg) is an efficient anti-arthritic agent that confers its effects via the cytokine pathway.
Collapse
Affiliation(s)
- Lavanya Ramamoorthi
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (DU), Porur, Chennai, India
| | - Srikanth Jeyabalan
- Department of Pharmacology, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (DU), Porur, Chennai, India
| | - Seethalakshmi Sankar
- Department of Pharmacology, ESIC Medical College & PGIMSR, KK Nagar, Chennai, India
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | | | - Mahendran Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, 71800 Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, School of Medical and Life Sciences, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
25
|
Chirivi M, Abou-Rjeileh U, Gandy J, Parales-Giron J, Panda V, Terrian L, Bhattacharya S, Lock AL, Contreras GA. Chromium and palmitic acid supplementation modulate adipose tissue insulin sensitivity in postpartum dairy cows. J Dairy Sci 2025; 108:1078-1091. [PMID: 39369891 DOI: 10.3168/jds.2024-24972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024]
Abstract
Periparturient dairy cows exhibit intense lipolysis driven by reduced DMI, enhanced energy needs, and the loss of adipose tissue (AT) insulin sensitivity. Extended periods of low insulin sensitivity and negative energy balance induce lipolysis dysregulation, leading to increased disease susceptibility and poor lactation performance. Chromium (Cr) supplementation improves systemic insulin sensitivity, whereas palmitic acid (PA) increases energy availability for milk production. However, the effect of supplementing Cr and PA alone or in combination on insulin sensitivity in AT is unknown. A total of 32 multiparous cows were used in a randomized complete block design experiment and randomly assigned to one of 4 diets fed from 1 to 24 DIM: a control diet with no supplementation (CON, n = 8); the Cr diet (Cr propionate at 0.45 mg/kg Cr/kg DM, n = 8); the PA diet (1.5% DM, n = 8); or Cr+PA (n = 8). Plasma samples were collected at -13 ± 5.1 d prepartum (PreP), and at 14.4 ± 1.9 d (PP1) and 21 ± 1.9 d (PP2) after calving for quantification of albumin, BHB, BUN, calcium, cholesterol, glucose, nonesterified fatty acids (NEFA), total protein, iron, transferrin, triglycerides, and oxylipids. Subcutaneous AT (SCAT) explants were collected at PreP, PP1, and PP2 and incubated in the presence of the lipolytic agent isoproterenol (ISO = 1 µM, BAS = 0 µM) for 3 h. The antilipolytic effect of insulin (1 µL/L) on SCAT explants was evaluated during ISO stimulation (ISO+INS). Lipolysis was quantified by glycerol release in the medium (nmol glycerol/mg AT). Macrophage infiltration and adipocyte size were measured using hematoxylin and eosin-stained AT sections and immunohistochemistry. The Cr diet tended to reduce postpartum NEFA concentrations when compared with CON, PA, and Cr+PA. Likewise, Cr increased the percentage of large adipocytes (>9,000 µm2) postpartum compared with other diets. In line with higher lipid content, Cr-fed cows had higher ex vivo BAS lipolysis at PP2 when compared with PA and Cr+PA. Isoproterenol induced higher lipolysis at PP1 and PP2, but it was not affected by Cr and PA. The ISO+INS treatment reduced lipolysis by 29.91% ± 11% in Cr compared with ISO. In contrast, ISO+INS did not affect ISO lipolysis in CON, PA, and Cr+PA. Plasma transferrin was reduced by Cr. At PP2, PA cows had 3.3-fold higher macrophage infiltration in SCAT when compared with CON and Cr. Plasma 9-hydroxyoctadecadienoic acid (HODE) and 9-oxo-octadecadienoic acid (oxoODE) were increased by Cr+PA. Palmitic acid increased plasma 13-oxoODE and Cr increased the ratio of 13-HODE to 13-oxoODE. Palmitic acid increased 5-iso prostaglandin F2α-VI. Our results demonstrate that supplementing Cr during the immediate postpartum enhances SCAT insulin sensitivity and lipid accumulation. Further studies should determine the effects and mechanisms of action of Cr and PA on AT lipogenesis, adipogenesis, and their impact on lactation performance.
Collapse
Affiliation(s)
- Miguel Chirivi
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Ursula Abou-Rjeileh
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Jeff Gandy
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Jair Parales-Giron
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824
| | - Vishal Panda
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824; Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824
| | - Leah Terrian
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824; Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824
| | - Sudin Bhattacharya
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824; Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824
| | - Adam L Lock
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824.
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824.
| |
Collapse
|
26
|
Cavaillon JM, Chaudry IH. Facing stress and inflammation: From the cell to the planet. World J Exp Med 2024; 14:96422. [PMID: 39713080 PMCID: PMC11551703 DOI: 10.5493/wjem.v14.i4.96422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 10/31/2024] Open
Abstract
As identified in 1936 by Hans Selye, stress is shaping diseases through the induction of inflammation. But inflammation display some yin yang properties. On one hand inflammation is merging with the innate immune response aimed to fight infectious or sterile insults, on the other hand inflammation favors chronic physical or psychological disorders. Nature has equipped the cells, the organs, and the individuals with mediators and mechanisms that allow them to deal with stress, and even a good stress (eustress) has been associated with homeostasis. Likewise, societies and the planet are exposed to stressful settings, but wars and global warming suggest that the regulatory mechanisms are poorly efficient. In this review we list some inducers of the physiological stress, psychologic stress, societal stress, and planetary stress, and mention some of the great number of parameters which affect and modulate the response to stress and render it different from an individual to another, from the cellular level to the societal one. The cell, the organ, the individual, the society, and the planet share many stressors of which the consequences are extremely interconnected ending in the domino effect and the butterfly effect.
Collapse
Affiliation(s)
| | - Irshad H Chaudry
- Department of Surgery, University of Alabama Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
27
|
Xu J, Chang L, Xiong Y, Peng Q. Chitosan-Based Hydrogels as Antibacterial/Antioxidant/Anti-Inflammation Multifunctional Dressings for Chronic Wound Healing. Adv Healthc Mater 2024; 13:e2401490. [PMID: 39036852 DOI: 10.1002/adhm.202401490] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/25/2024] [Indexed: 07/23/2024]
Abstract
Due to repeated microbial infection, persistent inflammation, excessive oxidative stress, and cell dysfunction, chronic wounds are difficult to heal, posing a serious threat to public health. Therefore, developing multifunctional wound dressings that can regulate the complex microenvironment of chronic wounds and enhance cellular function holds great significance. Recently, chitosan has emerged as a promising biopolymer for wound healing due to its excellent biocompatibility, biodegradability, and versatile bioactivity. The aim of this review is to provide a comprehensive understanding of the mechanisms of delayed chronic wound healing and discuss the healing-promoting properties of chitosan and its derivatives, such as good biocompatibility, antibacterial activity, hemostatic capacity, and the ability to promote tissue regeneration. On this basis, the potential applications of chitosan-based hydrogels are summarized in chronic wound healing, including providing a suitable microenvironment, eliminating bacterial infections, promoting hemostasis, inhibiting chronic inflammation, alleviating oxidative stress, and promoting tissue regeneration. In addition, the concerns and perspectives for the clinical application of chitosan-based hydrogels are also discussed.
Collapse
Affiliation(s)
- Jingchen Xu
- Department of Dental Medical Center, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Lili Chang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuhuan Xiong
- Department of Stomatology, The First People's Hospital of Longquanyi District, Chengdu, Sichuan, 610100, China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
28
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024; 42:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [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: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
29
|
Lacina L, Kolář M, Pfeiferová L, Gál P, Smetana K. Wound healing: insights into autoimmunity, ageing, and cancer ecosystems through inflammation and IL-6 modulation. Front Immunol 2024; 15:1403570. [PMID: 39676864 PMCID: PMC11638159 DOI: 10.3389/fimmu.2024.1403570] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/30/2024] [Indexed: 12/17/2024] Open
Abstract
Wound healing represents a complex and evolutionarily conserved process across vertebrates, encompassing a series of life-rescuing events. The healing process runs in three main phases: inflammation, proliferation, and maturation/remodelling. While acute inflammation is indispensable for cleansing the wound, removing infection, and eliminating dead tissue characterised by the prevalence of neutrophils, the proliferation phase is characterised by transition into the inflammatory cell profile, shifting towards the prevalence of macrophages. The proliferation phase involves development of granulation tissue, comprising fibroblasts, activated myofibroblasts, and inflammatory and endothelial cells. Communication among these cellular components occurs through intercellular contacts, extracellular matrix secretion, as well as paracrine production of bioactive factors and proteolytic enzymes. The proliferation phase of healing is intricately regulated by inflammation, particularly interleukin-6. Prolonged inflammation results in dysregulations during the granulation tissue formation and may lead to the development of chronic wounds or hypertrophic/keloid scars. Notably, pathological processes such as autoimmune chronic inflammation, organ fibrosis, the tumour microenvironment, and impaired repair following viral infections notably share morphological and functional similarities with granulation tissue. Consequently, wound healing emerges as a prototype for understanding these diverse pathological processes. The prospect of gaining a comprehensive understanding of wound healing holds the potential to furnish fundamental insights into modulation of the intricate dialogue between cancer cells and non-cancer cells within the cancer ecosystem. This knowledge may pave the way for innovative approaches to cancer diagnostics, disease monitoring, and anticancer therapy.
Collapse
Affiliation(s)
- Lukáš Lacina
- Institute of Anatomy, First Faculty of Medicine, Charles, University, Prague, Czechia
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Lucie Pfeiferová
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Peter Gál
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc., Košice, Slovakia
- Prague Burn Centre, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czechia
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Karel Smetana
- Institute of Anatomy, First Faculty of Medicine, Charles, University, Prague, Czechia
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
| |
Collapse
|
30
|
Zhao G, Hu Y. Mechanistic insights into intrauterine adhesions. Semin Immunopathol 2024; 47:3. [PMID: 39613882 DOI: 10.1007/s00281-024-01030-9] [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: 09/16/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024]
Abstract
Intrauterine adhesions (IUA), also known as Asherman's syndrome, arise from damage to the basal layer of the endometrium, frequently caused by intrauterine interventions. This damage leads to nonregenerative healing of endometrium resulting in replacement by fibrous connective tissue, which bring about the adherence of opposing endometrium to render the uterine cavity and/or cervical canal partially or completely obliterated. IUA is a common cause of the refractory uterine infertility. Hysteroscopy is the gold standard for diagnosis of IUA. However, the method of accurately predicting the likelihood of achieving a live birth in the future remains established. Classical treatments have shown limited success, particularly in severe cases. Therefore, utilizing new research methods to deepen the understanding of the pathogenesis of IUA will facilitate the new treatment approaches to be found. In this article we briefly described the advances in the pathogenesis of IUA, with focus on inflammation and parenchymal cellular homeostasis disruption, defects in autophagy and the role of ferroptosis, and we also outlined the progress in IUA therapy.
Collapse
Affiliation(s)
- Guangfeng Zhao
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yali Hu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
31
|
Hutcherson C, Luke B, Khader K, Dhaher YY. Unraveling the complex interplay of sex, endocrinology, and inflammation in post-Injury articular cartilage breakdown through in silico modeling. Sci Rep 2024; 14:28654. [PMID: 39562596 PMCID: PMC11576913 DOI: 10.1038/s41598-024-77730-x] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/21/2024] Open
Abstract
The onset of degenerative joint diseases such as post-traumatic osteoarthritis (PTOA) are associated with joint injury, biomechanical changes, and synovial biochemical anomalies. Sex and reproductive endocrinology have been emerging as potential risk factors, with epidemiological evidence revealing that female's exhibit higher PTOA risk and poorer outcomes post-injury compared to males. Sex hormones, including estradiol, progesterone, and testosterone, have been shown to regulate inflammatory signaling in immune and synovial cells, yet their collective impact on injury-induced joint inflammation and catabolism is poorly understood. Using an in silico kinetic model, we investigated the effects of sex-specific endocrine states on post-injury mechanisms in the human synovial joint. Our model results reveal that heightened estradiol levels in pre-menopausal females during the peri-ovulatory phase increase interleukin (IL)-1β expression and suppress IL-10 expression within the synovium after a simulated injury. Conversely, elevated testosterone levels in males decrease post-injury IL-1β, tumor necrosis factor alpha (TNF)-α, and stromelysin (MMP)-3 expression while increasing IL-10 production compared to females. Gaining insight into the effects of sex hormones on injury-induced inflammation and cartilage degradation provides a basis for designing future experimental and clinical studies to explore their effects on the synovial system, with a particular focus on the female sex.
Collapse
Affiliation(s)
- C Hutcherson
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - B Luke
- Department of Mechanical Engineering, Valparaiso University, Valparaiso, IN, USA
| | - K Khader
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Y Y Dhaher
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern, Dallas, TX, USA.
- Department of Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
32
|
Li Y, Chen W, Koo S, Liu H, Saiding Q, Xie A, Kong N, Cao Y, Abdi R, Serhan CN, Tao W. Innate immunity-modulating nanobiomaterials for controlling inflammation resolution. MATTER 2024; 7:3811-3844. [PMID: 40123651 PMCID: PMC11925551 DOI: 10.1016/j.matt.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The acute inflammatory response is an inherent protective mechanism, its unsuccessful resolution can contribute to disease pathogenesis and potentially lead to death. Innate immune cells are the first line of host defenders and play a substantial role in inflammation initiation, amplification, resolution, or subsequent disease progression. As the resolution of inflammation is an active and highly regulated process, modulating innate immune cells, including neutrophils, monocytes and macrophages, and endothelial cells, and their interactions offer opportunities to control excessive inflammation. Nanobiomaterials have shown superior therapeutic potential in inflammation-related diseases by manipulating inflammatory responses because nanobiomaterials can target and interact with innate immune cells. Versatile nanobiomaterials can be designed for targeted modulation of specific innate immune responses. Nanopro-resolving medicines have been prepared both with pro-resolving lipid mediators and peptides each demonstrated to active resolution of inflammation in animal disease models. Here, we review innovative nanobiomaterials for modulating innate immunity and alleviating inflammation. We summarise the strategies converging the design of nanobiomaterials and the nano-bio interaction in modulating innate immune profiles and propelling the advancement of nanobiomaterials for inflammatory disease treatments. We also propose the future perspectives and translational challenges of nanobiomaterials that need to be overcome in this swiftly rising field.
Collapse
Affiliation(s)
- Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haijun Liu
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angel Xie
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
33
|
Schoberleitner I, Lackner M, Coraça-Huber DC, Augustin A, Imsirovic A, Sigl S, Wolfram D. SMI-Capsular Fibrosis and Biofilm Dynamics: Molecular Mechanisms, Clinical Implications, and Antimicrobial Approaches. Int J Mol Sci 2024; 25:11675. [PMID: 39519227 PMCID: PMC11546664 DOI: 10.3390/ijms252111675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Silicone mammary implants (SMIs) frequently result in capsular fibrosis, which is marked by the overproduction of fibrous tissue surrounding the implant. This review provides a detailed examination of the molecular and immunological mechanisms driving capsular fibrosis, focusing on the role of foreign body responses (FBRs) and microbial biofilm formation. We investigate how microbial adhesion to implant surfaces and biofilm development contribute to persistent inflammation and fibrotic responses. The review critically evaluates antimicrobial strategies, including preoperative antiseptic protocols and antimicrobial-impregnated materials, designed to mitigate infection and biofilm-related complications. Additionally, advancements in material science, such as surface modifications and antibiotic-impregnated meshes, are discussed for their potential to reduce capsular fibrosis and prevent contracture of the capsule. By integrating molecular insights with clinical applications, this review aims to elucidate the current understanding of SMI-related fibrotic responses and highlight knowledge gaps. The synthesis of these findings aims to guide future research directions of improved antimicrobial interventions and implant materials, ultimately advancing the management of capsular fibrosis and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ines Schoberleitner
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Débora C. Coraça-Huber
- BIOFILM Lab, Department of Orthopedics and Traumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Angela Augustin
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Anja Imsirovic
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
34
|
Astrab LR, Skelton ML, Caliari SR. Direct M2 macrophage co-culture overrides viscoelastic hydrogel mechanics to promote fibroblast activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618034. [PMID: 39463963 PMCID: PMC11507682 DOI: 10.1101/2024.10.13.618034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Fibroblast activation drives fibrotic diseases such as pulmonary fibrosis. However, the complex interplay of how tissue mechanics and macrophage signals combine to influence fibroblast activation is not well understood. Here, we use hyaluronic acid hydrogels as a tunable cell culture system to mimic lung tissue stiffness and viscoelasticity. We applied this platform to investigate the influence of macrophage signaling on fibroblast activation. Fibroblasts cultured on stiff (50 kPa) hydrogels mimicking fibrotic tissue exhibit increased activation as measured by spreading as well as type I collagen and cadherin-11 expression compared to fibroblasts cultured on soft (1 kPa) viscoelastic hydrogels mimicking normal tissue. These trends were unchanged in fibroblasts cultured with macrophage-conditioned media. However, fibroblasts directly co-cultured with M2 macrophages show increased activation, even on soft viscoelastic hydrogels that normally suppress activation. Inhibition of interleukin 6 (IL6) signaling does not change activation in fibroblast-only cultures but ameliorates the pro-fibrotic effects of M2 macrophage co-culture. These results underscore the ability of direct M2 macrophage co-culture to override hydrogel viscoelasticity to promote fibroblast activation in an IL6-dependent manner. This work also highlights the utility of using hydrogels to deconstruct complex tissue microenvironments to better understand the interplay between microenvironmental mechanical and cellular cues.
Collapse
Affiliation(s)
- Leilani R. Astrab
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Mackenzie L. Skelton
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Steven R. Caliari
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
35
|
van de Vyver M, Benecke RM, van den Heuvel L, Kruger MJ, Powrie Y, Seedat S, Smith C. Posttraumatic stress disorder is characterized by functional dysregulation of dermal fibroblasts. Biochimie 2024; 225:10-18. [PMID: 38719136 DOI: 10.1016/j.biochi.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/24/2024]
Abstract
Incidence of mental health disorders are rising in modernity, with psychological stress linked to a propensity for developing various chronic diseases due to a relative inability of the body to counter the allostatic load on cellular level. Despite these high rates of comorbidities associated with posttraumatic stress disorder (PTSD), there is still a lack of understanding in terms of the peripheral effects of PTSD on tissue level. Therefore, the purpose of this study was to profile basal dermal fibroblast functional status in PTSD using a wide range of markers involved in the cell-to-cell communication facilitated by fibroblasts. Primary dermal fibroblasts derived from patients diagnosed with PTSD (n = 11) and matched trauma exposed controls (i.e. who did not develop PTSD, n = 10) were cultured using standard techniques. The patients and controls were matched based on age, sex, body-mass index (BMI) and lifestyle. The growth rate, population doubling time, cell surface marker expression (CD31, FNDC5) (flow cytometry), secretome (TIMP-2, MMP-9) (ELISAs), intracellular signalling capacity (Fluo-4 Ca2+ flux) and gene expression (IL-6, IL-10, PTX-3, iNOS, Arg1) were compared between groups. The data illustrated significant PTSD-associated fibroblast conditioning resulting in a blunted signalling capacity. This observation highlights the importance of including tissue-specific investigations in future studies focused on elucidating the association between PTSD and subsequent risk for somatic disease.
Collapse
Affiliation(s)
- M van de Vyver
- Experimental Medicine Research Group, Division Internal Medicine, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, South Africa
| | - R M Benecke
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, South Africa
| | - L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, University of Stellenbosch, Francie van Zijl Drive, Tygerberg, 7505, Cape Town, South Africa; South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - M J Kruger
- Experimental Medicine Research Group, Division Internal Medicine, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, South Africa
| | - Y Powrie
- Experimental Medicine Research Group, Division Internal Medicine, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, South Africa
| | - S Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, University of Stellenbosch, Francie van Zijl Drive, Tygerberg, 7505, Cape Town, South Africa; South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - C Smith
- Experimental Medicine Research Group, Division Internal Medicine, Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, South Africa.
| |
Collapse
|
36
|
Onoi Y, Matsumoto T, Anjiki K, Hayashi S, Nakano N, Kuroda Y, Tsubosaka M, Kamenaga T, Ikuta K, Tachibana S, Suda Y, Wada K, Maeda T, Saitoh A, Hiranaka T, Sobajima S, Iwaguro H, Matsushita T, Kuroda R. Human uncultured adipose-derived stromal vascular fraction shows therapeutic potential against osteoarthritis in immunodeficient rats via direct effects of transplanted M2 macrophages. Stem Cell Res Ther 2024; 15:325. [PMID: 39334434 PMCID: PMC11438128 DOI: 10.1186/s13287-024-03946-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The uncultured adipose-derived stromal vascular fraction (SVF), consisting of adipose-derived stromal cells (ADSCs), M2 macrophages (M2Φ) and others, has shown therapeutic potential against osteoarthritis (OA), however, the mechanisms underlying its therapeutic effects remain unclear. Therefore, this study investigated the effects of the SVF on OA in a human-immunodeficient rat xenotransplantation model. METHODS OA model was induced in the knees of female immunodeficient rats by destabilization of the medial meniscus. Immediately after the surgery, human SVF (1 × 105), ADSCs (1 × 104), or phosphate buffered saline as a control group were transplanted into the knees. At 4 and 8 weeks postoperatively, OA progression and synovitis were analyzed by macroscopic and histological analyses, and the expression of collagen II, SOX9, MMP-13, ADAMTS-5, F4/80, CD86 (M1), CD163 (M2), and human nuclear antigen (hNA) were evaluated immunohistochemically. In vitro, flow cytometry was performed to collect CD163-positive cells as M2Φ from the SVF. Chondrocyte pellets (1 × 105) were co-cultured with SVF (1 × 105), M2Φ (1 × 104), and ADSCs (1 × 104) or alone as a control group, and the pellet size was compared. TGF-β, IL-10 and MMP-13 concentrations in the medium were evaluated using enzyme-linked immunosorbent assay. RESULTS In comparison with the control and ADSC groups, the SVF group showed significantly slower OA progression and less synovitis with higher expression of collagen II and SOX9, lower expression of MMP-13 and ADAMTS-5, and lower F4/80 and M1/M2 ratio in the synovium. Only the SVF group showed partial expression of hNA-, CD163-, and F4/80-positive cells in the rat synovium. In vitro, the SVF, M2Φ, ADSC and control groups, in that order, showed larger pellet sizes, higher TGF-β and IL-10, and lower MMP-13 concentrations. CONCLUSIONS The M2Φ in the transplanted SVF directly affected recipient tissue, enhancing the secretion of growth factors and chondrocyte-protecting cytokines, and partially improving chondrocytes and joint homeostasis. These findings indicate that the SVF is as an effective option for regenerative therapy for OA, with mechanisms different from those of ADSCs.
Collapse
Affiliation(s)
- Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kemmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshihito Suda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kensuke Wada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takuma Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Akira Saitoh
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takafumi Hiranaka
- Department of Orthopaedic Surgery and Joint Surgery Center, Takatsuki General Hospital, Osaka, Japan
| | - Satoshi Sobajima
- Department of Orthopaedic Surgery, Sobajima Clinic, Osaka, Japan
| | - Hideki Iwaguro
- Department of Orthopaedic Surgery, Sobajima Clinic, Osaka, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
37
|
Tang M, Shen L, Tang M, Liu L, Rao Z, Wang Z, Wang Y, Yin S, Li S, Xu G, Zhang K. Human mesenchymal stromal cells ameliorate cisplatin-induced acute and chronic kidney injury via TSG-6. Stem Cells 2024; 42:848-859. [PMID: 38804841 DOI: 10.1093/stmcls/sxae037] [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: 03/25/2023] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Cisplatin is widely used in tumor chemotherapy, but nephrotoxicity is an unavoidable side effect of cisplatin. Several studies have demonstrated that mesenchymal stromal cells (MSCs) ameliorate cisplatin-induced kidney injury, but the underlying mechanisms are unknown. In this study, the cisplatin-induced kidney injury mouse model was established by subjecting a single intraperitoneal injection with cisplatin. One hour before cisplatin injection, the mice received human bone marrow MSCs (hBM-MSCs) with or without siRNA-transfection, recombinant human tumor necrosis factor-α-stimulated gene/protein 6 (rhTSG-6), or PBS through the tail vein. In addition, cisplatin-stimulated HK-2 cells were treated with hBM-MSCs or rhTSG-6. Human BM-MSCs treatment remarkably ameliorated cisplatin-induced acute and chronic kidney injury, as evidenced by significant reductions in serum creatinine (Scr), blood urea nitrogen, tubular injury, collagen deposition, α-smooth muscle actin accumulation, as well as inflammatory responses, and by remarkable increased anti-inflammatory factor expression and Treg cells infiltration in renal tissues. Furthermore, we found that only a few hBM-MSCs engrafted into damaged kidney and that the level of human TSG-6 in the serum of mice increased significantly following hBM-MSCs administration. Moreover, hBM-MSCs significantly increased the viability of damaged HK-2 cells and decreased the levels of inflammatory cytokines in the culture supernatant. However, the knockdown of the TSG-6 gene in hBM-MSCs significantly attenuated their beneficial effects in vivo and in vitro. On the contrary, treated with rhTSG-6 achieved similar beneficial effects of hBM-MSCs. Our results indicate that systemic administration of hBM-MSCs alleviates cisplatin-induced acute and chronic kidney injury in part by paracrine TSG-6 secretion.
Collapse
Affiliation(s)
- Ming Tang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Linguo Shen
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Maozhi Tang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Ling Liu
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Zhengsheng Rao
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Zhilin Wang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Yadi Wang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Supei Yin
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Shujing Li
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Guilian Xu
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Keqin Zhang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| |
Collapse
|
38
|
Zhao Y, Zhang X, Zhou X, Chen B, Duan X. IL-22/IL-22RA1 Promotes Human Tenon's Capsule Fibroblasts Proliferation and Regulates Fibrosis Through STAT3 Signaling Pathway. J Ocul Pharmacol Ther 2024; 40:435-444. [PMID: 39046932 DOI: 10.1089/jop.2023.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
Purpose: Although it is now understood that most antiglaucoma surgeries fail because of scarring of the filtering tract, the underlying mechanism remains to be elucidated. The present study investigated the mechanism by which the interleukin (IL)-22/IL-22 receptor alpha 1 (IL-22RA1) signaling pathway regulates scar formation in glaucoma patients. Method: A total of 31 glaucoma patients who underwent trabeculectomy surgery with uncontrollable intraocular pressure because of scarring and 19 strabismus patients as the control patient group were included in the present study. ELISA was performed to measure the content of IL-22 in peripheral blood. Serum from patients was used to incubate human Tenon's capsule fibroblasts (HTFs) cells and IL-22 antibody rescued the effect of IL-22 on the biological functions. qPCR and Western blot were performed to determine IL-22RA1 mRNA and protein expression levels. Flow cytometry was performed to assess the cell cycle distribution and the Cell Counting Kit-8 assay was used to analyze cell proliferation. Results: The ELISA assay revealed that the serum IL-22 level of glaucoma patients was significantly higher than the healthy group (29.80 ± 5.1 ng/µL vs. 5.21 ± 0.9 ng/µL). After incubation with patient serum, the proliferation and activation of human Tenon fibroblasts (HTFs) were promoted. IL-22 mediated the biological function of HTFs via directly binding IL-22RA1. Moreover, transfection of the siR-IL-22RA1 or IL-22RA1 gene resulted in significant antifibrosis or profibrosis in HTFs. When a signal transducer and activator of transcription (STAT) 3 inhibitor (BAY) was introduced to the IL-22RA1 overexpression group, IL-22-induced proliferation was reduced in HTFs. Additionally, glaucoma patients had increased levels of IL-22 expression following surgery. Conclusions: The IL-22/IL-22RA1/STAT3 signaling pathway promoted fibroblast cell proliferation and alpha-smooth muscle actin, potentially regulating fibrosis in glaucoma filtration tracts. Our results provide hitherto undocumented insights into the pathophysiology of postoperative scarring.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Glaucoma, Changsha Aier Eye Hospital, Changsha, China
- Department of Ophthalmology, The Second Xiangya Hospital, Changsha, China
| | - Xinyue Zhang
- Department of Glaucoma, Changsha Aier Eye Hospital, Changsha, China
- Department of Ophthalmology, The Second Xiangya Hospital, Changsha, China
| | - Xiaoyu Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Changsha, China
- Aier Glaucoma Institute, Aier Eye Hospital Group, Changsha, China
| | - Baihua Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Changsha, China
| | - Xuanchu Duan
- Department of Glaucoma, Changsha Aier Eye Hospital, Changsha, China
- Aier Glaucoma Institute, Aier Eye Hospital Group, Changsha, China
| |
Collapse
|
39
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
40
|
Liao T, Shi L, He C, Liu D, Wei Y, Ma Z, Wang P, Mao J, Wu P. Suppression of NUPR1 in fibroblast-like synoviocytes reduces synovial fibrosis via the Smad3 pathway. J Transl Med 2024; 22:715. [PMID: 39090667 PMCID: PMC11295884 DOI: 10.1186/s12967-024-05540-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Synovial fibrosis is a common complication of knee osteoarthritis (KOA), a pathological process characterized by myofibroblast activation and excessive extracellular matrix (ECM) deposition. Fibroblast-like synoviocytes (FLSs) are implicated in KOA pathogenesis, contributing to synovial fibrosis through diverse mechanisms. Nuclear protein 1 (NUPR1) is a recently identified transcription factor with crucial roles in various fibrotic diseases. However, its molecular determinants in KOA synovial fibrosis remain unknown. This study aims to investigate the role of NUPR1 in KOA synovial fibrosis through in vivo and in vitro experiments. METHODS We examined NUPR1 expression in the murine synovium and determined the impact of NUPR1 on synovial fibrosis by knockdown models in the destabilization of the medial meniscus (DMM)-induced KOA mouse model. TGF-β was employed to induce fibrotic response and myofibroblast activation in mouse FLSs, and the role and molecular mechanisms in synovial fibrosis were evaluated under conditions of NUPR1 downexpression. Additionally, the pharmacological effect of NUPR1 inhibitor in synovial fibrosis was assessed using a surgically induced mouse KOA model. RESULTS We found that NUPR1 expression increased in the murine synovium after DMM surgical operation. The adeno-associated virus (AAV)-NUPR1 shRNA promoted NUPR1 deficiency, attenuating synovial fibrosis, inhibiting synovial hyperplasia, and significantly reducing the expression of pro-fibrotic molecules. Moreover, the lentivirus-mediated NUPR1 deficiency alleviated synoviocyte proliferation and inhibited fibroblast to myofibroblast transition. It also decreased the expression of fibrosis markers α-SMA, COL1A1, CTGF, Vimentin and promoted the activation of the SMAD family member 3 (SMAD3) pathway. Importantly, trifluoperazine (TFP), a NUPR1 inhibitor, attenuated synovial fibrosis in DMM mice. CONCLUSIONS These findings indicate that NUPR1 is an antifibrotic modulator in KOA, and its effect on anti-synovial fibrosis is partially mediated by SMAD3 signaling. This study reveals a promising target for developing novel antifibrotic treatment.
Collapse
Affiliation(s)
- Taiyang Liao
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Lei Shi
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Chenglong He
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Deren Liu
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yibao Wei
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zhenyuan Ma
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Peimin Wang
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jun Mao
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| | - Peng Wu
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
41
|
Huyut Z, Yildizhan K, Altındağ F. The effects of berberine and curcumin on cardiac, lipid profile and fibrosis markers in cyclophosphamide-induced cardiac damage: The role of the TRPM2 channel. J Biochem Mol Toxicol 2024; 38:e23783. [PMID: 39056209 DOI: 10.1002/jbt.23783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/02/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Cyclophosphamide (CYP) is widely used to treat various types of cancer. In addition to the therapeutic properties of this drug, unfortunately, its side effects are still not fully understood. This study investigated the protective effect of curcumin (CURC) and berberine (BER) on CYP-induced cardiac damage. Thirty-six male rats were equally divided into the control, dimethyl sulfoxide (DMSO), CYP, CYP + CURC, CYP + BER and CYP + BER + CURC groups. Troponin-I, Creatine kinase-myocardial band (CK-MB), total cholesterol, triglyceride levels in serum samples, and reactive oxygen species (ROS), poly(ADP-ribose) polymerase-1 (PARP-1), and transient receptor potential melastatin 2 (TRPM2) channel levels in heart tissue were measured using an enzyme-linked immunoassay (ELISA) kit. In addition, histopathological examination and immunohistochemical investigation of the TRPM2 channel, fibroblast specific protein-1 (FSP1), transforming growth factor-beta- 1 (TGF-β1) and α-smooth muscle actin (α-SMA) expressions were determined in heart tissue. The CYP group's troponin-I, total cholesterol, triglyceride, CK-MB, ROS, PARP-1 and TRPM2 channel levels were higher than in the other groups in the ELISA measurements (p < 0.05). In contrast, these parameters in the group treated with CURC and BER together with CYP were lower than in the CYP group (p < 0.05). Additionally, CUR and BER reduced CYP-induced pathological damage, TRPM2, FSP1, TGF-β1 and α-SMA expressions. The data showed that CYP administration can cause cardiac damage by increasing the TRPM2 channel, TGF-β1, FSP1 and α-SMA expression levels. Therefore, we concluded that CURC and BER administration following CYP application may be used as therapeutic agents to prevent CYP-induced cardiac damage.
Collapse
Affiliation(s)
- Zübeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Kenan Yildizhan
- Department of Biophysics, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Fikret Altındağ
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
42
|
Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol 2024; 25:617-638. [PMID: 38589640 DOI: 10.1038/s41580-024-00716-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
The term 'fibroblast' often serves as a catch-all for a diverse array of mesenchymal cells, including perivascular cells, stromal progenitor cells and bona fide fibroblasts. Although phenotypically similar, these subpopulations are functionally distinct, maintaining tissue integrity and serving as local progenitor reservoirs. In response to tissue injury, these cells undergo a dynamic fibroblast-myofibroblast transition, marked by extracellular matrix secretion and contraction of actomyosin-based stress fibres. Importantly, whereas transient activation into myofibroblasts aids in tissue repair, persistent activation triggers pathological fibrosis. In this Review, we discuss the roles of mechanical cues, such as tissue stiffness and strain, alongside cell signalling pathways and extracellular matrix ligands in modulating myofibroblast activation and survival. We also highlight the role of epigenetic modifications and myofibroblast memory in physiological and pathological processes. Finally, we discuss potential strategies for therapeutically interfering with these factors and the associated signal transduction pathways to improve the outcome of dysregulated healing.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andrew E Miller
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Fabio M V Rossi
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada.
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
43
|
Shen Y, Pang L, Jiang C, Jin J, Zhang Y, Xing H, Li J, Wu H, Chen J, Guan M, Zhu T, Gao Z, Cui W, Wang Y. Extracellular Vesicles Functional “Brick‐Cement” Bio‐Integrated System for Annulus Fibrosus Repair. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202400066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Indexed: 01/06/2025]
Abstract
AbstractDue to the deficiency of mechanical supporting after discectomy and weak proliferative capacity of annulus fibrosus (AF) cells, the AF defect repair remains a clinical challenge. Herein, a myofibroblasts derived extracellular vesicles (M‐EVs) functional “brick‐cement” bio‐integrated system (M‐EVs@PGBgel) is developed to repair AF defect. The modified Poly(glycerol‐sebacate) (PGBS), “bio‐brick” layer, exhibited excellent support features on account of its elastomeric mechanical properties. The loaded M‐EVs in the “bio‐cement” layer activated ITGA6/PI3K/AKT pathway, regulated M2 macrophage polarization, thus synergistically promoting AF cell proliferation and migration. The “bio‐cement” layer integrated PGBS and remnant tissue at the defect through the Schiff base reaction and aided M‐EVs’ sustained release. This study demonstrated that M‐EVs@PGBgel significantly improved the disc's biological and mechanical properties in the AF defect microenvironments and promoted AF regeneration in vivo. The M‐EVs@PGBgel shows promise as an effective strategy to simultaneously address the mechanical imbalance and biological disruptions resulting from AF defect.
Collapse
Affiliation(s)
- Yifan Shen
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Libin Pang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Chao Jiang
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Jiale Jin
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Yijian Zhang
- Department of Orthopedics The First Affiliated Hospital of Soochow University Soochow University Suzhou 215006 P. R. China
| | - Hongyuan Xing
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Jiafeng Li
- Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Honghao Wu
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Jingyao Chen
- Core Facilities Zhejiang University School of Medicine Hangzhou 310058 P. R. China
| | - Ming Guan
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Tonghe Zhu
- School of Chemistry and Chemical Engineering Institute for Frontier Medical Technology Shanghai University of Engineering Science Shanghai 201620 P. R. China
| | - Zhongyang Gao
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| | - Wenguo Cui
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Yue Wang
- Spine lab, Department of Orthopedic Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310003 P. R. China
| |
Collapse
|
44
|
Patrick R, Janbandhu V, Tallapragada V, Tan SSM, McKinna EE, Contreras O, Ghazanfar S, Humphreys DT, Murray NJ, Tran YTH, Hume RD, Chong JJH, Harvey RP. Integration mapping of cardiac fibroblast single-cell transcriptomes elucidates cellular principles of fibrosis in diverse pathologies. SCIENCE ADVANCES 2024; 10:eadk8501. [PMID: 38905342 PMCID: PMC11192082 DOI: 10.1126/sciadv.adk8501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Single-cell technology has allowed researchers to probe tissue complexity and dynamics at unprecedented depth in health and disease. However, the generation of high-dimensionality single-cell atlases and virtual three-dimensional tissues requires integrated reference maps that harmonize disparate experimental designs, analytical pipelines, and taxonomies. Here, we present a comprehensive single-cell transcriptome integration map of cardiac fibrosis, which underpins pathophysiology in most cardiovascular diseases. Our findings reveal similarity between cardiac fibroblast (CF) identities and dynamics in ischemic versus pressure overload models of cardiomyopathy. We also describe timelines for commitment of activated CFs to proliferation and myofibrogenesis, profibrotic and antifibrotic polarization of myofibroblasts and matrifibrocytes, and CF conservation across mouse and human healthy and diseased hearts. These insights have the potential to inform knowledge-based therapies.
Collapse
Affiliation(s)
- Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | | | - Shannon S. M. Tan
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Emily E. McKinna
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Osvaldo Contreras
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - David T. Humphreys
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Nicholas J. Murray
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Yen T. H. Tran
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Robert D. Hume
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- School of Medical Science, The University of Sydney, Camperdown, NSW 2006, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Sydney, NSW 2042, Australia
| | - James J. H. Chong
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, NSW 2052, Australia
| |
Collapse
|
45
|
Damerau A, Rosenow E, Alkhoury D, Buttgereit F, Gaber T. Fibrotic pathways and fibroblast-like synoviocyte phenotypes in osteoarthritis. Front Immunol 2024; 15:1385006. [PMID: 38895122 PMCID: PMC11183113 DOI: 10.3389/fimmu.2024.1385006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.
Collapse
Affiliation(s)
- Alexandra Damerau
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Emely Rosenow
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Dana Alkhoury
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| |
Collapse
|
46
|
Salminen A, Kaarniranta K, Kauppinen A. Tissue fibroblasts are versatile immune regulators: An evaluation of their impact on the aging process. Ageing Res Rev 2024; 97:102296. [PMID: 38588867 DOI: 10.1016/j.arr.2024.102296] [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: 12/18/2023] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Fibroblasts are abundant stromal cells which not only control the integrity of extracellular matrix (ECM) but also act as immune regulators. It is known that the structural cells within tissues can establish an organ-specific immunity expressing many immune-related genes and closely interact with immune cells. In fact, fibroblasts can modify their immune properties to display both pro-inflammatory and immunosuppressive activities in a context-dependent manner. After acute insults, fibroblasts promote tissue inflammation although they concurrently recruit immunosuppressive cells to enhance the resolution of inflammation. In chronic pathological states, tissue fibroblasts, especially senescent fibroblasts, can display many pro-inflammatory and immunosuppressive properties and stimulate the activities of different immunosuppressive cells. In return, immunosuppressive cells, such as M2 macrophages and myeloid-derived suppressor cells (MDSC), evoke an excessive conversion of fibroblasts into myofibroblasts, thus aggravating the severity of tissue fibrosis. Single-cell transcriptome studies on fibroblasts isolated from aged tissues have confirmed that tissue fibroblasts express many genes coding for cytokines, chemokines, and complement factors, whereas they lose some fibrogenic properties. The versatile immune properties of fibroblasts and their close cooperation with immune cells indicate that tissue fibroblasts have a crucial role in the aging process and age-related diseases.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio FI-70211, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio FI-70211, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, KYS FI-70029, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, Kuopio FI-70211, Finland
| |
Collapse
|
47
|
Xu S, Jiemy WF, Brouwer E, Burgess JK, Heeringa P, van der Geest KSM, Alba-Rovira R, Corbera-Bellalta M, Boots AH, Cid MC, Sandovici M. Current evidence on the role of fibroblasts in large-vessel vasculitides: From pathogenesis to therapeutics. Autoimmun Rev 2024; 23:103574. [PMID: 38782083 DOI: 10.1016/j.autrev.2024.103574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Large-vessel vasculitides (LVV) comprise a group of chronic inflammatory diseases of the aorta and its major branches. The most common forms of LVV are giant cell arteritis (GCA) and Takayasu arteritis (TAK). Both GCA and TAK are characterized by granulomatous inflammation of the vessel wall accompanied by a maladaptive immune and vascular response that promotes vascular damage and remodeling. The inflammatory process in LVV starts in the adventitia where fibroblasts constitute the dominant cell population. Fibroblasts are traditionally recognized for synthesizing and renewing the extracellular matrix thereby being major players in maintenance of normal tissue architecture and in tissue repair. More recently, fibroblasts have emerged as a highly plastic cell population exerting various functions, including the regulation of local immune processes and organization of immune cells at the site of inflammation through production of cytokines, chemokines and growth factors as well as cell-cell interaction. In this review, we summarize and discuss the current knowledge on fibroblasts in LVV. Furthermore, we identify key questions that need to be addressed to fully understand the role of fibroblasts in the pathogenesis of LVV.
Collapse
Affiliation(s)
- Shuang Xu
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - William F Jiemy
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Elisabeth Brouwer
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Peter Heeringa
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Kornelis S M van der Geest
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Roser Alba-Rovira
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Corbera-Bellalta
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Annemieke H Boots
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Maria C Cid
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Sandovici
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands.
| |
Collapse
|
48
|
Azeredo PDS, Fan D, Murphy EA, Carver WE. Potential of Plant-Derived Compounds in Preventing and Reversing Organ Fibrosis and the Underlying Mechanisms. Cells 2024; 13:421. [PMID: 38474385 PMCID: PMC10930795 DOI: 10.3390/cells13050421] [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: 01/15/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Increased production of extracellular matrix is a necessary response to tissue damage and stress. In a normal healing process, the increase in extracellular matrix is transient. In some instances; however, the increase in extracellular matrix can persist as fibrosis, leading to deleterious alterations in organ structure, biomechanical properties, and function. Indeed, fibrosis is now appreciated to be an important cause of mortality and morbidity. Extensive research has illustrated that fibrosis can be slowed, arrested or even reversed; however, few drugs have been approved specifically for anti-fibrotic treatment. This is in part due to the complex pathways responsible for fibrogenesis and the undesirable side effects of drugs targeting these pathways. Natural products have been utilized for thousands of years as a major component of traditional medicine and currently account for almost one-third of drugs used clinically worldwide. A variety of plant-derived compounds have been demonstrated to have preventative or even reversal effects on fibrosis. This review will discuss the effects and the underlying mechanisms of some of the major plant-derived compounds that have been identified to impact fibrosis.
Collapse
Affiliation(s)
- Patrícia dos Santos Azeredo
- Laboratory of Atherosclerosis, Thrombosis and Cell Therapy, Institute of Biology, State University of Campinas—UNICAMP Campinas, Campinas 13083-970, Brazil;
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| | - E. Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| | - Wayne E. Carver
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| |
Collapse
|
49
|
Huang Y, Liu R, Meng T, Zhang B, Ma J, Liu X. The TGFβ1/SMADs/Snail1 signaling axis mediates pericyte-derived fibrous scar formation after spinal cord injury. Int Immunopharmacol 2024; 128:111482. [PMID: 38237223 DOI: 10.1016/j.intimp.2023.111482] [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: 09/27/2023] [Revised: 12/20/2023] [Accepted: 12/31/2023] [Indexed: 02/08/2024]
Abstract
AIMS The deposition of fibrous scars after spinal cord injury (SCI) affects axon regeneration and the recovery of sensorimotor function. It has been reported that microvascular pericytes in the neurovascular unit are the main source of myofibroblasts after SCI, but the specific molecular targets that regulate pericyte participation in the formation of fibrous scars remain to be clarified. METHODS In this study, a rat model of spinal cord dorsal hemisection injury was used. After SCI, epigallocatechin gallate (EGCG) was intraperitoneally injected to block the TGFβ1 signaling pathway or LV-Snail1-shRNA was immediately injected near the core of the injury using a microsyringe to silence Snail1 expression. Western blotting and RT-qPCR were used to analyze protein expression and transcription levels in tissues. Nissl staining and immunofluorescence analysis were used to analyze neuronal cell viability, scar tissue, and axon regeneration after SCI. Finally, the recovery of hind limb function after SCI was evaluated. RESULTS The results showed that targeted inhibition of Snail1 could block TGFβ1-induced pericyte-myofibroblast differentiation in vitro. In vivo experiments showed that timely blockade of Snail1 could reduce fibrous scar deposition after SCI, promote axon regeneration, improve neuronal survival, and facilitate the recovery of lower limb motor function. CONCLUSION In summary, Snail1 promotes the deposition of fibrous scars and inhibits axonal regeneration after SCI by inducing the differentiation of pericytes into myofibroblasts. Snail1 may be a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yan Huang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, People 's Republic of China
| | - Renzhong Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, People 's Republic of China
| | - Tingyang Meng
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, People 's Republic of China
| | - Bin Zhang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, People 's Republic of China
| | - Jingxing Ma
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, People 's Republic of China.
| | - Xuqiang Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, People 's Republic of China.
| |
Collapse
|
50
|
Wang X, Liu C, Li X, Shen T, Lian J, Shi J, Jiang Z, Qiu G, Wang Y, Meng E, Wei G. A novel electrospun polylactic acid silkworm fibroin mesh for abdominal wall hernia repair. Mater Today Bio 2024; 24:100915. [PMID: 38188648 PMCID: PMC10767193 DOI: 10.1016/j.mtbio.2023.100915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024] Open
Abstract
Objective Abdominal wall hernias are common abdominal diseases, and effective hernia repair is challenging. In clinical practice, synthetic meshes are widely applied for repairing abdominal wall hernias. However, postoperative complications, such as inflammation and adhesion, are prevalent. Although biological meshes can solve this problem to a certain extent, they face the problems of heterogeneity, rapid degradation rate, ordinary mechanical properties, and high-cost. Here, a novel electrospinning mesh composed of polylactic acid and silk fibroin (PLA-SF) for repairing abdominal wall hernias was manufactured with good physical properties, biocompatibility and low production cost. Materials and methods FTIR and EDS were used to demonstrate that the PLA-SF mesh was successfully synthesized. The physicochemical properties of PLA-SF were detected by swelling experiments and in vitro degradation experiments. The water contact angle reflected the hydrophilicity, and the stress‒strain curve reflected the mechanical properties. A rat abdominal wall hernia model was established to observe degradation, adhesion, and inflammation in vivo. In vitro cell mesh culture experiments were used to detect cytocompatibility and search for affected biochemical pathways. Results The PLA-SF mesh was successfully synthesized and did not swell or degrade over time in vitro. It had a high hydrophilicity and strength. The PLA-SF mesh significantly reduced abdominal inflammation and inhibited adhesion formation in rat models. The in vitro degradation rate of the PLA-SF mesh was slower than that of tissue remodeling. Coculture experiments suggested that the PLA-SF mesh reduced the expression of inflammatory factors secreted by fibroblasts and promoted fibroblast proliferation through the TGF-β1/Smad pathway. Conclusion The PLA-SF mesh had excellent physicochemical properties and biocompatibility, promoted hernia repair of the rat abdominal wall, and reduced postoperative inflammation and adhesion. It is a promising mesh and has potential for clinical application.
Collapse
Affiliation(s)
- Xingjie Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Changjun Liu
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, Hunan, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Tianli Shen
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jie Lian
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jing Shi
- Department of Respiratory and Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Zhengdong Jiang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Guanglin Qiu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yuanbo Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Er Meng
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, Hunan, China
| | - Guangbing Wei
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| |
Collapse
|