1
|
Pan Y, Li L, Cao N, Liao J, Chen H, Zhang M. Advanced nano delivery system for stem cell therapy for Alzheimer's disease. Biomaterials 2025; 314:122852. [PMID: 39357149 DOI: 10.1016/j.biomaterials.2024.122852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's Disease (AD) represents one of the most significant neurodegenerative challenges of our time, with its increasing prevalence and the lack of curative treatments underscoring an urgent need for innovative therapeutic strategies. Stem cells (SCs) therapy emerges as a promising frontier, offering potential mechanisms for neuroregeneration, neuroprotection, and disease modification in AD. This article provides a comprehensive overview of the current landscape and future directions of stem cell therapy in AD treatment, addressing key aspects such as stem cell migration, differentiation, paracrine effects, and mitochondrial translocation. Despite the promising therapeutic mechanisms of SCs, translating these findings into clinical applications faces substantial hurdles, including production scalability, quality control, ethical concerns, immunogenicity, and regulatory challenges. Furthermore, we delve into emerging trends in stem cell modification and application, highlighting the roles of genetic engineering, biomaterials, and advanced delivery systems. Potential solutions to overcome translational barriers are discussed, emphasizing the importance of interdisciplinary collaboration, regulatory harmonization, and adaptive clinical trial designs. The article concludes with reflections on the future of stem cell therapy in AD, balancing optimism with a pragmatic recognition of the challenges ahead. As we navigate these complexities, the ultimate goal remains to translate stem cell research into safe, effective, and accessible treatments for AD, heralding a new era in the fight against this devastating disease.
Collapse
Affiliation(s)
- Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| | - Long Li
- Department of Neurosurgery, First Hospital of China Medical University, Liaoning, 110001, China.
| | - Ning Cao
- Army Medical University, Chongqing, 400000, China
| | - Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Huiyue Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, 110001, China.
| | - Meng Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| |
Collapse
|
2
|
Huang L, Yang X, Feng Y, Huang HX, Hu JQ, Yan PY, Pan HD, Xie Y. ShaShen-MaiDong decoction attenuates bleomycin-induced pulmonary fibrosis by inhibiting TGF-β/smad3, AKT/MAPK, and YAP/TAZ pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118755. [PMID: 39209002 DOI: 10.1016/j.jep.2024.118755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary fibrosis (PF) is progressive and terminal lung disease, which is also the most common sequelae of Corona Virus Disease (2019) (COVID-19) survivors. Unfortunately, there is currently no cure for PF. ShaShen-MaiDong decoction (SMT), a traditional Chinese medicine, has been employed in treating various lung diseases, which may offer potential therapeutic benefits for PF. AIM OF THE STUDY To investigate the antifibrotic efficacy of SMT and its major active ingredients as well as the underlying mechanisms for treating PF. MATERIALS AND METHODS Fist, we build the UPLC-MS based qualitative and quantitative profiling for the quality control of SMT. Then, the antifibrotic efficacy of SMT was investigated in bleomycin (BLM)-induced PF mice model. Network pharmacology was used to predict the mechanism and active components of SMT for the treatment of PF, which was further verified in vitro and in vivo. RESULTS SMT improved the weight loss and attenuated hydroxyproline, inflammatory cytokines, and collagen deposition in BLM-induced PF mice model in a dose-dependent manner. Mechanistically, as predicted by network pharmacology analysis, SMT and its active compounds (kaempferol, quercetin, and isorhamnetin) regulated the mitogen-activated protein kinase (MAPK) signaling pathways, TGF-β/Smad signaling pathway, and YAP/TAZ signaling pathway, which was further verified in the PF mice and TGF-β-induced A549 cell model. Moreover, SMT balanced the proportions of increased CD4+ and decreased CD8+ T cells in the peripheral blood of PF mice model. CONCLUSIONS Considering the high mortality and complex pathogenesis of fibrotic diseases, our results provide novel evidence that SMT would be beneficial for pulmonary fibrosis therapy by modulating MAPK, TGF-β/Smad, and YAP/TAZ signaling pathways at same time.
Collapse
Affiliation(s)
- Li Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, China
| | - Xi Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 650201 Kunming, China
| | - Yi Feng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hua-Xue Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, China
| | - Jia-Qin Hu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Pei-Yu Yan
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, China.
| | - Hu-Dan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Ying Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Mao Q, Zhang X, Yang J, Kong Q, Cheng H, Yu W, Cao X, Li Y, Li C, Liu L, Ding Z. HSPA12A acts as a scaffolding protein to inhibit cardiac fibroblast activation and cardiac fibrosis. J Adv Res 2025; 67:217-229. [PMID: 38219869 DOI: 10.1016/j.jare.2024.01.012] [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: 09/19/2023] [Revised: 12/12/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024] Open
Abstract
INTRODUCTION Cardiac fibrosis is the main driver for adverse remodeling and progressive functional decline in nearly all types of heart disease including myocardial infarction (MI). The activation of cardiac fibroblasts (CF) into myofibroblasts is responsible for cardiac fibrosis. Unfortunately, no ideal approach for controlling CF activation currently exists. OBJECTIVES This study investigated the role of Heat shock protein A12A (HSPA12A), an atypical member of the HSP70 family, in CF activation and MI-induced cardiac fibrosis. METHODS Primary CF and Hspa12a knockout mice were used in the experiments. CF activation was indicated by the upregulation of myofibroblast characters including alpha-Smooth muscle actin (αSMA), Collagen, and Fibronectin. Cardiac fibrosis was illustrated by Masson's trichrome and picrosirius staining. Cardiac function was examined using echocardiography. Glycolytic activity was indicated by levels of extracellular lactate and the related protein expression. Protein stability was examined following cycloheximide and MG132 treatment. Protein-protein interaction was examined by immunoprecipitation-immunoblotting analysis. RESULTS HSPA12A displayed a high expression level in quiescent CF but showed a decreased expression in activated CF, while ablation of HSPA12A in mice promoted CF activation and cardiac fibrosis following MI. HSPA12A overexpression inhibited the activation of primary CF through inhibiting glycolysis, while HSPA12A knockdown showed the opposite effects. Moreover, HSPA12A upregulated the protein expression of transcription factor p53, by which mediated the HSPA12A-induced inhibition of glycolysis and CF activation. Mechanistically, this action of HSPA12A was achieved by acting as a scaffolding protein to bind p53 and ubiquitin specific protease 10 (USP10), thereby promoting the USP10-mediated p53 protein stability and the p53-medicated glycolysis inhibition. CONCLUSION The present study provided clear evidence that HSPA12A is a novel endogenous inhibitor of CF activation and cardiac fibrosis. Targeting HSPA12A in CF could represent a promising strategy for the management of cardiac fibrosis in patients.
Collapse
Affiliation(s)
- Qian Mao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinna Yang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Cheng
- Department of Anesthesiology, The First Affiliated Hospital with Wannan Medical College, Wuhu, China
| | - Wansu Yu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaofei Cao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
4
|
Xu J, Wang Y, Shao Z, Zhou Y, Bin X, Liu L, Huang W, Wang X, Hu Y, Li K. Adipose-derived stem cell exosomes attenuates myofibroblast transformation via inhibiting autophagy through TGF-β/Smad2 axis in oral submucosal fibrosis. J Nanobiotechnology 2024; 22:780. [PMID: 39702233 DOI: 10.1186/s12951-024-03067-2] [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: 10/13/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Oral submucous fibrosis (OSF) is a precancerous condition that poses substantial health risks. OSF is mainly caused by betel nut chewing behavior, but its pathogenesis is still unclear and there is no effective treatment strategy. The transformation of fibroblasts to myofibroblast is the key pathological change in the development of OSF. We isolated fibroblasts from human oral mucosa and induced them into myofibroblasts by arecoline, during which autophagy was significantly activated. Here, we found that adipose-derived stem cell exosomes (ADSCs-EXO) could inhibit autophagy to regulate myofibroblast phenotype, and transcriptome sequencing analysis suggested that this process is closely related to the TGF-β pathway. The interplay between autophagy and TGF-β pathway was examined through modulation the two with autophagy activators and inhibitors, TGF-β receptor activators and inhibitors. Results showed that in vitro, the TGF-β/Smad2 pathway augmented autophagy and promoted myofibroblast transformation. The transcriptome information of ADSCs-EXO showed that it contains a large number of miRNAs. Among them, miR-125a-5p could target Smad2. In vivo, injection of ADSCs-EXO alleviated OSF in mice, during which TGF-β and autophagy signals were inhibited. We suggested that ADSCs-EXO could inhibit myofibroblast transformation via inhibiting autophagy through TGF-β/Smad2 axis in OSF, providing new insights for autophagy-based intervention strategies.
Collapse
Affiliation(s)
- Jinhao Xu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Yujing Wang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Zifei Shao
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Yuxi Zhou
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Xin Bin
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Lian Liu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Weiman Huang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Xidi Wang
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Yanjia Hu
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China
| | - Kun Li
- Department of Oral & Maxillofacial Surgery, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, 410000, China.
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
5
|
Zhang H, Thai PN, Shivnaraine RV, Ren L, Wu X, Siepe DH, Liu Y, Tu C, Shin HS, Caudal A, Mukherjee S, Leitz J, Wen WTL, Liu W, Zhu W, Chiamvimonvat N, Wu JC. Multiscale drug screening for cardiac fibrosis identifies MD2 as a therapeutic target. Cell 2024; 187:7143-7163.e22. [PMID: 39413786 PMCID: PMC11645214 DOI: 10.1016/j.cell.2024.09.034] [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: 12/05/2023] [Revised: 08/13/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
Cardiac fibrosis impairs cardiac function, but no effective clinical therapies exist. To address this unmet need, we employed a high-throughput screening for antifibrotic compounds using human induced pluripotent stem cell (iPSC)-derived cardiac fibroblasts (CFs). Counter-screening of the initial candidates using iPSC-derived cardiomyocytes and iPSC-derived endothelial cells excluded hits with cardiotoxicity. This screening process identified artesunate as the lead compound. Following profibrotic stimuli, artesunate inhibited proliferation, migration, and contraction in human primary CFs, reduced collagen deposition, and improved contractile function in 3D-engineered heart tissues. Artesunate also attenuated cardiac fibrosis and improved cardiac function in heart failure mouse models. Mechanistically, artesunate targeted myeloid differentiation factor 2 (MD2) and inhibited MD2/Toll-like receptor 4 (TLR4) signaling pathway, alleviating fibrotic gene expression in CFs. Our study leverages multiscale drug screening that integrates a human iPSC platform, tissue engineering, animal models, in silico simulations, and multiomics to identify MD2 as a therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Hao Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | | | - Lu Ren
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuekun Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dirk H Siepe
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chengyi Tu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hye Sook Shin
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Jeremy Leitz
- Greenstone Biosciences, Palo Alto, CA 94305, USA
| | - Wilson Tan Lek Wen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wenqiang Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wenjuan Zhu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Department of Basic Medical Sciences and Translational Cardiovascular Research Center, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
Xiang L, Qin Y, Li L, Xiang X, Zhang W, Jiao Q, Shao Y, Huang X, Wu M, Zhou T, Lin Y, Chen Y. Targeting hyperactive mitochondria in activated HSCs and inhibition of liver fibrogenesis in mice using sorafenib complex micelles. Int J Pharm 2024; 669:125058. [PMID: 39653289 DOI: 10.1016/j.ijpharm.2024.125058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
Liver fibrosis is a pathological condition marked by the excessive buildup of extracellular matrix primarily resulting from the transformation of quiescent hepatic stellate cells (HSCs) to myofibroblastic (MF) phenotype and their resultant over-expansion. Activated HSCs completely rely on their hyperactive mitochondria to supply the energy and biomass for their rapid proliferation and collagen secretion, so an intervention targeting their mitochondria can effectively restrict their pathological amplification and contribution to liver fibrosis. Here we tried sorafenib, a drug that plays anticancer roles by inducing the disruption and loss of mitochondrial functions, to reach an antifibrotic goal. And a complex micellar system, VA-PEG-PCL/TPGS (VPP/TPGS), was specifically designed and fabricated to encapsulate and deliver sorafenib selectively to activated HSCs to overcome its application limitations in bioavailability, toxicity and intracellular stay, and eventually maximize its induction of mitochondrial dysfunction and therapeutically antifibrotic efficacy. The prepared sorafenib complex micelles not only exhibited a suitable particle size, uniform morphology, and nice stability, but also performed excellently in the biosafety and HSCs-targetability in vitro and in vivo. In human active HSC cell lines, they markedly attenuated mitochondrial hyperactivity, induced apoptosis, and downregulated fibrosis markers as expected; while in a CCl4-induced murine model of hepatic fibrosis, they effectively restricted the expansion of MF-HSCs, reduced collagen deposition, and promoted the healing of liver damage, showing a good potential in fibrosis curation. Collectively, our VPP/TPGS complex micelles provide an ideal drug delivery platform that has the potential to revolutionize the treatment of liver fibrosis via addressing its cellular and metabolic underpinnings and thus improve patient outcomes.
Collapse
Affiliation(s)
- Li Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China; Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Yuting Qin
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Lei Li
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Xianjing Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Wenhui Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yaru Shao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Xinqiong Huang
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Meichun Wu
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Tianle Zhou
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yukang Lin
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China; Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China; MOE Key Laboratory of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China.
| |
Collapse
|
7
|
Nagalingam RS, Jayousi F, Hamledari H, Dababneh S, Hosseini D, Lindsay C, Klein Geltink R, Lange PF, Dixon IM, Rose RA, Czubryt MP, Tibbits GF. Molecular and metabolomic characterization of hiPSC-derived cardiac fibroblasts transitioning to myofibroblasts. Front Cell Dev Biol 2024; 12:1496884. [PMID: 39698493 PMCID: PMC11653212 DOI: 10.3389/fcell.2024.1496884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Background Mechanical stress and pathological signaling trigger the activation of fibroblasts to myofibroblasts, which impacts extracellular matrix composition, disrupts normal wound healing, and can generate deleterious fibrosis. Myocardial fibrosis independently promotes cardiac arrhythmias, sudden cardiac arrest, and contributes to the severity of heart failure. Fibrosis can also alter cell-to-cell communication and increase myocardial stiffness which eventually may lead to lusitropic and inotropic cardiac dysfunction. Human induced pluripotent stem cell derived cardiac fibroblasts (hiPSC-CFs) have the potential to enhance clinical relevance in precision disease modeling by facilitating the study of patient-specific phenotypes. However, it is unclear whether hiPSC-CFs can be activated to become myofibroblasts akin to primary cells, and the key signaling mechanisms in this process remain unidentified. Objective We aim to explore the notable changes in fibroblast phenotype upon passage-mediated activation of hiPSC-CFs with increased mitochondrial metabolism, like primary cardiac fibroblasts. Methods We activated the hiPSC-CFs with serial passaging from passage 0 to 3 (P0 to P3) and treatment of P0 with TGFβ1. Results Passage-mediated activation of hiPSC-CFs was associated with a gradual induction of genes to initiate the activation of these cells to myofibroblasts, including collagen, periostin, fibronectin, and collagen fiber processing enzymes with concomitant downregulation of cellular proliferation markers. Most importantly, canonical TGFβ1 and Hippo signaling component genes including TAZ were influenced by passaging hiPSC-CFs. Seahorse assay revealed that passaging and TGFβ1 treatment increased mitochondrial respiration, consistent with fibroblast activation requiring increased energy production, whereas treatment with the glutaminolysis inhibitor BPTES completely attenuated this process. Conclusion Our study highlights that the hiPSC-CF passaging enhanced fibroblast activation, activated fibrotic signaling pathways, and enhanced mitochondrial metabolism approximating what has been reported in primary cardiac fibroblasts. Thus, hiPSC-CFs may provide an accurate in vitro preclinical model for the cardiac fibrotic condition, which may facilitate the identification of putative anti-fibrotic therapies, including patient-specific approaches.
Collapse
Affiliation(s)
- Raghu Sundaresan Nagalingam
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Farah Jayousi
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Homa Hamledari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dina Hosseini
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Chloe Lindsay
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Ramon Klein Geltink
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Colombia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Philipp F. Lange
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Colombia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Ian Michael Dixon
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Robert Alan Rose
- Department of Cardiac Sciences, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Michael Paul Czubryt
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Glen Findlay Tibbits
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
8
|
Carter K, Shah E, Waite J, Rana D, Zhao ZQ. Pathophysiology of Angiotensin II-Mediated Hypertension, Cardiac Hypertrophy, and Failure: A Perspective from Macrophages. Cells 2024; 13:2001. [PMID: 39682749 DOI: 10.3390/cells13232001] [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: 10/08/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Heart failure is a complex syndrome characterized by cardiac hypertrophy, fibrosis, and diastolic/systolic dysfunction. These changes share many pathological features with significant inflammatory responses in the myocardium. Among the various regulatory systems that impact on these heterogeneous pathological processes, angiotensin II (Ang II)-activated macrophages play a pivotal role in the induction of subcellular defects and cardiac adverse remodeling during the progression of heart failure. Ang II stimulates macrophages via its AT1 receptor to release oxygen-free radicals, cytokines, chemokines, and other inflammatory mediators in the myocardium, and upregulates the expression of integrin adhesion molecules on both monocytes and endothelial cells, leading to monocyte-endothelial cell-cell interactions. The transendothelial migration of monocyte-derived macrophages exerts significant biological effects on the proliferation of fibroblasts, deposition of extracellular matrix proteins, induction of perivascular/interstitial fibrosis, and development of hypertension, cardiac hypertrophy and heart failure. Inhibition of macrophage activation using Ang II AT1 receptor antagonist or depletion of macrophages from the peripheral circulation has shown significant inhibitory effects on Ang II-induced vascular and myocardial injury. The purpose of this review is to discuss the current understanding in Ang II-induced maladaptive cardiac remodeling and dysfunction, particularly focusing on molecular signaling pathways involved in macrophages-mediated hypertension, cardiac hypertrophy, fibrosis, and failure. In addition, the challenges remained in translating these findings to the treatment of heart failure patients are also addressed.
Collapse
Affiliation(s)
- Kelly Carter
- Cardiovascular Research Laboratory, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Eshan Shah
- Cardiovascular Research Laboratory, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Jessica Waite
- Cardiovascular Research Laboratory, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Dhruv Rana
- Cardiovascular Research Laboratory, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Zhi-Qing Zhao
- Cardiovascular Research Laboratory, Mercer University School of Medicine, Savannah, GA 31404, USA
| |
Collapse
|
9
|
Fleckner M, Döhmen NK, Salz K, Christophers T, Windolf J, Suschek CV, Oezel L. Exposure of Primary Human Skin Fibroblasts to Carbon Dioxide-Containing Solution Significantly Reduces TGF-β-Induced Myofibroblast Differentiation In Vitro. Int J Mol Sci 2024; 25:13013. [PMID: 39684728 DOI: 10.3390/ijms252313013] [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: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Wound healing as a result of a skin injury involves a series of dynamic physiological processes, leading to wound closure, re-epithelialization, and the remodeling of the extracellular matrix (ECM). The primary scar formed by the new ECM never fully regains the original tissue's strength or flexibility. Moreover, in some cases, due to dysregulated fibroblast activity, proliferation, and differentiation, the normal scarring can be replaced by pathological fibrotic tissue, leading to hypertrophic scars or keloids. These disorders can cause significant physical impairment and psychological stress and represent significant challenges in medical management in the wound-healing process. The present study aimed to investigate the therapeutic effects of exogenously applied carbon dioxide (CO2) on fibroblast behavior, focusing on viability, proliferation, migration, and differentiation to myofibroblasts. We found that CO2 exposure for up to 60 min did not significantly affect fibroblast viability, apoptosis rate, or proliferation and migration capacities. However, a notable finding was the significant reduction in α-smooth muscle actin (α-SMA) protein expression, indicative of myofibroblast differentiation inhibition, following CO2 exposure. This effect was specific to CO2 and concentration as well as time-dependent, with longer exposure durations leading to greater reductions in α-SMA expression. Furthermore, the inhibition of myofibroblast differentiation correlated with a statistically significantly reduced glycolytic and mitochondrial energy metabolism, and as a result, with a reduced ATP synthesis rate. This very noticeable decrease in cellular energy levels seemed to be specific to CO2 exposure and could not be observed in the control cultures using nitrogen (N2)-saturated solutions, indicating a unique and hypoxia-independent effect of CO2 on fibroblast metabolism. These findings suggest that exogenously applied CO2 may possess fibroblast differentiation-reducing properties by modulating fibroblast's energy metabolism and could offer new therapeutic options in the prevention of scar and keloid development.
Collapse
Affiliation(s)
- Maxine Fleckner
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Niklas K Döhmen
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Katharina Salz
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Till Christophers
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Joachim Windolf
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Christoph V Suschek
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Lisa Oezel
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| |
Collapse
|
10
|
Lunde IG, Rypdal KB, Van Linthout S, Diez J, González A. Myocardial fibrosis from the perspective of the extracellular matrix: Mechanisms to clinical impact. Matrix Biol 2024; 134:1-22. [PMID: 39214156 DOI: 10.1016/j.matbio.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and constitutes a central pathophysiological process that underlies tissue dysfunction, across organs, in multiple chronic diseases and during aging. Myocardial fibrosis is a key contributor to dysfunction and failure in numerous diseases of the heart and is a strong predictor of poor clinical outcome and mortality. The excess structural and matricellular ECM proteins deposited by cardiac fibroblasts, is found between cardiomyocytes (interstitial fibrosis), in focal areas where cardiomyocytes have died (replacement fibrosis), and around vessels (perivascular fibrosis). Although myocardial fibrosis has important clinical prognostic value, access to cardiac tissue biopsies for histological evaluation is limited. Despite challenges with sensitivity and specificity, cardiac magnetic resonance imaging (CMR) is the most applicable diagnostic tool in the clinic, and the scientific community is currently actively searching for blood biomarkers reflecting myocardial fibrosis, to complement the imaging techniques. The lack of mechanistic insights into specific pro- and anti-fibrotic molecular pathways has hampered the development of effective treatments to prevent or reverse myocardial fibrosis. Development and implementation of anti-fibrotic therapies is expected to improve patient outcomes and is an urgent medical need. Here, we discuss the importance of the ECM in the heart, the central role of fibrosis in heart disease, and mechanistic pathways likely to impact clinical practice with regards to diagnostics of myocardial fibrosis, risk stratification of patients, and anti-fibrotic therapy.
Collapse
Affiliation(s)
- Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway.
| | - Karoline B Rypdal
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
11
|
Wang Y, Zhang J, Shao C. Cytological changes in radiation-induced lung injury. Life Sci 2024; 358:123188. [PMID: 39481833 DOI: 10.1016/j.lfs.2024.123188] [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: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Radiation-induced lung injury (RILI) is a prevalent complication associated with radiotherapy for thoracic tumors. Based on the pathological progression, it can be categorized into two stages: early radiation pneumonitis and late radiation pulmonary fibrosis. The occurrence of RILI not only constrains the therapeutic dose that can be administered to the tumor target area but also significantly impairs patients' health and quality of life, thereby limiting the efficacy and applicability of radiotherapy. To effectively prevent and mitigate the development of RILI, it is crucial to disclose its underlying mechanisms. This review aims to elucidate the specific mechanisms involved in RILI and to examine the roles of various cell types, including lung parenchymal cells and different immune cells. The functions and interactions of lung epithelial cells, pulmonary vascular endothelial cells, a variety of immune cells, and fibroblasts during different stages of inflammation, tissue repair, and fibrosis following radiation-induced lung injury are analyzed. A comprehensive understanding of the dynamic changes in these cellular components is anticipated to offer new strategies for the prevention of RILI.
Collapse
Affiliation(s)
- Yun Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China.
| |
Collapse
|
12
|
Guo Q, Li P, Chen M, Yu Y, Wan Y, Zhang Z, Ren C, Shen L, Liu X, He D, Zhang Y, Wei G, Zhang D. Exosomes From Human Umbilical Cord Stem Cells Suppress Macrophage-to-myofibroblast Transition, Alleviating Renal Fibrosis. Inflammation 2024; 47:2094-2107. [PMID: 38662165 DOI: 10.1007/s10753-024-02027-0] [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: 03/12/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Renal fibrosis, a progressive scarring of the kidney, lacks effective treatment. Human umbilical cord mesenchymal stem cell-derived exosomes (HucMSC-Exos) hold promise for treating kidney diseases due to their anti-inflammatory properties. This study investigates their potential to lessen renal fibrosis by targeting macrophage-to-myofibroblast transformation (MMT), a key driver of fibrosis. We employed a mouse model of unilateral ureteral obstruction (UUO) and cultured cells exposed to transforming growth factor-β (TGF-β) to mimic MMT. HucMSC-Exos were administered to UUO mice, and their effects on kidney function and fibrosis were assessed. Additionally, RNA sequencing and cellular analysis were performed to elucidate the mechanisms by which HucMSC-Exos inhibit MMT. HucMSC-Exos treatment significantly reduced kidney damage and fibrosis in UUO mice. They downregulated markers of fibrosis (Collagen I, vimentin, alpha-smooth muscle actin) and suppressed MMT (α-SMA + F4/80 + cells). Furthermore, ARNTL, a specific molecule, emerged as a potential target of HucMSC-Exos in hindering MMT and consequently preventing fibrosis. HucMSC-Exos effectively lessen renal fibrosis by suppressing MMT, suggesting a novel therapeutic strategy for managing kidney damage and fibrosis.
Collapse
Affiliation(s)
- Qitong Guo
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Ping Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Meiling Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Yihang Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Yonghong Wan
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Zhaoxia Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Chunnian Ren
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Lianju Shen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Xing Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China
| | - Deying Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, 400014, China.
| |
Collapse
|
13
|
Alexandre-Santos B, Reis GDS, Medeiros GR, Stockler-Pinto MB, Oliveira NSC, Miranda-Alves L, Nóbrega ACLD, Magliano DC, Frantz EDC. Bisphenol S exposure induces cardiac remodeling and aggravates high-fat diet-induced cardiac hypertrophy in mice. ENVIRONMENTAL RESEARCH 2024; 261:119781. [PMID: 39142458 DOI: 10.1016/j.envres.2024.119781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Bisphenol S (BPS) is widely used in the manufacture products and increase the risk of cardiovascular diseases. The effect of the association between obesity and BPS on cardiac outcomes is still unknown. Male C57BL/6 mice were divided into standard chow diet (SC; 15 kJ/g), standard chow diet + BPS (SCB), high-fat diet (HF; 21 kJ/g), and high-fat diet + BPS (HFB). Over 12 weeks, the groups were exposed to BPS through drinking water (dose: 25 μg/kg/day) and/or a HF diet. We evaluated: body mass (BM), total cholesterol, systolic blood pressure (SBP), left ventricle (LV) mass, and cardiac remodeling. In the SCB group, BM, total cholesterol, and SBP increase were augmented in relation to the SC group. In the HF and HFB groups, these parameters were higher than in the SC and SCB groups. Cardiac hypertrophy was evidenced by augmented LV mass and wall thickness, and ANP protein expression in all groups in comparison to the SC group. Only the HFB group had a thicker LV wall than SCB and HF groups, and increased cardiomyocyte area when compared with SC and SCB groups. Concerning cardiac fibrosis, SCB, HF, and HFB groups presented higher interstitial collagen area, TGFβ, and α-SMA protein expression than the SC group. Perivascular collagen area was increased only in the HF and HFB groups than SC group. Higher IL-6, TNFα, and CD11c protein expression in all groups than the SC group evidenced inflammation. All groups had elevated CD36 and PPARα protein expression in relation to the SC group, but only HF and HFB groups promoted cardiac steatosis with increased perilipin 5 protein expression than the SC group. BPS exposure alone promoted cardiac remodeling with pathological concentric hypertrophy, fibrosis, and inflammation. Diet-induced remodeling is aggravated when associated with BPS, with marked hypertrophy, alongside fibrosis, inflammation, and lipid accumulation.
Collapse
Affiliation(s)
- Beatriz Alexandre-Santos
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Guilherme Dos Santos Reis
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Gabriela Rodrigues Medeiros
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Milena Barcza Stockler-Pinto
- Research Center on Nutrigenetics and Nutrigenomics, Faculty of Nutrition, Fluminense Federal University, Niteroi, RJ, Brazil
| | | | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology, Institute of Biomedical Science, Federal University of Rio de Janeiro, RJ, Brazil
| | | | - D'Angelo Carlo Magliano
- Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Laboratory of Experimental Endocrinology, Institute of Biomedical Science, Federal University of Rio de Janeiro, RJ, Brazil
| | - Eliete Dalla Corte Frantz
- Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil; Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
14
|
Santos F, Correia M, Dias R, Bola B, Noberini R, Ferreira RS, Trigo D, Domingues P, Teixeira J, Bonaldi T, Oliveira PJ, Bär C, de Jesus BB, Nóbrega-Pereira S. Age-associated metabolic and epigenetic barriers during direct reprogramming of mouse fibroblasts into induced cardiomyocytes. Aging Cell 2024:e14371. [PMID: 39540462 DOI: 10.1111/acel.14371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 08/28/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
Heart disease is the leading cause of mortality in developed countries, and novel regenerative procedures are warranted. Direct cardiac conversion (DCC) of adult fibroblasts can create induced cardiomyocytes (iCMs) for gene and cell-based heart therapy, and in addition to holding great promise, still lacks effectiveness as metabolic and age-associated barriers remain elusive. Here, by employing MGT (Mef2c, Gata4, Tbx5) transduction of mouse embryonic fibroblasts (MEFs) and adult (dermal and cardiac) fibroblasts from animals of different ages, we provide evidence that the direct reprogramming of fibroblasts into iCMs decreases with age. Analyses of histone posttranslational modifications and ChIP-qPCR revealed age-dependent alterations in the epigenetic landscape of DCC. Moreover, DCC is accompanied by profound mitochondrial metabolic adaptations, including a lower abundance of anabolic metabolites, network remodeling, and reliance on mitochondrial respiration. In vitro metabolic modulation and dietary manipulation in vivo improve DCC efficiency and are accompanied by significant alterations in histone marks and mitochondrial homeostasis. Importantly, adult-derived iCMs exhibit increased accumulation of oxidative stress in the mitochondria and activation of mitophagy or dietary lipids; they improve DCC and revert mitochondrial oxidative damage. Our study provides evidence that metaboloepigenetics plays a direct role in cell fate transitions driving DCC, highlighting the potential use of metabolic modulation to improve cardiac regenerative strategies.
Collapse
Affiliation(s)
- Francisco Santos
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Magda Correia
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Rafaela Dias
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Bárbara Bola
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Roberta Noberini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Rita S Ferreira
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Diogo Trigo
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Center, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- LAQV/REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - José Teixeira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Cantanhede, Portugal
| | - Tiziana Bonaldi
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milano, Milan, Italy
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Cantanhede, Portugal
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School (MHH), Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Bruno Bernardes de Jesus
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Sandrina Nóbrega-Pereira
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
15
|
Schoettler FI, Fatehi Hassanabad A, Jadli AS, Patel VB, Fedak PWM. Exploring the role of pericardial miRNAs and exosomes in modulating cardiac fibrosis. Cardiovasc Pathol 2024; 73:107671. [PMID: 38906439 DOI: 10.1016/j.carpath.2024.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/26/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
The potential of the pericardial space as a therapeutic delivery tool for cardiac fibrosis and heart failure (HF) treatment has yet to be elucidated. Recently, miRNAs and exosomes have been discovered to be present in human pericardial fluid (PF). Novel studies have shown characteristic human PF miRNA compositions associated with cardiac diseases and higher miRNA expressions in PF compared to peripheral blood. Five key studies found differentially expressed miRNAs in HF, angina pectoris, aortic stenosis, ventricular tachycardia, and congenital heart diseases with either atrial fibrillation or sinus rhythm. As miRNA-based therapeutics for cardiac fibrosis and HF showed promising results in several in vivo studies for multiple miRNAs, we hypothesize a potential role of miRNA-based therapeutics delivered through the pericardial cavity. This is underlined by the favorable results of the first phase 1b clinical trial in this emerging field. Presenting the first human miRNA antisense drug trial, inhibition of miR-132 by intravenous administration of a novel antisense oligonucleotide, CDR132L, established efficacy in reducing miR-132 in plasma samples in a dose-dependent manner. We screened the literature, provided an overview of the miRNAs and exosomes present in PF, and drew a connection to those miRNAs previously elucidated in cardiac fibrosis and HF. Further, we speculate about clinical implications and potential delivery methods.
Collapse
Affiliation(s)
- Friederike I Schoettler
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ali Fatehi Hassanabad
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cardiac Sciences, Section of Cardiac Surgery, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Anshul S Jadli
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul W M Fedak
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cardiac Sciences, Section of Cardiac Surgery, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
16
|
Niro F, Fernandes S, Cassani M, Apostolico M, Oliver-De La Cruz J, Pereira-Sousa D, Pagliari S, Vinarsky V, Zdráhal Z, Potesil D, Pustka V, Pompilio G, Sommariva E, Rovina D, Maione AS, Bersanini L, Becker M, Rasponi M, Forte G. Fibrotic extracellular matrix impacts cardiomyocyte phenotype and function in an iPSC-derived isogenic model of cardiac fibrosis. Transl Res 2024; 273:58-77. [PMID: 39025226 DOI: 10.1016/j.trsl.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
Cardiac fibrosis occurs following insults to the myocardium and is characterized by the abnormal accumulation of non-compliant extracellular matrix (ECM), which compromises cardiomyocyte contractile activity and eventually leads to heart failure. This phenomenon is driven by the activation of cardiac fibroblasts (cFbs) to myofibroblasts and results in changes in ECM biochemical, structural and mechanical properties. The lack of predictive in vitro models of heart fibrosis has so far hampered the search for innovative treatments, as most of the cellular-based in vitro reductionist models do not take into account the leading role of ECM cues in driving the progression of the pathology. Here, we devised a single-step decellularization protocol to obtain and thoroughly characterize the biochemical and micro-mechanical properties of the ECM secreted by activated cFbs differentiated from human induced pluripotent stem cells (iPSCs). We activated iPSC-derived cFbs to the myofibroblast phenotype by tuning basic fibroblast growth factor (bFGF) and transforming growth factor beta 1 (TGF-β1) signalling and confirmed that activated cells acquired key features of myofibroblast phenotype, like SMAD2/3 nuclear shuttling, the formation of aligned alpha-smooth muscle actin (α-SMA)-rich stress fibres and increased focal adhesions (FAs) assembly. Next, we used Mass Spectrometry, nanoindentation, scanning electron and confocal microscopy to unveil the characteristic composition and the visco-elastic properties of the abundant, collagen-rich ECM deposited by cardiac myofibroblasts in vitro. Finally, we demonstrated that the fibrotic ECM activates mechanosensitive pathways in iPSC-derived cardiomyocytes, impacting on their shape, sarcomere assembly, phenotype, and calcium handling properties. We thus propose human bio-inspired decellularized matrices as animal-free, isogenic cardiomyocyte culture substrates recapitulating key pathophysiological changes occurring at the cellular level during cardiac fibrosis.
Collapse
Affiliation(s)
- Francesco Niro
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Soraia Fernandes
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Marco Cassani
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Monica Apostolico
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Jorge Oliver-De La Cruz
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Daniel Pereira-Sousa
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic
| | - Stefania Pagliari
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Vladimir Vinarsky
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Zbyněk Zdráhal
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - David Potesil
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Vaclav Pustka
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy; Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Davide Rovina
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Angela Serena Maione
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | | | - Marco Rasponi
- Department of Electronics, Informatics and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK.
| |
Collapse
|
17
|
Liao P, Han L, Tao R, Li D, Zhang P, Xiao H. Specific peptides targeting the myocardiocyte are prognostic markers for heart attack: Function of α-SMA protein. Int J Biol Macromol 2024; 280:135793. [PMID: 39304042 DOI: 10.1016/j.ijbiomac.2024.135793] [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: 08/01/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Myocardial infarction (MI) is a serious cardiovascular disease that often results in a significant decline in heart function and associated complications. α-SMA (α-smooth muscle cell actin) is an important biomarker in the process of cardiac remodeling and repair, and its expression level is closely related to myocardial remodeling and prognosis. Therefore, the purpose of this study was to investigate the potential of nanoparticles containing cardiomyocyte targeting peptides in predicting prognosis and α-SMA protein expression after myocardial infarction, with a view to providing new therapeutic strategies and clinical guidelines. In this study, a novel targeting nanoparticle was constructed, using cardiomyocyte specific peptides as targeting ligands, and characterized by loading different drugs. Subsequently, a mouse model of myocardial infarction was used to systematically evaluate the effect of nanoparticles on α-SMA protein expression and prognosis prediction ability after MI. The expression level of α-SMA was analyzed by Western blot and immunohistochemistry, and the prognosis was evaluated by cardiac function assessment. The study found that nanoparticles containing cardiomyocyte targeting peptides significantly increased α-SMA expression levels and improved heart function in animal models of myocardial infarction. Compared with the control group, the application of targeted nanoparticles was closely related to the level of myocardial cell repair and fibrosis, and could effectively predict the prognosis after myocardial infarction. Therefore, nanoparticles containing cardiomyocyte targeting peptides can not only effectively improve the expression of α-SMA, but also serve as an important prognostic tool after myocardial infarction.
Collapse
Affiliation(s)
- Pengfei Liao
- Department of Cardiology, Minhang Hospital, Fudan University, No.179 Xinsong Road, Minhang District, Shanghai 201199, China
| | - Lu Han
- Department of Cardiology, Minhang Hospital, Fudan University, No.179 Xinsong Road, Minhang District, Shanghai 201199, China
| | - Ran Tao
- Department of Cardiology, Minhang Hospital, Fudan University, No.179 Xinsong Road, Minhang District, Shanghai 201199, China
| | - Dandan Li
- Department of Cardiology, Minhang Hospital, Fudan University, No.179 Xinsong Road, Minhang District, Shanghai 201199, China
| | - Peng Zhang
- Department of Cardiology, Minhang Hospital, Fudan University, No.179 Xinsong Road, Minhang District, Shanghai 201199, China.
| | - Hongbing Xiao
- Department of Cardiology, Minhang Hospital, Fudan University, No.179 Xinsong Road, Minhang District, Shanghai 201199, China.
| |
Collapse
|
18
|
Zhuang Z, Liu A, Zhang J, Han S, Tang L, Yu T, Shi Y, Li H, Yang H, Bai P, Tang Y. Hyperuricemia suppresses lumican, exacerbating adverse remodeling after myocardial infarction by promoting fibroblast phenotype transition. J Transl Med 2024; 22:983. [PMID: 39482719 PMCID: PMC11526644 DOI: 10.1186/s12967-024-05778-4] [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: 06/14/2024] [Accepted: 10/19/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Hyperuricemia is independently associated with a poor prognosis in patients with myocardial infarction (MI). Furthermore, MI induces activation of the repair response in local fibroblasts, resulting in extracellular matrix accumulation that generates a stable fibrotic scar in the infarcted area. However, researchers have not determined whether hyperuricemia affects fibroblast activation and its involvement in postinfarction cardiac remodeling. OBJECTIVES We aimed to trigger hyperuricemia by administering potassium oxonate in a mouse model of MI to evaluate the role of hyperuricemia in MI pathogenesis. METHODS Microarray datasets and single-cell sequencing data from gout patients, heart failure patients, and model mice were used to identify the underlying mechanisms responsible for the effect of hyperuricemia on MI progression. A hyperuricemia-related MI mouse model was established. Cardiac function was assessed, followed by sample collection and a uric acid assay. We conducted an enzyme-linked immunosorbent assay, histological detection, immunofluorescence, sequencing data processing, single-cell RNA-seq, and functional enrichment analysis. We then isolated and cultured cardiac fibroblasts and performed Western blotting, quantitative real-time polymerase chain reaction, and shRNA-mediated lumican knockdown assays. RESULTS Hyperuricemia decreased cardiac function, increased mortality, and aggravated adverse fibrosis remodeling in mice after MI. These outcomes were closely related to reduced levels of fibroblast-derived lumican. This reduction activated the TGF-β/SMAD signaling pathway to induce aberrant myofibroblast activation and extracellular matrix deposition in the infarcted area. Furthermore, lumican supplementation or uric acid-lowering therapy with allopurinol alleviated hyperuricemia-mediated abnormal cardiac remodeling. CONCLUSION Hyperuricemia aggravates postinfarction cardiac remodeling by reducing lumican expression and promoting fibroblast phenotype transition. We highlight the clinical importance of lowering uric acid levels in hyperuricemia-related MI to prevent adverse ventricular remodeling.
Collapse
Affiliation(s)
- Zehao Zhuang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Ao Liu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinghong Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shuangjian Han
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Lu Tang
- Department of Echocardiography, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
| | - Tingting Yu
- Department of Dermatology, Pudong New Area People's Hospital, Shanghai, China
| | - Yiping Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Li
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Heng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Peiyuan Bai
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| | - Yanhua Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
19
|
Zhang J, Zhao X, Tang J, Liu C, Zhang Y, Cai C, Du Q. Sleep restriction exacerbates cardiac dysfunction in diabetic mice by causing cardiomyocyte death and fibrosis through mitochondrial damage. Cell Death Discov 2024; 10:446. [PMID: 39433752 PMCID: PMC11494183 DOI: 10.1038/s41420-024-02214-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a cardiovascular complication of diabetes mellitus with a poor prognosis and is the leading cause of death in diabetic patients. Sleep deficiency is not only recognized as an important risk factor for the development of type 2 DM, but is also associated with increased morbidity and mortality of cardiovascular disease. The underlying role and mechanisms of sleep restriction (SR) in DCM are far from clear. The KK/Upj-Ay mouse model of T2 DM was used as a study subject, and the small animal ultrasound imaging system was used to detect the function of the heart; immunopathological staining was used to clarify the histo-structural pathological alterations of the heart; and TUNEL staining, qPCR, transmission electron microscopy (TEM), and ELISA kits were used to detect apoptosis, oxidative stress, inflammation, and mitochondrial damage, and related molecular alterations. SR led to a significant increase in mortality, cardiac hypertrophy, necrosis, glycogen deposition and fibrosis further deteriorated in DM KK mice. SR increased cardiomyocyte death in KK mice through the Bax/Bcl2 pathway. In addition to this, SR not only exacerbated the inflammatory response, but also aggravated mitochondrial damage and promoted oxidative stress in KK mice through the PRDM16-PGC-1α pathway. Overall, SR exacerbates structural alterations and dysfunction through inflammation, oxidative stress, and apoptosis in DM KK mice, increasing the risk of death. Clinicians and diabetic patients are prompted to pay attention to sleep habits to avoid accelerating the transition of DCM to heart failure and inducing death due to poor sleep habits.
Collapse
Affiliation(s)
- Jingyi Zhang
- Centre of General Practice, The Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Xu Zhao
- Centre of General Practice, The Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Jing Tang
- Centre of General Practice, The Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Ce Liu
- Department of Laboratory Medicine, The Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Yining Zhang
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Cheng Cai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Qingfeng Du
- Centre of General Practice, The Seventh Affiliated Hospital of Southern Medical University, Foshan, China.
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, China.
- Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China.
- Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, China.
| |
Collapse
|
20
|
Beikzadeh B, Khani M, Zarinehzadeh Y, Abedini Bakhshmand E, Sadeghizadeh M, Rabbani S, Soltani BM. Preventive and treatment efficiency of dendrosomal nano-curcumin against ISO-induced cardiac fibrosis in mouse model. PLoS One 2024; 19:e0311817. [PMID: 39388499 PMCID: PMC11469592 DOI: 10.1371/journal.pone.0311817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Cardiac fibrosis (c-fibrosis) is a critical factor in cardiovascular diseases, leading to impaired cardiac function and heart failure. This study aims to optimize the isoproterenol (ISO)-induced c-fibrosis model and evaluate the therapeutic efficacy of dendrosomal nano-curcumin (DNC) in both in-vitro and in-vivo conditions. Also, we were looking for the differentially expressed genes following the c-fibrosis induction. At the in-vitro condition, primary cardiac fibroblasts were exclusively cultured on collagen-coated or polystyrene plates and, were treated with ISO for fibrosis induction and post-treated or co-treated with DNC. RT-qPCR and flow cytometry analysis indicated that DNC treatment attenuated the fibrotic effect of ISO treatment in these cells. At the in-vivo condition, our findings demonstrated that ISO treatment effectively induces cardiac (and pulmonary) fibrosis, characterized by pro-fibrotic and pro-inflammatory gene expression and IHC (α-SMA, COL1A1, and TGFβ). Interestingly, fibrosis symptoms were reduced following the pretreatment, co-treatment, or post-treatment of DNC with ISO. Additionally, the intensive RNAseq analysis suggested the COMP gene is differentially expressed following the c-fibrosis and our RT-qPCR analysis suggested it as a novel potential marker. Overall, our results promise the application of DNC as a potential preventive or therapy agent before and after heart challenges that lead to c-fibrosis.
Collapse
Affiliation(s)
- Behnaz Beikzadeh
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mona Khani
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yasamin Zarinehzadeh
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Majid Sadeghizadeh
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram M. Soltani
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
21
|
Wang Z, Li L, Yang S, Li Z, Zhang P, Shi R, Zhou X, Tang X, Li Q. Possible mechanisms of SARS-CoV-2-associated myocardial fibrosis: reflections in the post-pandemic era. Front Microbiol 2024; 15:1470953. [PMID: 39444690 PMCID: PMC11497467 DOI: 10.3389/fmicb.2024.1470953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Since December 2019, coronavirus disease 2019 (COVID-19) has been spreading worldwide with devastating immediate or long-term effects on people's health. Although the lungs are the primary organ affected by COVID-19, individuals infected with SARS-CoV-2 also develop systemic lesions involving multiple organs throughout the body, such as the cardiovascular system. Emerging evidence reveals that COVID-19 could generate myocardial fibrosis, termed "COVID-19-associated myocardial fibrosis." It can result from the activation of fibroblasts via the renin-angiotensin-aldosterone system (RAAS), transforming growth factor-β1 (TGF-β1), microRNAs, and other pathways, and can also occur in other cellular interactions with SARS-CoV-2, such as immunocytes, endothelial cells. Nonetheless, to gain a more profound insight into the natural progression of COVID-19-related myocardial fibrosis, additional investigations are necessary. This review delves into the underlying mechanisms contributing to COVID-19-associated myocardial fibrosis while also examining the antifibrotic potential of current COVID-19 treatments, thereby offering guidance for future clinical trials of these medications. Ultimately, we propose future research directions for COVID-19-associated myocardial fibrosis in the post-COVID-19 era, such as artificial intelligence (AI) telemedicine. We also recommend that relevant tests be added to the follow-up of COVID-19 patients to detect myocardial fibrosis promptly.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luwei Li
- Department of Pediatric Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Third Clinical Medical College of Zhengzhou University, Zhengzhou, China
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojuan Tang
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Pasvanis Z, Kong RCK, Shah MH, Chan EC, Fan Gaskin JC. 3',4'-Dihydroxyflavonol Inhibits Fibrotic Response in a Rabbit Model of Glaucoma Filtration Surgery. Int J Mol Sci 2024; 25:10767. [PMID: 39409096 PMCID: PMC11476621 DOI: 10.3390/ijms251910767] [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: 07/19/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Post-operative fibrosis of the filtering bleb limits the success of glaucoma filtration surgery (GFS). To minimise subconjunctival scarring following GFS, treatment with antimetabolites such as Mitomycin C (MMC) has become standard practice; however, their use is associated with considerable side effects. This study aimed to investigate the anti-scarring properties of 3',4'-dihydroxyflavonol (DiOHF). GFS was performed in New Zealand white rabbits who received eye drops of DiOHF three times daily and vehicle eye drops after surgery (n = 5) or a single intraoperative treatment of MMC (n = 5). Blebs were imaged immediately following surgery and on days 7, 15, 21, and 28 for clinical examination. On day 28, eyes were harvested to assess collagen deposition, expression of α-SMA, oxidative stress, angiogenesis, fibroblast activity, and inflammation in the conjunctiva/Tenon's layer. At 7 and 28 days post-GFS, MMC-treated blebs were more ischaemic than DiOHF- or vehicle-treated blebs. On day 28, DiOHF treatment significantly suppressed collagen accumulation, CD31 expression, Vimentin expression, and CD45 expression compared to the vehicle control. No difference was observed in 3-Nitrotyrosine or αSMA expression between treatment groups. Treatment with DiOHF reduced conjunctival scarring and angiogenesis in rabbits with GFS, which was comparable to MMC. DiOHF may be a safer and more effective wound-modulating agent than conventional antifibrotic therapy in GFS.
Collapse
Affiliation(s)
- Zoe Pasvanis
- Ophthalmology, Department of Surgery, University of Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Roy C. K. Kong
- Ophthalmology, Department of Surgery, University of Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Manisha H. Shah
- Ophthalmology, Department of Surgery, University of Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Elsa C. Chan
- Ophthalmology, Department of Surgery, University of Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
- Department of Medicine, St Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Jennifer C. Fan Gaskin
- Ophthalmology, Department of Surgery, University of Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
- Glaucoma Research and Investigation Unit, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| |
Collapse
|
23
|
Zhu YF, Wan MC, Gao P, Shen MJ, Zhu YN, Hao JX, Lu WC, Wang CY, Tay F, Ehrlich H, Niu LN, Jiao K. Fibrocyte: A missing piece in the pathogenesis of fibrous epulis. Oral Dis 2024; 30:4376-4389. [PMID: 38148479 DOI: 10.1111/odi.14847] [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: 09/09/2023] [Revised: 11/18/2023] [Accepted: 12/10/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVES To explore the role of fibrocytes in the recurrence and calcification of fibrous epulides. METHODS Different subtypes of fibrous epulides and normal gingival tissue specimens were first collected for histological and immunofluorescence analyses to see if fibrocytes were present and whether they differentiated into myofibroblasts and osteoblasts upon stimulated by transforming growth factor-β1 (TGF-β1). Electron microscopy and elemental analysis were used to characterize the extracellular microenvironment in different subtypes of fibrous epulides. Human peripheral blood mononuclear cells (PBMCs) were subsequently isolated from in vitro models to mimic the microenvironment in fibrous epulides to identify whether TGF-β1 as well as the calcium and phosphorus ion concentration in the extracellular matrix (ECM) of a fibrous epulis trigger fibrocyte differentiation. RESULTS Fibrous epulides contain fibrocytes that accumulate in the local inflammatory environment and have the ability to differentiate into myofibroblasts or osteoblasts. TGF-β1 promotes fibrocytes differentiation into myofibroblasts in a concentration-dependent manner, while TGF-β1 stimulates the fibrocytes to differentiate into osteoblasts when combined with a high calcium and phosphorus environment. CONCLUSIONS Our study revealed fibrocytes play an important role in the fibrogenesis and osteogenesis in fibrous epulis, and might serve as a therapeutic target for the inhibition of recurrence of fibrous epulides.
Collapse
Affiliation(s)
- Yi-Fei Zhu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Mei-Chen Wan
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Peng Gao
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Min-Juan Shen
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yi-Na Zhu
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jia-Xin Hao
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Wei-Cheng Lu
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Chen-Yu Wang
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Franklin Tay
- The Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hermann Ehrlich
- Institute of Electronic and Sensor Materials, Freiberg, Germany
| | - Li-Na Niu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
24
|
Xu K, Saaoud F, Shao Y, Lu Y, Yang Q, Jiang X, Wang H, Yang X. A new paradigm in intracellular immunology: Mitochondria emerging as leading immune organelles. Redox Biol 2024; 76:103331. [PMID: 39216270 PMCID: PMC11402145 DOI: 10.1016/j.redox.2024.103331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Mitochondria, traditionally recognized as cellular 'powerhouses' due to their pivotal role in energy production, have emerged as multifunctional organelles at the intersection of bioenergetics, metabolic signaling, and immunity. However, the understanding of their exact contributions to immunity and inflammation is still developing. This review first introduces the innovative concept of intracellular immunity, emphasizing how mitochondria serve as critical immune signaling hubs. They are instrumental in recognizing and responding to pathogen and danger signals, and in modulating immune responses. We also propose mitochondria as the leading immune organelles, drawing parallels with the broader immune system in their functions of antigen presentation, immune regulation, and immune response. Our comprehensive review explores mitochondrial immune signaling pathways, their therapeutic potential in managing inflammation and chronic diseases, and discusses cutting-edge methodologies for mitochondrial research. Targeting a broad readership of both experts in mitochondrial functions and newcomers to the field, this review sets forth new directions that could transform our understanding of intracellular immunity and the integrated immune functions of intracellular organelles.
Collapse
Affiliation(s)
- Keman Xu
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | - Fatma Saaoud
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | - Ying Shao
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | - Yifan Lu
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | | | - Xiaohua Jiang
- Lemole Center for Integrated Lymphatics and Vascular Research, USA; Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xiaofeng Yang
- Lemole Center for Integrated Lymphatics and Vascular Research, USA; Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
25
|
Zhang B, Wu Y, Zhou C, Xie J, Zhang Y, Yang X, Xiao J, Wang DW, Shan C, Zhou X, Xiang Y, Yang B. Hyperactivation of ATF4/TGF-β1 signaling contributes to the progressive cardiac fibrosis in Arrhythmogenic cardiomyopathy caused by DSG2 Variant. BMC Med 2024; 22:361. [PMID: 39227800 PMCID: PMC11373413 DOI: 10.1186/s12916-024-03593-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiomyopathy characterized with progressive cardiac fibrosis and heart failure. However, the exact mechanism driving the progression of cardiac fibrosis and heart failure in ACM remains elusive. This study aims to investigate the underlying mechanisms of progressive cardiac fibrosis in ACM caused by newly identified Desmoglein-2 (DSG2) variation. METHODS We identified homozygous DSG2F531C variant in a family with 8 ACM patients using whole-exome sequencing and generated Dsg2F536C knock-in mice. Neonatal and adult mouse ventricular myocytes isolated from Dsg2F536C knock-in mice were used. We performed functional, transcriptomic and mass spectrometry analyses to evaluate the mechanisms of ACM caused by DSG2F531C variant. RESULTS All eight patients with ACM were homozygous for DSG2F531C variant. Dsg2F536C/F536C mice displayed cardiac enlargement, dysfunction, and progressive cardiac fibrosis in both ventricles. Mechanistic investigations revealed that the variant DSG2-F536C protein underwent misfolding, leading to its recognition by BiP within the endoplasmic reticulum, which triggered endoplasmic reticulum stress, activated the PERK-ATF4 signaling pathway and increased ATF4 levels in cardiomyocytes. Increased ATF4 facilitated the expression of TGF-β1 in cardiomyocytes, thereby activating cardiac fibroblasts through paracrine signaling and ultimately promoting cardiac fibrosis in Dsg2F536C/F536C mice. Notably, inhibition of the PERK-ATF4 signaling attenuated progressive cardiac fibrosis and cardiac systolic dysfunction in Dsg2F536C/F536C mice. CONCLUSIONS Hyperactivation of the ATF4/TGF-β1 signaling in cardiomyocytes emerges as a novel mechanism underlying progressive cardiac fibrosis in ACM. Targeting the ATF4/TGF-β1 signaling may be a novel therapeutic target for managing ACM.
Collapse
Affiliation(s)
- Baowei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Yizhang Wu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Chunjiang Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Jiaxi Xie
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P.R. China
| | - Youming Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Xingbo Yang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Jing Xiao
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Dao Wu Wang
- State Key Laboratory of Reproductive Medicine, the Centre for Clinical Reproductive Medicine, Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P.R. China
| | - Congjia Shan
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Xiujuan Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P.R. China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China.
| | - Bing Yang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China.
| |
Collapse
|
26
|
Miguel V, Alcalde-Estévez E, Sirera B, Rodríguez-Pascual F, Lamas S. Metabolism and bioenergetics in the pathophysiology of organ fibrosis. Free Radic Biol Med 2024; 222:85-105. [PMID: 38838921 DOI: 10.1016/j.freeradbiomed.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Fibrosis is the tissue scarring characterized by excess deposition of extracellular matrix (ECM) proteins, mainly collagens. A fibrotic response can take place in any tissue of the body and is the result of an imbalanced reaction to inflammation and wound healing. Metabolism has emerged as a major driver of fibrotic diseases. While glycolytic shifts appear to be a key metabolic switch in activated stromal ECM-producing cells, several other cell types such as immune cells, whose functions are intricately connected to their metabolic characteristics, form a complex network of pro-fibrotic cellular crosstalk. This review purports to clarify shared and particular cellular responses and mechanisms across organs and etiologies. We discuss the impact of the cell-type specific metabolic reprogramming in fibrotic diseases in both experimental and human pathology settings, providing a rationale for new therapeutic interventions based on metabolism-targeted antifibrotic agents.
Collapse
Affiliation(s)
- Verónica Miguel
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| | - Elena Alcalde-Estévez
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain; Department of Systems Biology, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Belén Sirera
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Fernando Rodríguez-Pascual
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
27
|
Jeon KI, Kumar A, Brookes PS, Nehrke K, Huxlin KR. Manipulating mitochondrial pyruvate carrier function causes metabolic remodeling in corneal myofibroblasts that ameliorates fibrosis. Redox Biol 2024; 75:103235. [PMID: 38889622 PMCID: PMC11231598 DOI: 10.1016/j.redox.2024.103235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Myofibroblasts are key cellular effectors of corneal wound healing from trauma, surgery, or infection. However, their persistent deposition of disorganized extracellular matrix can also cause corneal fibrosis and visual impairment. Recent work showed that the PPARγ agonist Troglitazone can mitigate established corneal fibrosis, and parallel in vitro data suggested this occurred through inhibition of the mitochondrial pyruvate carrier (MPC) rather than PPARγ. In addition to oxidative phosphorylation (Ox-Phos), pyruvate and other mitochondrial metabolites provide carbon for the synthesis of biological macromolecules. However, it is currently unclear how these roles selectively impact fibrosis. Here, we performed bioenergetic, metabolomic, and epigenetic analyses of corneal fibroblasts treated with TGF-β1 to stimulate myofibroblast trans-differentiation, with further addition of Troglitazone or the MPC inhibitor UK5099, to identify MPC-dependencies that may facilitate remodeling and loss of the myofibroblast phenotype. Our results show that a shift in energy metabolism is associated with, but not sufficient to drive cellular remodeling. Metabolites whose abundances were sensitive to MPC inhibition suggest that sustained carbon influx into the Krebs' cycle is prioritized over proline synthesis to fuel collagen deposition. Furthermore, increased abundance of acetyl-CoA and increased histone H3 acetylation suggest that epigenetic mechanisms downstream of metabolic remodeling may reinforce cellular phenotypes. Overall, our results highlight a novel molecular target and metabolic vulnerability that affects myofibroblast persistence in the context of corneal wounding.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Ankita Kumar
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Paul S Brookes
- Dept. Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Keith Nehrke
- Dept. Medicine-Nephrology Division, University of Rochester, Rochester, NY, USA
| | - Krystel R Huxlin
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
28
|
Zhang X, Wang Y, Li H, Wang DW, Chen C. Insights into the post-translational modifications in heart failure. Ageing Res Rev 2024; 100:102467. [PMID: 39187021 DOI: 10.1016/j.arr.2024.102467] [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: 10/25/2023] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Heart failure (HF), as the terminal manifestation of multiple cardiovascular diseases, causes a huge socioeconomic burden worldwide. Despite the advances in drugs and medical-assisted devices, the prognosis of HF remains poor. HF is well-accepted as a myriad of subcellular dys-synchrony related to detrimental structural and functional remodelling of cardiac components, including cardiomyocytes, fibroblasts, endothelial cells and macrophages. Through the covalent chemical process, post-translational modifications (PTMs) can coordinate protein functions, such as re-localizing cellular proteins, marking proteins for degradation, inducing interactions with other proteins and tuning enzyme activities, to participate in the progress of HF. Phosphorylation, acetylation, and ubiquitination predominate in the currently reported PTMs. In addition, advanced HF is commonly accompanied by metabolic remodelling including enhanced glycolysis. Thus, glycosylation induced by disturbed energy supply is also important. In this review, firstly, we addressed the main types of HF. Then, considering that PTMs are associated with subcellular locations, we summarized the leading regulation mechanisms in organelles of distinctive cell types of different types of HF, respectively. Subsequently, we outlined the aforementioned four PTMs of key proteins and signaling sites in HF. Finally, we discussed the perspectives of PTMs for potential therapeutic targets in HF.
Collapse
Affiliation(s)
- Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yan Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
29
|
Zhu W, Guo S, Sun J, Zhao Y, Liu C. Lactate and lactylation in cardiovascular diseases: current progress and future perspectives. Metabolism 2024; 158:155957. [PMID: 38908508 DOI: 10.1016/j.metabol.2024.155957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Cardiovascular diseases (CVDs) are often linked to structural and functional impairments, such as heart defects and circulatory dysfunction, leading to compromised peripheral perfusion and heightened morbidity risks. Metabolic remodeling, particularly in the context of cardiac fibrosis and inflammation, is increasingly recognized as a pivotal factor in the pathogenesis of CVDs. Metabolic syndromes further predispose individuals to these conditions, underscoring the need to elucidate the metabolic underpinnings of CVDs. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that connects cellular metabolism with the regulation of cellular activity. The transport of lactate between different cells is essential for metabolic homeostasis and signal transduction. Disruptions to lactate dynamics are implicated in various CVDs. Furthermore, lactylation, a novel post-translational modification, has been identified in cardiac cells, where it influences protein function and gene expression, thereby playing a significant role in CVD pathogenesis. In this review, we summarized recent advancements in understanding the role of lactate and lactylation in CVDs, offering fresh insights that could guide future research directions and therapeutic interventions. The potential of lactate metabolism and lactylation as innovative therapeutic targets for CVD is a promising avenue for exploration.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| | - Siyu Guo
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Junyi Sun
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Yudan Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, PR China.
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| |
Collapse
|
30
|
Wei Q, Gan C, Sun M, Xie Y, Liu H, Xue T, Deng C, Mo C, Ye T. BRD4: an effective target for organ fibrosis. Biomark Res 2024; 12:92. [PMID: 39215370 PMCID: PMC11365212 DOI: 10.1186/s40364-024-00641-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Fibrosis is an excessive wound-healing response induced by repeated or chronic external stimuli to tissues, significantly impacting quality of life and primarily contributing to organ failure. Organ fibrosis is reported to cause 45% of all-cause mortality worldwide. Despite extensive efforts to develop new antifibrotic drugs, drug discovery has not kept pace with the clinical demand. Currently, only pirfenidone and nintedanib are approved by the FDA to treat pulmonary fibrotic illness, whereas there are currently no available antifibrotic drugs for hepatic, cardiac or renal fibrosis. The development of fibrosis is closely related to epigenetic alterations. The field of epigenetics primarily studies biological processes, including chromatin modifications, epigenetic readers, DNA transcription and RNA translation. The bromodomain and extra-terminal structural domain (BET) family, a class of epigenetic readers, specifically recognizes acetylated histone lysine residues and promotes the formation of transcriptional complexes. Bromodomain-containing protein 4 (BRD4) is one of the most well-researched proteins in the BET family. BRD4 is implicated in the expression of genes related to inflammation and pro-fibrosis during fibrosis. Inhibition of BRD4 has shown promising anti-fibrotic effects in preclinical studies; however, no BRD4 inhibitor has been approved for clinical use. This review introduces the structure and function of BET proteins, the research progress on BRD4 in organ fibrosis, and the inhibitors of BRD4 utilized in fibrosis. We emphasize the feasibility of targeting BRD4 as an anti-fibrotic strategy and discuss the therapeutic potential and challenges associated with BRD4 inhibitors in treating fibrotic diseases.
Collapse
Affiliation(s)
- Qun Wei
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cailing Gan
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Sun
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuting Xie
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongyao Liu
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Taixiong Xue
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Conghui Deng
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Tinghong Ye
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Ningxia Medical University, Yin Chuan, 640100, China.
| |
Collapse
|
31
|
Wen J, Guan Y, Niu H, Dang Y, Guan J. Targeting cardiac resident CCR2+ macrophage-secreted MCP-1 to attenuate inflammation after myocardial infarction. Acta Biomater 2024:S1742-7061(24)00469-0. [PMID: 39182804 DOI: 10.1016/j.actbio.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/26/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
After myocardial infarction (MI), cardiac resident CCR2+ macrophages release various cytokines and chemokines, notably monocyte chemoattractant protein-1 (MCP-1). MCP-1 is instrumental in recruiting CCR2+ monocytes to the damaged region. The excessive arrival of these monocytes, which then become macrophages, perpetuates inflammation at the site of injury. This continuous inflammation leads to adverse tissue remodeling and compromises cardiac function over time. We hypothesized that neutralizing the MCP-1 secreted by cardiac resident CCR2+ macrophages can mitigate post-MI inflammation by curtailing the recruitment of monocytes and their differentiation into macrophages. In this work, we developed nanoparticles that target the infarcted heart, specifically accumulating in the damaged area after intravenous (IV) administration, and docking onto CCR2+ macrophages. These nanoparticles were designed to slowly release an MCP-1 binding peptide, HSWRHFHTLGGG (HSW), which neutralizes the upregulated MCP-1. We showed that the HSW reduced monocyte migration, inhibited pro-inflammatory cytokine upregulation, and suppressed myofibroblast differentiation in vitro. After IV delivery, the released HSW significantly decreased monocyte recruitment and pro-inflammatory macrophage density, increased cardiac cell survival, attenuated cardiac fibrosis, and improved cardiac function. Taken together, our findings support the strategy of MCP-1 neutralization at the acute phase of MI as a promising way to alleviate post-MI inflammation. STATEMENT OF SIGNIFICANCE: After a myocardial infarction (MI), CCR2+ macrophages resident in the heart release various cytokines and chemokines, notably monocyte chemoattractant protein-1 (MCP-1). MCP-1 is instrumental in attracting CCR2+ monocytes to the damaged region. The excessive arrival of these monocytes, which then become macrophages, perpetuates inflammation at the site of injury. This continuous inflammation leads to adverse tissue remodeling and compromises cardiac function over time. In this work, we tested the hypothesis that neutralizing the MCP-1 secreted by cardiac CCR2+ macrophages can mitigate post-MI inflammation by curtailing the recruitment of monocytes.
Collapse
Affiliation(s)
- Jiaxing Wen
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Ya Guan
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Hong Niu
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Yu Dang
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA
| | - Jianjun Guan
- Institute of Materials Science and Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis. St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis. St. Louis, MO 63130, USA.
| |
Collapse
|
32
|
Müller WEG, Schepler H, Neufurth M, Dobmeyer R, Batel R, Schröder HC, Wang X. Energy level as a theranostic factor for successful therapy of tissue injuries with polyphosphate: the triad metabolic energy - mechanical energy - heat. Theranostics 2024; 14:5262-5280. [PMID: 39267793 PMCID: PMC11388067 DOI: 10.7150/thno.100622] [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: 07/07/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Rationale: Tissue regeneration of skin and bone is an energy-intensive, ATP-consuming process that, if impaired, can lead to the development of chronic clinical pictures. ATP levels in the extracellular space including the exudate of wounds, especially chronic wounds, are low. This deficiency can be compensated by inorganic polyphosphate (polyP) supplied via the blood platelets to the regenerating site. Methods: The contribution of the different forms of energy derived from polyP (metabolic energy, mechanical energy and heat) to regeneration processes was dissected and studied both in vitro and in patients. ATP is generated metabolically during the enzymatic cleavage of the energy-rich anhydride bonds between the phosphate units of polyP, involving the two enzymes alkaline phosphatase (ALP) and adenylate kinase (ADK). Exogenous polyP was administered after incorporation into compressed collagen or hydrogel wound coverages to evaluate its regenerative activity for chronic wound healing. Results: In a proof-of-concept study, fast healing of chronic wounds was achieved with the embedded polyP, supporting the crucial regeneration-promoting activity of ATP. In the presence of Ca2+ in the wound exudate, polyP undergoes a coacervation process leading to a conversion of fibroblasts into myofibroblasts, a crucial step supporting cell migration during regenerative tissue repair. During coacervation, a switch from an endothermic to an exothermic, heat-generating process occurs, reflecting a shift from an entropically- to an enthalpically-driven thermodynamic reaction. In addition, mechanical forces cause the appearance of turbulent flows and vortices during liquid-liquid phase separation. These mechanical forces orient the cellular and mineralic (hydroxyapatite crystallite) components, as shown using mineralizing SaOS-2 cells as a model. Conclusion: Here we introduce the energetic triad: metabolic energy (ATP), thermal energy and mechanical energy as a novel theranostic biomarker, which contributes essentially to a successful application of polyP for regeneration processes.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, GERMANY
| | - Hadrian Schepler
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, GERMANY
| | - Meik Neufurth
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, GERMANY
| | - Rita Dobmeyer
- Galenus GH AG, Rainstrasse 7, 6052 Hergiswil, Switzerland
| | - Renato Batel
- Faculty of Natural Sciences, Juraj Dobrila University, Zagrebačka 30, 52100 Pula, Croatia
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, GERMANY
| | - Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at the Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128 Mainz, GERMANY
| |
Collapse
|
33
|
Gao Z, Yan L, Meng J, Lu Z, Ge K, Jiang Z, Feng T, Wang H, Liu C, Tang J, Zhang H. Targeting cardiac fibrosis with chimeric antigen receptor macrophages. Cell Discov 2024; 10:86. [PMID: 39134538 PMCID: PMC11319452 DOI: 10.1038/s41421-024-00718-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Affiliation(s)
- Zibei Gao
- School of Life Science and Technology & Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai, China
| | - Lei Yan
- School of Life Science and Technology & Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai, China
| | - Jufeng Meng
- School of Life Science and Technology & Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai, China
| | - Zhengkai Lu
- State Key Laboratory of Cardiovascular Disease and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Kaixin Ge
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhen Jiang
- School of Life Science and Technology & Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai, China
| | - Teng Feng
- School of Life Science and Technology & Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai, China
| | - Haopeng Wang
- School of Life Science and Technology & Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai, China
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China
| | - Chen Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Juan Tang
- State Key Laboratory of Cardiovascular Disease and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai, China.
| | - Hui Zhang
- School of Life Science and Technology & Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai, China.
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
34
|
Kruithof BPT, Mousavi Gourabi B, van de Merbel AF, DeRuiter MC, Goumans MJ. A New Ex Vivo Model to Study Cardiac Fibrosis in Whole Mouse Hearts. JACC Basic Transl Sci 2024; 9:1005-1022. [PMID: 39297130 PMCID: PMC11405901 DOI: 10.1016/j.jacbts.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 09/21/2024]
Abstract
Fibrosis is a characteristic of many cardiac diseases for which no effective treatment exists. We have developed an ex vivo flow system, which allows induction of cardiac fibrosis in intact adult mouse hearts. Lineage-tracing studies indicated that the collagen-producing myofibroblasts originated from the resident fibroblasts. The extent of fibrosis was flow rate dependent, and pharmacological inhibition of the transforming growth factor beta signaling pathway prevented fibrosis. Therefore, in this powerful system, the cellular and molecular mechanisms underlying cardiac fibrosis can be studied. In addition, new targets can be tested on organ level for their ability to inhibit fibrosis.
Collapse
Affiliation(s)
- Boudewijn P T Kruithof
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Babak Mousavi Gourabi
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Marco C DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
35
|
Du L, Wang X, Guo Y, Tao T, Wu H, Xu X, Zhang B, Chen T, Xu Q, Guo X. Altered lipid metabolism promoting cardiac fibrosis is mediated by CD34 + cell-derived FABP4 + fibroblasts. Exp Mol Med 2024; 56:1869-1886. [PMID: 39198543 PMCID: PMC11372182 DOI: 10.1038/s12276-024-01309-9] [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: 10/09/2023] [Revised: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 09/01/2024] Open
Abstract
Hyperlipidemia and hypertension might play a role in cardiac fibrosis, in which a heterogeneous population of fibroblasts seems important. However, it is unknown whether CD34+ progenitor cells are involved in the pathogenesis of heart fibrosis. This study aimed to explore the mechanism of CD34+ cell differentiation in cardiac fibrosis during hyperlipidemia. Through the analysis of transcriptomes from 50,870 single cells extracted from mouse hearts and 76,851 single cells from human hearts, we have effectively demonstrated the evolving cellular landscape throughout cardiac fibrosis. Disturbances in lipid metabolism can accelerate the development of fibrosis. Through the integration of bone marrow transplantation models and lineage tracing, our study showed that hyperlipidemia can expedite the differentiation of non-bone marrow-derived CD34+ cells into fibroblasts, particularly FABP4+ fibroblasts, in response to angiotensin II. Interestingly, the partial depletion of CD34+ cells led to a notable reduction in triglycerides in the heart, mitigated fibrosis, and improved cardiac function. Furthermore, immunostaining of human heart tissue revealed colocalization of CD34+ cells and fibroblasts. Mechanistically, our investigation of single-cell RNA sequencing data through pseudotime analysis combined with in vitro cellular studies revealed the crucial role of the PPARγ/Akt/Gsk3β pathway in orchestrating the differentiation of CD34+ cells into FABP4+ fibroblasts. Through our study, we generated valuable insights into the cellular landscape of CD34+ cell-derived cells in the hypertrophic heart with hyperlipidemia, indicating that the differentiation of non-bone marrow-derived CD34+ cells into FABP4+ fibroblasts during this process accelerates lipid accumulation and promotes heart failure via the PPARγ/Akt/Gsk3β pathway.
Collapse
Affiliation(s)
- Luping Du
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Tao
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Wu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodong Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bohuan Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Health care, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
36
|
Hughes DM, Won T, Talor MV, Kalinoski HM, Jurčová I, Szárszoi O, Stříž I, Čurnová L, Bracamonte-Baran W, Melenovský V, Čiháková D. The protective role of GATA6 + pericardial macrophages in pericardial inflammation. iScience 2024; 27:110244. [PMID: 39040070 PMCID: PMC11260870 DOI: 10.1016/j.isci.2024.110244] [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: 10/22/2023] [Revised: 03/18/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Prior research has suggested that GATA6+ pericardial macrophages may traffic to the myocardium to prevent interstitial fibrosis after myocardial infarction (MI), while subsequent literature claims that they do not. We demonstrate that GATA6+ pericardial macrophages are critical for preventing IL-33 induced pericarditis and attenuate trafficking of inflammatory monocytes and granulocytes to the pericardial cavity after MI. However, absence of GATA6+ macrophages did not affect myocardial inflammation due to MI or coxsackievirus-B3 induced myocarditis, or late-stage cardiac fibrosis and cardiac function post MI. GATA6+ macrophages are significantly less transcriptionally active following stimulation in vitro compared to bone marrow-derived macrophages and do not induce upregulation of inflammatory markers in fibroblasts. This suggests that GATA6+ pericardial macrophages attenuate inflammation through their interactions with surrounding cells. We therefore conclude that GATA6+ pericardial macrophages are critical in modulating pericardial inflammation, but do not play a significant role in controlling myocardial inflammation or fibrosis.
Collapse
Affiliation(s)
- David M. Hughes
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Monica V. Talor
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hannah M. Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ivana Jurčová
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Ondrej Szárszoi
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Ilja Stříž
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Lenka Čurnová
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | | | - Vojtěch Melenovský
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
37
|
Lazaropoulos MP, Gibb AA, Chapski DJ, Nair AA, Reiter AN, Roy R, Eaton DM, Bedi KC, Margulies KB, Wellen KE, Estarás C, Vondriska TM, Elrod JW. Nuclear ATP-citrate lyase regulates chromatin-dependent activation and maintenance of the myofibroblast gene program. NATURE CARDIOVASCULAR RESEARCH 2024; 3:869-882. [PMID: 39196175 PMCID: PMC11358007 DOI: 10.1038/s44161-024-00502-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 05/31/2024] [Indexed: 08/29/2024]
Abstract
Differentiation of cardiac fibroblasts to myofibroblasts is necessary for matrix remodeling and fibrosis in heart failure. We previously reported that mitochondrial calcium signaling drives α-ketoglutarate-dependent histone demethylation, promoting myofibroblast formation. Here we investigate the role of ATP-citrate lyase (ACLY), a key enzyme for acetyl-CoA biosynthesis, in histone acetylation regulating myofibroblast fate and persistence in cardiac fibrosis. We show that inactivation of ACLY prevents myofibroblast differentiation and reverses myofibroblasts towards quiescence. Genetic deletion of Acly in post-activated myofibroblasts prevents fibrosis and preserves cardiac function in pressure-overload heart failure. TGFβ stimulation enhances ACLY nuclear localization and ACLY-SMAD2/3 interaction, and increases H3K27ac at fibrotic gene loci. Pharmacological inhibition of ACLY or forced nuclear expression of a dominant-negative ACLY mutant prevents myofibroblast formation and H3K27ac. Our data indicate that nuclear ACLY activity is necessary for myofibroblast differentiation and persistence by maintaining histone acetylation at TGFβ-induced myofibroblast genes. These findings provide targets to prevent and reverse pathological fibrosis.
Collapse
Affiliation(s)
- Michael P Lazaropoulos
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Andrew A Gibb
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Douglas J Chapski
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Abheya A Nair
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Allison N Reiter
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Rajika Roy
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Deborah M Eaton
- Cardiovascular Institute and Cardiovascular Medicine Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth C Bedi
- Cardiovascular Institute and Cardiovascular Medicine Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth B Margulies
- Cardiovascular Institute and Cardiovascular Medicine Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Conchi Estarás
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Thomas M Vondriska
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Departments Medicine/Cardiology and Physiology, and Molecular Biology Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - John W Elrod
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Frangogiannis NG. Targeting metabolically activated fibroblasts in the failing heart. NATURE CARDIOVASCULAR RESEARCH 2024; 3:782-784. [PMID: 39196174 DOI: 10.1038/s44161-024-00500-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York City, NY, USA.
| |
Collapse
|
39
|
Ma J, Wang X, Jia Y, Tan F, Yuan X, Du J. The roles of B cells in cardiovascular diseases. Mol Immunol 2024; 171:36-46. [PMID: 38763105 DOI: 10.1016/j.molimm.2024.05.002] [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: 01/16/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 05/21/2024]
Abstract
Damage to the heart can start the repair process and cause cardiac remodeling. B cells play an important role in this process. B cells are recruited to the injured place and activate cardiac remodeling through secreting antibodies and cytokines. Different types of B cells showed specific functions in the heart. Among all types of B cells, heart-associated B cells play a vital role in the heart by secreting TGFβ1. B cells participate in the activation of fibroblasts and promote cardiac fibrosis. Four subtypes of B cells in the heart revealed the relationship between the B cells' heterogeneity and cardiac remodeling. Many cardiovascular diseases like atherosclerosis, heart failure (HF), hypertension, myocardial infarction (MI), and dilated cardiomyopathy (DCM) are related to B cells. The primary mechanisms of these B cell-related activities will be discussed in this review, which may also suggest potential novel therapeutic targets.
Collapse
Affiliation(s)
- Jian Ma
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaotong Wang
- Department of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuewang Jia
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangyan Tan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
40
|
Xie C, Zhong L, Feng H, Wang R, Shi Y, Lv Y, Hu Y, Li J, Xiao D, Liu S, Chen Q, Tao Y. Exosomal miR-17-5p derived from epithelial cells is involved in aberrant epithelium-fibroblast crosstalk and induces the development of oral submucosal fibrosis. Int J Oral Sci 2024; 16:48. [PMID: 38897993 PMCID: PMC11187069 DOI: 10.1038/s41368-024-00302-2] [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: 10/11/2023] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 06/21/2024] Open
Abstract
Oral submucous fibrosis (OSF) is a chronic and inflammatory mucosal disease caused by betel quid chewing, which belongs to oral potentially malignant disorders. Abnormal fibroblast differentiation leading to disordered collagen metabolism is the core process underlying OSF development. The epithelium, which is the first line of defense against the external environment, can convert external signals into pathological signals and participate in the remodeling of the fibrotic microenvironment. However, the specific mechanisms by which the epithelium drives fibroblast differentiation remain unclear. In this study, we found that Arecoline-exposed epithelium communicated with the fibrotic microenvironment by secreting exosomes. MiR-17-5p was encapsulated in epithelial cell-derived exosomes and absorbed by fibroblasts, where it promoted cell secretion, contraction, migration and fibrogenic marker (α-SMA and collagen type I) expression. The underlying molecular mechanism involved miR-17-5p targeting Smad7 and suppressing the degradation of TGF-β receptor 1 (TGFBR1) through the E3 ubiquitination ligase WWP1, thus facilitating downstream TGF-β pathway signaling. Treatment of fibroblasts with an inhibitor of miR-17-5p reversed the contraction and migration phenotypes induced by epithelial-derived exosomes. Exosomal miR-17-5p was confirmed to function as a key regulator of the phenotypic transformation of fibroblasts. In conclusion, we demonstrated that Arecoline triggers aberrant epithelium-fibroblast crosstalk and identified that epithelial cell-derived miR-17-5p mediates fibroblast differentiation through the classical TGF-β fibrotic pathway, which provided a new perspective and strategy for the diagnosis and treatment of OSF.
Collapse
Affiliation(s)
- Changqing Xie
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Liang Zhong
- Hospital of Stomatology and Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Feng
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Rifu Wang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yuxin Shi
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yonglin Lv
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yanjia Hu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Jing Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Desheng Xiao
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qianming Chen
- Hospital of Stomatology and Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China.
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine Sciences, Central South University, Changsha, China.
| |
Collapse
|
41
|
Li Z, Williams H, Jackson ML, Johnson JL, George SJ. WISP-1 Regulates Cardiac Fibrosis by Promoting Cardiac Fibroblasts' Activation and Collagen Processing. Cells 2024; 13:989. [PMID: 38891121 PMCID: PMC11172092 DOI: 10.3390/cells13110989] [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: 04/12/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Hypertension induces cardiac fibrotic remodelling characterised by the phenotypic switching of cardiac fibroblasts (CFs) and collagen deposition. We tested the hypothesis that Wnt1-inducible signalling pathway protein-1 (WISP-1) promotes CFs' phenotypic switch, type I collagen synthesis, and in vivo fibrotic remodelling. The treatment of human CFs (HCFs, n = 16) with WISP-1 (500 ng/mL) induced a phenotypic switch (α-smooth muscle actin-positive) and type I procollagen cleavage to an intermediate form of collagen (pC-collagen) in conditioned media after 24h, facilitating collagen maturation. WISP-1-induced collagen processing was mediated by Akt phosphorylation via integrin β1, and disintegrin and metalloproteinase with thrombospondin motifs 2 (ADAMTS-2). WISP-1 wild-type (WISP-1+/+) mice and WISP-1 knockout (WISP-1-/-) mice (n = 5-7) were subcutaneously infused with angiotensin II (AngII, 1000 ng/kg/min) for 28 days. Immunohistochemistry revealed the deletion of WISP-1 attenuated type I collagen deposition in the coronary artery perivascular area compared to WISP-1+/+ mice after a 28-day AngII infusion, and therefore, the deletion of WISP-1 attenuated AngII-induced cardiac fibrosis in vivo. Collectively, our findings demonstrated WISP-1 is a critical mediator in cardiac fibrotic remodelling, by promoting CFs' activation via the integrin β1-Akt signalling pathway, and induced collagen processing and maturation via ADAMTS-2. Thereby, the modulation of WISP-1 levels could provide potential therapeutic targets in clinical treatment.
Collapse
Affiliation(s)
- Ze Li
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Helen Williams
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Molly L. Jackson
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Jason L. Johnson
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Sarah J. George
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
- Bristol Heart Institute, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Upper Maudlin St, Bristol BS2 8HW, UK
| |
Collapse
|
42
|
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
|
43
|
Haase M, Comlekoglu T, Petrucciani A, Peirce SM, Blemker SS. Agent-based model demonstrates the impact of nonlinear, complex interactions between cytokinces on muscle regeneration. eLife 2024; 13:RP91924. [PMID: 38828844 PMCID: PMC11147512 DOI: 10.7554/elife.91924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Muscle regeneration is a complex process due to dynamic and multiscale biochemical and cellular interactions, making it difficult to identify microenvironmental conditions that are beneficial to muscle recovery from injury using experimental approaches alone. To understand the degree to which individual cellular behaviors impact endogenous mechanisms of muscle recovery, we developed an agent-based model (ABM) using the Cellular-Potts framework to simulate the dynamic microenvironment of a cross-section of murine skeletal muscle tissue. We referenced more than 100 published studies to define over 100 parameters and rules that dictate the behavior of muscle fibers, satellite stem cells (SSCs), fibroblasts, neutrophils, macrophages, microvessels, and lymphatic vessels, as well as their interactions with each other and the microenvironment. We utilized parameter density estimation to calibrate the model to temporal biological datasets describing cross-sectional area (CSA) recovery, SSC, and fibroblast cell counts at multiple timepoints following injury. The calibrated model was validated by comparison of other model outputs (macrophage, neutrophil, and capillaries counts) to experimental observations. Predictions for eight model perturbations that varied cell or cytokine input conditions were compared to published experimental studies to validate model predictive capabilities. We used Latin hypercube sampling and partial rank correlation coefficient to identify in silico perturbations of cytokine diffusion coefficients and decay rates to enhance CSA recovery. This analysis suggests that combined alterations of specific cytokine decay and diffusion parameters result in greater fibroblast and SSC proliferation compared to individual perturbations with a 13% increase in CSA recovery compared to unaltered regeneration at 28 days. These results enable guided development of therapeutic strategies that similarly alter muscle physiology (i.e. converting extracellular matrix [ECM]-bound cytokines into freely diffusible forms as studied in cancer therapeutics or delivery of exogenous cytokines) during regeneration to enhance muscle recovery after injury.
Collapse
Affiliation(s)
- Megan Haase
- University of VirginiaCharlottesvilleUnited States
| | | | | | | | | |
Collapse
|
44
|
Lv B, Yang L, Gao Y, Li G. Epoxyeicosatrienoic Acids Attenuate LPS-Induced NIH/3T3 Cell Fibrosis through the A 2AR and PI3K/Akt Signaling Pathways. Bull Exp Biol Med 2024; 177:185-189. [PMID: 39090469 DOI: 10.1007/s10517-024-06153-5] [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: 04/28/2022] [Indexed: 08/04/2024]
Abstract
Inflammation plays a crucial role in progression of fibrosis. Epoxyeicosatrienoic acids (EET) have multiple protective effects in different diseases, but their ability to inhibit the development of LPS-induced fibrosis remains unknown. The potential therapeutic effects of 11,12-EET were studied in in vitro model of LPS-induced fibrosis. Mouse embryonic fibroblast cells NIH/3T3 were pre-incubated with 1 μM 11,12-EET and/or a structural analogue and selective EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid before exposing to LPS. The effect of EET was evaluated by the protein and mRNA expression of NF-κB, collagens I and III, and α-smooth muscle actin by Western blotting and quantitative reverse transcription PCR, respectively. LPS provoked inflammation and fibrosis-like changes accompanied by elevated expression of NF-κB and collagens in NIH/3T3 cells. We also studied the effects of 11,12-EET on the A2AR and PI3K/Akt signaling pathways in intact and LPS-treated NIH/3T3 cells. 11,12-EET prevented inflammation and fibrosis-like changes through up-regulation of A2AR and PI3K/Akt signaling pathways. Our findings demonstrate the potential antifibrotic effects of 11,12-EET, which can be natural antagonists of tissue fibrosis.
Collapse
Affiliation(s)
- B Lv
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Diseases, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - L Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Diseases, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Y Gao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Diseases, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - G Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Diseases, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
45
|
Patil M, Singh S, Dubey PK, Tousif S, Umbarkar P, Zhang Q, Lal H, Sewell-Loftin MK, Umeshappa CS, Ghebre YT, Pogwizd S, Zhang J, Krishnamurthy P. Fibroblast-Specific Depletion of Human Antigen R Alleviates Myocardial Fibrosis Induced by Cardiac Stress. JACC Basic Transl Sci 2024; 9:754-770. [PMID: 39070272 PMCID: PMC11282885 DOI: 10.1016/j.jacbts.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 07/30/2024]
Abstract
Cardiac fibrosis can be mitigated by limiting fibroblast-to-myofibroblast differentiation and proliferation. Human antigen R (HuR) modulates messenger RNA stability and expression of multiple genes. However, the direct role of cardiac myofibroblast HuR is unknown. Myofibroblast-specific deletion of HuR limited cardiac fibrosis and preserved cardiac functions in pressure overload injury. Knockdown of HuR in transforming growth factor-β1-treated cardiac fibroblasts suppressed myofibroblast differentiation and proliferation. HuR deletion abrogated the expression and messenger RNA stability of cyclins D1 and A2, suggesting a potential mechanism by which HuR promotes myofibroblast proliferation. Overall, these data suggest that inhibition of HuR could be a potential therapeutic approach to limit cardiac fibrosis.
Collapse
Affiliation(s)
- Mallikarjun Patil
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sarojini Singh
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Praveen Kumar Dubey
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sultan Tousif
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qinkun Zhang
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hind Lal
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Yohannes T. Ghebre
- Department of Radiation Oncology, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Steven Pogwizd
- Comprehensive Cardiovascular Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
46
|
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
|
47
|
Zhao X, Li Y, Yu J, Teng H, Wu S, Wang Y, Zhou H, Li F. Role of mitochondria in pathogenesis and therapy of renal fibrosis. Metabolism 2024; 155:155913. [PMID: 38609039 DOI: 10.1016/j.metabol.2024.155913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Renal fibrosis, specifically tubulointerstitial fibrosis, represents the predominant pathological consequence observed in the context of progressive chronic kidney conditions. The pathogenesis of renal fibrosis encompasses a multifaceted interplay of mechanisms, including but not limited to interstitial fibroblast proliferation, activation, augmented production of extracellular matrix (ECM) components, and impaired ECM degradation. Notably, mitochondria, the intracellular organelles responsible for orchestrating biological oxidation processes in mammalian cells, assume a pivotal role within this intricate milieu. Mitochondrial dysfunction, when manifest, can incite a cascade of events, including inflammatory responses, perturbed mitochondrial autophagy, and associated processes, ultimately culminating in the genesis of renal fibrosis. This comprehensive review endeavors to furnish an exegesis of mitochondrial pathophysiology and biogenesis, elucidating the precise mechanisms through which mitochondrial aberrations contribute to the onset and progression of renal fibrosis. We explored how mitochondrial dysfunction, mitochondrial cytopathy and mitochondrial autophagy mediate ECM deposition and renal fibrosis from a multicellular perspective of mesangial cells, endothelial cells, podocytes, macrophages and fibroblasts. Furthermore, it succinctly encapsulates the most recent advancements in the realm of mitochondrial-targeted therapeutic strategies aimed at mitigating renal fibrosis.
Collapse
Affiliation(s)
- Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Jinyu Yu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Haolin Teng
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
48
|
Rajalekshmi R, Agrawal DK. Understanding Fibrous Tissue in the Effective Healing of Rotator Cuff Injury. JOURNAL OF SURGERY AND RESEARCH 2024; 7:215-228. [PMID: 38872898 PMCID: PMC11174978 DOI: 10.26502/jsr.10020363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The rotator cuff is a crucial group of muscles and tendons in the shoulder complex that plays a significant role in the stabilization of the glenohumeral joint and enabling a wide range of motion. Rotator cuff tendon tears can occur due to sudden injuries or degenerative processes that develop gradually over time, whether they are partial or full thickness. These injuries are common causes of shoulder pain and functional impairment, and their complex nature highlights the essential role of the rotator cuff in shoulder function. Scar formation is a crucial aspect of the healing process initiated following a rotator cuff tendon tear, but excessive fibrous tissue development can potentially lead to stiffness, discomfort, and movement limitations. Age is a critical risk factor, with the prevalence of these tears increasing among older individuals. This comprehensive review aims to delve deeper into the anatomy and injury mechanisms of the rotator cuff. Furthermore, it will inspect the signaling pathways involved in fibrous tissue development, evaluate the various factors affecting the healing environment, and discuss proactive measures aimed at reducing excessive fibrous tissue formation. Lastly, this review identifed gaps within existing knowledge to advance methods for better management of rotator cuff tendon injuries.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| | - Devendra K Agrawal
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| |
Collapse
|
49
|
Xu C, Zhang H, Yang C, Wang Y, Wang K, Wang R, Zhang W, Li C, Tian C, Han C, Li M, Liu X, Wang Y, Li Y, Zhang J, Li Y, Luo L, Shang Y, Zhang L, Chen Y, Shen K, Hu D. miR-125b-5p delivered by adipose-derived stem cell exosomes alleviates hypertrophic scarring by suppressing Smad2. BURNS & TRAUMA 2024; 12:tkad064. [PMID: 38765787 PMCID: PMC11102599 DOI: 10.1093/burnst/tkad064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 05/22/2024]
Abstract
Background Hypertrophic scarring is the most serious and unmet challenge following burn and trauma injury and often leads to pain, itching and even loss of function. However, the demand for ideal scar prevention and treatment is difficult to satisfy. We aimed to discover the effects and mechanisms of adipose-derived stem cell (ADSC) exosomes in hypertrophic scarring. Methods ADSC exosomes were isolated from the culture supernatant of ADSCs and identified by nanoparticle tracking analysis, transmission electron microscopy and western blotting. The effect of ADSC exosomes on wound healing and scar formation was detected by the wound model of BALB/c mice. We isolated myofibroblasts from hypertrophic scar tissue and detected the cell viability, proliferation and migration of myofibroblasts. In addition, collagen formation and fibrosis-related molecules were also detected. To further disclose the mechanism of ADSC exosomes on fibrosis in myofibroblasts, we detected the expression of Smad2 in hypertrophic scar tissue and normal skin and the regulatory mechanism of ADSC exosomes on Smad2. Injection of bleomycin was performed in male BALB/c mice to establish an in vivo fibrosis model while ADSC exosomes were administered to observe their protective effect. The tissue injury of mice was observed via hematoxylin and eosin and Masson staining and related testing. Results In this study, we found that ADSC exosomes could not only speed up wound healing and improve healing quality but also prevent scar formation. ADSC exosomes inhibited expression of fibrosis-related molecules such as α-smooth muscle actin, collagen I (COL1) and COL3 and inhibited the transdifferentiation of myofibroblasts. In addition, we verified that Smad2 is highly expressed in both hypertrophic scar tissue and hypertrophic fibroblasts, while ADSC exosomes downregulated the expression of Smad2 in hypertrophic fibroblasts. Further regulatory mechanism analysis revealed that microRNA-125b-5p (miR-125b-5p) is highly expressed in ADSC exosomes and binds to the 3' untranslated region of Smad2, thus inhibiting its expression. In vivo experiments also revealed that ADSC exosomes could alleviate bleomycin-induced skin fibrosis and downregulate the expression of Smad2. Conclusions We found that ADSC exosomes could alleviate hypertrophic scars via the suppression of Smad2 by the specific delivery of miR-125b-5p.
Collapse
Affiliation(s)
- Chaolei Xu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Hao Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Chen Yang
- Department of Plastic Surgery, Burns and Cosmetology, The First Affiliated Hospital of Xi’an Medical University, Xi’an 710032, China
| | - Ying Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Kejia Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Rui Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Wei Zhang
- Department of Plastic Surgery, Burns and Cosmetology, The First Affiliated Hospital of Xi’an Medical University, Xi’an 710032, China
| | - Chao Li
- Department of Plastic Surgery, Burns and Cosmetology, The First Affiliated Hospital of Xi’an Medical University, Xi’an 710032, China
| | - Chenyang Tian
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Chao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Mengyang Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Xu Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yunwei Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Jian Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yu Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yage Shang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Lixia Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Yuxi Chen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
50
|
Acharya P, Parkins S, Tranter M. RNA binding proteins as mediators of pathological cardiac remodeling. Front Cell Dev Biol 2024; 12:1368097. [PMID: 38818408 PMCID: PMC11137256 DOI: 10.3389/fcell.2024.1368097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
RNA binding proteins (RBPs) play a central in the post-transcriptional regulation of gene expression, which can account for up to 50% of all variations in protein expression within a cell. Following their binding to target RNAs, RBPs most typically confer changes in gene expression through modulation of alternative spicing, RNA stabilization/degradation, or ribosome loading/translation rate. All of these post-transcriptional regulatory processes have been shown to play a functional role in pathological cardiac remodeling, and a growing body of evidence is beginning to identify the mechanistic contribution of individual RBPs and their cardiac RNA targets. This review highlights the mechanisms of RBP-dependent post-transcriptional gene regulation in cardiomyocytes and fibroblasts and our current understanding of how RNA binding proteins functionally contribute to pathological cardiac remodeling.
Collapse
Affiliation(s)
- Pooja Acharya
- Department of Molecular Medicine and Therapeutics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sharon Parkins
- Department of Molecular Medicine and Therapeutics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michael Tranter
- Department of Molecular Medicine and Therapeutics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|