1
|
Docshin P, Panshin D, Malashicheva A. Molecular Interplay in Cardiac Fibrosis: Exploring the Functions of RUNX2, BMP2, and Notch. Rev Cardiovasc Med 2024; 25:368. [PMID: 39484128 PMCID: PMC11522771 DOI: 10.31083/j.rcm2510368] [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: 04/30/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 11/03/2024] Open
Abstract
Cardiac fibrosis, characterized by the excessive deposition of extracellular matrix proteins, significantly contributes to the morbidity and mortality associated with cardiovascular diseases. This article explores the complex interplay between Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2), and Notch signaling pathways in the pathogenesis of cardiac fibrosis. Each of these pathways plays a crucial role in the regulation of cellular functions and interactions that underpin fibrotic processes in the heart. Through a detailed review of current research, we highlight how the crosstalk among RUNX2, BMP2, and Notch not only facilitates our understanding of the fibrotic mechanisms but also points to potential biomolecular targets for intervention. This article delves into the regulatory networks, identifies key molecular mediators, and discusses the implications of these signaling pathways in cardiac structural remodeling. By synthesizing findings from recent studies, we provide insights into the cellular and molecular mechanisms that could guide future research directions, aiming to uncover new therapeutic strategies to manage and treat cardiac fibrosis effectively.
Collapse
Affiliation(s)
- Pavel Docshin
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| | - Daniil Panshin
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| |
Collapse
|
2
|
Winters J, Kawczynski MJ, Gilbers MD, Isaacs A, Zeemering S, Bidar E, Maesen B, Rienstra M, van Gelder I, Verheule S, Maessen JG, Schotten U. Circulating BMP10 Levels Associate With Late Postoperative Atrial Fibrillation and Left Atrial Endomysial Fibrosis. JACC Clin Electrophysiol 2024; 10:1326-1340. [PMID: 38639699 DOI: 10.1016/j.jacep.2024.03.003] [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: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Serum bone morphogenetic protein 10 (BMP10) blood levels are a marker for history of atrial fibrillation (AF) and for major adverse cardiovascular events in patients with AF, including stroke, AF recurrences after catheter ablations, and mortality. The predictive value of BMP10 in patients undergoing cardiac surgery and association with morphologic properties of atrial tissues are unknown. OBJECTIVES This study sought to study the correlation between BMP10 levels and preoperative clinical traits, occurrence of early and late postoperative atrial fibrillation (POAF), and atrial fibrosis in patients undergoing cardiac surgery. METHODS Patients with and without preoperative AF history undergoing first cardiac surgery were included (RACE V, n = 147). Preoperative blood biomarkers were analyzed, left (n = 114) and right (n = 125) atrial appendage biopsy specimens were histologically investigated after WGA staining, and postoperative rhythm was monitored continuously with implantable loop recorders (n = 133, 2.5 years). RESULTS Adjusted multinomial logistic regression indicated that BMP10 accurately reflected a history of persistent AF (OR: 1.24, 95% CI: 1.10-1.40, P = 0.001), similar to NT-pro-BNP. BMP10 levels were associated with increased late POAF90 occurrence after adjustment for age, sex, AF history, and early POAF occurrence (HR: 1.07 [per 0.1 ng/mL increase], 95% CI: 1.00-1.14, P = 0.041). Left atrial endomysial fibrosis (standardized β = 0.22, P = 0.041) but not overall fibrosis (standardized Β = 0.12, P = 0.261) correlated with circulating BMP10 after adjustment for age, sex, AF history, reduced LVF, and valvular surgery indication. CONCLUSIONS Increased BMP10 levels were associated with persistent AF history, increased late POAF incidence, and LAA endomysial fibrosis in a diverse sample of patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Joris Winters
- Department of Physiology, Maastricht University, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Michal J Kawczynski
- Department of Physiology, Maastricht University, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Department of Cardiothoracic Surgery, Heart and Vascular Centre Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Martijn D Gilbers
- Department of Physiology, Maastricht University, Maastricht, the Netherlands
| | - Aaron Isaacs
- Department of Physiology, Maastricht University, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Maastricht Centre for Systems Biology, University Maastricht, Maastricht, the Netherlands
| | - Stef Zeemering
- Department of Physiology, Maastricht University, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Elham Bidar
- Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Department of Cardiothoracic Surgery, Heart and Vascular Centre Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Bart Maesen
- Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Department of Cardiothoracic Surgery, Heart and Vascular Centre Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Centre, Groningen, the Netherlands
| | - Isabelle van Gelder
- Department of Cardiology, University of Groningen, University Medical Centre, Groningen, the Netherlands
| | - Sander Verheule
- Department of Physiology, Maastricht University, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Jos G Maessen
- Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; Department of Cardiothoracic Surgery, Heart and Vascular Centre Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Ulrich Schotten
- Department of Physiology, Maastricht University, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands.
| |
Collapse
|
3
|
Shen J, Liang J, Rejiepu M, Ma Z, Zhao J, Li J, Zhang L, Yuan P, Wang J, Tang B. Analysis of immunoinfiltration and EndoMT based on TGF-β signaling pathway-related genes in acute myocardial infarction. Sci Rep 2024; 14:5183. [PMID: 38431730 PMCID: PMC10908777 DOI: 10.1038/s41598-024-55613-5] [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: 09/17/2023] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
Acute myocardial infarction (AMI), a critical manifestation of coronary heart disease, presents a complex and not entirely understood etiology. This study investigates the potential role of immune infiltration and endothelial-mesenchymal transition (EndoMT) in AMI pathogenesis. We conducted an analysis of the GSE24519 and MSigDB datasets to identify differentially expressed genes associated with the TGF-β signaling pathway (DE-TSRGs) and carried out a functional enrichment analysis. Additionally, we evaluated immune infiltration in AMI and its possible link to myocardial fibrosis. Key genes were identified using machine learning and LASSO logistic regression. The expression of MEOX1 in the ventricular muscles and endothelial cells of Sprague-Dawley rats was assessed through RT-qPCR, immunohistochemical and immunofluorescence assays, and the effect of MEOX1 overexpression on EndoMT was investigated. Our study identified five DE-TSRGs, among which MEOX1, SMURF1, and SPTBN1 exhibited the most significant associations with AMI. Notably, we detected substantial immune infiltration in AMI specimens, with a marked increase in neutrophils and macrophages. MEOX1 demonstrated consistent expression patterns in rat ventricular muscle tissue and endothelial cells, and its overexpression induced EndoMT. Our findings suggest that the TGF-β signaling pathway may contribute to AMI progression by activating the immune response. MEOX1, linked to the TGF-β signaling pathway, appears to facilitate myocardial fibrosis via EndoMT following AMI. These novel insights into the mechanisms of AMI pathogenesis could offer promising therapeutic targets for intervention.
Collapse
Affiliation(s)
- Jun Shen
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China.
| | - Junqing Liang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Manzeremu Rejiepu
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhiqin Ma
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Jixian Zhao
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Jia Li
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Ling Zhang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| | - Ping Yuan
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China.
| | - Jianing Wang
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China.
| | - Baopeng Tang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
4
|
Chen X, Ma J, Wang ZW, Wang Z. The E3 ubiquitin ligases regulate inflammation in cardiovascular diseases. Semin Cell Dev Biol 2024; 154:167-174. [PMID: 36872193 DOI: 10.1016/j.semcdb.2023.02.008] [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: 02/03/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023]
Abstract
Accumulating evidence has illustrated that the E3 ubiquitin ligases critically participate in the development and progression of cardiovascular diseases. Dysregulation of E3 ubiquitin ligases exacerbates cardiovascular diseases. Blockade or activation of E3 ubiquitin ligases mitigates cardiovascular performance. Therefore, in this review, we mainly introduced the critical role and underlying molecular mechanisms of E3 ubiquitin ligase NEDD4 family in governing the initiation and progression of cardiovascular diseases, including ITCH, WWP1, WWP2, Smurf1, Smurf2, Nedd4-1 and Nedd4-2. Moreover, the functions and molecular insights of other E3 ubiquitin ligases, such as F-box proteins, in cardiovascular disease development and malignant progression are described. Furthermore, we illustrate several compounds that alter the expression of E3 ubiquitin ligases to alleviate cardiovascular diseases. Therefore, modulation of E3 ubiquitin ligases could be a novel and promising strategy for improvement of therapeutic efficacy of deteriorative cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jia Ma
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Zhi-Wei Wang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Zhiting Wang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
5
|
Polopalli S, Saha A, Niri P, Kumar M, Das P, Kamboj DV, Chattopadhyay P. ROCK Inhibitors as an Alternative Therapy for Corneal Grafting: A Systematic Review. J Ocul Pharmacol Ther 2023; 39:585-599. [PMID: 37738326 DOI: 10.1089/jop.2023.0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023] Open
Abstract
Currently, corneal blindness is affecting >10 million individuals worldwide, and there is a significant unmet medical need because only 1.5% of transplantation needs are met globally due to a lack of high-quality grafts. In light of this global health disaster, researchers are developing corneal substitutes that can resemble the human cornea in vivo and replace human donor tissue. Thus, this review examines ROCK (Rho-associated coiled-coil containing protein kinases) inhibitors as a potential corneal wound-healing (CWH) therapy by reviewing the existing clinical and nonclinical findings. The systematic review was done from PubMed, Scopus, Web of Science, and Google Scholar for CWH, corneal injury, corneal endothelial wound healing, ROCK inhibitors, Fasudil, Netarsudil, Ripasudil, Y-27632, clinical trial, clinical study, case series, case reports, preclinical study, in vivo, and in vitro studies. After removing duplicates, all downloaded articles were examined. The literature search included the data till January 2023. This review summarized the results of ROCK inhibitors in clinical and preclinical trials. In a clinical trial, various ROCK inhibitors improved CWH in individuals with open-angle glaucoma, cataract, iris cyst, ocular hypertension, and other ocular diseases. ROCK inhibitors also improved ocular wound healing by increasing cell adhesion, migration, and proliferation in vitro and in vivo. ROCK inhibitors have antifibrotic, antiangiogenic, anti-inflammatory, and antiapoptotic characteristics in CWH, according to the existing research. ROCK inhibitors were effective topical treatments for corneal infections. Ripasudil, Y-27632, H-1152, Y-39983, and AMA0526 are a few new ROCK inhibitors that may help CWH and replace human donor tissue.
Collapse
Affiliation(s)
- Subramanyam Polopalli
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Pakter Niri
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Mohit Kumar
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| | - Parikshit Das
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| | - Dev Vrat Kamboj
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| |
Collapse
|
6
|
Mahmood N, Sefat E, Roberts D, Gilger BC, Gluck JM. Application of Noggin-Coated Electrospun Scaffold in Corneal Wound Healing. Transl Vis Sci Technol 2023; 12:15. [PMID: 37594449 PMCID: PMC10445176 DOI: 10.1167/tvst.12.8.15] [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: 12/16/2022] [Accepted: 07/16/2023] [Indexed: 08/19/2023] Open
Abstract
Purpose The objective of this study is to develop and characterize electrospun corneal bandage infused with Noggin protein and evaluate its therapeutic potential in the treatment of superficial nonhealing corneal ulceration. Methods Electrospun nanofibrous scaffolds were created with different blend ratios of polycaprolactone and gelatin and coated with different concentrations of Noggin protein. Morphologic, mechanical, degradation, and surface chemistry of the developed scaffold was assessed. Biocompatibility of the developed scaffold with corneal epithelial cells was evaluated by looking at cell viability, proliferation, and immunostaining. In vitro wound healing in the presence of Noggin-coated scaffold was evaluated by measuring wound closure rate after scratch. Results Uniform nanofibrous scaffolds coated with Noggin were constructed through optimization of electrospinning parameters and demonstrated mechanical properties better than or similar to commercially available contact lenses used in corneal wound healing. In the presence of Noggin-coated scaffold, corneal epithelial cells showed higher proliferation and wound-healing rate. Conclusions This Noggin-coated electrospun scaffold represents a step toward, expanding treatment options for patients with indolent corneal ulcers. Translational Relevance In this study, the feasibility of Noggin-coated electrospun scaffold as a therapeutic for indolent corneal ulcer was evaluated. This study also provides a better perspective for understanding electrospun scaffolds as a tunable platform to infuse topical therapeutics and use as a corneal bandage.
Collapse
Affiliation(s)
- Nasif Mahmood
- Department of Textile Engineering, Chemistry, and Science, Wilson College of Textiles, North Carolina State University, Raleigh, NC, USA
| | - Eelya Sefat
- Department of Textile Engineering, Chemistry, and Science, Wilson College of Textiles, North Carolina State University, Raleigh, NC, USA
| | - Darby Roberts
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Brian C. Gilger
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Jessica M. Gluck
- Department of Textile Engineering, Chemistry, and Science, Wilson College of Textiles, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
7
|
Bertaud A, Joshkon A, Heim X, Bachelier R, Bardin N, Leroyer AS, Blot-Chabaud M. Signaling Pathways and Potential Therapeutic Strategies in Cardiac Fibrosis. Int J Mol Sci 2023; 24:ijms24021756. [PMID: 36675283 PMCID: PMC9866199 DOI: 10.3390/ijms24021756] [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: 12/19/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
Cardiac fibrosis constitutes irreversible necrosis of the heart muscle as a consequence of different acute (myocardial infarction) or chronic (diabetes, hypertension, …) diseases but also due to genetic alterations or aging. Currently, there is no curative treatment that is able to prevent or attenuate this phenomenon that leads to progressive cardiac dysfunction and life-threatening outcomes. This review summarizes the different targets identified and the new strategies proposed to fight cardiac fibrosis. Future directions, including the use of exosomes or nanoparticles, will also be discussed.
Collapse
|
8
|
Pisciotta A, Di Tinco R, Bertani G, Orlandi G, Bertoni L, Pignatti E, Orciani M, Sena P, Bertacchini J, Salvarani C, Carnevale G. Human dental pulp stem cells (hDPSCs) promote the lipofibroblast transition in the early stage of a fibro-inflammatory process. Front Cell Dev Biol 2023; 11:1196023. [PMID: 37206922 PMCID: PMC10189147 DOI: 10.3389/fcell.2023.1196023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction: In autoimmune diseases, particularly in systemic sclerosis and chronic periaortitis, a strict correlation between chronic inflammation and fibrosis exists. Since the currently used drugs prove mostly effective in suppressing inflammation, a better comprehension of the molecular mechanisms exerted by cell types implicated in fibro-inflammation is needed to develop novel therapeutic strategies. Mesenchymal stromal/stem cells (MSCs) are being matter of deep investigation to unveil their role in the evolution of fibrogenetic process. Several findings pointed out the controversial implication of MSCs in these events, with reports lining at a beneficial effect exerted by external MSCs and others highlighting a direct contribution of resident MSCs in fibrosis progression. Human dental pulp stem cells (hDPSCs) have demonstrated to hold promise as potential therapeutic tools due to their immunomodulatory properties, which strongly support their contribution to tissue regeneration. Methods: Our present study evaluated hDPSCs response to a fibro-inflammatory microenvironment, mimicked in vitro by a transwell co-culture system with human dermal fibroblasts, at early and late culture passages, in presence of TGF-β1, a master promoter of fibrogenesis. Results and Discussion: We observed that hDPSCs, exposed to acute fibro-inflammatory stimuli, promote a myofibroblast-to-lipofibroblast transition, likely based on BMP2 dependent pathways. Conversely, when a chronic fibro-inflammatory microenvironment is generated, hDPSCs reduce their anti-fibrotic effect and acquire a pro-fibrotic phenotype. These data provide the basis for further investigations on the response of hDPSCs to varying fibro-inflammatory conditions.
Collapse
Affiliation(s)
- Alessandra Pisciotta
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- *Correspondence: Alessandra Pisciotta,
| | - Rosanna Di Tinco
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Bertani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Orlandi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Bertoni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Pignatti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Paola Sena
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Jessika Bertacchini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Salvarani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Rheumatology, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
9
|
Zhu H, Ji H, Chen W, Han L, Yu L. Integrin subunit β-like 1 mediates angiotensin II-induced myocardial fibrosis by regulating the forkhead box Q1/Snail axis. Arch Biochem Biophys 2022; 730:109422. [DOI: 10.1016/j.abb.2022.109422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022]
|
10
|
Tan Z, Si Y, Yu Y, Ding J, Huang L, Xu Y, Zhang H, Lu Y, Wang C, Yu B, Yuan L. Yi-Shen-Hua-Shi Granule Alleviates Adriamycin-Induced Glomerular Fibrosis by Suppressing the BMP2/Smad Signaling Pathway. Front Pharmacol 2022; 13:917428. [PMID: 35784691 PMCID: PMC9240271 DOI: 10.3389/fphar.2022.917428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a common clinical condition with manifestations of nephrotic syndrome and fibrosis of the glomeruli and interstitium. Yi-Shen-Hua-Shi (YSHS) granule has been shown to have a good effect in alleviating nephrotic syndrome (NS) in clinical and in animal models of FSGS, but whether it can alleviate renal fibrosis in FSGS and its mechanism and targets are not clear. In this study, we explored the anti-fibrotic effect and the targets of the YSHS granule in an adriamycin (ADR)-induced FSGS model and found that the YSHS granule significantly improved the renal function of ADR-induced FSGS model mice and also significantly reduced the deposition of collagen fibers and the expression of mesenchymal cell markers FN, vimentin, and α-SMA in the glomeruli of ADR-induced FSGS mice, suggesting that the YSHS granule inhibited the fibrosis of sclerotic glomeruli. Subsequently, a network pharmacology-based approach was used to identify the potential targets of the YSHS granule for the alleviation of glomerulosclerosis in FSGS, and the results showed that the YSHS granule down-regulated the expressions of BMP2, GSTA1, GATS3, BST1, and S100A9 and up-regulated the expressions of TTR and GATM in ADR-induced FSGS model mice. We also proved that the YSHS granule inhibited the fibrosis in the glomeruli of ADR-induced FSGS model mice through the suppression of the BMP2/Smad signaling pathway.
Collapse
Affiliation(s)
- Zhuojing Tan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yachen Si
- Department of Internal Medicine, No. 944 Hospital of Joint Logistics Support Force, Jiuquan, China
| | - Yan Yu
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiarong Ding
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Linxi Huang
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Xu
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hongxia Zhang
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yihan Lu
- Nanjing Medical University, Nanjing, China
| | - Chao Wang
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
- *Correspondence: Li Yuan, ; Bing Yu, ; Chao Wang,
| | - Bing Yu
- Department of Cell Biology, Naval Medical University (Second Military Medical University), Shanghai, China
- *Correspondence: Li Yuan, ; Bing Yu, ; Chao Wang,
| | - Li Yuan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Li Yuan, ; Bing Yu, ; Chao Wang,
| |
Collapse
|
11
|
A probabilistic Boolean model on hair follicle cell fate regulation by TGF-β. Biophys J 2022; 121:2638-2652. [PMID: 35714600 DOI: 10.1016/j.bpj.2022.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
Hair follicles (HFs) are mini skin organs that undergo cyclic growth. Various signals regulate HF cell fate decisions jointly. Recent experimental results suggest that transforming growth factor beta (TGF-β) exhibits a dual role in HF cell fate regulation that can be either anti- or pro-apoptosis. To understand the underlying mechanisms of HF cell fate control, we develop a novel probabilistic Boolean network (pBN) model on the HF epithelial cell gene regulation dynamics. First, the model is derived from literature, then refined using single-cell RNA sequencing data. Using the model, we both explore the mechanisms underlying HF cell fate decisions and make predictions that could potentially guide future experiments: 1) we propose that a threshold-like switch in the TGF-β strength may necessitate the dual roles of TGF-β in either activating apoptosis or cell proliferation, in cooperation with Bmp and tumor necrosis factor (TNF) and at different stages of a follicle growth cycle; 2) our model shows concordance with the high-activator-low-inhibitor theory of anagen initiation; 3) we predict that TNF may be more effective in catagen initiation than TGF-β, and they may cooperate in a two-step fashion; 4) finally, predictions of gene knockout and overexpression reveal the roles in HF cell fate regulations of each gene. Attractor and motif analysis from the associated Boolean networks reveal the relations between the topological structure of the gene regulation network and the cell fate regulation mechanism. A discrete spatial model equipped with the pBN illustrates how TGF-β and TNF cooperate in initiating and driving the apoptosis wave during catagen.
Collapse
|
12
|
Liu Q, Li HY, Wang SJ, Huang SQ, Yue Y, Maihemuti A, Zhang Y, Huang L, Luo L, Feng KN, Wu ZK. Belumosudil, ROCK2-Specific Inhibitor, alleviates cardiac fibrosis by inhibiting cardiac fibroblasts activation. Am J Physiol Heart Circ Physiol 2022; 323:H235-H247. [PMID: 35657612 DOI: 10.1152/ajpheart.00014.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac fibrosis is thought to be the hallmark of pathological hypertrophic remodeling, of which the myofibroblasts transdifferentiation is the key cell biological event. However, there is still no specific and effective therapeutic agent approved for cardiac fibrosis. To investigate the effects of Belumosudil, the first ROCK2-specific inhibitor, on cardiac hypertrophy, fibrosis and dysfunction induced by pressure overload, the transverse aortic constriction (TAC) or sham operation was carried out on wild-type C57BL/6 mice (male, 6-8 week old) under pentobarbital anesthesia. After that, mice were randomly divided into three groups: sham operation + vehicle, TAC + vehicle, TAC + 50 mg·kg-1·d-1 Belumosudil. We found that Belumosudil effectively ameliorated cardiac hypertrophy, fibrosis and dysfunction in TAC mice. To elucidate the underlying mechanism, we inhibited the expression of ROCK2 in vitro by either Belumosudil or siRNA. We showed that the inhibition of ROCK2 by either Belumosudil or knockdown suppressed cardiac fibroblasts activation and proliferation significantly induced by Transforming Growth Factor-β1 (TGF-β1). Furthermore, our study confirmed ROCK2 mediates cardiac fibrosis by interacting with Transforming Growth Factor-β1 (TGF-β1)/mothers against decapentaplegic homolog (Smad2) pathway. Taken together, we demonstrated that Belumosudil ameliorates cardiac hypertrophy and fibrosis induced by TAC via inhibiting cardiac fibroblasts activation. In conclusion, Belumosudil may be a promising therapeutic drug for cardiac hypertrophy and fibrosis induced by myocardial pressure overload.
Collapse
Affiliation(s)
- Quan Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Hua-Yang Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Shun-Jun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Sui-Qing Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yuan Yue
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Adilai Maihemuti
- Department of Operating Room, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yi Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Lin Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Li Luo
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Kang-Ni Feng
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhong-Kai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
13
|
Su CJ, Murugan A, Linton JM, Yeluri A, Bois J, Klumpe H, Langley MA, Antebi YE, Elowitz MB. Ligand-receptor promiscuity enables cellular addressing. Cell Syst 2022; 13:408-425.e12. [PMID: 35421362 PMCID: PMC10897978 DOI: 10.1016/j.cels.2022.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 11/08/2021] [Accepted: 03/16/2022] [Indexed: 12/24/2022]
Abstract
In multicellular organisms, secreted ligands selectively activate, or "address," specific target cell populations to control cell fate decision-making and other processes. Key cell-cell communication pathways use multiple promiscuously interacting ligands and receptors, provoking the question of how addressing specificity can emerge from molecular promiscuity. To investigate this issue, we developed a general mathematical modeling framework based on the bone morphogenetic protein (BMP) pathway architecture. We find that promiscuously interacting ligand-receptor systems allow a small number of ligands, acting in combinations, to address a larger number of individual cell types, defined by their receptor expression profiles. Promiscuous systems outperform seemingly more specific one-to-one signaling architectures in addressing capability. Combinatorial addressing extends to groups of cell types, is robust to receptor expression noise, grows more powerful with increases in the number of receptor variants, and is maximized by specific biochemical parameter relationships. Together, these results identify design principles governing cellular addressing by ligand combinations.
Collapse
Affiliation(s)
- Christina J Su
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Arvind Murugan
- Department of Physics, University of Chicago, Chicago, IL 60637, USA
| | - James M Linton
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Akshay Yeluri
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Justin Bois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Heidi Klumpe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Matthew A Langley
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yaron E Antebi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
14
|
Qi Y, Tang Y, Yin L, Ding K, Zhao C, Yan W, Yao Y. miR-129-5p restores cardiac function in rats with chronic heart failure by targeting the E3 ubiquitin ligase Smurf1 and promoting PTEN expression. Bioengineered 2022; 13:2371-2386. [PMID: 35034538 PMCID: PMC8974089 DOI: 10.1080/21655979.2021.2024335] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chronic heart failure (CHF) is a prevalent health concern with complex pathogenesis. This current study set out to estimate the function of the miR-129-5p/Smurf1/PTEN axis on cardiac function injury in CHF. The model of CHF in rats was established. The cardiac function indexes, myocardial tissue damage, and oxidative stress-related factors in CHF rats were evaluated after the interference of Smurf1/miR-129-5p/PTEN. The targeting relationships between miR-129-5p and Smurf1 and between PTEN and Smurf1 were verified. It was found that that after modeling, cardiac functions were impaired, heart/left ventricular/lung weight and the myocardial structure was destroyed, and the degree of fibrosis of myocardial tissue was increased. After Smurf1 knockdown, the cardiac function, myocardial structure, and oxidative stress were improved, and the fibrosis in myocardial tissue was decreased. Smurf1 was a target of miR-129-5p. miR-129-5p could annul the protective effect of Smurf1 silencing on CHF rats. Smurf1 inhibited PTEN expression by promoting PTEN ubiquitination, while miR-129-5p enhanced PTEN expression by inhibiting Smurf1. Meanwhile, overexpression of PTEN annulled the cardiac dysfunction in CHF rats induced by Smurf1. In conclusion, miR-129-5p targeted Smurf1 and repressed the ubiquitination of PTEN, and promoted PTEN expression, thus improving the cardiac function of CHF rats.
Collapse
Affiliation(s)
- Yuan Qi
- Department of Cardiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Yan Tang
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Lu Yin
- Department of Cardiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Keke Ding
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Cuimei Zhao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Wenwen Yan
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Yi'an Yao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
15
|
Oouchi Y, Watanabe M, Ida Y, Ohguro H, Hikage F. Rosiglitasone and ROCK Inhibitors Modulate Fibrogenetic Changes in TGF-β2 Treated Human Conjunctival Fibroblasts (HconF) in Different Manners. Int J Mol Sci 2021; 22:ijms22147335. [PMID: 34298955 PMCID: PMC8307967 DOI: 10.3390/ijms22147335] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/16/2021] [Accepted: 07/02/2021] [Indexed: 02/08/2023] Open
Abstract
Purpose: The effects of Rho-associated coiled-coil containing protein kinase (ROCK) 1 and 2 inhibitor, ripasudil hydrochloride hydrate (Rip), ROCK2 inhibitor, KD025 or rosiglitazone (Rosi) on two-dimension (2D) and three-dimension (3D) cultured human conjunctival fibroblasts (HconF) treated by transforming growth factor (TGFβ2) were studied. Methods: Two-dimension and three-dimension cultured HconF were examined by transendothelial electrical resistance (TEER, 2D), size and stiffness (3D), and the expression of the extracellular matrix (ECM) including collagen1 (COL1), COL4 and COL6, fibronectin (FN), and α-smooth muscle actin (αSMA) by quantitative PCR (2D, 3D) in the presence of Rip, KD025 or Rosi. Results: TGFβ2 caused a significant increase in (1) the TEER values (2D) which were greatly reduced by Rosi, (2) the stiffness of the 3D organoids which were substantially reduced by Rip or KD025, and (3) TGFβ2 induced a significant up-regulation of all ECMs, except for COL6 (2D) or αSMA (3D), and down-regulation of COL6 (2D). Rosi caused a significant up-regulation of COL1, 4 and 6 (3D), and down-regulation of COL6 (2D) and αSMA (3D). Most of these TGFβ2-induced expressions in the 2D and αSMA in the 3D were substantially inhibited by KD025, but COL4 and αSMA in 2D were further enhanced by Rip. Conclusion: The findings reported herein indicate that TGFβ2 induces an increase in fibrogenetic changes on the plane and in the spatial space, and are inhibited by Rosi and ROCK inhibitors, respectively.
Collapse
|
16
|
The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int J Mol Sci 2021; 22:ijms22116065. [PMID: 34199773 PMCID: PMC8199989 DOI: 10.3390/ijms22116065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.
Collapse
|
17
|
Assembly of the Cardiac Pacemaking Complex: Electrogenic Principles of Sinoatrial Node Morphogenesis. J Cardiovasc Dev Dis 2021; 8:jcdd8040040. [PMID: 33917972 PMCID: PMC8068396 DOI: 10.3390/jcdd8040040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/31/2021] [Accepted: 04/05/2021] [Indexed: 11/24/2022] Open
Abstract
Cardiac pacemaker cells located in the sinoatrial node initiate the electrical impulses that drive rhythmic contraction of the heart. The sinoatrial node accounts for only a small proportion of the total mass of the heart yet must produce a stimulus of sufficient strength to stimulate the entire volume of downstream cardiac tissue. This requires balancing a delicate set of electrical interactions both within the sinoatrial node and with the downstream working myocardium. Understanding the fundamental features of these interactions is critical for defining vulnerabilities that arise in human arrhythmic disease and may provide insight towards the design and implementation of the next generation of potential cellular-based cardiac therapeutics. Here, we discuss physiological conditions that influence electrical impulse generation and propagation in the sinoatrial node and describe developmental events that construct the tissue-level architecture that appears necessary for sinoatrial node function.
Collapse
|
18
|
Horlock D, Kaye DM, Winbanks CE, Gao XM, Kiriazis H, Donner DG, Gregorevic P, McMullen JR, Bernardo BC. Old Drug, New Trick: Tilorone, a Broad-Spectrum Antiviral Drug as a Potential Anti-Fibrotic Therapeutic for the Diseased Heart. Pharmaceuticals (Basel) 2021; 14:ph14030263. [PMID: 33804032 PMCID: PMC7998193 DOI: 10.3390/ph14030263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiac fibrosis is associated with most forms of cardiovascular disease. No reliable therapies targeting cardiac fibrosis are available, thus identifying novel drugs that can resolve or prevent fibrosis is needed. Tilorone, an antiviral agent, can prevent fibrosis in a mouse model of lung disease. We investigated the anti-fibrotic effects of tilorone in human cardiac fibroblasts in vitro by performing a radioisotopic assay for [3H]-proline incorporation as a proxy for collagen synthesis. Exploratory studies in human cardiac fibroblasts treated with tilorone (10 µM) showed a significant reduction in transforming growth factor-β induced collagen synthesis compared to untreated fibroblasts. To determine if this finding could be recapitulated in vivo, mice with established pathological remodelling due to four weeks of transverse aortic constriction (TAC) were administered tilorone (50 mg/kg, i.p) or saline every third day for eight weeks. Treatment with tilorone was associated with attenuation of fibrosis (assessed by Masson's trichrome stain), a favourable cardiac gene expression profile and no further deterioration of cardiac systolic function determined by echocardiography compared to saline treated TAC mice. These data demonstrate that tilorone has anti-fibrotic actions in human cardiac fibroblasts and the adult mouse heart, and represents a potential novel therapy to treat fibrosis associated with heart failure.
Collapse
Affiliation(s)
- Duncan Horlock
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
| | - David M. Kaye
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Department of Cardiology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Medicine, Monash University, Clayton, VIC 3800, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Catherine E. Winbanks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Disease in Central Asia, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi 830054, China
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Daniel G. Donner
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Julie R. McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Department of Medicine, Monash University, Clayton, VIC 3800, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia
- Department of Physiology, Monash University, Clayton, VIC 3800, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Bianca C. Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (D.H.); (D.M.K.); (C.E.W.); (X.-M.G.); (H.K.); (D.G.D.); (P.G.); (J.R.M.)
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia
- Department of Paediatrics, University of Melbourne, VIC 3010, Australia
- Correspondence: ; Tel.: +61-(0)3-8532-1167; Fax: +61-(0)3-8532-1100
| |
Collapse
|
19
|
Frohlich J, Vinciguerra M. Candidate rejuvenating factor GDF11 and tissue fibrosis: friend or foe? GeroScience 2020; 42:1475-1498. [PMID: 33025411 PMCID: PMC7732895 DOI: 10.1007/s11357-020-00279-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Growth differentiation factor 11 (GDF11 or bone morphogenetic protein 11, BMP11) belongs to the transforming growth factor-β superfamily and is closely related to other family member-myostatin (also known as GDF8). GDF11 was firstly identified in 2004 due to its ability to rejuvenate the function of multiple organs in old mice. However, in the past few years, the heralded rejuvenating effects of GDF11 have been seriously questioned by many studies that do not support the idea that restoring levels of GDF11 in aging improves overall organ structure and function. Moreover, with increasing controversies, several other studies described the involvement of GDF11 in fibrotic processes in various organ setups. This review paper focuses on the GDF11 and its pro- or anti-fibrotic actions in major organs and tissues, with the goal to summarize our knowledge on its emerging role in regulating the progression of fibrosis in different pathological conditions, and to guide upcoming research efforts.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, UK.
| |
Collapse
|
20
|
Li J, Huang B, Dong L, Zhong Y, Huang Z. WJ‑MSCs intervention may relieve intrauterine adhesions in female rats via TGF‑β1‑mediated Rho/ROCK signaling inhibition. Mol Med Rep 2020; 23:8. [PMID: 33179074 PMCID: PMC7673328 DOI: 10.3892/mmr.2020.11646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen is a commonly used hormone in the adjuvant treatment of intrauterine adhesion (IUA), which can promote endometrial growth. Stem cell transplantation has also been reported to promote endometrial regeneration in IUA due to its potential differentiative capacity. Human Wharton's jelly mesenchymal stem cells (WJ-MSCs) are isolated from the umbilical cord, possess strong self-renewal and proliferative abilities, and are hypo-immunogenic and non-tumorigenic. Therefore, the present study aimed to investigate the therapeutic effects and underlying mechanism of WJ-MSCs transplantation with estrogen treatment, separately or as a combined therapy, on IUA. The IUA model was established using the ethanol damage method. A total of 50 Sprague-Dawley female rats were randomly divided into the control, IUA model, WJ-MSCs treatment, estrogen treatment and WJ-MSCs+ estrogen treatment groups (n=10/group). WJ-MSCs were injected three times at 5-day intervals. IUA rats in the estrogen group received 0.2 mg/kg estrogen through intragastric administration, once every 2 days for 8 weeks. Morphological changes were evaluated by hematoxylin-eosin staining. Immunohistochemical evaluations of pan-keratin, vimentin, transforming growth factor (TGF)-β1, RhoA, RhoB, RhoC, Rho-associated coiled-coil-containing protein kinase (ROCK)I, and ROCKII expression were performed in uterine tissue. After treatment, the uterine specimens were observed to have increased uterine thickness and gland numbers in all treatment groups compared with the IUA group; however, the degree of restoration in the independent WJ-MSCs and estrogen treatment groups was better than in the combined treatment group. Immunohistochemical analysis demonstrated that pan-keratin expression was increased, and RhoA, ROCKI and TGF-β1 expression was significantly inhibited in the WJ-MSCs and WJ-MSCs + estrogen treatment groups compared with the IUA group; however, the expression levels of these proteins were similar among all treatment groups. No change in vimentin expression was detected in any treatment group. The expression levels of RhoB, RhoC and ROCKII were clearly not affected by WJ-MSCs intervention alone. In conclusion, transplantation of WJ-MSCs may repair endometrial damage in IUA rats via TGF-β1-mediated inhibition of RhoA/ROCKI signaling.
Collapse
Affiliation(s)
- Jun Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo Huang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lan Dong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yajuan Zhong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhixin Huang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
21
|
Azar F, Courtet K, Dekky B, Bonnier D, Dameron O, Colige A, Legagneux V, Théret N. Integration of miRNA-regulatory networks in hepatic stellate cells identifies TIMP3 as a key factor in chronic liver disease. Liver Int 2020; 40:2021-2033. [PMID: 32306499 DOI: 10.1111/liv.14476] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Activation of hepatic stellate cells (HSC) is a critical process involved in liver fibrosis. Several miRNAs are implicated in gene regulation during this process but their exact and respective contribution is still incompletely understood. Here we propose an integrative approach of miRNA-regulatory networks to predict new targets. METHODS miRNA regulatory networks in activated HSCs were built using lists of validated miRNAs and the CyTargetLinker tool. The resulting graphs were filtered according to public transcriptomic data and the reduced graphs were analysed through GO annotation. A miRNA network regulating the expression of TIMP3 was further studied in human liver samples, isolated hepatic cells and mouse model of liver fibrosis. RESULTS Within the up-regulated miRNAs, we identified a subnetwork of five miRNAs (miR-21-5p, miR-222-3p, miR-221-3p miR-181b-5p and miR-17-5p) that target TIMP3. We demonstrated that TIMP3 expression is inversely associated with inflammatory activity and IL1-ß expression in vivo. We further showed that IL1-ß inhibits TIMP3 expression in HSC-derived LX-2 cells. Using data from The Cancer Genome Atlas (TCGA), we showed that, in hepatocellular carcinoma (HCC), TIMP3 expression is associated with survival (P < .001), while miR-221 (P < .05), miR-222 (P < .01) and miR-181b (P < .01) are markers for a poor prognosis. CONCLUSIONS Several miRNAs targeting TIMP3 are up-regulated in activated HSCs and down-regulation of TIMP3 expression is associated with inflammatory activity in liver fibrosis and poor prognosis in HCC. The regulatory network including specific miRNAs and TIMP3 is therefore central for the evolution of chronic liver disease.
Collapse
Affiliation(s)
- Fida Azar
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Kevin Courtet
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France.,University Rennes, CNRS, IRISA (Institut de recherche en informatique et système aléatoire, Rennes, France
| | - Bassil Dekky
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Dominique Bonnier
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Olivier Dameron
- University Rennes, CNRS, IRISA (Institut de recherche en informatique et système aléatoire, Rennes, France
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-R, University of Liege, Sart Tilman, Belgium
| | - Vincent Legagneux
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Nathalie Théret
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France.,University Rennes, CNRS, IRISA (Institut de recherche en informatique et système aléatoire, Rennes, France
| |
Collapse
|
22
|
You T, Yuan S, Bai L, Zhang X, Chen P, Zhang W. Benzyl alcohol accelerates recovery from Achilles tendon injury, potentially via TGF-β1/Smad2/3 pathway. Injury 2020; 51:1515-1521. [PMID: 32409188 DOI: 10.1016/j.injury.2020.03.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/21/2020] [Accepted: 03/29/2020] [Indexed: 02/02/2023]
Abstract
Benzyl alcohol (BnOH) is a natural colorless liquid organic compound that plays an important role in bacteriostatic and anesthetic processes. It is also used to relieve the nerve and ganglionic pain. In this study, we assessed the effect of topical application of BnOH on the Achilles tendon healing process. Sprague Dawley rats were subjected to an experimentally induced wound in the tendon area and then randomized into four groups. Normal saline (0.5 mL) was applied to rats in control group, and BnOH at the concentrations of 0.5 mL 0.075%, 0.15%, 0.3% were applied to the BnOH treatment groups, respectively. Wound treatment with BnOH led to significantly faster functional recovery than with saline. Moreover, treatment of wounds with 0.3% BnOH accelerated the healing process faster than with 0.075% and 0.15% BnOH. Histological analysis of healed wounds that had been treated with BnOH showed more collagen and blood capillaries and fewer inflammatory cells compared to the control. To study the mechanism of the process, the expression of mRNA of TGF-β1, Smad2/3 and Smad7 and protein of TGF-β1, p-Smad2/3 and Smad7 were quantified by real-time PCR and Western blotting, respectively. Results of this study showed that wounds treated with BnOH significantly enhanced the expression of TGF-β1 and Smad2/3 and reduced the expression of Smad7. In general, the current study demonstrated that BnOH improved the recovery process of tendon healing through the promotion of collagen with angiogenesis and showed that TGF-β plays a role in BnOH treatment of tendon healing.
Collapse
Affiliation(s)
- Tian You
- Sports Medicine Department, Peking University Shenzhen Hospital, 1120th, Lianhua Road, Futian District Shenzhen 518000, Guangdong, China
| | - Shufang Yuan
- Ultrasonic imaging Department, Peking University Shenzhen Hospital, Shenzhen 518000, Guangdong, China
| | - Lu Bai
- Sports Medicine Department, Peking University Shenzhen Hospital, 1120th, Lianhua Road, Futian District Shenzhen 518000, Guangdong, China
| | - Xintao Zhang
- Sports Medicine Department, Peking University Shenzhen Hospital, 1120th, Lianhua Road, Futian District Shenzhen 518000, Guangdong, China
| | - Peng Chen
- Sports Medicine Department, Peking University Shenzhen Hospital, 1120th, Lianhua Road, Futian District Shenzhen 518000, Guangdong, China
| | - Wentao Zhang
- Sports Medicine Department, Peking University Shenzhen Hospital, 1120th, Lianhua Road, Futian District Shenzhen 518000, Guangdong, China
| |
Collapse
|
23
|
Yu B, Yu M, Zhang H, Xie D, Nie W, Shi K. Suppression of miR-143-3p contributes to the anti-fibrosis effect of atorvastatin on myocardial tissues via the modulation of Smad2 activity. Exp Mol Pathol 2020; 112:104346. [DOI: 10.1016/j.yexmp.2019.104346] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/20/2019] [Accepted: 11/19/2019] [Indexed: 01/17/2023]
|
24
|
Ren Q, Lin P, Wang Q, Zhang B, Feng L. Chronic peripheral ghrelin injection exerts antifibrotic effects by increasing growth differentiation factor 15 in rat hearts with myocardial fibrosis induced by isoproterenol. Physiol Res 2019; 69:439-450. [PMID: 31852204 DOI: 10.33549/physiolres.934183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
This study aimed to investigate the anti-fibrotic effects of ghrelin in isoproterenol (ISO)-induced myocardial fibrosis and the underlying mechanism. Sprague-Dawley rats were randomized to control, ISO, and ISO + ghrelin groups. ISO (2 mg/kg per day, subcutaneous) or vehicle was administered once daily for 7 days, then ghrelin (100 microg/kg per day, subcutaneous) was administered once daily for the next 3 weeks. Ghrelin treatment greatly improved the cardiac function of ISO-treated rats. Ghrelin also decreased plasma brain natriuretic peptide level and ratios of heart weight to body weight and left ventricular weight to body weight. Ghrelin significantly reduced myocardial collagen area and hydroxyproline content, accompanied by decreased mRNA levels of collagen type I and III. Furthermore, ghrelin increased plasma level of growth differentiation factor 15 (GDF15) and GDF15 mRNA and protein levels in heart tissues, which were significantly decreased with ISO alone. The phosphorylation of Akt at Ser473 and GSK-3beta at Ser9 was decreased with ISO, and ghrelin significantly reversed the downregulation of p-Akt and p-GSK-3beta. Mediated by GDF15, ghrelin could attenuate ISO-induced myocardial fibrosis via Akt-GSK-3beta signaling.
Collapse
Affiliation(s)
- Q Ren
- Geriatric Department of the Third Hospital of Hangzhou, Hangzhou, China.
| | | | | | | | | |
Collapse
|
25
|
Dituri F, Cossu C, Mancarella S, Giannelli G. The Interactivity between TGFβ and BMP Signaling in Organogenesis, Fibrosis, and Cancer. Cells 2019; 8:E1130. [PMID: 31547567 PMCID: PMC6829314 DOI: 10.3390/cells8101130] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
The Transforming Growth Factor beta (TGFβ) and Bone Morphogenic Protein (BMP) pathways intersect at multiple signaling hubs and cooperatively or counteractively participate to bring about cellular processes which are critical not only for tissue morphogenesis and organogenesis during development, but also for adult tissue homeostasis. The proper functioning of the TGFβ/BMP pathway depends on its communication with other signaling pathways and any deregulation leads to developmental defects or diseases, including fibrosis and cancer. In this review we explore the cellular and physio-pathological contexts in which the synergism or antagonism between the TGFβ and BMP pathways are crucial determinants for the normal developmental processes, as well as the progression of fibrosis and malignancies.
Collapse
Affiliation(s)
- Francesco Dituri
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Carla Cossu
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| |
Collapse
|
26
|
Ning J, Zhao Y, Ye Y, Yu J. Opposing roles and potential antagonistic mechanism between TGF-β and BMP pathways: Implications for cancer progression. EBioMedicine 2019; 41:702-710. [PMID: 30808576 PMCID: PMC6442991 DOI: 10.1016/j.ebiom.2019.02.033] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 02/08/2023] Open
Abstract
The transforming growth factor β (TGF-β) superfamily participates in tumour proliferation, apoptosis, differentiation, migration, invasion, immune evasion and extracellular matrix remodelling. Genetic deficiency in distinct components of TGF-β and BMP-induced signalling pathways or their excessive activation has been reported to regulate the development and progression of some cancers. As more in-depth studies about this superfamily have been conducted, more evidence suggests that the TGF-β and BMP pathways play an opposing role. The cross-talk of these 2 pathways has been widely studied in kidney disease and bone formation, and the opposing effects have also been observed in some cancers. However, the antagonistic mechanisms are still insufficiently investigated in cancer. In this review, we aim to display more evidences and possible mechanisms accounting for the antagonism between these 2 pathways, which might provide some clues for further study in cancer. Describe the basics of TGF-β and BMP signalling Summarize the potential mechanisms accounting for the antagonism between TGF-β and BMP pathways Provide some evidence about the antagonistic effects between pathways observed in some cancers
Collapse
Affiliation(s)
- Junya Ning
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Yi Zhao
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, State Key Laboratory of Computer Architecture, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, PR China
| | - Yingnan Ye
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| |
Collapse
|
27
|
Wang Y, Wu X. SMOC1 silencing suppresses the angiotensin II-induced myocardial fibrosis of mouse myocardial fibroblasts via affecting the BMP2/Smad pathway. Oncol Lett 2018; 16:2903-2910. [PMID: 30127878 PMCID: PMC6096163 DOI: 10.3892/ol.2018.8989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/27/2018] [Indexed: 12/28/2022] Open
Abstract
SPARC-related modular calcium binding 1 (SMOC1) represents a vital member of the SPARC matricellular protein family that regulates cell matrix interaction through binding to cell-surface receptors. The present study aimed to investigate the roles and molecular mechanisms of SMOC1 silencing on the fibrosis of myocardial fibroblasts (MFBs). Cell Counting kit-8 and flow cytometry assays were performed to determine cell viability and reactive oxygen species (ROS) content, respectively. ELISA was performed to detect the expression of associated cytokines and matrix proteins. Western blot analysis and reverse transcription-quantitative polymerase chain reaction assays were used to evaluate the expression of associated proteins and mRNAs, respectively. The results revealed that SMOC1 silencing suppressed the cell viability of angiotensin II (Ang II)-treated MFBs. SMOC1 silencing reduced the ROS content and oxidative stress in MFBs in response to Ang II. Furthermore, SMOC1 silencing downregulated the expression levels of fibrosis-associated proteins in Ang II-treated MFBs. SMOC1 silencing affected the bone morphogenetic protein 2 (BMP2)/Smad signaling pathway in Ang II-treated MFBs. In conclusion, the results of the present study suggested that SMOC1 silencing suppressed the Ang II-induced myocardial fibrosis of mouse MFBs through affecting the BMP2/Smad signaling pathway.
Collapse
Affiliation(s)
- Yize Wang
- Department of Cardiology, Yiyang Central Hospital, Yiyang, Hunan 413000, P.R. China
| | - Xianming Wu
- Department of Cardiology, Yiyang Central Hospital, Yiyang, Hunan 413000, P.R. China
| |
Collapse
|
28
|
Shah TA, Rogers MB. Unanswered Questions Regarding Sex and BMP/TGF-β Signaling. J Dev Biol 2018; 6:jdb6020014. [PMID: 29914150 PMCID: PMC6027345 DOI: 10.3390/jdb6020014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/02/2018] [Accepted: 06/14/2018] [Indexed: 01/23/2023] Open
Abstract
Crosstalk between the BMP and TGF-β signaling pathways regulates many complex developmental processes from the earliest stages of embryogenesis throughout adult life. In many situations, the two signaling pathways act reciprocally. For example, TGF-β signaling is generally pro-fibrotic, whereas BMP signaling is anti-fibrotic and pro-calcific. Sex-specific differences occur in many diseases including cardiovascular pathologies. Differing ratios of fibrosis and calcification in stenotic valves suggests that BMP/TGF-β signaling may vary in men and women. In this review, we focus on the current understanding of the interplay between sex and BMP/TGF-β signaling and pose several unanswered questions.
Collapse
Affiliation(s)
- Tapan A Shah
- Rutgers-New Jersey Medical School, Microbiology, Biochemistry, & Molecular Genetics, Newark, NJ 07103, USA.
| | - Melissa B Rogers
- Rutgers-New Jersey Medical School, Microbiology, Biochemistry, & Molecular Genetics, Newark, NJ 07103, USA.
| |
Collapse
|
29
|
Frangogiannis NG. Fibroblasts and the extracellular matrix in right ventricular disease. Cardiovasc Res 2018; 113:1453-1464. [PMID: 28957531 DOI: 10.1093/cvr/cvx146] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/01/2017] [Indexed: 12/17/2022] Open
Abstract
Right ventricular failure predicts adverse outcome in patients with pulmonary hypertension (PH), and in subjects with left ventricular heart failure and is associated with interstitial fibrosis. This review manuscript discusses the cellular effectors and molecular mechanisms implicated in right ventricular fibrosis. The right ventricular interstitium contains vascular cells, fibroblasts, and immune cells, enmeshed in a collagen-based matrix. Right ventricular pressure overload in PH is associated with the expansion of the fibroblast population, myofibroblast activation, and secretion of extracellular matrix proteins. Mechanosensitive transduction of adrenergic signalling and stimulation of the renin-angiotensin-aldosterone cascade trigger the activation of right ventricular fibroblasts. Inflammatory cytokines and chemokines may contribute to expansion and activation of macrophages that may serve as a source of fibrogenic growth factors, such as transforming growth factor (TGF)-β. Endothelin-1, TGF-βs, and matricellular proteins co-operate to activate cardiac myofibroblasts, and promote synthesis of matrix proteins. In comparison with the left ventricle, the RV tolerates well volume overload and ischemia; whether the right ventricular interstitial cells and matrix are implicated in these favourable responses remains unknown. Expansion of fibroblasts and extracellular matrix protein deposition are prominent features of arrhythmogenic right ventricular cardiomyopathies and may be implicated in the pathogenesis of arrhythmic events. Prevailing conceptual paradigms on right ventricular remodelling are based on extrapolation of findings in models of left ventricular injury. Considering the unique embryologic, morphological, and physiologic properties of the RV and the clinical significance of right ventricular failure, there is a need further to dissect RV-specific mechanisms of fibrosis and interstitial remodelling.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B Bronx, 10461 NY, USA
| |
Collapse
|
30
|
Yang J, Xu T, Gomez DR, Yuan X, Nguyen QN, Jeter M, Song Y, Hahn S, Liao Z. Polymorphisms in BMP2/BMP4, with estimates of mean lung dose, predict radiation pneumonitis among patients receiving definitive radiotherapy for non-small cell lung cancer. Oncotarget 2018; 8:43080-43090. [PMID: 28574846 PMCID: PMC5522129 DOI: 10.18632/oncotarget.17904] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/07/2017] [Indexed: 12/17/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) in TGFβ1 can predict the risk of radiation pneumonitis (RP) in patients with non-small cell lung cancer (NSCLC) after definitive radiotherapy. Here we investigated whether SNPs in TGFβ superfamily members BMP2 and BMP4 are associated with RP in such patients. In total, we retrospectively analyzed 663 patients given ≥ 60 Gy for NSCLC. We randomly assigned 323 patients to the training cohort and 340 patients to the validation cohort. Potentially functional and tagging SNPs of BMP2 (rs170986, rs1979855, rs1980499, rs235768, rs3178250) and BMP4 (rs17563, rs4898820, rs762642) were genotyped. The median of mean lung dose (MLD) was 17.9 Gy (range, 0.15–32.74 Gy). Higher MLD was strongly associated with increased risk of grade ≥ 2 RP (hazard ratio [HR]=2.191, 95% confidence interval [CI] = 1.680–2.856, P < 0.001) and grade ≥ 3 RP (HR = 4.253, 95% CI = 2.493–7.257, P < 0.001). In multivariate analyses, BMP2 rs235768 AT/TT was associated with higher risk of grade ≥ 2 RP (HR = 1.866, 95% CI = 1.221–2.820, P = 0.004 vs. AA) both in training cohort and validation cohort. Similar results were observed for BMP2 rs1980499. BMP2 rs3178250 CT/TT was associated with lower risk of grade ≥ 3 RP (HR = 0.406, 95% CI = 0.175–0.942, P = 0.036 vs. CC) in the pooled analysis. Adding the rs235768 and rs1980499 SNPs to a model comprising age, performance status, and MLD raised the Harrell's C for predicting grade ≥ 2 RP from 0.6117 to 0.6235 (P = 0.0105). SNPs in BMP2 can predict grade ≥ 2 or 3 RP after radiotherapy for NSCLC and improve the predictive power of MLD model. Validation is underway through an ongoing prospective trial.
Collapse
Affiliation(s)
- Ju Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Ting Xu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Daniel R Gomez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Melenda Jeter
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yipeng Song
- Department of Radiation Oncology, Yuhuangding Hospital, Shandong, 264000, China
| | - Stephen Hahn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhongxing Liao
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
31
|
Segers VFM, Brutsaert DL, De Keulenaer GW. Cardiac Remodeling: Endothelial Cells Have More to Say Than Just NO. Front Physiol 2018; 9:382. [PMID: 29695980 PMCID: PMC5904256 DOI: 10.3389/fphys.2018.00382] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022] Open
Abstract
The heart is a highly structured organ consisting of different cell types, including myocytes, endothelial cells, fibroblasts, stem cells, and inflammatory cells. This pluricellularity provides the opportunity of intercellular communication within the organ, with subsequent optimization of its function. Intercellular cross-talk is indispensable during cardiac development, but also plays a substantial modulatory role in the normal and failing heart of adults. More specifically, factors secreted by cardiac microvascular endothelial cells modulate cardiac performance and either positively or negatively affect cardiac remodeling. The role of endothelium-derived small molecules and peptides—for instance NO or endothelin-1—has been extensively studied and is relatively well defined. However, endothelial cells also secrete numerous larger proteins. Information on the role of these proteins in the heart is scattered throughout the literature. In this review, we will link specific proteins that modulate cardiac contractility or cardiac remodeling to their expression by cardiac microvascular endothelial cells. The following proteins will be discussed: IL-6, periostin, tenascin-C, thrombospondin, follistatin-like 1, frizzled-related protein 3, IGF-1, CTGF, dickkopf-3, BMP-2 and−4, apelin, IL-1β, placental growth factor, LIF, WISP-1, midkine, and adrenomedullin. In the future, it is likely that some of these proteins can serve as markers of cardiac remodeling and that the concept of endothelial function and dysfunction might have to be redefined as we learn more about other factors secreted by ECs besides NO.
Collapse
Affiliation(s)
- Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Dirk L Brutsaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Middelheim Hospital, Antwerp, Belgium
| |
Collapse
|
32
|
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor (TGF)-β family of ligands and exert most of their effects through the canonical effectors Smad1, 5, and 8. Appropriate regulation of BMP signaling is critical for the development and homeostasis of numerous human organ systems. Aberrations in BMP pathways or their regulation are increasingly associated with diverse human pathologies, and there is an urgent and growing need to develop effective approaches to modulate BMP signaling in the clinic. In this review, we provide a wide perspective on diseases and/or conditions associated with dysregulated BMP signal transduction, outline the current strategies available to modulate BMP pathways, highlight emerging second-generation technologies, and postulate prospective avenues for future investigation.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, Indiana 46222
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts 02115
| |
Collapse
|
33
|
Altara R, Zouein FA, Brandão RD, Bajestani SN, Cataliotti A, Booz GW. In Silico Analysis of Differential Gene Expression in Three Common Rat Models of Diastolic Dysfunction. Front Cardiovasc Med 2018; 5:11. [PMID: 29556499 PMCID: PMC5850854 DOI: 10.3389/fcvm.2018.00011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/05/2018] [Indexed: 12/13/2022] Open
Abstract
Standard therapies for heart failure with preserved ejection fraction (HFpEF) have been unsuccessful, demonstrating that the contribution of the underlying diastolic dysfunction pathophysiology differs from that of systolic dysfunction in heart failure and currently is far from being understood. Complicating the investigation of HFpEF is the contribution of several comorbidities. Here, we selected three established rat models of diastolic dysfunction defined by three major risk factors associated with HFpEF and researched their commonalities and differences. The top differentially expressed genes in the left ventricle of Dahl salt sensitive (Dahl/SS), spontaneous hypertensive heart failure (SHHF), and diabetes 1 induced HFpEF models were derived from published data in Gene Expression Omnibus and used for a comprehensive interpretation of the underlying pathophysiological context of each model. The diversity of the underlying transcriptomic of the heart of each model is clearly observed by the different panel of top regulated genes: the diabetic model has 20 genes in common with the Dahl/SS and 15 with the SHHF models. Advanced analytics performed in Ingenuity Pathway Analysis (IPA®) revealed that Dahl/SS heart tissue transcripts triggered by upstream regulators lead to dilated cardiomyopathy, hypertrophy of heart, arrhythmia, and failure of heart. In the heart of SHHF, a total of 26 genes were closely linked to cardiovascular disease including cardiotoxicity, pericarditis, ST-elevated myocardial infarction, and dilated cardiomyopathy. IPA Upstream Regulator analyses revealed that protection of cardiomyocytes is hampered by inhibition of the ERBB2 plasma membrane-bound receptor tyrosine kinases. Cardioprotective markers such as natriuretic peptide A (NPPA), heat shock 27 kDa protein 1 (HSPB1), and angiogenin (ANG) were upregulated in the diabetes 1 induced model; however, the model showed a different underlying mechanism with a majority of the regulated genes involved in metabolic disorders. In conclusion, our findings suggest that multiple mechanisms may contribute to diastolic dysfunction and HFpEF, and thus drug therapies may need to be guided more by phenotypic characteristics of the cardiac remodeling events than by the underlying molecular processes.
Collapse
Affiliation(s)
- Raffaele Altara
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, Oslo, Norway.,Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Fouad A Zouein
- Faculty of Medicine, Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Rita Dias Brandão
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Saeed N Bajestani
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States.,Department of Ophthalmology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, Oslo, Norway
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
34
|
TGF-β signaling inhibits canonical BMP signaling pathway during palate development. Cell Tissue Res 2017; 371:283-291. [PMID: 29247325 DOI: 10.1007/s00441-017-2757-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 11/24/2017] [Indexed: 12/20/2022]
Abstract
During early palate development, gene expression and regulation exhibit heterogeneity along the anterior-posterior axis. Transforming growth factor-β (TGF-β) and bone morphogenetic protein (BMP) signaling pathways play essential roles in secondary palatal formation but the exact relationship between the TGF-β and BMP pathways in palate development remains unknown. Here, we demonstrate that, during early secondary palate development, phospho-(p)Smad1/5/8 is highly expressed in the anterior palate but relatively lowly expressed in the posterior palate. Conversely, pSmad2/3 has a lower expression level in the anterior palate than in the posterior palate. With the BRE-Gal reporter, we found that the canonical BMP signaling pathway was not activated in the anterior palate but exhibited a moderate level in the posterior palate. Co-immunoprecipitation revealed that Smad4 bound to pSmad1/5/8 only in the posterior palate and not in the anterior palate. Knocking-out of Tgfbr2 (Wnt1-Cre;Tgfbr2 f/f;BRE) in the palatal mesenchyme enhanced canonical BMP activity in the posterior palate but not in the anterior palate, because of decreased pSmad2/3. pSmad1/5/8-Smad4 complexes were found to be dramatically increased in posterior palatal mesenchymal cells at embryonic day 13.5 cultured in the presence of SB525334. Proximity ligation assay also showed pSmad1/5/8-Smad4 complexes were increased in the posterior palate of Wnt1-Cre;Tgfbr2 f/f mice. Therefore, the reduction of pSmad2/3 level in the palatal mesenchyme of Wnt1-Cre;Tgfbr2 f/f;BRE mice contributes primarily to the increase of pSmad1/5/8-Smad4 complexes leading to enhanced canonical BMP activity in the posterior palate. Moreover, during early development, canonical BMP signaling operates in the posterior palate but is completely absent in the anterior palate. Canonical TGF-β signaling suppresses canonical BMP signaling activity in the posterior palate by competing limited Smad4.
Collapse
|
35
|
Hudnall AM, Arthur JW, Lowery JW. Clinical Relevance and Mechanisms of Antagonism Between the BMP and Activin/TGF-β Signaling Pathways. J Osteopath Med 2017; 116:452-61. [PMID: 27367950 DOI: 10.7556/jaoa.2016.089] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The transforming growth factor β (TGF-β) superfamily is a large group of signaling molecules that participate in embryogenesis, organogenesis, and tissue homeostasis. These molecules are present in all animal genomes. Dysfunction in the regulation or activity of this superfamily's components underlies numerous human diseases and developmental defects. There are 2 distinct arms downstream of the TGF-β superfamily ligands-the bone morphogenetic protein (BMP) and activin/TGF-β signaling pathways-and these 2 responses can oppose one another's effects, most notably in disease states. However, studies have commonly focused on a single arm of the TGF-β superfamily, and the antagonism between these pathways is unknown in most physiologic and pathologic contexts. In this review, the authors summarize the clinically relevant scenarios in which the BMP and activin/TGF-β pathways reportedly oppose one another and identify several molecular mechanisms proposed to mediate this interaction. Particular attention is paid to experimental findings that may be informative to human pathology to highlight potential therapeutic approaches for future investigation.
Collapse
|
36
|
Yu X, Gu P, Huang Z, Fang X, Jiang Y, Luo Q, Li X, Zhu X, Zhan M, Wang J, Fan L, Chen R, Yu J, Gu Y, Liang A, Yi X. Reduced expression of BMP3 contributes to the development of pulmonary fibrosis and predicts the unfavorable prognosis in IIP patients. Oncotarget 2017; 8:80531-80544. [PMID: 29113323 PMCID: PMC5655218 DOI: 10.18632/oncotarget.20083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 07/25/2017] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) and idiopathic nonspecific interstitial pneumonia (INSIP) are two related diseases involving varying degrees of pulmonary fibrosis with no effective cure. Bone morphogenetic protein 3 (BMP3) is a member of the transforming growth factor-β (TGF-β) super-family, which has not been implicated in pulmonary fibrosis previously. In this study, we aimed to investigate the potential role of BMP3 playing in pulmonary fibrosis from clinical diagnosis to molecular signaling regulation. RNA sequencing was performed to explore the potential biomarker of IIP patients. The expression of BMP3 was evaluated in 83 cases of IPF and INSIP by immunohistochemistry. The function of BMP3 was investigated in both fibroblast cells and a bleomycin-induced murine pulmonary fibrosis model. The clinical relevance of BMP3 expression were analyzed in 47 IIP patients, which were included in 83 cases and possess more than five-year follow-up data. Both RNA-sequencing and immunohistochemistry staining revealed that BMP3 was significantly down-regulated in lung tissues of patients with IPF and INSIP. Consistently, lower expression of BMP3 also was found in pulmonary fibrotic tissues of bleomycin-induced mice model. Up-regulation of BMP3 prevented pulmonary fibrosis processing through inhibiting cellular proliferation of fibroblasts as well as TGF-β1 signal transduction. Finally, the relatively higher expression of BMP3 in IPF patients was associated with less/worse mortality. Intravenous injection of recombinant BMP3. Taken together, our results suggested that the low expression level of BMP3 may indicate the unfavorable prognosis of IPF patients, targeting BMP3 may represent a novel potential therapeutic method for pulmonary fibrosis management.
Collapse
Affiliation(s)
- Xiaoting Yu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Pan Gu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ziling Huang
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Xia Fang
- Department of Biotherapy, Tongji Hosptial, Tongji University School of Medicine, Shanghai 200065, China
| | - Ying Jiang
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Qun Luo
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Xia Li
- Department of Respiratory, Shanghai Pulmonary Hospital, Tongji Universiy School of Medicine, Shanghai 200433, China
| | - Xuyou Zhu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Mengna Zhan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbang Wang
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Lichao Fan
- Department of Respiratory, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Rongchang Chen
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Juehua Yu
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Yingying Gu
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Aibin Liang
- Department of Biotherapy, Tongji Hosptial, Tongji University School of Medicine, Shanghai 200065, China
| | - Xianghua Yi
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
37
|
Flevaris P, Khan SS, Eren M, Schuldt AJT, Shah SJ, Lee DC, Gupta S, Shapiro AD, Burridge PW, Ghosh AK, Vaughan DE. Plasminogen Activator Inhibitor Type I Controls Cardiomyocyte Transforming Growth Factor-β and Cardiac Fibrosis. Circulation 2017; 136:664-679. [PMID: 28588076 DOI: 10.1161/circulationaha.117.028145] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/15/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND Fibrosis is the pathological consequence of stress-induced tissue remodeling and matrix accumulation. Increased levels of plasminogen activator inhibitor type I (PAI-1) have been shown to promote fibrosis in multiple organ systems. Paradoxically, homozygous genetic deficiency of PAI-1 is associated with spontaneous age-dependent, cardiac-selective fibrosis in mice. We have identified a novel PAI-1-dependent mechanism that regulates cardiomyocyte-derived fibrogenic signals and cardiac transcriptional pathways during injury. METHODS Cardiac fibrosis in subjects with homozygous mutation in SERPINE-1 was evaluated with late gadolinium-enhanced cardiac magnetic resonance imaging. A murine cardiac injury model was performed by subcutaneous infusion of either saline or Angiotensin II by osmotic minipumps. We evaluated blood pressure, cardiac function (by echocardiography), fibrosis (with Masson Trichrome staining), and apoptosis (with TUNEL staining), and we performed transcriptome analysis (with RNA sequencing). We further evaluated fibrotic signaling in isolated murine primary ventricular myocytes. RESULTS Cardiac fibrosis was detected in 2 otherwise healthy humans with complete PAI-1 deficiency because of a homozygous frameshift mutation in SERPINE-1. In addition to its suppressive role during spontaneous cardiac fibrosis in multiple species, we hypothesized that PAI-1 also regulates fibrosis during cardiac injury. Treatment of young PAI-1-/- mice with Angiotensin II induced extensive hypertrophy and fibrotic cardiomyopathy, with increased cardiac apoptosis and both reactive and replacement fibrosis. Although Angiotensin II-induced hypertension was blunted in PAI-1-/- mice, cardiac hypertrophy was accelerated. Furthermore, ventricular myocytes were found to be an important source of cardiac transforming growth factor-β (TGF-β) and PAI-1 regulated TGF-β synthesis by cardiomyocytes in vitro as well as in vivo during cardiac injury. Transcriptome analysis of ventricular RNA after Angiotensin II treatment confirmed that PAI-1 deficiency significantly enhanced multiple TGF-β signaling elements and transcriptional targets, including genes for extracellular matrix components, mediators of extracellular matrix remodeling, matricellular proteins, and cardiac integrins compared with wild-type mice. CONCLUSIONS PAI-1 is an essential repressor of cardiac fibrosis in mammals. We define a novel cardiomyocyte-specific regulatory mechanism for TGF-β production by PAI-1, which explains the paradoxical effect of PAI-1 deficiency in promoting cardiac-selective fibrosis. Thus, PAI-1 is a molecular switch that controls the cardiac TGF-β axis and its early transcriptional effects that lead to myocardial fibrosis.
Collapse
Affiliation(s)
- Panagiotis Flevaris
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Sadiya S Khan
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Mesut Eren
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Adam J T Schuldt
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Sanjiv J Shah
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Daniel C Lee
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Sweta Gupta
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Amy D Shapiro
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Paul W Burridge
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Asish K Ghosh
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.)
| | - Douglas E Vaughan
- From Division of Cardiology, Department of Medicine (P.F., S.S.K., M.E., A.J.T.S., S.J.S., D.C.L., A.K.G., D.E.V.); Feinberg Cardiovascular Research Institute (P.F., S.S.K., S.J.S., D.C.L., A.K.G., D.E.V.), Department of Pharmacology (A.J.T.S., P.W.B.), Northwestern University Feinberg School of Medicine, Chicago, IL; and Indiana Hemophilia and Thrombosis Center, Indianapolis (S.G., A.D.S.).
| |
Collapse
|
38
|
Ma Y, Zou H, Zhu XX, Pang J, Xu Q, Jin QY, Ding YH, Zhou B, Huang DS. Transforming growth factor β: A potential biomarker and therapeutic target of ventricular remodeling. Oncotarget 2017; 8:53780-53790. [PMID: 28881850 PMCID: PMC5581149 DOI: 10.18632/oncotarget.17255] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/11/2017] [Indexed: 12/15/2022] Open
Abstract
Transforming growth factor β (TGF-β) is a multifunctional cytokine that is synthesized by many types of cells and regulates the cell cycle. Increasing evidence has led to TGF-β receiving increased and deserved attention in recent years because it may play a potentially novel and critical role in the development and progression of myocardial fibrosis and the subsequent progress of ventricular remodeling (VR). Numerous studies have highlighted a crucial role of TGF-β in VR and suggest potential therapeutic targets of the TGF-β signaling pathways for VR. Changes in TGF-β activity may elicit anti-VR activity and may serve as a novel therapeutic target for VR therapy. This review we discusses the smad-dependent signaling pathway, such as TGF-β/Smads, TGF-β/Sirtuins, TGF-β/BMP, TGF-β/miRNAs, TGF-β/MAPK, and Smad-independent signaling pathway of TGF-β, such as TGF-β/PI3K/Akt, TGF-β/Rho/ROCK,TGF-β/Wnt/β-catenin in the cardiac fibrosis and subsequent progression of VR. Furthermore, agonists and antagonists of TGF-β as potential therapeutic targets in VR are also described.
Collapse
Affiliation(s)
- Yuan Ma
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Hai Zou
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xing-Xing Zhu
- Department of Nephrology, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jie Pang
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Qiang Xu
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Qin-Yang Jin
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ya-Hui Ding
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Bing Zhou
- Department of Cardiac Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Dong-Sheng Huang
- Department of Hepatobiliary Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
39
|
Gilda JE, Gomes AV. Proteasome dysfunction in cardiomyopathies. J Physiol 2017; 595:4051-4071. [PMID: 28181243 DOI: 10.1113/jp273607] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a critical role in removing unwanted intracellular proteins and is involved in protein quality control, signalling and cell death. Because the heart is subject to continuous metabolic and mechanical stress, the proteasome plays a particularly important role in the heart, and proteasome dysfunction has been suggested as a causative factor in cardiac dysfunction. Proteasome impairment has been detected in cardiomyopathies, heart failure, myocardial ischaemia, and hypertrophy. Proteasome inhibition is also sufficient to cause cardiac dysfunction in healthy pigs, and patients using a proteasome inhibitor for cancer therapy have a higher incidence of heart failure. In this Topical Review we discuss the experimental data which suggest UPS dysfunction is a common feature of cardiomyopathies, with an emphasis on hypertrophic cardiomyopathy caused by sarcomeric mutations. We also propose potential mechanisms by which cardiomyopathy-causing mutations may lead to proteasome impairment, such as altered calcium handling and increased oxidative stress due to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jennifer E Gilda
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA.,Department of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
40
|
Heat shock transcription factor 1 protects against pressure overload-induced cardiac fibrosis via Smad3. J Mol Med (Berl) 2017; 95:445-460. [PMID: 28091697 PMCID: PMC5357304 DOI: 10.1007/s00109-016-1504-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 12/17/2016] [Accepted: 12/30/2016] [Indexed: 12/31/2022]
Abstract
Abstract Fibrotic cardiac muscle exhibits high stiffness and low compliance which are major risk factors of heart failure. Although heat shock transcription factor 1 (HSF1) was identified as an intrinsic cardioprotective factor, the role that HSF1 plays in cardiac fibrosis remains unclear. Our study aims to investigate the role of HSF1 in pressure overload-induced cardiac fibrosis and the underlying mechanism. HSF1 phosphorylation was significantly downregulated in transverse aortic constriction (TAC)-treated mouse hearts and mechanically stretched cardiac fibroblasts (cFBs). HSF1 transgenic (TG) mice, HSF1 deficient heterozygote (KO) mice, and their wild-type littermates were subjected to sham or TAC surgery for 4 weeks. HSF1 overexpression significantly attenuated pressure overload-induced cardiac fibrosis and dysfunction. Conversely, HSF1 KO mice showed deteriorated fibrotic response and cardiac dysfunction upon TAC. Moreover, we uncovered that overexpression of HSF1 protected against fibrotic response of cFBs to pressure overload. Mechanistically, we observed that the phosphorylation and the nuclear distribution of the Smad family member 3 (Smad3) were significantly decreased in HSF1-overexpressing mouse hearts, while being greatly increased in HSF1 KO mouse hearts upon TAC, compared to the control hearts, respectively. Similar alteration of Smad3 phosphorylation and nuclear distribution were found in isolated mouse cardiac fibroblasts and mechanically stretched cFBs. Constitutively active Smad3 blocked the anti-fibrotic effect of HSF1 in cFBs. Furthermore, we found a direct binding of phosphorylated HSF1 and Smad3, which can be suppressed by mechanical stress. In conclusion, the present study demonstrated for the first time that HSF1 acts as a novel negative regulator of cardiac fibrosis by blocking Smad3 activation. Key messages HSF1 activity is decreased in fibrotic hearts. HSF1 overexpression attenuates pressure overload-induced cardiac fibrosis and dysfunction. Deficiency of HSF1 deteriorates fibrotic response and cardiac dysfunction upon TAC. HSF1 inhibits phosphorylation and nuclear distribution of Smad3 via direct binding to Smad3. Active Smad3 blocks the anti-fibrotic effect of HSF1.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-016-1504-2) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Cossette SM, Bhute VJ, Bao X, Harmann LM, Horswill MA, Sinha I, Gastonguay A, Pooya S, Bordas M, Kumar SN, Mirza SP, Palecek SP, Strande JL, Ramchandran R. Sucrose Nonfermenting-Related Kinase Enzyme-Mediated Rho-Associated Kinase Signaling is Responsible for Cardiac Function. ACTA ACUST UNITED AC 2016; 9:474-486. [PMID: 27780848 DOI: 10.1161/circgenetics.116.001515] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 09/28/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiac metabolism is critical for the functioning of the heart, and disturbance in this homeostasis is likely to influence cardiac disorders or cardiomyopathy. Our laboratory has previously shown that SNRK (sucrose nonfermenting related kinase) enzyme, which belongs to the AMPK (adenosine monophosphate-activated kinase) family, was essential for cardiac metabolism in mammals. Snrk global homozygous knockout (KO) mice die at postnatal day 0, and conditional deletion of Snrk in cardiomyocytes (Snrk cmcKO) leads to cardiac failure and death by 8 to 10 months. METHODS AND RESULTS We performed additional cardiac functional studies using echocardiography and identified further cardiac functional deficits in Snrk cmcKO mice. Nuclear magnetic resonance-based metabolomics analysis identified key metabolic pathway deficits in SNRK knockdown cardiomyocytes in vitro. Specifically, metabolites involved in lipid metabolism and oxidative phosphorylation are altered, and perturbations in these pathways can result in cardiac function deficits and heart failure. A phosphopeptide-based proteomic screen identified ROCK (Rho-associated kinase) as a putative substrate for SNRK, and mass spec-based fragment analysis confirmed key amino acid residues on ROCK that are phosphorylated by SNRK. Western blot analysis on heart lysates from Snrk cmcKO adult mice and SNRK knockdown cardiomyocytes showed increased ROCK activity. In addition, in vivo inhibition of ROCK partially rescued the in vivo Snrk cmcKO cardiac function deficits. CONCLUSIONS Collectively, our data suggest that SNRK in cardiomyocytes is responsible for maintaining cardiac metabolic homeostasis, which is mediated in part by ROCK, and alteration of this homeostasis influences cardiac function in the adult heart.
Collapse
Affiliation(s)
- Stephanie M Cossette
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Vijesh J Bhute
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Xiaoping Bao
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Leanne M Harmann
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Mark A Horswill
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Indranil Sinha
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Adam Gastonguay
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Shabnam Pooya
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Michelle Bordas
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Suresh N Kumar
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Shama P Mirza
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Sean P Palecek
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Jennifer L Strande
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.)
| | - Ramani Ramchandran
- From the Department of Pediatrics (S.M.C., A.G., S. Pooya, M.B., R.R.), OBGYN, Developmental Vascular Biology Program, Children's Research Institute (R.R.), Division of Cardiovascular Medicine, Cardiovascular Center, Clinical and Translational Science Institute (L.M.H.), Division of Cardiovascular Medicine, Department of Cell Biology, Neurobiology and Anatomy, Cardiovascular Center, Clinical and Translational Science Institute (J.L.S.), and Division of Pediatric Pathology, Department of Pathology (S.N.K.), Medical College of Wisconsin, Milwaukee; Department of Chemical and Biological Engineering (V.J.B., X.B., S. Palecek), Morgridge Institute for Research (M.A.H.), University of Wisconsin-Madison; Marginalen Bank, Stockholm, Sweden (I.S.); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee (S.P.M.).
| |
Collapse
|
42
|
Furumatsu T, Maehara A, Ozaki T. ROCK inhibition stimulates SOX9/Smad3-dependent COL2A1 expression in inner meniscus cells. J Orthop Sci 2016; 21:524-529. [PMID: 27113646 DOI: 10.1016/j.jos.2016.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/07/2016] [Accepted: 03/27/2016] [Indexed: 02/09/2023]
Abstract
BACKGROUND Proper functioning of the meniscus depends on the composition and organization of its fibrocartilaginous extracellular matrix. We previously demonstrated that the avascular inner meniscus has a more chondrocytic phenotype compared with the outer meniscus. Inhibition of the Rho family GTPase ROCK, the major regulator of the actin cytoskeleton, stimulates the chondrogenic transcription factor Sry-type HMG box (SOX) 9-dependent α1(II) collagen (COL2A1) expression in inner meniscus cells. However, the crosstalk between ROCK inhibition, SOX9, and other transcription modulators on COL2A1 upregulation remains unclear in meniscus cells. The aim of this study was to investigate the role of SOX9-related transcriptional complex on COL2A1 expression under the inhibition of ROCK in human meniscus cells. METHODS Human inner and outer meniscus cells were prepared from macroscopically intact lateral menisci. Cells were cultured in the presence or absence of ROCK inhibitor (ROCKi, Y27632). Gene expression, collagen synthesis, and nuclear translocation of SOX9 and Smad2/3 were analyzed. RESULTS Treatment of ROCKi increased the ratio of type I/II collagen double positive cells derived from the inner meniscus. In real-time PCR analyses, expression of SOX9 and COL2A1 genes was stimulated by ROCKi treatment in inner meniscus cells. ROCKi treatment also induced nuclear translocation of SOX9 and phosphorylated Smad2/3 in immunohistological analyses. Complex formation between SOX9 and Smad3 was increased by ROCKi treatment in inner meniscus cells. Chromatin immunoprecipitation analyses revealed that association between SOX9/Smad3 transcriptional complex with the COL2A1 enhancer region was increased by ROCKi treatment. CONCLUSIONS This study demonstrated that ROCK inhibition stimulated SOX9/Smad3-dependent COL2A1 expression through the immediate nuclear translocation of Smad3 in inner meniscus cells. Our results suggest that ROCK inhibition can stimulates type II collagen synthesis through the cooperative activation of Smad3 in inner meniscus cells. ROCKi treatment may be useful to promote the fibrochondrocytic healing of the injured inner meniscus.
Collapse
Affiliation(s)
- Takayuki Furumatsu
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan.
| | - Ami Maehara
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| |
Collapse
|
43
|
Wang S, Huang G, Hu Q, Zou Q. A network-based method for the identification of putative genes related to infertility. Biochim Biophys Acta Gen Subj 2016; 1860:2716-24. [PMID: 27102279 DOI: 10.1016/j.bbagen.2016.04.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/02/2016] [Accepted: 04/08/2016] [Indexed: 01/18/2023]
Abstract
BACKGROUND Infertility has become one of the major health problems worldwide, with its incidence having risen markedly in recent decades. There is an urgent need to investigate the pathological mechanisms behind infertility and to design effective treatments. However, this is made difficult by the fact that various biological factors have been identified to be related to infertility, including genetic factors. METHODS A network-based method was established to identify new genes potentially related to infertility. A network constructed using human protein-protein interactions based on previously validated infertility-related genes enabled the identification of some novel candidate genes. These genes were then filtered by a permutation test and their functional and structural associations with infertility-related genes. RESULTS Our method identified 23 novel genes, which have strong functional and structural associations with previously validated infertility-related genes. CONCLUSIONS Substantial evidence indicates that the identified genes are strongly related to dysfunction of the four main biological processes of fertility: reproductive development and physiology, gametogenesis, meiosis and recombination, and hormone regulation. GENERAL SIGNIFICANCE The newly discovered genes may provide new directions for investigating infertility. This article is part of a Special Issue entitled "System Genetics" Guest Editor: Dr. Yudong Cai and Dr. Tao Huang.
Collapse
Affiliation(s)
- ShaoPeng Wang
- College of Life Science, Shanghai University, Shanghai 200444, China.
| | - GuoHua Huang
- College of Life Science, Shanghai University, Shanghai 200444, China.
| | - Qinghua Hu
- School of Computer Science and Technology, Tianjin University, Tianjin 300072, China; State Key Laboratory of System Bioengineering of the Ministry of Education, Tianjin University, Tianjin 300072, China.
| | - Quan Zou
- School of Computer Science and Technology, Tianjin University, Tianjin 300072, China; State Key Laboratory of Medicinal Chemical Biology, NanKai University, Tianjin 300071, China.
| |
Collapse
|
44
|
Bai T, Wang F, Mellen N, Zheng Y, Cai L. Diabetic cardiomyopathy: role of the E3 ubiquitin ligase. Am J Physiol Endocrinol Metab 2016; 310:E473-83. [PMID: 26732687 DOI: 10.1152/ajpendo.00467.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022]
Abstract
Diabetic cardiomyopathy (DCM) is the leading cause of mortality in diabetes. As the number of cases of diabetes continues to rise, it is urgent to develop new strategies to protect against DCM, which is characterized by cardiac hypertrophy, increased apoptosis, fibrosis, and altered insulin metabolism. The E3 ubiquitin ligases (E3s), one component of the ubiquitin-proteasome system, play vital roles in all of the features of DCM listed above. They also modulate the activity of several transcription factors involved in the pathogenesis of DCM. In addition, the E3s degrade both insulin receptor and insulin receptor substrates and also regulate insulin gene transcription, leading to insulin resistance and insulin deficiency. Therefore, the E3s may be a driving force for DCM. This review summarizes currently available studies to analyze the roles of the E3s in DCM, enriches our knowledge of how DCM develops, and provides a novel strategy to protect heart from diabetes.
Collapse
Affiliation(s)
- Tao Bai
- Cardiovascular Center, First Hospital of Jilin University, Changchun, China; Kosair Children's Hospital Research Institute, Departments of Pediatrics and Radiation Oncology, University of Louisville, Louisville, Kentucky
| | - Fan Wang
- Internal Medicine, People's Hospital of Jilin Province, Changchun, China; and
| | - Nicholas Mellen
- Kosair Children's Hospital Research Institute, Departments of Pediatrics and Radiation Oncology, University of Louisville, Louisville, Kentucky
| | - Yang Zheng
- Cardiovascular Center, First Hospital of Jilin University, Changchun, China;
| | - Lu Cai
- Kosair Children's Hospital Research Institute, Departments of Pediatrics and Radiation Oncology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
45
|
Hydrogen sulfide suppresses transforming growth factor-β1-induced differentiation of human cardiac fibroblasts into myofibroblasts. SCIENCE CHINA-LIFE SCIENCES 2015; 58:1126-34. [PMID: 26246380 DOI: 10.1007/s11427-015-4904-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
Abstract
In heart disease, transforming growth factor-β1 (TGF-β1) converts fibroblasts into myofibroblasts, which synthesize and secrete fibrillar type I and III collagens. The purpose of the present study was to investigate how hydrogen sulfide (H2S) suppresses TGF-β1-induced differentiation of human cardiac fibroblasts to myofibroblasts. Human cardiac fibroblasts were serum-starved in fibroblast medium for 16 h before exposure to TGF-β1 (10 ng mL(-1)) for 24 h with or without sodium hydrosulfide (NaHS, 100 µmol L(-1), 30 min pretreatment) treatment. NaHS, an exogenous H2S donor, potently inhibited the proliferation and migration of TGF-β1-induced human cardiac fibroblasts and regulated their cell cycle progression. Furthermore, NaHS treatment led to suppression of fibroblast differentiation into myofibroblasts, and reduced the levels of collagen, TGF-β1, and activated Smad3 in TGF-β1-induced human cardiac fibroblasts in vitro. We therefore conclude that H2S suppresses TGF-β1-stimulated conversion of fibroblasts to myofibroblasts by inhibiting the TGF-β1/Smad3 signaling pathway, as well as by inhibiting the proliferation, migration, and cell cycle progression of human cardiac myofibroblasts. These effects of H2S may play significant roles in cardiac remodeling associated with heart failure.
Collapse
|
46
|
Abstract
Corneal wound healing is a complex process involving cell death, migration, proliferation, differentiation, and extracellular matrix remodeling. Many similarities are observed in the healing processes of corneal epithelial, stromal and endothelial cells, as well as cell-specific differences. Corneal epithelial healing largely depends on limbal stem cells and remodeling of the basement membrane. During stromal healing, keratocytes get transformed to motile and contractile myofibroblasts largely due to activation of transforming growth factor-β (TGF-β) system. Endothelial cells heal mostly by migration and spreading, with cell proliferation playing a secondary role. In the last decade, many aspects of wound healing process in different parts of the cornea have been elucidated, and some new therapeutic approaches have emerged. The concept of limbal stem cells received rigorous experimental corroboration, with new markers uncovered and new treatment options including gene and microRNA therapy tested in experimental systems. Transplantation of limbal stem cell-enriched cultures for efficient re-epithelialization in stem cell deficiency and corneal injuries has become reality in clinical setting. Mediators and course of events during stromal healing have been detailed, and new treatment regimens including gene (decorin) and stem cell therapy for excessive healing have been designed. This is a very important advance given the popularity of various refractive surgeries entailing stromal wound healing. Successful surgical ways of replacing the diseased endothelium have been clinically tested, and new approaches to accelerate endothelial healing and suppress endothelial-mesenchymal transformation have been proposed including Rho kinase (ROCK) inhibitor eye drops and gene therapy to activate TGF-β inhibitor SMAD7. Promising new technologies with potential for corneal wound healing manipulation including microRNA, induced pluripotent stem cells to generate corneal epithelium, and nanocarriers for corneal drug delivery are discussed. Attention is also paid to problems in wound healing understanding and treatment, such as lack of specific epithelial stem cell markers, reliable identification of stem cells, efficient prevention of haze and stromal scar formation, lack of data on wound regulating microRNAs in keratocytes and endothelial cells, as well as virtual lack of targeted systems for drug and gene delivery to select corneal cells.
Collapse
Affiliation(s)
- Alexander V Ljubimov
- Eye Program, Board of Governors Regenerative Medicine Institute, Departments of Biomedical Sciences and Neurosurgery, Cedars-Sinai Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Mehrnoosh Saghizadeh
- Eye Program, Board of Governors Regenerative Medicine Institute, Departments of Biomedical Sciences and Neurosurgery, Cedars-Sinai Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
47
|
Luo JY, Zhang Y, Wang L, Huang Y. Regulators and effectors of bone morphogenetic protein signalling in the cardiovascular system. J Physiol 2015; 593:2995-3011. [PMID: 25952563 DOI: 10.1113/jp270207] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/27/2015] [Indexed: 12/22/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) play key roles in the regulation of cell proliferation, differentiation and apoptosis in various tissues and organs, including the cardiovascular system. BMPs signal through both Smad-dependent and -independent cascades to exert a wide spectrum of biological activities. Cardiovascular disorders such as abnormal angiogenesis, atherosclerosis, pulmonary hypertension and cardiac hypertrophy have been linked to aberrant BMP signalling. To correct the dysregulated BMP signalling in cardiovascular pathogenesis, it is essential to get a better understanding of how the regulators and effectors of BMP signalling control cardiovascular function and how the dysregulated BMP signalling contributes to cardiovascular dysfunction. We hence highlight several key regulators of BMP signalling such as extracellular regulators of ligands, mechanical forces, microRNAs and small molecule drugs as well as typical BMP effectors like direct downstream target genes, mitogen-activated protein kinases, reactive oxygen species and microRNAs. The insights into these molecular processes will help target both the regulators and important effectors to reverse BMP-associated cardiovascular pathogenesis.
Collapse
Affiliation(s)
- Jiang-Yun Luo
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Zhang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Li Wang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
48
|
Zhang X, Ma Y, You T, Tian X, Zhang H, Zhu Q, Zhang W. Roles of TGF-β/Smad signaling pathway in pathogenesis and development of gluteal muscle contracture. Connect Tissue Res 2015; 56:9-17. [PMID: 25207745 PMCID: PMC4438420 DOI: 10.3109/03008207.2014.964400] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE OF THE STUDY Gluteal muscle contracture (GMC) is a chronic fibrotic disease of gluteal muscles which is characterized by excessive deposition of collagen in the extracellular matrix. Transforming growth factor (TGF)-βs have been shown to play an important role in the progression of GMC. However, the underlying mechanisms are not entirely clear. We sought to explore the expression of TGF-β/Smad pathway proteins and their downstream targets in gluteal muscle contracture disease. MATERIALS AND METHODS The expression levels of collagens type I/III, TGF-β1, Smad2/3/4/7 and PAI-1 (plasminogen activator inhibitor type 1) in gluteal muscle contraction (GMC) patients were measured using immunohistochemistry, reverse transcription and polymerase chain reaction (RT-PCR) and western blot assays. RESULTS The expressions of collagens type I/III and TGF-β1 were significantly increased in the contraction band compared with unaffected muscle. In addition, R-Smad phosphorylation and Smad4 protein expression in the contraction band were also elevated, while the expression of Smad7 was significantly decreased in the fibrotic muscle of the GMC patients compared to the unaffected adjacent muscle. The protein and mRNA levels of PAI-1 were also remarkably increased in the contraction band compared with adjacent muscle. Immunohistochemical analysis also demonstrated that the expression levels of TGF-β1 and PAI-1 were higher in contraction band than those in the adjacent muscle. CONCLUSION Our data confirm the stimulating effects of the TGF-β/Smad pathway in gluteal muscle contracture disease and reveal the internal changes of TGF-β/Smad pathway proteins and their corresponding targets in gluteal muscle contracture patients.
Collapse
Affiliation(s)
- Xintao Zhang
- Department of Sports Medicine and Rehabilitation, Peking University Shen Zhen Hospital, ShenZhen, China
| | - Yukun Ma
- Department of Pediatric Surgery, Linyi People's Hospital, Shandong Province, China
| | - Tian You
- Department of Sports Medicine and Rehabilitation, Peking University Shen Zhen Hospital, ShenZhen, China
| | - Xiaopeng Tian
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Honglei Zhang
- Department of Sports Medicine and Rehabilitation, Peking University Shen Zhen Hospital, ShenZhen, China
| | - Qi Zhu
- Orthopaedics Hong Kong University, Shen Zhen Hospital, ShenZhen, China
| | - Wentao Zhang
- Department of Sports Medicine and Rehabilitation, Peking University Shen Zhen Hospital, ShenZhen, China,Correspondence: Wentao Zhang, Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, No. 1120, Lianhua, 518036 ShenZhen, China. Tel: 075583923333-6135(6137). E-mail:
| |
Collapse
|
49
|
McDonald ME, Li C, Bian H, Smith BD, Layne MD, Farmer SR. Myocardin-related transcription factor A regulates conversion of progenitors to beige adipocytes. Cell 2015; 160:105-18. [PMID: 25579684 DOI: 10.1016/j.cell.2014.12.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 09/17/2014] [Accepted: 11/19/2014] [Indexed: 10/24/2022]
Abstract
Adipose tissue is an essential regulator of metabolic homeostasis. In contrast with white adipose tissue, which stores excess energy in the form of triglycerides, brown adipose tissue is thermogenic, dissipating energy as heat via the unique expression of the mitochondrial uncoupling protein UCP1. A subset of UCP1+ adipocytes develops within white adipose tissue in response to physiological stimuli; however, the developmental origin of these "brite" or "beige" adipocytes is unclear. Here, we report the identification of a BMP7-ROCK signaling axis regulating beige adipocyte formation via control of the G-actin-regulated transcriptional coactivator myocardin-related transcription factor A, MRTFA. White adipose tissue from MRTFA(-/-) mice contains more multilocular adipocytes and expresses enhanced levels of brown-selective proteins, including UCP1. MRTFA(-/-) mice also show improved metabolic profiles and protection from diet-induced obesity and insulin resistance. Our study hence unravels a central pathway driving the development of physiologically functional beige adipocytes.
Collapse
Affiliation(s)
- Meghan E McDonald
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Chendi Li
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hejiao Bian
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Barbara D Smith
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Matthew D Layne
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Stephen R Farmer
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
50
|
Tokola H, Rysä J, Pikkarainen S, Hautala N, Leskinen H, Kerkelä R, Ilves M, Aro J, Vuolteenaho O, Ritvos O, Ruskoaho H. Bone morphogenetic protein-2--a potential autocrine/paracrine factor in mediating the stretch activated B-type and atrial natriuretic peptide expression in cardiac myocytes. Mol Cell Endocrinol 2015; 399:9-21. [PMID: 25218476 DOI: 10.1016/j.mce.2014.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 08/25/2014] [Accepted: 09/03/2014] [Indexed: 12/31/2022]
Abstract
Hemodynamic overload exposes the heart to variety of neural, humoral and mechanical stresses. Even without the neurohumoral control of the entire organism cardiac myocytes have the ability to sense mechanical stretch and convert it into adaptive intracellular signals. This process is controlled by several growth factors. Here we show that mechanical stretch in vitro and hemodynamic overload in vivo activated the expression of bone morphogenetic protein-2 (BMP-2), while expression of BMP-4 was temporarily attenuated by stretch. BMP-2 and BMP-4 alone stimulated B-type and atrial natriuretic peptide (BNP and ANP) expression and protein synthesis, and activated transcription factor GATA-4 resembling the effects of mechanical stretch of cultured cardiac myocytes. Further, BMP antagonist Noggin was able to inhibit stretch and hypertrophic agonist induced BNP and ANP expression. Together these data provide evidence for BMP-2 as a new autocrine/paracrine factor that regulates cardiomyocyte mechanotransduction and adaptation to increased mechanical stretch.
Collapse
Affiliation(s)
- Heikki Tokola
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Pathology, Institute of Diagnostics, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Pathology, Oulu University Hospital, P.O. BOX 50, Oulu FI-90029 OYS, Finland
| | - Jaana Rysä
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio FIN-70211, Finland
| | - Sampsa Pikkarainen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Medicine, Division of Gastroenterology, Helsinki University Central Hospital, P.O. BOX 340, Helsinki FI-00029 HUS, Finland
| | - Nina Hautala
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Ophthalmology, Oulu University Hospital, P.O. BOX 21, Oulu FI-90029 OYS, Finland
| | - Hanna Leskinen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Risto Kerkelä
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Mika Ilves
- Department of Physiology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Jani Aro
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Olli Vuolteenaho
- Department of Physiology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Olli Ritvos
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki FI-00014, Finland
| | - Heikki Ruskoaho
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. BOX 56, Viikinkaari 5E, Helsinki FI-00014, Finland.
| |
Collapse
|