1
|
Bois A, Grandela C, Gallant J, Mummery C, Menasché P. Revitalizing the heart: strategies and tools for cardiomyocyte regeneration post-myocardial infarction. NPJ Regen Med 2025; 10:6. [PMID: 39843488 PMCID: PMC11754855 DOI: 10.1038/s41536-025-00394-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Myocardial infarction (MI) causes the loss of millions of cardiomyocytes, and current treatments do not address this root issue. New therapies focus on stimulating cardiomyocyte division in the adult heart, inspired by the regenerative capacities of lower vertebrates and neonatal mice. This review explores strategies for heart regeneration, offers insights into cardiomyocyte proliferation, evaluates in vivo models, and discusses integrating in vitro human cardiac models to advance cardiac regeneration research.
Collapse
Affiliation(s)
- Axelle Bois
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
- Department of Cardiovascular Surgery, Université Paris Cité, INSERM U970, PARCC Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - Catarina Grandela
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - James Gallant
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Université Paris Cité, INSERM U970, PARCC Hôpital Européen Georges Pompidou, 75015, Paris, France
| |
Collapse
|
2
|
Matovic V, Ljujic B, Radojevic I, Đelic G, Kovacevic MM, Zivanovic S, Papic M, Milivojevic N, Petrovic I, Gazdic Jankovic M. Trapa natans L. Extract Attenuates Inflammation and Oxidative Damage in Cisplatin-Induced Cardiotoxicity in Rats by Promoting M2 Macrophage Polarization. Mediators Inflamm 2025; 2025:6587305. [PMID: 39886549 PMCID: PMC11779992 DOI: 10.1155/mi/6587305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/14/2024] [Indexed: 02/01/2025] Open
Abstract
Background: Trapa natans L. fruits and leaf extracts have a broad range of immunomodulatory, anti-inflammatory, and antioxidant effects; however, their effects on cardiac protection have not been investigated. Objective: The study aims to test the biological activity of Trapa natans L. extract (TNE) in cisplatin (CDDP)-induced cardiotoxicity. Methods: Wistar albino rats received a single dose of CDDP intraperitoneally and TNE ones per day for 2 weeks orally. Cardiac inflammation, necrosis, and fibrosis were determined by histological and immunohistochemical analyses. Cytokines in rat sera and cardiac tissue were detected by enzyme-linked immunosorbent assay (ELISA) and quantitative real-time (qRT)-PCR. Rat macrophages cultured in the presence of TNE for 48 h were harvested for flow cytometry, while supernatants were collected for cytokine and reactive oxygen species (ROS) measurement. Results: Application of TNE significantly attenuated CDDP induced cardiotoxicity as demonstrated by biochemical and histopathological analysis. Administration of TNE once daily for 14 days decreased level of proinflammatory (TNF-α, IFN-γ, and IL-6) and prooxidative parameters (NO2, O2, and H2O2), while increased level of immunosuppressive IL-10 and antioxidative glutathione (GSH), catalase (CAT) and uperoxide dismutase (SOD) in the systemic circulation. TNE treatment resulted in attenuated heart inflammation and fibrosis accompanied with the reduced infiltration of macrophages and reduced expression of proinflammatory and profibrotic genes in heart tissue of CDDP-treated animals. In vitro lipopolysaccharide (LPS)-stimulated macrophages cultured in the presence of TNE adopted immunosuppressive phenotype characterized by decreased production of proinflammatory cytokines and prooxidative mediators. Conclusion: Our study provides the evidence that TNE ameliorates cisplatin-induced cardiotoxicity in rats by reducing inflammation and oxidative stress via promoting M2 macrophage polarization.
Collapse
Affiliation(s)
- Vesna Matovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
| | - Biljana Ljujic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
- Faculty of Medical Sciences, Center for Harm Reduction of Biological and Chemical Hazards, University of Kragujevac, Kragujevac, Serbia
| | - Ivana Radojevic
- Faculty of Science, Department of Biology and Ecology, University of Kragujevac, Radoja Domanovica 12, Kragujevac 34000, Serbia
| | - Gorica Đelic
- Faculty of Science, Department of Biology and Ecology, University of Kragujevac, Radoja Domanovica 12, Kragujevac 34000, Serbia
| | - Marina Miletic Kovacevic
- Faculty of Medical Sciences, Center for Harm Reduction of Biological and Chemical Hazards, University of Kragujevac, Kragujevac, Serbia
- Faculty of Medical Sciences, Department of Histology and Embryology, University of Kragujevac, Kragujevac, Serbia
| | - Suzana Zivanovic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Milos Papic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena Milivojevic
- Laboratory for Bioengineering, Institute of Information Technologies Kragujevac, University of Kragujevac, Kragujevac, Serbia
| | - Ivica Petrovic
- Faculty of Medical Sciences, Department of Pathophysiology, University of Kragujevac, Kragujevac, Serbia
| | - Marina Gazdic Jankovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
- Faculty of Medical Sciences, Center for Harm Reduction of Biological and Chemical Hazards, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
3
|
Chaudhary S, Das U, Jabbar S, Gangisetty O, Rousseau B, Hanft S, Sarkar DK. Developmental pluripotency-associated 4 increases aggressiveness of pituitary neuroendocrine tumors by enhancing cell stemness. Neuro Oncol 2025; 27:123-139. [PMID: 39093695 PMCID: PMC11726338 DOI: 10.1093/neuonc/noae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Pituitary neuroendocrine tumors, PitNETs, are often aggressive and precipitate in distant metastases that are refractory to current therapies. However, the molecular mechanism in PitNETs' aggressiveness is not well understood. Developmental pluripotency-associated 4 (DPPA4) is known as a stem cell regulatory gene and overexpressed in certain cancers, but its function in the context of PitNETs' aggressiveness is not known. METHODS We employed both rat and human models of PitNETs. In the rat pituitary tumor model, we used prenatal-alcohol-exposed (PAE) female Fischer rats which developed aggressive PitNETs following estrogen treatment, while in the human pituitary tumor model, we used aggressively proliferative cells from pituitary tumors of patients undergone surgery. Various molecular, cellular, and epigenetic techniques were used to determine the role of DPPA4 in PitNETs' aggressiveness. RESULTS We show that DPPA4 is overexpressed in association with increased cell stemness factors in aggressive PitNETs of PAE rats and of human patients. Gene-editing experiments demonstrate that DPPA4 increases the expression of cell stemness and tumor aggressiveness genes and promotes proliferation, colonization, migration, and tumorigenic potential of PitNET cells. ChIP assays and receptor antagonism studies reveal that DPPA4 binds to canonical WINTs promoters and increases directly or indirectly the WNT/β-CATENIN control of cell stemness, tumor growth, and aggressiveness of PitNETs. Epigenetic studies show the involvement of histone methyltransferase in alcohol activation of DPPA4. CONCLUSIONS These findings support a role of DPPA4 in tumor stemness and aggressiveness and provide a preclinical rationale for modulating this stemness regulator for the treatment of PitNETs.
Collapse
Affiliation(s)
- Shaista Chaudhary
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Ujjal Das
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Shaima Jabbar
- Endocrinology and Animal Biosciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Omkaram Gangisetty
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Bénédicte Rousseau
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Simon Hanft
- Pituitary Tumor Program, Rutgers Cancer Institute of New Jersey, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Dipak K Sarkar
- Endocrinology and Animal Biosciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
4
|
Jakobsen TS, Adsersen RL, Askou AL, Corydon TJ. Functional Roles of Pigment Epithelium-Derived Factor in Retinal Degenerative and Vascular Disorders: A Scoping Review. Invest Ophthalmol Vis Sci 2024; 65:41. [PMID: 39728690 PMCID: PMC11684118 DOI: 10.1167/iovs.65.14.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose This review explores the role of pigment epithelium-derived factor (PEDF) in retinal degenerative and vascular disorders and assesses its potential both as an adjunct to established vascular endothelial growth factor inhibiting treatments for retinal vascular diseases and as a neuroprotective therapeutic agent. Methods A comprehensive literature review was conducted, focusing on the neuroprotective and anti-angiogenic properties of PEDF. The review evaluated its effects on retinal health, its dysregulation in ocular disorders, and its therapeutic application in preclinical models. Advances in drug delivery, including gene therapy, were also examined. Results PEDF, initially identified for promoting neuronal differentiation, is also a potent endogenous angiogenesis inhibitor. Strong anti-angiogenic and neuroprotective effects are observed in preclinical studies. It has pro-apoptotic and antiproliferative effects on endothelial cells thereby reducing neovascularization. Although promising, clinical development is limited with only a single conducted phase I clinical trial for macular neovascularization. Development of PEDF-derived peptides enhances potency and specificity, and emerging gene therapy approaches offer sustained PEDF expression for long-term treatment. However, questions regarding dosage, durability, and efficacy remain, particularly in large animal models. Conclusions PEDF shows significant therapeutic potential in preclinical models of retinal degeneration and vascular disorders. Despite inconclusive evidence on PEDF downregulation as a primary disease driver, many studies highlight its therapeutic benefits and favorable safety profile. Advances in gene therapy could enable long-acting PEDF-based treatments, but further research is needed to optimize dosage and durability, potentially leading to clinical trials and expanding treatment options for retinal disorders.
Collapse
Affiliation(s)
- Thomas Stax Jakobsen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark
| | | | - Anne Louise Askou
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
5
|
Wei YR, Hou YL, Yin YJ, Li Z, Liu Y, Han NX, Wang ZX, Liu L, Wang XQ, Hao YJ, Ma K, Gu JJ, Jia ZH. Tongxinluo Activates PI3K/AKT Signaling Pathway to Inhibit Endothelial Mesenchymal Transition and Attenuate Myocardial Fibrosis after Ischemia-Reperfusion in Mice. Chin J Integr Med 2024; 30:608-615. [PMID: 38386252 DOI: 10.1007/s11655-024-3652-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 02/23/2024]
Abstract
OBJECTIVE To investigate the potential role of Tongxinluo (TXL) in attenuating myocardial fibrosis after myocardial ischemia-reperfusion injury (MIRI) in mice. METHODS A MIRI mouse model was established by left anterior descending coronary artery ligation for 45 min. According to a random number table, 66 mice were randomly divided into 6 groups (n=11 per group): the sham group, the model group, the LY-294002 group, the TXL group, the TXL+LY-294002 group and the benazepril (BNPL) group. The day after modeling, TXL and BNPL were administered by gavage. Intraperitoneal injection of LY-294002 was performed twice a week for 4 consecutive weeks. Echocardiography was used to measure cardiac function in mice. Masson staining was used to evaluate the degree of myocardial fibrosis in mice. Qualitative and quantitative analysis of endothelial mesenchymal transition (EndMT) after MIRI was performed by immunohistochemistry, immunofluorescence staining and flow cytometry, respectively. The protein expressions of platelet endothelial cell adhesion molecule-1 (CD31), α-smoth muscle actin (α-SMA), phosphatidylinositol-3-kinase (PI3K) and phospho protein kinase B (p-AKT) were assessed using Western blot. RESULTS TXL improved cardiac function in MIRI mice, reduced the degree of myocardial fibrosis, increased the expression of CD31 and inhibited the expression of α-SMA, thus inhibited the occurrence of EndMT (P<0.05 or P<0.01). TXL significantly increased the protein expressions of PI3K and p-AKT (P<0.05 or P<0.01). There was no significant difference between TXL and BNPL group (P>0.05). In addition, the use of the PI3K/AKT pathway-specific inhibitor LY-294002 to block this pathway and combination with TXL intervention, eliminated the protective effect of TXL, further supporting the protective effect of TXL. CONCLUSION TXL activated the PI3K/AKT signaling pathway to inhibit EndMT and attenuated myocardial fibrosis after MIRI in mice.
Collapse
Affiliation(s)
- Ya-Ru Wei
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Yun-Long Hou
- Shijiazhuang Yiling Pharmaceutical New Drug Evaluation Center, Shijiazhuang, 050035, China
| | - Yu-Jie Yin
- Hebei Institute of Integrated Traditional and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Department of Cardiology, Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Zhen Li
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Ning-Xin Han
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zi-Xuan Wang
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Lu Liu
- Hebei Institute of Integrated Traditional and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Department of Cardiology, Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Xiao-Qi Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Yuan-Jie Hao
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Kun Ma
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Jiao-Jiao Gu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Zhen-Hua Jia
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China.
- Hebei Institute of Integrated Traditional and Western Medicine, Shijiazhuang, 050035, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
- Department of Cardiology, Hebei Yiling Hospital, Shijiazhuang, 050091, China.
| |
Collapse
|
6
|
Miao H, Hui H, Fan W, Lin Y, Li H, Li D, Luo M, Qiu F, Jiang B, Zhang Y. Overexpressed pigment epithelium-derived factor alleviates pulmonary hypertension in two rat models induced by monocrotaline and SU5416/hypoxia. Biomed Pharmacother 2024; 172:116303. [PMID: 38377738 DOI: 10.1016/j.biopha.2024.116303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive and fatal cardiopulmonary disease characterized by vascular remodeling and is associated with endothelial-to-mesenchymal transition (EndoMT). The pigment epithelium-derived factor (PEDF), a secretory protein widely distributed in multiple organs, has been shown to demonstrate anti-EndoMT activity in cardiovascular diseases. In the present study, the role of PEDF in PH was investigated. METHODS For PEDF overexpression, Sprague Dawley rats were infected with an adeno-associated virus through injection via the internal jugular vein. To establish PH models, the animals were subjected to monocrotaline or Sugen/hypoxia. Four weeks later, pulmonary artery angiography was performed, and hemodynamic parameters, right ventricular function, and vascular remodeling were evaluated. EndoMT and cell proliferation in the pulmonary arteries were assessed via immunofluorescence staining. Moreover, pulmonary artery endothelial cells (PAECs) isolated from experimental PH rats were cultured to investigate the underlying molecular mechanisms involved. RESULTS PEDF expression was significantly downregulated in PAECs from PH patients and PH model rats. Overexpressed PEDF alleviated the development of PH by improving pulmonary artery morphology and perfusion, reducing pulmonary artery pressure, improving right ventricular function, and alleviating vascular remodeling. PEDF inhibits EndoMT and reduces excessive PAEC proliferation. Moreover, PEDF overexpression reduced EndoMT in cultured PAECs by competitively inhibiting the binding of wnt to LRP6 and downregulating phosphorylation at the 1490 site of LRP6. CONCLUSIONS Our findings suggest that PEDF may be a potential therapeutic target for PH. We also found that PEDF can inhibit EndoMT in PAECs and may exert these effects by inhibiting the Wnt/LRP6/β-catenin pathway.
Collapse
Affiliation(s)
- Haoran Miao
- Department of Thoracic Cardiovascular Surgery, China
| | - Hongliang Hui
- Department of Thoracic Cardiovascular Surgery, China
| | - Wenbin Fan
- Department of Thoracic Cardiovascular Surgery, China
| | - Yangui Lin
- Department of Thoracic Cardiovascular Surgery, China
| | - Huaming Li
- Department of Thoracic Cardiovascular Surgery, China
| | - Dan Li
- Community Health Center, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Min Luo
- Department of Thoracic Cardiovascular Surgery, China
| | - Fan Qiu
- Department of Thoracic Cardiovascular Surgery, China.
| | - Bo Jiang
- Department of Thoracic Cardiovascular Surgery, China.
| | - Yiqian Zhang
- Department of Thoracic Cardiovascular Surgery, China.
| |
Collapse
|
7
|
Hui HL, Jiang B, Zhou YY, Qiu F, Lin YG, Li HM, Li D, Luo M, Miao HR, Ong SB, Zhang YQ. PEDF inhibits VEGF-induced vascular leakage through binding to VEGFR2 in acute myocardial infarction. J Biomol Struct Dyn 2024:1-13. [PMID: 38345053 DOI: 10.1080/07391102.2024.2314260] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/30/2024] [Indexed: 03/11/2025]
Abstract
Pigment epithelium-derived factor (PEDF) could bind to vascular endothelial growth factor receptor 2 (VEGFR2) and inhibit its activation induced by VEGF. But how PEDF affects VEGFR2 pathway is still poorly understood. In this study, we elucidated the precise mechanism underlying the interaction between PEDF and VEGFR2, and subsequently corroborated our findings using a rat AMI model. PEDF prevented endocytosis of VE-cadherin induced by hypoxia, thereby protecting the endothelium integrity. A three-dimensional model of the VEGFR2-PEDF complex was constructed by protein-protein docking method. The results showed that the VEGFR2-PEDF complex was stable during the simulation. Hydrogen bonds, binding energy and binding modes were analyzed during molecular dynamics simulations, which indicated that hydrogen bonds and hydrophobic interactions were important for the recognition of VEGFR2 with PEDF. In addition, the results from exudation of fibrinogen suggested that PEDF inhibits vascular leakage in acute myocardial infarction and confirmed the critical role of key amino acids in the regulation of endothelial cell permeability. This observation is also supported by echocardiography studies showing that the 34mer peptide sustained cardiac function during acute myocardial infarction. Besides, PEDF and 34mer could inhibit the aggregation of myofiber in the heart and promoted the formation of a dense cell layer in cardiomyocytes, which suggested that PEDF and 34mer peptide protect against AMI-induced cardiac dysfunction. These results suggest that PEDF inhibits the phosphorylation of downstream proteins, thereby preventing vascular leakage, which provides a new therapeutic direction for the treatment of acute myocardial infarction.
Collapse
Affiliation(s)
- Hong-Liang Hui
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bo Jiang
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yan-Ying Zhou
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Fan Qiu
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yan-Gui Lin
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Hua-Ming Li
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Dan Li
- Community Health Center, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Min Luo
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Hao-Ran Miao
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics (MEDT), Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, CUHK, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), Hong Kong Children's Hospital (HKCH), Kowloon Bay, Hong Kong SAR, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- CUHK Shenzhen Research Institute (SZRI), Shenzhen, China
- Neural, Vascular, and Metabolic Biology Thematic Research Program, School of Biomedical Sciences (SBS), Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Yi-Qian Zhang
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
8
|
Kim S, Lee H, Moon H, Kim R, Kim M, Jeong S, Kim H, Kim SH, Hwang SS, Lee MY, Kim J, Song BW, Chang W. Epigallocatechin-3-Gallate Attenuates Myocardial Dysfunction via Inhibition of Endothelial-to-Mesenchymal Transition. Antioxidants (Basel) 2023; 12:1059. [PMID: 37237925 PMCID: PMC10215739 DOI: 10.3390/antiox12051059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/22/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiac tissue damage following ischemia leads to cardiomyocyte apoptosis and myocardial fibrosis. Epigallocatechin-3-gallate (EGCG), an active polyphenol flavonoid or catechin, exerts bioactivity in tissues with various diseases and protects ischemic myocardium; however, its association with the endothelial-to-mesenchymal transition (EndMT) is unknown. Human umbilical vein endothelial cells (HUVECs) pretreated with transforming growth factor β2 (TGF-β2) and interleukin 1β (IL-1β) were treated with EGCG to verify cellular function. In addition, EGCG is involved in RhoA GTPase transmission, resulting in reduced cell mobility, oxidative stress, and inflammation-related factors. A mouse myocardial infarction (MI) model was used to confirm the association between EGCG and EndMT in vivo. In the EGCG-treated group, ischemic tissue was regenerated by regulating proteins involved in the EndMT process, and cardioprotection was induced by positively regulating apoptosis and fibrosis of cardiomyocytes. Furthermore, EGCG can reactivate myocardial function due to EndMT inhibition. In summary, our findings confirm that EGCG is an impact activator controlling the cardiac EndMT process derived from ischemic conditions and suggest that supplementation with EGCG may be beneficial in the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Sejin Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea; (S.K.); (H.L.); (R.K.); (M.K.)
| | - Hyunjae Lee
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea; (S.K.); (H.L.); (R.K.); (M.K.)
| | - Hanbyeol Moon
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea; (H.M.); (S.J.); (H.K.)
| | - Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea; (S.K.); (H.L.); (R.K.); (M.K.)
| | - Minsuk Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea; (S.K.); (H.L.); (R.K.); (M.K.)
| | - Seongtae Jeong
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea; (H.M.); (S.J.); (H.K.)
| | - Hojin Kim
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea; (H.M.); (S.J.); (H.K.)
| | - Sang Hyeon Kim
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.H.K.); (S.S.H.)
- Chronic Intractable Disease Systems Medical Research Center, Institute of Genetic Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Soo Seok Hwang
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.H.K.); (S.S.H.)
- Chronic Intractable Disease Systems Medical Research Center, Institute of Genetic Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women’s University, Seoul 04310, Republic of Korea;
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea; (H.M.); (S.J.); (H.K.)
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea; (S.K.); (H.L.); (R.K.); (M.K.)
| |
Collapse
|
9
|
Jones IC, Dass CR. Roles of pigment epithelium-derived factor in cardiomyocytes: implications for use as a cardioprotective therapeutic. J Pharm Pharmacol 2023:7146108. [PMID: 37104852 DOI: 10.1093/jpp/rgad037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/12/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVES Cardiovascular diseases are the leading cause of death worldwide, with patients having limited options for treatment. Pigment epithelium-derived factor (PEDF) is an endogenous multifunctional protein with several mechanisms of action. Recently, PEDF has emerged as a potential cardioprotective agent in response to myocardial infarction. However, PEDF is also associated with pro-apoptotic effects, complicating its role in cardioprotection. This review summarises and compares knowledge of PEDF's activity in cardiomyocytes with other cell types and draws links between them. Following this, the review offers a novel perspective of PEDF's therapeutic potential and recommends future directions to understand the clinical potential of PEDF better. KEY FINDINGS PEDF's mechanisms as a pro-apoptotic and pro-survival protein are not well understood, despite PEDF's implication in several physiological and pathological activities. However, recent evidence suggests that PEDF may have significant cardioprotective properties mediated by key regulators dependent on cell type and context. CONCLUSIONS While PEDF's cardioprotective activity shares some key regulators with its apoptotic activity, cellular context and molecular features likely allow manipulation of PEDF's cellular activity, highlighting the importance of further investigation into its activities and its potential to be applied as a therapeutic to mitigate damage from a range of cardiac pathologies.
Collapse
Affiliation(s)
- Isobel C Jones
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| |
Collapse
|
10
|
Lou C, Li T. Long non-coding RNA SENCR alleviates endothelial-to-mesenchymal transition via targeting miR-126a. Arch Med Sci 2023; 19:180-188. [PMID: 36817675 PMCID: PMC9897100 DOI: 10.5114/aoms.2020.97991] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/21/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Long non-coding RNAs (lncRNAs) constitute a growing class of non-coding genes with diverse cellular function. Recent studies have reported that lncRNA smooth muscle and endothelial cell-enriched (SENCR) was associated with the phenotype switch of vascular smooth muscle cells and participated in vascular homeostasis. However, the potential role of SENCR in endothelial-to-mesenchymal transition (EndMT) and the underlying mechanism remain unknown. MATERIAL AND METHODS Human carotid plaque samples and human coronary endothelial cells (HACECs) were collected to examine the expression of SENCR. Quantitative PCR and immunoblots were performed to evaluate the expression of SENCR and miR-126a in HACECs in response to TGF-β1 and transfected with small interfering RNA. RESULTS We found that SENCR was significantly decreased in carotid plaques as compared to normal carotids. Knockdown of SENCR in HACECs aggravated the expression of smooth muscle markers α-SMA and calponin induced by TGF-β1 but repressed the expression of endothelial markers platelet/endothelial cell adhesion molecule 1 (PECAM1) and VE-cadherin down-regulated by TGF-β1. Through bioinformatic analysis and Luciferase assay, miR-126a was identified as the direct target of SENCR. Further mechanistic experiments revealed that overexpression of miR-126a bound to the 3'UTR region of SMURF2 and inhibited the expression of SMURF2, which was considered as the negative regulator of TGF-β/Smad signaling. Finally, overexpression of miR-126a did not restore the decreased expression of the smooth muscle markers α-SMA and calponin under the condition of SMURF2 depletion, suggesting that the effect of miR-126a on EndMT progression is SMURF2 dependent. CONCLUSIONS SENCR alleviates TGF-β-induced EndMT and sponges miR-126a expression via direct inhibition of the negative regulator of TGF-β/Smad signaling SMURF2.
Collapse
Affiliation(s)
- Chuang Lou
- Department of Cardiology, AnKang Hospital of Traditional Chinese Medicine, Ankang, China
| | - Tao Li
- Department of Cardiology, AnKang Hospital of Traditional Chinese Medicine, Ankang, China
| |
Collapse
|
11
|
Mahapatra S, Sharma MVR, Brownson B, Gallicano VE, Gallicano GI. Cardiac inducing colonies halt fibroblast activation and induce cardiac/endothelial cells to move and expand via paracrine signaling. Mol Biol Cell 2022; 33:ar96. [PMID: 35653297 DOI: 10.1091/mbc.e22-02-0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Myocardial fibrosis (MF), a common event that develops after myocardial infarction, initially is a reparative process but eventually leads to heart failure and sudden cardiac arrest. In MF, the infarct area is replaced by a collagenous-based scar induced by "excessive" collagen deposition from activated cardiac fibroblasts. The scar prevents ventricular wall thinning; however, over time it expands to noninfarcted myocardium. Therapies to prevent fibrosis include reperfusion, anti-fibrotic agents, and ACE inhibitors. Paracrine factor (PF)/stem cell research has recently gained significance as a therapy. We consistently find that cardiac inducing colonies (CiCs) (derived from human germline pluripotent stem cells) secrete PFs at physiologically relevant concentrations that suppress cardiac fibroblast activation and excessive extracellular matrix protein secretion. These factors also affect human cardiomyocytes and endothelial cells by inducing migration/proliferation of both populations into a myocardial wound model. Finally, CiC factors modulate matrix turnover and proinflammation. Taking the results together, we show that CiCs could help tip the balance from fibrosis toward repair.
Collapse
Affiliation(s)
- Samiksha Mahapatra
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145
| | | | - Breanna Brownson
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145.,Rye High School, Rye, NY 10580
| | - Vaughn E Gallicano
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145.,Thomas Edison High School, Alexandria, VA 22310
| | - G Ian Gallicano
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145
| |
Collapse
|
12
|
Qin X, Jia C, Liang J, Chen J, Liu X, Chao Z, Qin H, Yuan Y, Liu Z, Zhang Z, Dong H, Zhang H. PEDF is an antifibrosis factor that inhibits the activation of fibroblasts in a bleomycin-induced pulmonary fibrosis rat model. Respir Res 2022; 23:100. [PMID: 35459189 PMCID: PMC9027047 DOI: 10.1186/s12931-022-02027-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 04/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a highly heterogeneous and fatal lung disease. In addition to dense fibrous tissue, abnormal angiogenesis is also an important feature of IPF. Pigment epithelium-derived factor (PEDF) is an angiogenesis inhibitor and a potential anti-fibrous factor. The purpose of this experiment is to observe the effect of PEDF on bleomycin (BLM)-induced pulmonary fibrosis in rats. Methods In vivo, pathological examination and detection of related factors were performed on pulmonary fibrosis induced by BLM in rats, and the temporal and spatial distribution of PEDF was investigated. Furthermore, lung gene delivery (PEDF-adeno-associated virus) was performed to investigate the effect of PEDF on pulmonary fibrosis. In vitro, lentiviral vectors were used to construct PEDF over-expression or knock out primary rat lung (PRL) fibroblasts. The effect of PEDF on fibroblast activation under TGF-β1 stimulation was evaluated, and the activation of TGF-β1/smad pathway and PPAR-γ expression (in the presence or absence of PPAR-γ inhibitors) were analyzed. Results In vivo results showed that PEDF expression decreased during the inflammatory phase and increased during the fibrotic phase. PEDF could inhibit the progression of pulmonary fibrosis in rats. In vitro results showed that PEDF could effectively inhibit TGF-β1-stimulated fibroblast activation and reduce the production of α-SMA and collagen-I. PEDF could inhibit the TGF-β1/smad pathway by up-regulating the activity of PPAR-γ. Conclusions PEDF can act as an anti-fibrotic factor, inhibit fibroblast activation by upregulating PPAR-γ activity and reduce BLM-induced pulmonary fibrosis in rats. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02027-4.
Collapse
Affiliation(s)
- Xichun Qin
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China.,Department of Thoracic and Cardiovascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Caili Jia
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Jingtian Liang
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Jiali Chen
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Zhixiang Chao
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Hao Qin
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Yanliang Yuan
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Zhiwei Liu
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Zhongming Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China
| | - Hongyan Dong
- Morphological Research Experiment Center, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| | - Hao Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, China.
| |
Collapse
|
13
|
Lin B, Zheng W, Jiang X. Crosstalk between Circulatory Microenvironment and Vascular Endothelial Cells in Acute Myocardial Infarction. J Inflamm Res 2021; 14:5597-5610. [PMID: 34744446 PMCID: PMC8565985 DOI: 10.2147/jir.s316414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/29/2021] [Indexed: 12/03/2022] Open
Abstract
Background The reason of high mortality of acute myocardial infarction (AMI) was the lack of exploring the cellular and molecular mechanism of AMI. Therefore, we explored the crosstalk among cells, as well as its potential molecular mechanism of mediating AMI. Methods The gene expression profile of peripheral blood, endothelial, platelets and mononuclear cells were applied to differentially expressed genes (DEGs) analysis. ClusterProfiler and the package of gene set enrichment analysis (GSEA) were applied to explore the potential functional pathways of DEGs in 3 types of intravascular cells (endothelial, platelets and mononuclear cells) and peripheral blood. Subsequently, we extracted the surface receptors, secreted proteins and extracellular matrix from the up-regulated DEGs to explore their potential interactions mechanism of AMI by crosstalk and pivot analysis. Findings A total 11 common regulated DEGs (CDEGs) were identified, which might be potential biomarkers for AMI diagnosis. The abnormal pathways involved in DEGs of 3 types of intravascular cells and peripheral blood were shown, which also verified by GSEA. Afterwards, it was found that there was crosstalk in 3 types of intravascular cells and peripheral blood. Furthermore, we constructed a cell–cell interaction map among cells in AMI regulated by exosome lncRNA, which was involved in the development of AMI. Finally, we identified 8 hub genes, which might be potential biomarkers of AMI. Interpretation The result of this study can not only be used as a reference for subsequent experiments and further exploration, but also contribute to the development of novel cell and molecular therapies.
Collapse
Affiliation(s)
- Beiyou Lin
- Department of Cardiology, Zhuhai People's Hospital, (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, People's Republic of China
| | - Weiwei Zheng
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital & Zhengzhou University People's Hospital & Henan University People's Hospital, Zhengzhou, 450003, Henan, People's Republic of China
| | - Xiaofei Jiang
- Department of Cardiology, Zhuhai People's Hospital, (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, People's Republic of China
| |
Collapse
|
14
|
Zhang H, Li Z, Quan X, Liu X, Sun T, Wei T, Pan J, Liu Z, Wang M, Dong H, Zhang Z. Strategies to Attenuate Myocardial Infarction and No-Reflow Through Preservation of Vascular Integrity by Pigment Epithelium-Derived Factor. Hum Gene Ther 2021; 33:330-345. [PMID: 34278806 DOI: 10.1089/hum.2021.068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The phenomenon of no-reflow seriously limits the therapeutic value of coronary recanalization and leads to poor prognosis. Recent studies have demonstrated the potential role of pigment epithelium-derived factor (PEDF) in stabilizing endothelial cell junction, reducing vascular permeability and maintaining a quiescent vasculature. In this study, intramyocardial gene delivery was performed 5 days before the acute myocardial infarction/recanalization experiment in male rats. Positron emission tomography perfusion imaging with 13N-NH3 indicated PEDF to promote microvascular reperfusion significantly 4 h postcoronary occlusion. PEDF was observed to maintain the stability of endothelial adherens junctions (AJs), thus preventing the occurrence of no-reflow. PEDF reduced the hypoxia-induced vascular endothelial (VE)-cadherin endocytosis through PEDF/LR/Src/VE-cadherin S665 axis in vitro, which was remarkably observed to maintain endothelial AJs. Generally, PEDF might function as a relevant target for therapeutic vasculoprotection by way of regulating the phosphorylation level of VE-cadherin according to our data, thus being crucial for preventing no-reflow.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhimin Li
- Department of Cardiothoracic Surgery, Xuzhou Cancer Hospital, Xuzhou, China
| | - Xiaoyu Quan
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Thoracic Surgery, Tongji University School of Medicine, Shanghai Pulmonary Hospital, Shanghai, China
| | - Teng Sun
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tengteng Wei
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jiajun Pan
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhiwei Liu
- Morphological Research Experiment Center, Xuzhou Medical University, Xuzhou, China
| | - Meng Wang
- Morphological Research Experiment Center, Xuzhou Medical University, Xuzhou, China
| | - Hongyan Dong
- Morphological Research Experiment Center, Xuzhou Medical University, Xuzhou, China
| | - Zhongming Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
15
|
Tomin T, Schittmayer M, Sedej S, Bugger H, Gollmer J, Honeder S, Darnhofer B, Liesinger L, Zuckermann A, Rainer PP, Birner-Gruenberger R. Mass Spectrometry-Based Redox and Protein Profiling of Failing Human Hearts. Int J Mol Sci 2021; 22:1787. [PMID: 33670142 PMCID: PMC7916846 DOI: 10.3390/ijms22041787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress contributes to detrimental functional decline of the myocardium, leading to the impairment of the antioxidative defense, dysregulation of redox signaling, and protein damage. In order to precisely dissect the changes of the myocardial redox state correlated with oxidative stress and heart failure, we subjected left-ventricular tissue specimens collected from control or failing human hearts to comprehensive mass spectrometry-based redox and quantitative proteomics, as well as glutathione status analyses. As a result, we report that failing hearts have lower glutathione to glutathione disulfide ratios and increased oxidation of a number of different proteins, including constituents of the contractile machinery as well as glycolytic enzymes. Furthermore, quantitative proteomics of failing hearts revealed a higher abundance of proteins responsible for extracellular matrix remodeling and reduced abundance of several ion transporters, corroborating contractile impairment. Similar effects were recapitulated by an in vitro cell culture model under a controlled oxygen atmosphere. Together, this study provides to our knowledge the most comprehensive report integrating analyses of protein abundance and global and peptide-level redox state in end-stage failing human hearts as well as oxygen-dependent redox and global proteome profiles of cultured human cardiomyocytes.
Collapse
Affiliation(s)
- Tamara Tomin
- Faculty of Technical Chemistry, Institute of Chemical Technologies and Analytics, Vienna University of Technology-TU Wien, Getreidemarkt 9/164, 1060 Vienna, Austria;
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Matthias Schittmayer
- Faculty of Technical Chemistry, Institute of Chemical Technologies and Analytics, Vienna University of Technology-TU Wien, Getreidemarkt 9/164, 1060 Vienna, Austria;
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Simon Sedej
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
| | - Johannes Gollmer
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
| | - Sophie Honeder
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Barbara Darnhofer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Laura Liesinger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Andreas Zuckermann
- Cardiac Transplantation, Department of Cardiac Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria;
| | - Peter P. Rainer
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
| | - Ruth Birner-Gruenberger
- Faculty of Technical Chemistry, Institute of Chemical Technologies and Analytics, Vienna University of Technology-TU Wien, Getreidemarkt 9/164, 1060 Vienna, Austria;
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| |
Collapse
|
16
|
Cheng W, Li X, Liu D, Cui C, Wang X. Endothelial-to-Mesenchymal Transition: Role in Cardiac Fibrosis. J Cardiovasc Pharmacol Ther 2020; 26:3-11. [PMID: 32851865 DOI: 10.1177/1074248420952233] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT) is a complex biological process by which endothelial cells lose their endothelial cell characteristics and acquire mesenchymal cell properties under certain physiological or pathological conditions. Recently, it has been found that EndMT plays an important role in the occurrence and development of fibrotic cardiovascular diseases. In this review, we first summarize the main induction pathways involved in EndMT process. In addition, we discuss the role of EndMT in fibrotic cardiovascular diseases and its potential implication in new therapeutic interventions.
Collapse
Affiliation(s)
- Weijia Cheng
- Henan Key Laboratory of Medical Tissue Regeneration, 91593Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiao Li
- Henan Key Laboratory of Medical Tissue Regeneration, 91593Xinxiang Medical University, Xinxiang, Henan, China
| | - Dongling Liu
- Henan Key Laboratory of Medical Tissue Regeneration, 91593Xinxiang Medical University, Xinxiang, Henan, China
| | - Chaochu Cui
- Henan Key Laboratory of Medical Tissue Regeneration, 91593Xinxiang Medical University, Xinxiang, Henan, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, 91593Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
17
|
Ma D, Chen L, Shi J, Zhao Y, Vasani S, Chen K, Romana‐Souza B, Henkin J, DiPietro LA. Pigment epithelium‐derived factor attenuates angiogenesis and collagen deposition in hypertrophic scars. Wound Repair Regen 2020; 28:684-695. [DOI: 10.1111/wrr.12828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Da Ma
- Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital Guanghua School of Stomatology, SunYat‐sen University Guangzhou Guangdong China
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Junhe Shi
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Yan Zhao
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Shruti Vasani
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Kevin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Bruna Romana‐Souza
- Tissue Repair Laboratory State University of Rio de Janeiro Rio de Janeiro Brazil
| | - Jack Henkin
- Center for Developmental Therapeutics and Department of Chemistry Northwestern University Evanston Illinois USA
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| |
Collapse
|
18
|
Pigment Epithelium-Derived Factor as a Possible Treatment Agent for Choroidal Neovascularization. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8941057. [PMID: 32215180 PMCID: PMC7079215 DOI: 10.1155/2020/8941057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/28/2020] [Indexed: 02/06/2023]
Abstract
Choroidal neovascularization (CNV) is a sight-threatening disease and is characterized by the formation of pathological neovascularization in the choroid which extends into the subretinal space. Exudative age-related macular degeneration (AMD) is the formation of CNV in the macular area which leads to irreversible blindness. Continuous leakage and hemorrhage of the CNV lesion may eventually result in scarring or later fibrosis, which could result in photoreceptor cell atrophy. The current strategy for treating CNV is the use of antivascular endothelial growth factor (VEGF) agents. Many studies have demonstrated the efficacy of intravitreal anti-VEGF therapy. Other studies have also reported the side effects of single anti-VEGF treatment. And long-term inhibition of a single system may result in collateral damage to other visual elements. Pigment epithelium-derived factor (PEDF) is a 50 kDa protein that was first isolated from the conditioned medium of human RPE cells. PEDF has both antiangiogenesis and neuroprotective functions for photoreceptor cells. It may be a potential ocular antiangiogenic agent. This review outlines the distribution of PEDF in the eye, the mechanism of antiangiogenesis, the protective effect on the retina, and the relationship between PEDF and VEGF.
Collapse
|
19
|
Piera-Velazquez S, Jimenez SA. Endothelial to Mesenchymal Transition: Role in Physiology and in the Pathogenesis of Human Diseases. Physiol Rev 2019; 99:1281-1324. [PMID: 30864875 DOI: 10.1152/physrev.00021.2018] [Citation(s) in RCA: 402] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Numerous studies have demonstrated that endothelial cells are capable of undergoing endothelial to mesenchymal transition (EndMT), a newly recognized type of cellular transdifferentiation. EndMT is a complex biological process in which endothelial cells adopt a mesenchymal phenotype displaying typical mesenchymal cell morphology and functions, including the acquisition of cellular motility and contractile properties. Endothelial cells undergoing EndMT lose the expression of endothelial cell-specific proteins such as CD31/platelet-endothelial cell adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin and initiate the expression of mesenchymal cell-specific genes and the production of their encoded proteins including α-smooth muscle actin, extra domain A fibronectin, N-cadherin, vimentin, fibroblast specific protein-1, also known as S100A4 protein, and fibrillar type I and type III collagens. Transforming growth factor-β1 is considered the main EndMT inducer. However, EndMT involves numerous molecular and signaling pathways that are triggered and modulated by multiple and often redundant mechanisms depending on the specific cellular context and on the physiological or pathological status of the cells. EndMT participates in highly important embryonic development processes, as well as in the pathogenesis of numerous genetically determined and acquired human diseases including malignant, vascular, inflammatory, and fibrotic disorders. Despite intensive investigation, many aspects of EndMT remain to be elucidated. The identification of molecules and regulatory pathways involved in EndMT and the discovery of specific EndMT inhibitors should provide novel therapeutic approaches for various human disorders mediated by EndMT.
Collapse
Affiliation(s)
- Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
20
|
PEDF decreases cardiomyocyte edema during oxygen‑glucose deprivation and recovery via inhibiting lactate accumulation and expression of AQP1. Int J Mol Med 2019; 43:1979-1990. [PMID: 30864707 PMCID: PMC6445592 DOI: 10.3892/ijmm.2019.4132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/28/2019] [Indexed: 01/13/2023] Open
Abstract
Myocardial edema is divided into cellular edema and interstitial edema; however, the dynamic change of cardiomyocyte edema has not been described in detail. Pigment epithelium-derived factor (PEDF) is known for its protective effects on ischemic cardiomyocytes; however, the association between PEDF and cardiomyocyte edema remains to be fully elucidated. In the present study, rat neonatal left ventricular cardiomyocytes were isolated and treated with oxygen-glucose deprivation (OGD) and recovery. During OGD and recovery, the cardiomyocytes exhibited significant edema following 30 min of OGD (OGD 30 min) and OGD 30 min with recovery for 6 h. PEDF significantly decreased the lactate content and extracellular acidification rate of the OGD-treated cardiomyocytes, thereby reducing cellular osmotic gradients and preventing the occurrence of cell edema. In addition, the glycolytic agonist, fructose-1, 6-diphosphate, eliminated the effect of PEDF on inhibiting edema in the OGD-treated cardiomyocytes. Furthermore, PEDF reduced the protein and mRNA expression of aquaporin 1 (AQP1), and thus downregulated cardiomyocyte edema during the OGD/recovery period. The addition of AQP1 agonist, arginine vasopressin, inhibited the inhibitory effect of PEDF on cardiomyocyte edema during OGD/recovery. In conclusion, the present study revealed a novel mechanism for the regulation of cardiomyocyte edema by PEDF involving lactate levels and the expression of AQP1 during OGD/recovery. The reduction of lactate content during OGD was associated with a decrease in the protein level of AQP1 during OGD/recovery; therefore, PEDF decreased cardiomyocyte edema and cellular apoptosis, prolonging the viability of the cells.
Collapse
|
21
|
Chen H, Liu Y, Gui Q, Zhu X, Zeng L, Meng J, Qing J, Gao L, Jackson AO, Feng J, Li Y, He J, Yin K. Ghrelin attenuates myocardial fibrosis after acute myocardial infarction via inhibiting endothelial-to mesenchymal transition in rat model. Peptides 2019; 111:118-126. [PMID: 30218693 DOI: 10.1016/j.peptides.2018.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/08/2018] [Accepted: 09/10/2018] [Indexed: 12/17/2022]
Abstract
Ghrelin, a peptide hormone produced in the gastrointestinal tract, has recently been found to be associated with the onset of myocardial fibrosis (MF). The exact mechanism, however, remains elusive. This study sought to identify the function and mechanism of ghrelin on MF after acute myocardial infarction (AMI). AMI was established in Spraque-Dawley rats by ligation of the left anterior descending (LAD). Ghrelin or saline was intraperitoneally injected two times per day for 8 weeks after ligation. The weight of heart (mg) and the weight ratio of heart to body (mg/g) as well as the fibrotic area were increased, while serum level of ghrelin was decreased after AMI. Ghrelin significantly ameliorated MF and decreased deposition of collagens in perivascular fibrosis area. In addition, ghrelin inhibited Endothelial-to-mesenchymal transition (EndMT), a crucial process for MF, in perivascular fibrosis area and TGF-β1-induced human coronary artery endothelial cells (HCAECs). Mechanistically, ghrelin persistently decreased the phosphorylation of Smad2/3 and enhanced the expression of Smad7 and p-AMPK in vivo and in vitro. After the abolition of Smad7, GHSR-1a and AMPK pathway, the effect of ghrelin on EndMT was significantly inhibited. In conclusion, these results presented a novel finding that ghrelin attenuated MF after AMI via regulation EndMT in a GHSR-1a/AMPK/Smad7- dependent manner.
Collapse
Affiliation(s)
- Hainan Chen
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; Institute of Cardiovascular Research, Key Laboratory Atherosclerology of Hunan Province, University of South China, Hengyang 421001, China
| | - Yijian Liu
- The Third Hospital of Changsha, Changsha 410000, China
| | - Qingjun Gui
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Xiao Zhu
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; Institute of Cardiovascular Research, Key Laboratory Atherosclerology of Hunan Province, University of South China, Hengyang 421001, China
| | - Lin Zeng
- Department of Neurology, First Affiliated Hospital of University of South China, University of South China, Hengyang 421001, China
| | - Jun Meng
- Functional Department, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Jina Qing
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Ling Gao
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Ampadu O Jackson
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; International College, University of South China, Hengyang 421001, China
| | - Juling Feng
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Yi Li
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China
| | - Jin He
- Functional Department, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China.
| | - Kai Yin
- Research Lab for Clinical & Translational Medicine, Medical school, University of South China, Hengyang 421001, China; Institute of Cardiovascular Research, Key Laboratory Atherosclerology of Hunan Province, University of South China, Hengyang 421001, China.
| |
Collapse
|
22
|
Abstract
Ischaemic heart disease is a leading cause of death worldwide. Injury to the heart is followed by loss of the damaged cardiomyocytes, which are replaced with fibrotic scar tissue. Depletion of cardiomyocytes results in decreased cardiac contraction, which leads to pathological cardiac dilatation, additional cardiomyocyte loss, and mechanical dysfunction, culminating in heart failure. This sequential reaction is defined as cardiac remodelling. Many therapies have focused on preventing the progressive process of cardiac remodelling to heart failure. However, after patients have developed end-stage heart failure, intervention is limited to heart transplantation. One of the main reasons for the dramatic injurious effect of cardiomyocyte loss is that the adult human heart has minimal regenerative capacity. In the past 2 decades, several strategies to repair the injured heart and improve heart function have been pursued, including cellular and noncellular therapies. In this Review, we discuss current therapeutic approaches for cardiac repair and regeneration, describing outcomes, limitations, and future prospects of preclinical and clinical trials of heart regeneration. Substantial progress has been made towards understanding the cellular and molecular mechanisms regulating heart regeneration, offering the potential to control cardiac remodelling and redirect the adult heart to a regenerative state.
Collapse
Affiliation(s)
- Hisayuki Hashimoto
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
23
|
Wang Z, Perez M, Lee ES, Kojima S, Griffin M. The functional relationship between transglutaminase 2 and transforming growth factor β1 in the regulation of angiogenesis and endothelial-mesenchymal transition. Cell Death Dis 2017; 8:e3032. [PMID: 28880274 PMCID: PMC5636968 DOI: 10.1038/cddis.2017.399] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/20/2017] [Accepted: 05/23/2017] [Indexed: 01/29/2023]
Abstract
The importance of transglutaminase 2 (TG2) in angiogenesis has been highlighted in recent studies, but other roles of this multi-functional enzyme in endothelial cell (EC) function still remains to be fully elucidated. We previously showed that the extracellular TG2 is involved in maintaining tubule formation in ECs by a mechanism involving matrix-bound vascular endothelial growth factor (VEGF) signalling. Here, by using the ECs and fibroblast co-culture and ECs 3D culture models, we demonstrate a further role for TG2 in both endothelial tubule formation and in tubule loss, which involves its role in the regulation of transforming growth factor β1 (TGFβ1) and Smad signalling. We demonstrate that inhibition of tubule formation by TG2 inhibitors can be restored by add-back of exogenous TGFβ1 at pg/ml levels and show that TG2 -/- mouse ECs are unable to form tubules in 3D culture and display negligible Smad signalling compared to wild-type cells. Loss of tubule formation in the TG2 -/- ECs can be reconstituted by transduction with TG2. We demonstrate that extracellular TG2 also has an important role in TGFβ1-induced transition of ECs into myofibroblast-like cells (endothelial-mesenchymal transition), resulting in loss of EC tubules and tubule formation. Our data also indicate that TG2 may have a role in regulating TGFβ signalling through entrapment of active TGFβ1 into the extracellular matrix. In conclusion, our work demonstrates that TG2 has multi-functional roles in ECs where its ability to fine-tune of TGFβ1 signalling means it can be involved in both endothelial tubule formation and tubule rarefaction.
Collapse
Affiliation(s)
- Zhuo Wang
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, UK
| | - Mileidys Perez
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, UK
| | - Eun-Seo Lee
- Micro-Signaling Regulation Technology Unit, Imaging Application Group, Division of Bio-function Dynamics Imaging, Center for Life Science Technology, RIKEN 2-1 Hirosawa, Wako 351-0198, Japan
| | - Soichi Kojima
- Micro-Signaling Regulation Technology Unit, Imaging Application Group, Division of Bio-function Dynamics Imaging, Center for Life Science Technology, RIKEN 2-1 Hirosawa, Wako 351-0198, Japan
| | - Martin Griffin
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, UK
| |
Collapse
|
24
|
Autophagy attenuates endothelial-to-mesenchymal transition by promoting Snail degradation in human cardiac microvascular endothelial cells. Biosci Rep 2017; 37:BSR20171049. [PMID: 28811357 PMCID: PMC5587916 DOI: 10.1042/bsr20171049] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/09/2017] [Accepted: 08/15/2017] [Indexed: 01/06/2023] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) mainly exists in cardiovascular development and disease progression, and is well known to contribute to cardiac fibrosis. Recent studies indicated that autophagy also participates in the regulation of cardiac fibrosis. However, the precise role of autophagy in cardiac fibrosis and the underlying molecular mechanism remain unclear. The present study aimed to explore the role of autophagy in EndMT, reveal the underlying molecular mechanism, and seek new therapy for cardiac fibrosis. In the present study, we found that EndMT and autophagy were induced simultaneously by hypoxia in human cardiac microvascular endothelial cells (HCMECs). Rapamycin, an autophagy enhancer, attenuated EndMT with promoting angiogenesis, while 3-methyladenine (3-MA) and chloroquine (CQ), agents that inhibit autophagy, accelerated the progression accompanied by the decrease in counts of tube formation under hypoxia conditions. Interestingly, intervening autophagy by rapamycin, 3-MA, or CQ did not affect hypoxia-induced autocrine TGFβ signaling, but changed the expression of Snail protein without alterations in the expression of Snail mRNA. Furthermore, the colocalization of LC3 and Snail indicated that autophagy might mediate Snail degradation under hypoxia conditions in HCMECs. Interaction of p62, the substrate of autophagy, with Snail by co-immunoprecipitation especially in hypoxia-incubated cells confirmed the hypothesis. In conclusion, autophagy serves as a cytoprotective mechanism against EndMT to promote angiogenesis by degrading Snail under hypoxia conditions, suggesting that autophagy targetted therapeutic strategies may be applicable for cardiac fibrosis by EndMT.
Collapse
|