1
|
Chong A, Joshua J, Raheb S, Pires A, Colpitts M, Caswell JL, Fonfara S. Evaluation of potential novel biomarkers for feline hypertrophic cardiomyopathy. Res Vet Sci 2024; 180:105430. [PMID: 39395261 DOI: 10.1016/j.rvsc.2024.105430] [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: 06/03/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common cardiomyopathy in cats. The diagnosis can be difficult, requiring advanced echocardiographic skills. Additionally, circulating biomarkers (N-terminal pro-B type natriuretic peptide and cardiac troponin I) have several limitations when used for HCM screening. In previous work, we identified interleukin 18 (IL-18), insulin-like growth factor binding protein 2 (IGFBP-2), brain-type glycogen phosphorylase B (PYGB), and WNT Family Member 5 A (WNT5A) as myocardial genes that show significant differential expression between cats with HCM and healthy cats. The products of these genes are released into the circulation, and we hypothesized that IL-18, IGFBP-2, PYGB, and WNT5A serum RNA and protein concentrations differ between healthy cats, cats with subclinical HCM, and those with HCM and congestive heart failure (HCM + CHF). Reverse transcriptase quantitative polymerase chain reaction (RTqPCR) and enzyme-linked immunosorbent assay (ELISA) were applied to evaluate gene and protein expression, respectively, in the serum of eight healthy controls, eight cats with subclinical HCM, and six cats with HCM + CHF. Serum IGFBP-2 RNA concentrations were significantly different among groups and were highest in cats with subclinical HCM. Compared to healthy controls, serum IL-18 and WNT5A gene expression were significantly higher in cats with HCM + CHF, and WNT5A was higher in cats with subclinical HCM. No differences were observed for PYGB. These results indicate that further investigation via large scale clinical studies for IGFBP-2, WNT5A, and IL-18 may be valuable in diagnosing and staging feline HCM.
Collapse
Affiliation(s)
- Andrew Chong
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Jessica Joshua
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada; Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road E., Guelph, ON N1G 2W1, Canada
| | - Shari Raheb
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Ananda Pires
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Michelle Colpitts
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Jeff L Caswell
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road E., Guelph, ON N1G 2W1, Canada
| | - Sonja Fonfara
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
2
|
Fu Y, Zhang X, Wu H, Zhang P, Liu S, Guo T, Shan H, Liang Y, Chen H, Xie J, Duan Y. HOXA3 functions as the on-off switch to regulate the development of hESC-derived third pharyngeal pouch endoderm through EPHB2-mediated Wnt pathway. Front Immunol 2024; 14:1258074. [PMID: 38259452 PMCID: PMC10800530 DOI: 10.3389/fimmu.2023.1258074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
Objectives Normal commitment of the endoderm of the third pharyngeal pouch (3PP) is essential for the development and differentiation of the thymus. The aim of this study was to investigate the role of transcription factor HOXA3 in the development and differentiation of 3PP endoderm (3PPE) from human embryonic stem cells (hESCs). Methods The 3PPE was differentiated from hESC-derived definitive endoderm (DE) by mimicking developmental queues with Activin A, WNT3A, retinoic acid and BMP4. The function of 3PPE was assessed by further differentiating into functional thymic epithelial cells (TECs). The effect of HOXA3 inhibition on cells of 3PPE was subsequently investigated. Results A highly efficient approach for differentiating 3PPE cells was developed and these cells expressed 3PPE related genes HOXA3, SIX1, PAX9 as well as EpCAM. 3PPE cells had a strong potential to develop into TECs which expressed both cortical TEC markers K8 and CD205, and medullary TEC markers K5 and AIRE, and also promoted the development and maturation of T cells. More importantly, transcription factor HOXA3 not only regulated the differentiation of 3PPE, but also had a crucial role for the proliferation and migration of 3PPE cells. Our further investigation revealed that HOXA3 controlled the commitment and function of 3PPE through the regulation of Wnt signaling pathway by activating EPHB2. Conclusion Our results demonstrated that HOXA3 functioned as the on-off switch to regulate the development of hESC-derived 3PPE through EPHB2-mediated Wnt pathway, and our findings will provide new insights into studying the development of 3PP and thymic organ in vitro and in vivo.
Collapse
Affiliation(s)
- Yingjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xueyan Zhang
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Pingping Zhang
- Department of Laboratory Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Tingting Guo
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huanhuan Shan
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yan Liang
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jinghe Xie
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- The Innovation Centre of Ministry of Education for Development and Diseases, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
3
|
Madè A, Bibi A, Garcia-Manteiga JM, Tascini AS, Piella SN, Tikhomirov R, Voellenkle C, Gaetano C, Leszek P, Castelvecchio S, Menicanti L, Martelli F, Greco S. circRNA-miRNA-mRNA Deregulated Network in Ischemic Heart Failure Patients. Cells 2023; 12:2578. [PMID: 37947656 PMCID: PMC10648415 DOI: 10.3390/cells12212578] [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: 09/15/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Noncoding RNAs (ncRNAs), which include circular RNAs (circRNAs) and microRNAs (miRNAs), regulate the development of cardiovascular diseases (CVD). Notably, circRNAs can interact with miRNAs, influencing their specific mRNA targets' levels and shaping a competing endogenous RNAs (ceRNA) network. However, these interactions and their respective functions remain largely unexplored in ischemic heart failure (IHF). This study is aimed at identifying circRNA-centered ceRNA networks in non-end-stage IHF. Approximately 662 circRNA-miRNA-mRNA interactions were identified in the heart by combining state-of-the-art bioinformatics tools with experimental data. Importantly, KEGG terms of the enriched mRNA indicated CVD-related signaling pathways. A specific network centered on circBPTF was validated experimentally. The levels of let-7a-5p, miR-18a-3p, miR-146b-5p, and miR-196b-5p were enriched in circBPTF pull-down experiments, and circBPTF silencing inhibited the expression of HDAC9 and LRRC17, which are targets of miR-196b-5p. Furthermore, as suggested by the enriched pathway terms of the circBPTF ceRNA network, circBPTF inhibition elicited endothelial cell cycle arrest. circBPTF expression increased in endothelial cells exposed to hypoxia, and its upregulation was confirmed in cardiac samples of 36 end-stage IHF patients compared to healthy controls. In conclusion, circRNAs act as miRNA sponges, regulating the functions of multiple mRNA targets, thus providing a novel vision of HF pathogenesis and laying the theoretical foundation for further experimental studies.
Collapse
Affiliation(s)
- Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Alessia Bibi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jose Manuel Garcia-Manteiga
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
| | - Anna Sofia Tascini
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Santiago Nicolas Piella
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Roman Tikhomirov
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, National Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Serenella Castelvecchio
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Lorenzo Menicanti
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| |
Collapse
|
4
|
Meyer IS, Li X, Meyer C, Voloshanenko O, Pohl S, Boutros M, Katus HA, Frey N, Leuschner F. Blockade of Wnt Secretion Attenuates Myocardial Ischemia-Reperfusion Injury by Modulating the Inflammatory Response. Int J Mol Sci 2022; 23:ijms232012252. [PMID: 36293109 PMCID: PMC9602582 DOI: 10.3390/ijms232012252] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Wnt (a portmanteau of Wingless and Int-1) signaling in the adult heart is largely quiescent. However, there is accumulating evidence that it gets reactivated during the healing process after myocardial infarction (MI). We here tested the therapeutic potential of the Wnt secretion inhibitor LGK-974 on MI healing. Ischemia/reperfusion (I/R) injury was induced in mice and Wnt signaling was inhibited by oral administration of the porcupine inhibitor LGK-974. The transcriptome was analyzed from infarcted tissue by using RNA sequencing analysis. The inflammatory response after I/R was evaluated by flow cytometry. Heart function was assessed by echocardiography and fibrosis by Masson's trichrome staining. Transcriptome and gene set enrichment analysis revealed a modulation of the inflammatory response upon administration of the Wnt secretion inhibitor LGK-974 following I/R. In addition, LGK-974-treated animals showed an attenuated inflammatory response and improved heart function. In an in vitro model of hypoxic cardiomyocyte and monocyte/macrophage interaction, LGK974 inhibited the activation of Wnt signaling in monocytes/macrophages and reduced their pro-inflammatory phenotype. We here show that Wnt signaling affects inflammatory processes after MI. The Wnt secretion inhibitor LGK-974 appears to be a promising compound for future immunomodulatory approaches to improve cardiac remodeling after MI.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Xue Li
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Carina Meyer
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | | - Susann Pohl
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hugo Albert Katus
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Norbert Frey
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
| | - Florian Leuschner
- Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg-Mannheim, 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
5
|
Kania K, Ahmed A, Ahmed S, Rådegran G. Elevated plasma WIF-1 levels are associated with worse prognosis in heart failure with pulmonary hypertension. ESC Heart Fail 2022; 9:4139-4149. [PMID: 36082780 PMCID: PMC9773778 DOI: 10.1002/ehf2.14148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 01/19/2023] Open
Abstract
AIMS Heart failure (HF) is a progressive condition that is becoming more prevalent in the ageing population. Pulmonary hypertension is a common complicating factor in HF and negatively impacts survival. Plasma biomarkers are a potential method for determining the prognosis of patients with left heart failure with pulmonary hypertension (LHF-PH). We aimed to analyse the prognostic capability of 33 proteins related to, among other pathways, inflammation, coagulation, and Wnt signalling in LHF-PH. METHODS Plasma levels of 33 proteins were analysed using proximity extension assay from the plasma of 20 controls and 67 LHF-PH patients, whereof 19 underwent heart transplantation (HT). Haemodynamics in the patients were assessed using right heart catheterization. RESULTS Eleven proteins had elevated plasma levels in LHF-PH compared with controls (P < 0.01), which decreased towards the controls' levels after HT (P < 0.01). Survival analysis of these proteins showed that elevated plasma levels of growth hormone, programmed cell death 1 ligand 2, tissue factor pathway inhibitor 2, and Wnt inhibitory factor 1 (WIF-1) were associated with worse transplantation-free survival in LHF-PH (P < 0.05). When adjusted for age, sex and N-terminal pro-brain natriuretic peptide (NT-proBNP) levels using multivariable cox regressions, only WIF-1 remained prognostic [hazard ratio (95% confidence interval)] [1.013 (1.001-1.024)]. WIF-1 levels in LHF-PH patients also correlated with the mean right atrial pressure (rs = 0.42; P < 0.01), stroke volume index (rs = 0.41; P < 0.01), cardiac index (rs = -0.42; P < 0.01), left ventricular stroke work index (rs = -0.41; P < 0.01), and NT-proBNP (rs = 0.63; P < 0.01). CONCLUSIONS The present study demonstrated that LHF-PH patients have higher plasma WIF-1 levels than healthy controls, suggesting that plasma WIF-1 may be a potential future prognostic biomarker in LHF-PH. Its prognostic capability could be further refined by including it in a multi-marker panel. Further studies are needed to establish the potential role of WIF-1 in LHF-PH pathophysiology in larger cohorts to determine its clinical applicability.
Collapse
Affiliation(s)
- Kriss Kania
- Department of Clinical Sciences Lund, CardiologyLund UniversityLundSweden,The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung MedicineSkåne University HospitalLundSweden
| | - Abdulla Ahmed
- Department of Clinical Sciences Lund, CardiologyLund UniversityLundSweden,The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung MedicineSkåne University HospitalLundSweden
| | - Salaheldin Ahmed
- Department of Clinical Sciences Lund, CardiologyLund UniversityLundSweden,The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung MedicineSkåne University HospitalLundSweden
| | - Göran Rådegran
- Department of Clinical Sciences Lund, CardiologyLund UniversityLundSweden,The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung MedicineSkåne University HospitalLundSweden
| |
Collapse
|
6
|
A New Hypothetical Concept in Metabolic Understanding of Cardiac Fibrosis: Glycolysis Combined with TGF-β and KLF5 Signaling. Int J Mol Sci 2022; 23:ijms23084302. [PMID: 35457114 PMCID: PMC9027193 DOI: 10.3390/ijms23084302] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 12/16/2022] Open
Abstract
The accumulation of fibrosis in cardiac tissues is one of the leading causes of heart failure. The principal cellular effectors in cardiac fibrosis are activated fibroblasts and myofibroblasts, which serve as the primary source of matrix proteins. TGF-β signaling pathways play a prominent role in cardiac fibrosis. The control of TGF-β by KLF5 in cardiac fibrosis has been demonstrated for modulating cardiovascular remodeling. Since the expression of KLF5 is reduced, the accumulation of fibrosis diminishes. Because the molecular mechanism of fibrosis is still being explored, there are currently few options for effectively reducing or reversing it. Studying metabolic alterations is considered an essential process that supports the explanation of fibrosis in a variety of organs and especially the glycolysis alteration in the heart. However, the interplay among the main factors involved in fibrosis pathogenesis, namely TGF-β, KLF5, and the metabolic process in glycolysis, is still indistinct. In this review, we explain what we know about cardiac fibroblasts and how they could help with heart repair. Moreover, we hypothesize and summarize the knowledge trend on the molecular mechanism of TGF-β, KLF5, the role of the glycolysis pathway in fibrosis, and present the future therapy of cardiac fibrosis. These studies may target therapies that could become important strategies for fibrosis reduction in the future.
Collapse
|
7
|
Guo Q, Lai Y, Chu J, Chen X, Gao M, Sang C, Dong J, Pu J, Ma C. LRP6 Polymorphisms Is Associated With Sudden Cardiac Death in Patients With Chronic Heart Failure in the Chinese Han Population. Front Cardiovasc Med 2022; 8:815595. [PMID: 35187114 PMCID: PMC8854291 DOI: 10.3389/fcvm.2021.815595] [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: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6) plays a critical role in cardiovascular homeostasis. The deficiency of LRP6 is associated with a high risk of arrhythmias. However, the association between genetic variations of LRP6 and sudden cardiac death (SCD) remains unknown. This study aims to explore the association between common variants of LRP6 and the prognosis of chronic heart failure (CHF) patients. From July 2005 to December 2009, patients with CHF were enrolled from 10 hospitals in China. The single-nucleotide polymorphism (SNP) rs2302684 was selected for the evaluation of the effect of LRP6 polymorphisms on the survival in patients with CHF. A total of 1,437 patients with CHF were finally included for the analysis. During a median follow-up of 61 months (range 0.4–129 months), a total of 546 (38.0%) patients died, including 201 (36.8%) cases with SCD and 345 (63.2%) cases with non-SCD. Patients carrying A allele of rs2302684 had an increased risk of all-cause death (adjusted HR 1.452, 95% CI 1.189–1.706; P < 0.001) and SCD (adjusted HR 1.783, 95% CI 1.337–2.378; P < 0.001). Therefore, the SNP rs2302684 T>A in LRP6 indicated higher risks of all-cause death and SCD in patients with CHF. LRP6 could be added as a novel predictor of SCD and might be a potential therapeutic target in the prevention of SCD in the CHF population.
Collapse
Affiliation(s)
- Qi Guo
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yiwei Lai
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jianmin Chu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuhua Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingyang Gao
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Caihua Sang
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jianzeng Dong
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jielin Pu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Shanghai East Hospital, Tongji University, Shanghai, China
- *Correspondence: Jielin Pu
| | - Changsheng Ma
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Changsheng Ma
| |
Collapse
|
8
|
Yin C, Ye Z, Wu J, Huang C, Pan L, Ding H, Zhong L, Guo L, Zou Y, Wang X, Wang Y, Gao P, Jin X, Yan X, Zou Y, Huang R, Gong H. Elevated Wnt2 and Wnt4 activate NF-κB signaling to promote cardiac fibrosis by cooperation of Fzd4/2 and LRP6 following myocardial infarction. EBioMedicine 2021; 74:103745. [PMID: 34911029 PMCID: PMC8669316 DOI: 10.1016/j.ebiom.2021.103745] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/29/2022] Open
Abstract
Background Acute myocardial infarction (AMI)-induced excessive myocardial fibrosis exaggerates cardiac dysfunction. However, serum Wnt2 or Wnt4 level in AMI patients, and the roles in cardiac fibrosis are largely unkown. Methods AMI and non-AMI patients were enrolled to examine serum Wnt2 and Wnt4 levels by ELISA analysis. The AMI patients were followed-up for one year. MI mouse model was built by ligation of left anterior descending branch (LAD). Findings Serum Wnt2 or Wnt4 level was increased in patients with AMI, and the elevated Wnt2 and Wnt4 were correlated to adverse outcome of these patients. Knockdown of Wnt2 and Wnt4 significantly attenuated myocardial remodeling and cardiac dysfunction following experimental MI. In vitro, hypoxia enhanced the secretion and expression of Wnt2 and Wnt4 in neonatal rat cardiac myocytes (NRCMs) or fibroblasts (NRCFs). Mechanistically, the elevated Wnt2 or Wnt4 activated β-catenin /NF-κB signaling to promote pro-fibrotic effects in cultured NRCFs. In addition, Wnt2 or Wnt4 upregulated the expression of these Wnt co-receptors, frizzled (Fzd) 2, Fzd4 and (ow-density lipoprotein receptor-related protein 6 (LRP6). Further analysis revealed that Wnt2 or Wnt4 activated β-catenin /NF-κB by the co-operation of Fzd4 or Fzd2 and LRP6 signaling, respectively. Interpretation Elevated Wnt2 and Wnt4 activate β-catenin/NF-κB signaling to promote cardiac fibrosis by cooperation of Fzd4/2 and LRP6 in fibroblasts, which contributes to adverse outcome of patients with AMI, suggesting that systemic inhibition of Wnt2 and Wnt4 may improve cardiac dysfunction after MI.
Collapse
Affiliation(s)
- Chao Yin
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zhishuai Ye
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China
| | - Jian Wu
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Chenxing Huang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Le Pan
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Huaiyu Ding
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Zhong
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Guo
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yan Zou
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiang Wang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Wang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Pan Gao
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xuejuan Jin
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoxiang Yan
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunzeng Zou
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Rongchong Huang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China.
| | - Hui Gong
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
9
|
Bracco Gartner TCL, Stein JM, Muylaert DEP, Bouten CVC, Doevendans PA, Khademhosseini A, Suyker WJL, Sluijter JPG, Hjortnaes J. Advanced In Vitro Modeling to Study the Paradox of Mechanically Induced Cardiac Fibrosis. Tissue Eng Part C Methods 2021; 27:100-114. [PMID: 33407000 DOI: 10.1089/ten.tec.2020.0298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In heart failure, cardiac fibrosis is the result of an adverse remodeling process. Collagen is continuously synthesized in the myocardium in an ongoing attempt of the heart to repair itself. The resulting collagen depositions act counterproductively, causing diastolic dysfunction and disturbing electrical conduction. Efforts to treat cardiac fibrosis specifically have not been successful and the molecular etiology is only partially understood. The differentiation of quiescent cardiac fibroblasts to extracellular matrix-depositing myofibroblasts is a hallmark of cardiac fibrosis and a key aspect of the adverse remodeling process. This conversion is induced by a complex interplay of biochemical signals and mechanical stimuli. Tissue-engineered 3D models to study cardiac fibroblast behavior in vitro indicate that cyclic strain can activate a myofibroblast phenotype. This raises the question how fibroblast quiescence is maintained in the healthy myocardium, despite continuous stimulation of ultimately profibrotic mechanotransductive pathways. In this review, we will discuss the convergence of biochemical and mechanical differentiation signals of myofibroblasts, and hypothesize how these affect this paradoxical quiescence. Impact statement Mechanotransduction pathways of cardiac fibroblasts seem to ultimately be profibrotic in nature, but in healthy human myocardium, cardiac fibroblasts remain quiescent, despite continuous mechanical stimulation. We propose three hypotheses that could explain this paradoxical state of affairs. Furthermore, we provide suggestions for future research, which should lead to a better understanding of fibroblast quiescence and activation, and ultimately to new strategies for the prevention and treatment of cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Thomas C L Bracco Gartner
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jeroen M Stein
- Division of Heart and Lungs, Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Dimitri E P Muylaert
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Carlijn V C Bouten
- Division of Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Pieter A Doevendans
- Division of Heart and Lungs, Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands.,Central Military Hospital, Utrecht, the Netherlands
| | - Ali Khademhosseini
- Department of Bioengineering, Radiology, Chemical and Biomolecular Engineering, Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California, USA
| | - Willem J L Suyker
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Division of Heart and Lungs, Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands
| | - Jesper Hjortnaes
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands.,University Utrecht, Utrecht, the Netherlands
| |
Collapse
|
10
|
Lithium Reduces Migration and Collagen Synthesis Activity in Human Cardiac Fibroblasts by Inhibiting Store-Operated Ca 2+ Entry. Int J Mol Sci 2021; 22:ijms22020842. [PMID: 33467715 PMCID: PMC7830715 DOI: 10.3390/ijms22020842] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/13/2021] [Accepted: 01/13/2021] [Indexed: 01/08/2023] Open
Abstract
Cardiac fibrosis plays a vital role in the pathogenesis of heart failure. Fibroblast activity is enhanced by increases in store-operated Ca2+ entry (SOCE) and calcium release-activated calcium channel protein 1 (Orai1) levels. Lithium regulates SOCE; however, whether therapeutic concentrations of lithium can be used to inhibit cardiac fibrogenesis is unknown. Migration and proliferation assays, Western blotting, real-time reverse-transcription polymerase chain reaction analysis, and calcium fluorescence imaging were performed in human cardiac fibroblasts treated with or without LiCl at 1.0 mM (i.e., therapeutic peak level) or 0.1 mM (i.e., therapeutic trough level) for 24 h. Results showed that LiCl (0.1 mM, but not 1.0 mM) inhibited the migration and collagen synthesis ability of cardiac fibroblasts. Additionally, thapsigargin-induced SOCE was reduced in fibroblasts treated with LiCl (0.1 mM). The expression level of Orai1 was lower in LiCl (0.1 mM)-treated fibroblasts relative to the fibroblasts without LiCl treatment. Fibroblasts treated with a combination of LiCl (0.1 mM) and 2-APB (10 μM, an Orai1 inhibitor) demonstrated similar migration and collagen synthesis abilities as those in LiCl (0.1 mM)-treated fibroblasts. Altogether, lithium at therapeutic trough levels reduced the migration and collagen synthesis abilities of human cardiac fibroblasts by inhibiting SOCE and Orai1 expression.
Collapse
|
11
|
Li T, Weng X, Cheng S, Wang D, Cheng G, Gao H, Li Y. Wnt3a upregulation is involved in TGFβ1-induced cardiac hypertrophy. Cytokine 2020; 138:155376. [PMID: 33243628 DOI: 10.1016/j.cyto.2020.155376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/06/2020] [Accepted: 11/16/2020] [Indexed: 01/03/2023]
Abstract
Pathological cardiac hypertrophy, characterized by enlarged cell size and fetal gene reactivation, ultimately leads to cardiac dysfunction and heart failure. The expression of transforming growth factor beta 1 (TGFβ1) is often elevated in experimental models of cardiac hypertrophy. In the present study, we observed the activation of Wnt/β-catenin signaling in TGFβ1-induced cardiac hypertrophy. TGFβ1 stimulation decreased the phosphorylation levels of β-catenin and triggered the nuclear accumulation of β-catenin. In turn, TGFβ1 enhanced the expression of c-Myc, which is a transcriptional target of canonical Wnt/β-catenin pathway. Knockdown of β-catenin completely blocked TGFβ1-induced c-Myc upregulation. Wnt3a is an important Wnt ligand associated with cardiac fibrosis and hypertrophy. Further investigation revealed that TGFβ1 can upregulate Wnt3a expression in an ALK5-Smad2/3-dependent manner. A consensus Smad binding sequence is located within the Wnt3a promoter, and TGFβ1 stimulation enhanced recruitment of Smad2/3 onto the Wnt3a promoter. Meanwhile, Wnt3a overexpression also stimulated TGFβ1 expression. Chemical inhibition of Wnt/β-catenin signaling partially attenuated TGFβ1-induced hypertrophic responses. These findings suggest crosstalk between TGFβ1 and canonical Wnt/β-catenin pathways in cardiac hypertrophy.
Collapse
Affiliation(s)
- Tao Li
- School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xiaofei Weng
- School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China
| | - Siya Cheng
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng, Henan Province 475000, China
| | - Dongxing Wang
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng, Henan Province 475000, China
| | - Guanchang Cheng
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng, Henan Province 475000, China
| | - Hai Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Yanming Li
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng, Henan Province 475000, China.
| |
Collapse
|
12
|
Li Z, Zhu S, Liu Q, Wei J, Jin Y, Wang X, Zhang L. Polystyrene microplastics cause cardiac fibrosis by activating Wnt/β-catenin signaling pathway and promoting cardiomyocyte apoptosis in rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:115025. [PMID: 32806417 DOI: 10.1016/j.envpol.2020.115025] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 05/23/2023]
Abstract
Microplastics (MPs) are new persistent organic pollutants derived from the degradation of plastics. They can accumulate along the food chain and enter the human body through oral administration, inhalation and dermal exposure. To identify the impact of Polystyrene (PS) MPs on the cardiovascular system and the underlying toxicological mechanism, 32 male Wister rats were divided into control group and three model groups, which were exposed to 0.5 μm PS MPs at 0.5, 5 and 50 mg/L for 90 days. Our results suggested that PS MPs exposure increased Troponin I and creatine kinase-MB (CK-MB) levels in serum, resulted in structure damage and apoptosis of myocardium, and led to collagen proliferation of heart. Moreover, PS MPs could induce oxidative stress and thus activate fibrosis-related Wnt/β-catenin signaling pathway. These results suggested that PS MPs could lead to cardiovascular toxicity by inducing cardiac fibrosis via activating Wnt/β-catenin pathway and myocardium apoptosis triggered by oxidative stress. The present study provided some novelty evidence to elucidate the potential mechanism of cardiovascular toxicity induced by PS MPs.
Collapse
Affiliation(s)
- Zekang Li
- College of Clinical Medicine, Bin Zhou Medical University, Yan Tai, PR China
| | - Shuxiang Zhu
- College of Clinical Medicine, Bin Zhou Medical University, Yan Tai, PR China
| | - Qian Liu
- College of Clinical Medicine, Bin Zhou Medical University, Yan Tai, PR China
| | - Jialiu Wei
- Key Laboratory of Cardiovascular Epidemiology & Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinchuan Jin
- Department of Medical Psychology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, China
| | - Xifeng Wang
- Department of Critical Care Medicine, Yu Huang Ding Hospital, Qingdao University, Yantai, PR China
| | - Lianshuang Zhang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, PR China.
| |
Collapse
|
13
|
Soliman H, Rossi FMV. Cardiac fibroblast diversity in health and disease. Matrix Biol 2020; 91-92:75-91. [PMID: 32446910 DOI: 10.1016/j.matbio.2020.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
The cardiac stroma plays essential roles in health and following cardiac damage. The major player of the stroma with respect to extracellular matrix deposition, maintenance and remodeling is the poorly defined fibroblast. It has long been recognized that there is considerable variability to the fibroblast phenotype. With the advent of new, high throughput analytical methods our understanding and appreciation of this heterogeneity has grown dramatically. This review aims to explore the diversity of cardiac fibroblasts and highlights new insights into the diverse nature of these cells and their progenitors as revealed by single cell sequencing and fate mapping studies. We propose that at least in part the observed heterogeneity is related to the existence of a differentiation cascade within stromal cells. Beyond in-organ heterogeneity, we also discuss how the stromal response to damage differs between non-regenerating organs such as the heart and regenerating organs such as skeletal muscle. In exploring possible causes for these differences, we outline that although fibrogenic cells from different organs overlap in many properties, they still possess organ-specific transcriptional signatures and differentiation biases that make them functionally distinct.
Collapse
Affiliation(s)
- Hesham Soliman
- Biomedical Research Centre, University of British Columbia, Vancouver, Canada; School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada; Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Fabio M V Rossi
- Biomedical Research Centre, University of British Columbia, Vancouver, Canada; School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada.
| |
Collapse
|
14
|
Abstract
Cardiovascular disease (CVD) is still a factor of mortality in the whole world. Through canonical and noncanonical pathways and with different receptors, the Wnt/β-catenin signaling pathway plays an essential role in response to heart injuries. Wnt regulates the mobilization and proliferation of cells in endothelium and epicardium in an infarcted heart. Therefore, with its profibrotic effects as well as its antagonism with other proteins, Wnt/β-catenin signaling pathway leads to beneficial effects on fibrosis and cardiac remodeling in myocardium. In addition, Wnt increases the proliferation and differentiation of cardiac progenitors in an ischemic heart. Complex interactions and dual activity of Wnt, the changes in its expression, and mutations that can change its activity during heart development have an adverse effect on cardiac myocardium after injury. However, targeting the Wnt in myocardium with cellular and molecular pathways can be suggested to improve and repair ischemic heart. Given these challenges, in this review article, we deal with the role of Wnt/β-catenin signaling pathway as well as its interactions with other cells and molecules in an ischemic myocardium.
Collapse
|
15
|
Padwal M, Liu L, Margetts PJ. The role of WNT5A and Ror2 in peritoneal membrane injury. J Cell Mol Med 2020; 24:3481-3491. [PMID: 32052562 PMCID: PMC7131918 DOI: 10.1111/jcmm.15034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/20/2019] [Accepted: 01/06/2020] [Indexed: 01/07/2023] Open
Abstract
Patients on peritoneal dialysis are at risk of developing peritoneal fibrosis and angiogenesis, which can lead to dysfunction of the peritoneal membrane. Recent evidence has identified cross‐talk between transforming growth factor beta (TGFB) and the WNT/β‐catenin pathway to induce fibrosis and angiogenesis. Limited evidence exists describing the role of non‐canonical WNT signalling in peritoneal membrane injury. Non‐canonical WNT5A is suggested to have different effects depending on the receptor environment. WNT5A has been implicated in antagonizing canonical WNT/β‐catenin signalling in the presence of receptor tyrosine kinase‐like orphan receptor (Ror2). We co‐expressed TGFB and WNT5A using adenovirus and examined its role in the development of peritoneal fibrosis and angiogenesis. Treatment of mouse peritoneum with AdWNT5A decreased the submesothelial thickening and angiogenesis induced by AdTGFB. WNT5A appeared to block WNT/β‐catenin signalling by inhibiting phosphorylation of glycogen synthase kinase 3 beta (GSK3B) and reducing levels of total β‐catenin and target proteins. To examine the function of Ror2, we silenced Ror2 in a human mesothelial cell line. We treated cells with AdWNT5A and observed a significant increase in fibronectin compared with AdWNT5A alone. We also analysed fibronectin and vascular endothelial growth factor (VEGF) in a TGFB model of mesothelial cell injury. Both fibronectin and VEGF were significantly increased in response to Ror2 silencing when cells were exposed to TGFB. Our results suggest that WNT5A inhibits peritoneal injury and this is associated with a decrease in WNT/β‐catenin signalling. In human mesothelial cells, Ror2 is involved in regulating levels of fibronectin and VEGF.
Collapse
Affiliation(s)
- Manreet Padwal
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Limin Liu
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Peter J Margetts
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
16
|
Meyer IS, Leuschner F. The role of Wnt signaling in the healing myocardium: a focus on cell specificity. Basic Res Cardiol 2018; 113:44. [PMID: 30327885 DOI: 10.1007/s00395-018-0705-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/01/2018] [Accepted: 10/09/2018] [Indexed: 12/18/2022]
Abstract
Various cell types are involved in the healing process after myocardial infarction (MI). Besides cardiac resident cells (such as cardiomyocytes, fibroblasts and endothelial cells) already present at the lesion site, a massive influx of leukocytes (mainly monocytes and neutrophils) is observed within hours after the ischemic event. So far, little is known about modes of interaction of these cells. Wnt signaling is an evolutionary conserved signaling cassette known to play an important role in cell-cell communication. While the overall reactivation of Wnt signaling upon ischemic injury is well described, the precise expression pattern of Wnt proteins, however, is far from understood. We here describe known Wnt components that partake in MI healing and differentiate cell-specific aspects. The secretion of Wnt proteins and their antagonists in the context of cardiac inflammation after MI appear to be tightly regulated in a spatial-temporal manner. Overall, we aim to stress the importance of elucidating not only Wnt component-specific aspects, but also their sometimes contradicting effects in different target cells. A better understanding of Wnt signaling in MI healing may eventually lead to the development of successful therapeutic approaches in an often considered "un-druggable" pathway.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Florian Leuschner
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
17
|
Orgeur M, Martens M, Leonte G, Nassari S, Bonnin MA, Börno ST, Timmermann B, Hecht J, Duprez D, Stricker S. Genome-wide strategies identify downstream target genes of chick connective tissue-associated transcription factors. Development 2018; 145:dev.161208. [PMID: 29511024 DOI: 10.1242/dev.161208] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/24/2018] [Indexed: 12/18/2022]
Abstract
Connective tissues support organs and play crucial roles in development, homeostasis and fibrosis, yet our understanding of their formation is still limited. To gain insight into the molecular mechanisms of connective tissue specification, we selected five zinc-finger transcription factors - OSR1, OSR2, EGR1, KLF2 and KLF4 - based on their expression patterns and/or known involvement in connective tissue subtype differentiation. RNA-seq and ChIP-seq profiling of chick limb micromass cultures revealed a set of common genes regulated by all five transcription factors, which we describe as a connective tissue core expression set. This common core was enriched with genes associated with axon guidance and myofibroblast signature, including fibrosis-related genes. In addition, each transcription factor regulated a specific set of signalling molecules and extracellular matrix components. This suggests a concept whereby local molecular niches can be created by the expression of specific transcription factors impinging on the specification of local microenvironments. The regulatory network established here identifies common and distinct molecular signatures of limb connective tissue subtypes, provides novel insight into the signalling pathways governing connective tissue specification, and serves as a resource for connective tissue development.
Collapse
Affiliation(s)
- Mickael Orgeur
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Thielallee 63, 14195 Berlin, Germany.,Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany.,Sorbonne Universités, UPMC Univ. Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, 9 Quai Saint-Bernard, 75005 Paris, France
| | - Marvin Martens
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, 9 Quai Saint-Bernard, 75005 Paris, France
| | - Georgeta Leonte
- Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany.,Freie Universität Berlin, Institute of Biology, Königin-Luise-Str. 1-3, 14195 Berlin, Germany
| | - Sonya Nassari
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, 9 Quai Saint-Bernard, 75005 Paris, France
| | - Marie-Ange Bonnin
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, 9 Quai Saint-Bernard, 75005 Paris, France
| | - Stefan T Börno
- Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Bernd Timmermann
- Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Jochen Hecht
- Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitatsmedizin, Augustenburger Platz 1, 13353 Berlin, Germany.,Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.,Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Delphine Duprez
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, 9 Quai Saint-Bernard, 75005 Paris, France
| | - Sigmar Stricker
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Thielallee 63, 14195 Berlin, Germany .,Max Planck Institute for Molecular Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| |
Collapse
|
18
|
Uitterdijk A, Springeling T, Hermans KCM, Merkus D, de Beer VJ, Gorsse-Bakker C, Mokelke E, Daskalopoulos EP, Wielopolski PA, Cleutjens JPM, Blankesteijn WM, Prinzen FW, van der Giessen WJ, van Geuns RJM, Duncker DJ. Intermittent pacing therapy favorably modulates infarct remodeling. Basic Res Cardiol 2017; 112:28. [PMID: 28386775 PMCID: PMC5383690 DOI: 10.1007/s00395-017-0616-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
Despite early revascularization, remodeling and dysfunction of the left ventricle (LV) after acute myocardial infarction (AMI) remain important therapeutic targets. Intermittent pacing therapy (IPT) of the LV can limit infarct size, when applied during early reperfusion. However, the effects of IPT on post-AMI LV remodeling and infarct healing are unknown. We therefore investigated the effects of IPT on global LV remodeling and infarct geometry in swine with a 3-day old AMI. For this purpose, fifteen pigs underwent 2 h ligation of the left circumflex coronary artery followed by reperfusion. An epicardial pacing lead was implanted in the peri-infarct zone. After three days, global LV remodeling and infarct geometry were assessed using magnetic resonance imaging (MRI). Animals were stratified into MI control and IPT groups. Thirty-five days post-AMI, follow-up MRI was obtained and myofibroblast content, markers of extracellular matrix (ECM) turnover and Wnt/frizzled signaling in infarct and non-infarct control tissue were studied. Results showed that IPT had no significant effect on global LV remodeling, function or infarct mass, but modulated infarct healing. In MI control pigs, infarct mass reduction was principally due to a 26.2 ± 4.4% reduction in infarct thickness (P ≤ 0.05), whereas in IPT pigs it was mainly due to a 35.7 ± 4.5% decrease in the number of infarct segments (P ≤ 0.05), with no significant change in infarct thickness. Myofibroblast content of the infarct zone was higher in IPT (10.9 ± 2.1%) compared to MI control (5.4 ± 1.6%; P ≤ 0.05). Higher myofibroblast presence did not coincide with alterations in expression of genes involved in ECM turnover or Wnt/frizzled signaling at 5 weeks follow-up. Taken together, IPT limited infarct expansion and altered infarct composition, showing that IPT influences remodeling of the infarct zone, likely by increasing regional myofibroblast content.
Collapse
Affiliation(s)
- André Uitterdijk
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Tirza Springeling
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| | - Kevin C M Hermans
- Department of Pharmacology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Daphne Merkus
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Vincent J de Beer
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Charlotte Gorsse-Bakker
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Eric Mokelke
- Boston Scientific Corporation, St. Paul, MN, USA.,Medical Products Division, W.L. Gore and Associates, Flagstaff, AZ, USA
| | | | | | - Jack P M Cleutjens
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | | | - Frits W Prinzen
- Department of Physiology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Willem J van der Giessen
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Robert-Jan M van Geuns
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Radiology, Erasmus MC, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Ee-2351, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
Palevski D, Levin-Kotler LP, Kain D, Naftali-Shani N, Landa N, Ben-Mordechai T, Konfino T, Holbova R, Molotski N, Rosin-Arbesfeld R, Lang RA, Leor J. Loss of Macrophage Wnt Secretion Improves Remodeling and Function After Myocardial Infarction in Mice. J Am Heart Assoc 2017; 6:JAHA.116.004387. [PMID: 28062479 PMCID: PMC5523630 DOI: 10.1161/jaha.116.004387] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background Macrophages and Wnt proteins (Wnts) are independently involved in cardiac development, response to cardiac injury, and repair. However, the role of macrophage‐derived Wnts in the healing and repair of myocardial infarction (MI) is unknown. We sought to determine the role of macrophage Wnts in infarct repair. Methods and Results We show that the Wnt pathway is activated after MI in mice. Furthermore, we demonstrate that isolated infarct macrophages express distinct Wnt pathway components and are a source of noncanonical Wnts after MI. To determine the effect of macrophage Wnts on cardiac repair, we evaluated mice lacking the essential Wnt transporter Wntless (Wls) in myeloid cells. Significantly, Wntless‐deficient macrophages presented a unique subset of M2‐like macrophages with anti‐inflammatory, reparative, and angiogenic properties. Serial echocardiography studies revealed that mice lacking macrophage Wnt secretion showed improved function and less remodeling 30 days after MI. Finally, mice lacking macrophage‐Wntless had increased vascularization near the infarct site compared with controls. Conclusions Macrophage‐derived Wnts are implicated in adverse cardiac remodeling and dysfunction after MI. Together, macrophage Wnts could be a new therapeutic target to improve infarct healing and repair.
Collapse
Affiliation(s)
- Dahlia Palevski
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - La-Paz Levin-Kotler
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - David Kain
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - Nili Naftali-Shani
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - Natalie Landa
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - Tammy Ben-Mordechai
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - Tal Konfino
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - Radka Holbova
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - Natali Molotski
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| | - Rina Rosin-Arbesfeld
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Richard A Lang
- The Visual Systems Group Divisions of Pediatric Ophthalmology and Developmental Biology, Cincinnati Children's Hospital Medical Center Research Foundation, Cincinnati, OH
| | - Jonathan Leor
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel .,Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sheba Center for Regenerative Medicine, Stem Cells and Tissue Engineering, Tel-Hashomer, Israel
| |
Collapse
|
20
|
Sun LY, Bie ZD, Zhang CH, Li H, Li LD, Yang J. MiR-154 directly suppresses DKK2 to activate Wnt signaling pathway and enhance activation of cardiac fibroblasts. Cell Biol Int 2016; 40:1271-1279. [PMID: 27542661 DOI: 10.1002/cbin.10655] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/14/2016] [Indexed: 12/26/2022]
Abstract
Excessive proliferation of cardiac fibroblasts (CFs) and their transdifferentiation into myofibroblasts leads to expression of α-smooth muscle actin (α-SMA), as well as excessive synthesis and secretion of collagens. This process represents an important pathological basis for myocardial fibrosis (MF). MicroRNA (miR)-154 and the Wnt signaling pathway play key roles in the above process, although their specific interactions are poorly understood. After transfecting CFs with miR-154 mimics or inhibitors, miR-154 was found to inhibit the expression of Dickkopf-related protein 2 (DKK2), while miR-154 inhibitors upregulated DKK2 expression in a Western blot analysis. In a subsequent dual-luciferase activity assay, direct binding of miR-154 to DKK2 was detected. Further experiments demonstrated that transfection of DKK2 siRNA or miR-154 resulted in increased levels of β-catenin, α-SMA, and collagens I and III. Moreover, these changes were observed in association with increases in CF proliferation and migration, and reduced apoptosis. Conversely, transfection of miR-154 inhibitors or DKK2 overexpression vector resulted in lower expression levels of β-catenin, α-SMA, and collagens I and III, suppressed cell proliferation and migration, and enhanced apoptosis. Furthermore, in each assay, when the DKK2 overexpression vector and miR-154 mimics were co-transfected, the functions of each component were counteracted by the other. Therefore, in CFs, targeting of DKK2 by miR-154 leads to upregulation of β-catenin expression and activation of the classical Wnt signaling pathway and CFs. These results suggest new targets for the clinical treatment of MF and ischemic heart disease.
Collapse
Affiliation(s)
- Li-Ye Sun
- Shandong University School of Medicine, Jinan, 250012, Shandong, China.,Department of Geratology, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Zi-Dong Bie
- Shandong University School of Medicine, Jinan, 250012, Shandong, China.,Department of Cardiology, Weihai Central Hospital, Weihai, 264423, Shandong, China
| | - Chuan-Huan Zhang
- Department of Cardiology, Yantai Yuhuangding Hospital, 20# Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Hua Li
- Department of Geratology, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Liu-Dong Li
- Department of Cardiology, Yantai Yuhuangding Hospital, 20# Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, 20# Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| |
Collapse
|
21
|
Awada HK, Hwang MP, Wang Y. Towards comprehensive cardiac repair and regeneration after myocardial infarction: Aspects to consider and proteins to deliver. Biomaterials 2016; 82:94-112. [PMID: 26757257 PMCID: PMC4872516 DOI: 10.1016/j.biomaterials.2015.12.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/15/2015] [Accepted: 12/19/2015] [Indexed: 12/13/2022]
Abstract
Ischemic heart disease is a leading cause of death worldwide. After the onset of myocardial infarction, many pathological changes take place and progress the disease towards heart failure. Pathologies such as ischemia, inflammation, cardiomyocyte death, ventricular remodeling and dilation, and interstitial fibrosis, develop and involve the signaling of many proteins. Proteins can play important roles in limiting or countering pathological changes after infarction. However, they typically have short half-lives in vivo in their free form and can benefit from the advantages offered by controlled release systems to overcome their challenges. The controlled delivery of an optimal combination of proteins per their physiologic spatiotemporal cues to the infarcted myocardium holds great potential to repair and regenerate the heart. The effectiveness of therapeutic interventions depends on the elucidation of the molecular mechanisms of the cargo proteins and the spatiotemporal control of their release. It is likely that multiple proteins will provide a more comprehensive and functional recovery of the heart in a controlled release strategy.
Collapse
Affiliation(s)
- Hassan K Awada
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Mintai P Hwang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Yadong Wang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
22
|
Lighthouse JK, Small EM. Transcriptional control of cardiac fibroblast plasticity. J Mol Cell Cardiol 2016; 91:52-60. [PMID: 26721596 PMCID: PMC4764462 DOI: 10.1016/j.yjmcc.2015.12.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/15/2015] [Accepted: 12/20/2015] [Indexed: 12/11/2022]
Abstract
Cardiac fibroblasts help maintain the normal architecture of the healthy heart and are responsible for scar formation and the healing response to pathological insults. Various genetic, biomechanical, or humoral factors stimulate fibroblasts to become contractile smooth muscle-like cells called myofibroblasts that secrete large amounts of extracellular matrix. Unfortunately, unchecked myofibroblast activation in heart disease leads to pathological fibrosis, which is a major risk factor for the development of cardiac arrhythmias and heart failure. A better understanding of the molecular mechanisms that control fibroblast plasticity and myofibroblast activation is essential to develop novel strategies to specifically target pathological cardiac fibrosis without disrupting the adaptive healing response. This review highlights the major transcriptional mediators of fibroblast origin and function in development and disease. The contribution of the fetal epicardial gene program will be discussed in the context of fibroblast origin in development and following injury, primarily focusing on Tcf21 and C/EBP. We will also highlight the major transcriptional regulatory axes that control fibroblast plasticity in the adult heart, including transforming growth factor β (TGFβ)/Smad signaling, the Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF) axis, and Calcineurin/transient receptor potential channel (TRP)/nuclear factor of activated T-Cell (NFAT) signaling. Finally, we will discuss recent strategies to divert the fibroblast transcriptional program in an effort to promote cardiomyocyte regeneration. This article is a part of a Special Issue entitled "Fibrosis and Myocardial Remodeling".
Collapse
Affiliation(s)
- Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Eric M Small
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA.
| |
Collapse
|
23
|
Pahnke A, Conant G, Huyer LD, Zhao Y, Feric N, Radisic M. The role of Wnt regulation in heart development, cardiac repair and disease: A tissue engineering perspective. Biochem Biophys Res Commun 2015; 473:698-703. [PMID: 26626076 DOI: 10.1016/j.bbrc.2015.11.060] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/14/2015] [Indexed: 01/08/2023]
Abstract
Wingless-related integration site (Wnt) signaling has proven to be a fundamental mechanism in cardiovascular development as well as disease. Understanding its particular role in heart formation has helped to develop pluripotent stem cell differentiation protocols that produce relatively pure cardiomyocyte populations. The resultant cardiomyocytes have been used to generate heart tissue for pharmaceutical testing, and to study physiological and disease states. Such protocols in combination with induced pluripotent stem cell technology have yielded patient-derived cardiomyocytes that exhibit some of the hallmarks of cardiovascular disease and are therefore being used to model disease states. While FDA approval of new treatments typically requires animal experiments, the burgeoning field of tissue engineering could act as a replacement. This would necessitate the generation of reproducible three-dimensional cardiac tissues in a well-controlled environment, which exhibit native heart properties, such as cellular density, composition, extracellular matrix composition, and structure-function. Such tissues could also enable the further study of Wnt signaling. Furthermore, as Wnt signaling has been found to have a mechanistic role in cardiac pathophysiology, e.g. heart attack, hypertrophy, atherosclerosis, and aortic stenosis, its strategic manipulation could provide a means of generating reproducible and specific, physiological and pathological cardiac models.
Collapse
Affiliation(s)
- Aric Pahnke
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Genna Conant
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Locke Davenport Huyer
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Nicole Feric
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Abstract
The ability to repair damaged or lost tissues varies significantly among vertebrates. The regenerative ability of the heart is clinically very relevant, because adult teleost fish and amphibians can regenerate heart tissue, but we mammals cannot. Interestingly, heart regeneration is possible in neonatal mice, but this ability is lost within 7 days after birth. In zebrafish and neonatal mice, lost cardiomyocytes are regenerated via proliferation of spared, differentiated cardiomyocytes. While some cardiomyocyte turnover occurs in adult mammals, the cardiomyocyte production rate is too low in response to injury to regenerate the heart. Instead, mammalian hearts respond to injury by remodeling of spared tissue, which includes cardiomyocyte hypertrophy. Wnt/β-catenin signaling plays important roles during vertebrate heart development, and it is re-activated in response to cardiac injury. In this review, we discuss the known functions of this signaling pathway in injured hearts, its involvement in cardiac fibrosis and hypertrophy, and potential therapeutic approaches that might promote cardiac repair after injury by modifying Wnt/β-catenin signaling. Regulation of cardiac remodeling by this signaling pathway appears to vary depending on the injury model and the exact stages that have been studied. Thus, conflicting data have been published regarding a potential role of Wnt/β-catenin pathway in promotion of fibrosis and cardiomyocyte hypertrophy. In addition, the Wnt inhibitory secreted Frizzled-related proteins (sFrps) appear to have Wnt-dependent and Wnt-independent roles in the injured heart. Thus, while the exact functions of Wnt/β-catenin pathway activity in response to injury still need to be elucidated in the non-regenerating mammalian heart, but also in regenerating lower vertebrates, manipulation of the pathway is essential for creation of therapeutically useful cardiomyocytes from stem cells in culture. Hopefully, a detailed understanding of the in vivo role of Wnt/β-catenin signaling in injured mammalian and non-mammalian hearts will also contribute to the success of current efforts towards developing regenerative therapies.
Collapse
Affiliation(s)
- Gunes Ozhan
- Izmir Biomedicine and Genome Center (iBG-izmir), Dokuz Eylul University, Inciralti-Balcova, 35340 Izmir, Turkey ; Department of Medical Biology and Genetics, Dokuz Eylul University Medical School, Inciralti-Balcova, 35340 Izmir, Turkey
| | - Gilbert Weidinger
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
25
|
Roche PL, Filomeno KL, Bagchi RA, Czubryt MP. Intracellular Signaling of Cardiac Fibroblasts. Compr Physiol 2015; 5:721-60. [DOI: 10.1002/cphy.c140044] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
26
|
Lee K, Yu P, Lingampalli N, Kim HJ, Tang R, Murthy N. Peptide-enhanced mRNA transfection in cultured mouse cardiac fibroblasts and direct reprogramming towards cardiomyocyte-like cells. Int J Nanomedicine 2015; 10:1841-54. [PMID: 25834424 PMCID: PMC4358644 DOI: 10.2147/ijn.s75124] [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] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The treatment of myocardial infarction is a major challenge in medicine due to the inability of heart tissue to regenerate. Direct reprogramming of endogenous cardiac fibroblasts into functional cardiomyocytes via the delivery of transcription factor mRNAs has the potential to regenerate cardiac tissue and to treat heart failure. Even though mRNA delivery to cardiac fibroblasts has the therapeutic potential, mRNA transfection in cardiac fibroblasts has been challenging. Herein, we develop an efficient mRNA transfection in cultured mouse cardiac fibroblasts via a polyarginine-fused heart-targeting peptide and lipofectamine complex, termed C-Lipo and demonstrate the partial direct reprogramming of cardiac fibroblasts towards cardiomyocyte cells. C-Lipo enabled the mRNA-induced direct cardiac reprogramming due to its efficient transfection with low toxicity, which allowed for multiple transfections of Gata4, Mef2c, and Tbx5 (GMT) mRNAs for a period of 2 weeks. The induced cardiomyocyte-like cells had α-MHC promoter-driven GFP expression and striated cardiac muscle structure from α-actinin immunohistochemistry. GMT mRNA transfection of cultured mouse cardiac fibroblasts via C-Lipo significantly increased expression of the cardiomyocyte marker genes, Actc1, Actn2, Gja1, Hand2, and Tnnt2, after 2 weeks of transfection. Moreover, this study provides the first direct evidence that the stoichiometry of the GMT reprogramming factors influence the expression of cardiomyocyte marker genes. Our results demonstrate that mRNA delivery is a potential approach for cardiomyocyte generation.
Collapse
Affiliation(s)
- Kunwoo Lee
- Department of Bioengineering, University of California, Berkeley, CA, USA ; UC Berkeley and UCSF Joint Graduate Program in Bioengineering, Berkeley/San Francisco, CA, USA
| | - Pengzhi Yu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
| | - Nithya Lingampalli
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | - Hyun Jin Kim
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | - Richard Tang
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | - Niren Murthy
- Department of Bioengineering, University of California, Berkeley, CA, USA ; UC Berkeley and UCSF Joint Graduate Program in Bioengineering, Berkeley/San Francisco, CA, USA
| |
Collapse
|
27
|
Primary cilium-associated genes mediate bone marrow stromal cell response to hypoxia. Stem Cell Res 2014; 13:284-99. [PMID: 25171775 DOI: 10.1016/j.scr.2014.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 12/26/2022] Open
Abstract
Currently there is intense interest in using mesenchymal stem cells (MSC) for therapeutic interventions in many diseases and conditions. To accelerate the therapeutic use of stem cells we must understand how they sense their environment. Primary cilia are an extracellular sensory organelle present on most growth arrested cells that transduce information about the cellular environment into cells, triggering signaling cascades that have profound effects on development, cell cycle, proliferation, differentiation and migration. Migrating cells likely encounter differing oxygen tensions, therefore we investigated the effect of oxygen tension on cilia. Using bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) we found that oxygen tension significantly affected the length of cilia in primary BMSCs. Chronic exposure to hypoxia specifically down-regulated genes involved in hedgehog signaling and re-localized the Smo and Gli2 proteins to cilia. Investigating the effects of chemotactic migration on cilia, we observed significantly longer cilia in migrating cells which was again, strongly influenced by oxygen tension. Finally, using computational modeling we identified links between migration and ciliation signaling pathways, characterizing the novel role of HSP90 and PI3K signaling in regulating BMSC cilia length. These findings enhance our current understanding of BMSC adaptions to hypoxia and advance our knowledge of BMSC biology and cilia regulation.
Collapse
|
28
|
de Oliveira GP, Maximino JR, Maschietto M, Zanoteli E, Puga RD, Lima L, Carraro DM, Chadi G. Early gene expression changes in skeletal muscle from SOD1(G93A) amyotrophic lateral sclerosis animal model. Cell Mol Neurobiol 2014; 34:451-62. [PMID: 24442855 DOI: 10.1007/s10571-014-0029-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/07/2014] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by loss of motor neurons. Familial ALS is strongly associated to dominant mutations in the gene for Cu/Zn superoxide dismutase (SOD1). Recent evidences point to skeletal muscle as a primary target in the ALS mouse model. Wnt/PI3 K signaling pathways and epithelial-mesenchymal transition (EMT) have important roles in maintenance and repair of skeletal muscle. Wnt/PI3 K pathways and EMT gene expression profile were investigated in gastrocnemius muscle from SOD1(G93A) mouse model and age-paired wild-type control in the presymptomatic ages of 40 and 80 days aiming the early neuromuscular abnormalities that precede motor neuron death in ALS. A customized cDNA microarray platform containing 326 genes of Wnt/PI3 K and EMT was used and results revealed eight up-regulated (Loxl2, Pik4ca, Fzd9, Cul1, Ctnnd1, Snf1lk, Prkx, Dner) and nine down-regulated (Pik3c2a, Ripk4, Id2, C1qdc1, Eif2ak2, Rac3, Cds1, Inppl1, Tbl1x) genes at 40 days, and also one up-regulated (Pik3ca) and five down-regulated (Cd44, Eef2 k, Fzd2, Crebbp, Piki3r1) genes at 80 days. Also, protein-protein interaction networks grown from the differentially expressed genes of 40 and 80 days old mice have identified Grb2 and Src genes in both presymptomatic ages, thus playing a potential central role in the disease mechanisms. mRNA and protein levels for Grb2 and Src were found to be increased in 80 days old ALS mice. Gene expression changes in the skeletal muscle of transgenic ALS mice at presymptomatic periods of disease gave further evidence of early neuromuscular abnormalities that precede motor neuron death. The results were discussed in terms of initial triggering for neuronal degeneration and muscle adaptation to keep function before the onset of symptoms.
Collapse
Affiliation(s)
- Gabriela P de Oliveira
- Neuroregeneration Center, Department of Neurology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 2nd Floor, Room 2119, São Paulo, 01246-903, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Habu M, Koyama H, Kishida M, Kamino M, Iijima M, Fuchigami T, Tokimura H, Ueda M, Tokudome M, Koriyama C, Hirano H, Arita K, Kishida S. Ryk is essential for Wnt-5a-dependent invasiveness in human glioma. J Biochem 2014; 156:29-38. [PMID: 24621529 DOI: 10.1093/jb/mvu015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma is characterized by marked invasiveness, but little is known about the mechanism of invasion in glioblastoma cells. Wnts are secreted ligands that regulate cell proliferation, differentiation, motility and fate at various developmental stages. In adults, misregulation of the Wnt pathway is associated with several diseases. Recently, we reported that Wnt-5a was overexpressed and correlated with cell motility and infiltrative activity through the regulation of matrix metalloproteinase (MMP)-2 in glioma-derived cells. Although several receptors for Wnt-5a were identified, the receptors of Wnt-5a that mediate cellular responses of glioma were not clearly identified. Knockdown of receptor-like tyrosine kinase (Ryk) but not that of Ror2 suppressed the activity of MMP-2 and Wnt-5a-dependent invasive activity in glioma cells. These results suggest that Ryk is important for the Wnt-5a-dependent induction of MMP-2 and invasive activity in glioma-derived cells and that Ryk might have a novel patho-physiological function in adult cancer invasion. Furthermore, not only the expression of Wnt-5a but also that of Frizzled (Fz)-2 and Ryk was correlated with the WHO histological grade in 38 human glioma tissues. Taking these findings together, Fz-2 and Ryk could be therapeutic or pharmacological target molecules for the control of Wnt-5a-dependent invasion of human glioma in the near future.
Collapse
Affiliation(s)
- Mika Habu
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, JapanDepartment of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hirofumi Koyama
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Michiko Kishida
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masayuki Kamino
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, JapanDepartment of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mikio Iijima
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takao Fuchigami
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, JapanDepartment of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiroshi Tokimura
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masahiro Ueda
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, JapanDepartment of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mai Tokudome
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Chihaya Koriyama
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hirofumi Hirano
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kazunori Arita
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shosei Kishida
- Department of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, JapanDepartment of Biochemistry and Genetics; Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences; Department of Pharmacy, Kagoshima Prefectural Satunan Hospital; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences; Natural Science Centre for Research and Education, Kagoshima University; and Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
30
|
Fujiu K, Nagai R. Fibroblast-mediated pathways in cardiac hypertrophy. J Mol Cell Cardiol 2014; 70:64-73. [PMID: 24492068 DOI: 10.1016/j.yjmcc.2014.01.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 01/20/2014] [Accepted: 01/23/2014] [Indexed: 12/26/2022]
Abstract
Under normal physiological conditions, cardiac fibroblasts are the primary producers of extracellular matrix and supply a mechanical scaffold for efficacious heart contractions induced by cardiomyocytes. In the hypertrophic heart, cardiac fibroblasts provide a pivotal contribution to cardiac remodeling. Many growth factors and extracellular matrix components secreted by cardiac fibroblasts induce and modify cardiomyocyte hypertrophy. Recent evidence revealed that cardiomyocyte-cardiac fibroblast communications are complex and multifactorial. Many growth factors and molecules contribute to cardiac hypertrophy via different roles that include induction of hypertrophy and the feedback hypertrophic response, fine-tuning of adaptive hypertrophy, limitation of left ventricular dilation, and modification of interstitial changes. This review focuses on recent work and topics and provides a mechanistic insight into cardiomyocyte-cardiac fibroblast communication in cardiac hypertrophy. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium ".
Collapse
Affiliation(s)
- Katsuhito Fujiu
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan; Translational Systems Biology and Medicine Initiative (TSBMI), The University of Tokyo, Tokyo, Japan.
| | - Ryozo Nagai
- Funding Program for World-Leading Innovative R&D on Science and Technology (FIRST Program), Tokyo, Japan; Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
31
|
Singh K, Agrawal NK, Gupta SK, Singh K. Association of variant rs7903146 (C/T) single nucleotide polymorphism of TCF7L2 gene with impairment in wound healing among north Indian type 2 diabetes population: a case-control study. INT J LOW EXTR WOUND 2013; 12:310-5. [PMID: 24214952 DOI: 10.1177/1534734613504435] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The variants of transcription factor 7-like 2 (TCF7L2) gene have been shown to be associated with type 2 diabetes mellitus (T2DM) and its several secondary complications. Here, we aimed to examine the possible role of one of the common variant of this gene, rs7903146 (C/T), with impairment of wound healing in cases with T2DM. A total of 750 individuals, including 322 patients with T2DM and 120 patients with diabetic foot ulcers (DFUs) and 308 controls, were analyzed for rs7903146 variant of the TCF7L2 gene. Genotyping was done by polymerase chain reaction-restriction fragment length polymorphism. For rs7903146 variant, TT genotype frequency in patients with DFU was 10.8% and in controls was 5.2%. Risk genotype (TT) frequencies showed statistically significant difference between the DFU patients versus non-DM control group (odds ratio = 2.44, P = .037, 95% confidence interval = 1.05-5.64) compared with nonrisk genotype (CC + CT). In the present study, a positive significant association between DFU and the TT genotype of rs7903146 (C/T) variant of TCF7L2 gene was found.
Collapse
Affiliation(s)
- Kanhaiya Singh
- 1Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi, India
| | | | | | | |
Collapse
|
32
|
Immunoreactivity of Wnt5a, Fzd2, Fzd6, and Ryk in glioblastoma: evaluative methodology for DAB chromogenic immunostaining. Brain Tumor Pathol 2013; 31:85-93. [PMID: 23748645 DOI: 10.1007/s10014-013-0153-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 05/19/2013] [Indexed: 10/26/2022]
Abstract
The aim of this study was to determine the influence of Wnt5a and its receptors on the survival of glioblastoma patients and to determine reliable evaluation methods for immunohistochemistry. Diagnostic specimens from 41 histopathologically confirmed primary glioblastoma patients whose Gd-enhanced tumors had been totally removed were immunohistochemically stained for Wnt5a, Fzd2, Fzd6, and Ryk. The immunoreactivity was evaluated using the following methods: (A) grayscale optical density after color deconvolution, (B) percentage of stained cells, (C) density of stained cells, (D) staining amount (multiplication product of B and C), and (E) staining rank. The data sets of A to E were statistically evaluated by correlation matrix analysis and regression analysis. The influence of the expression of the markers on survival was analyzed using a proportional hazard model. The results of color deconvolution (A) were well correlated with the results of the staining rank (E). In the semiquantitative results (B, C, and D), the staining amount (D) tended to show a better correlation with results of color deconvolution (A). Among all data sets, color deconvolution (A) demonstrated the most preferable fit in a proportional hazard model, and the expression of Fzd2 and Fzd6 was associated with poor prognosis in glioblastoma patients.
Collapse
|
33
|
Turner NA, Porter KE. Function and fate of myofibroblasts after myocardial infarction. FIBROGENESIS & TISSUE REPAIR 2013; 6:5. [PMID: 23448358 PMCID: PMC3599637 DOI: 10.1186/1755-1536-6-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/10/2013] [Indexed: 01/27/2023]
Abstract
The importance of cardiac fibroblasts in the regulation of myocardial remodelling following myocardial infarction (MI) is becoming increasingly recognised. Studies over the last few decades have reinforced the concept that cardiac fibroblasts are much more than simple homeostatic regulators of extracellular matrix turnover, but are integrally involved in all aspects of the repair and remodelling of the heart that occurs following MI. The plasticity of fibroblasts is due in part to their ability to undergo differentiation into myofibroblasts. Myofibroblasts are specialised cells that possess a more contractile and synthetic phenotype than fibroblasts, enabling them to effectively repair and remodel the cardiac interstitium to manage the local devastation caused by MI. However, in addition to their key role in cardiac restoration and healing, persistence of myofibroblast activation can drive pathological fibrosis, resulting in arrhythmias, myocardial stiffness and progression to heart failure. The aim of this review is to give an appreciation of both the beneficial and detrimental roles of the myofibroblast in the remodelling heart, to describe some of the major regulatory mechanisms controlling myofibroblast differentiation including recent advances in the microRNA field, and to consider how this cell type could be exploited therapeutically.
Collapse
Affiliation(s)
- Neil A Turner
- Division of Cardiovascular and Diabetes Research, and Multidisciplinary Cardiovascular Research Centre, School of Medicine, University of Leeds, Leeds LS2 9JT, UK.
| | | |
Collapse
|
34
|
Ruiz-Villalba A, Ziogas A, Ehrbar M, Pérez-Pomares JM. Characterization of epicardial-derived cardiac interstitial cells: differentiation and mobilization of heart fibroblast progenitors. PLoS One 2013; 8:e53694. [PMID: 23349729 PMCID: PMC3548895 DOI: 10.1371/journal.pone.0053694] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 12/03/2012] [Indexed: 11/19/2022] Open
Abstract
The non-muscular cells that populate the space found between cardiomyocyte fibers are known as ‘cardiac interstitial cells’ (CICs). CICs are heterogeneous in nature and include different cardiac progenitor/stem cells, cardiac fibroblasts and other cell types. Upon heart damage CICs soon respond by initiating a reparative response that transforms with time into extensive fibrosis and heart failure. Despite the biomedical relevance of CICs, controversy remains on the ontogenetic relationship existing between the different cell kinds homing at the cardiac interstitium, as well as on the molecular signals that regulate their differentiation, maturation, mutual interaction and role in adult cardiac homeostasis and disease. Our work focuses on the analysis of epicardial-derived cells, the first cell type that colonizes the cardiac interstitium. We present here a characterization and an experimental analysis of the differentiation potential and mobilization properties of a new cell line derived from mouse embryonic epicardium (EPIC). Our results indicate that these cells express some markers associated with cardiovascular stemness and retain part of the multipotent properties of embryonic epicardial derivatives, spontaneously differentiating into smooth muscle, and fibroblast/myofibroblast-like cells. Epicardium-derived cells are also shown to initiate a characteristic response to different growth factors, to display a characteristic proteolytic expression profile and to degrade biological matrices in 3D in vitro assays. Taken together, these data indicate that EPICs are relevant to the analysis of epicardial-derived CICs, and are a god model for the research on cardiac fibroblasts and the role these cells play in ventricular remodeling in both ischemic or non/ischemic myocardial disease.
Collapse
Affiliation(s)
- Adrián Ruiz-Villalba
- Department of Animal Biology, Faculty of Science, University of Málaga, Málaga, Spain
| | - Algirdas Ziogas
- Department of Obstetrics, University Hospital Zürich, Zürich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zürich, Zürich, Switzerland
| | - José M. Pérez-Pomares
- Department of Animal Biology, Faculty of Science, University of Málaga, Málaga, Spain
- * E-mail:
| |
Collapse
|
35
|
Abstract
Myocardial infarction is one of the major causes of left ventricular dilatation, frequently leading to heart failure. In the last decade, the wound healing process that takes place in the infarct area after infarction has been recognized as a novel therapeutic target to attenuate left ventricular dilatation and preserve an adequate cardiac function. In this chapter, we discuss the role of Wnt signaling in the wound healing process after infarction, with a specific focus on its modulating effect on myofibroblast characteristics.
Collapse
|
36
|
Targeting the Wnt/frizzled signaling pathway after myocardial infarction: a new tool in the therapeutic toolbox? Trends Cardiovasc Med 2012; 23:121-7. [PMID: 23266229 DOI: 10.1016/j.tcm.2012.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 09/24/2012] [Accepted: 09/25/2012] [Indexed: 11/20/2022]
Abstract
Wnt/frizzled signaling in the adult heart is quiescent under normal conditions; however it is reactivated after myocardial infarction (MI). Any intervention at the various levels of this pathway can modulate its signaling. Several studies have targeted Wnt/frizzled signaling after MI with the majority of them indicating that the inhibition of the pathway is beneficial since it improves infarct healing and prevents heart failure. This suggests that blocking the Wnt/frizzled signaling pathway could be a potential novel therapeutic target to prevent the adverse cardiac remodeling after MI.
Collapse
|
37
|
Dawson K, Aflaki M, Nattel S. Role of the Wnt-Frizzled system in cardiac pathophysiology: a rapidly developing, poorly understood area with enormous potential. J Physiol 2012. [PMID: 23207593 DOI: 10.1113/jphysiol.2012.235382] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract The Wnt-Frizzled (Fzd) G-protein-coupled receptor system, involving 19 distinct Wnt ligands and 10 Fzd receptors, plays key roles in the development and functioning of many organ systems. There is increasing evidence that Wnt-Fzd signalling is important in regulating cardiac function. Wnt-Fzd signalling primarily involves a canonical pathway, with dishevelled-1-dependent nuclear translocation of β-catenin that derepresses Wnt-sensitive gene transcription, but can also include non-canonical pathways via phospholipase-C/Ca(2+) mobilization and dishevelled-protein activation of small GTPases. Wnt-Fzd effects vary with specific ligand/receptor interactions and associated downstream pathways. This paper reviews the biochemistry and physiology of the Wnt-Fzd complex, and presents current knowledge of Wnt signalling in cardiac remodelling processes such as hypertrophy and fibrosis, as well as disease states such as myocardial infarction (MI), heart failure and arrhythmias. Wnt signalling is activated during hypertrophy; inhibiting Wnt signalling by activating glycogen synthase kinase attenuates the hypertrophic response. Wnt signalling has complex and time-dependent actions post-MI, so that either beneficial or harmful effects might result from Wnt-directed interventions. Stem cell biology, a promising area for therapeutic intervention, is highly regulated by Wnt signalling. The Wnt system regulates fibroblast function, and is prominently altered in arrhythmogenic ventricular cardiomyopathy, a familial disease involving excess deposition of fibroadipose tissue. Wnt signalling controls connexin43 expression, thereby contributing to the regulation of cardiac electrical stability and arrhythmia generation. Although much has been learned about Wnt-Fzd signalling in hypertrophy and infarction, its role is poorly understood for a broad range of other heart disorders. Much more needs to be learned for its contributions to be fully appreciated, and to permit more effective exploitation of its enormous potential in therapeutic development.
Collapse
Affiliation(s)
- Kristin Dawson
- S. Nattel: 5000 Belanger St. E, Montreal, Quebec, Canada H1T 1C8.
| | | | | |
Collapse
|
38
|
Hermans KC, Daskalopoulos EP, Blankesteijn WM. Interventions in Wnt signaling as a novel therapeutic approach to improve myocardial infarct healing. FIBROGENESIS & TISSUE REPAIR 2012; 5:16. [PMID: 22967504 PMCID: PMC3472244 DOI: 10.1186/1755-1536-5-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/20/2012] [Indexed: 01/12/2023]
Abstract
Following myocardial infarction, wound healing takes place in the infarct area where the non-viable cardiac tissue is replaced by a scar. Inadequate wound healing or insufficient maintenance of the extracellular matrix in the scar can lead to excessive dilatation of the ventricles, one of the hallmarks of congestive heart failure. Therefore, it is important to better understand the wound-healing process in the heart and to develop new therapeutic agents that target the infarct area in order to maintain an adequate cardiac function. One of these potential novel therapeutic targets is Wnt signaling. Wnt signaling plays an important role in embryonic myocardial development but in the adult heart the pathway is thought to be silent. However, there is increasing evidence that components of the Wnt pathway are re-expressed during cardiac repair, implying a regulatory role. Recently, several studies have been published where the effect of interventions in Wnt signaling on infarct healing has been studied. In this review, we will summarize the results of these studies and discuss the effects of these interventions on the different cell types that are involved in the wound healing process.
Collapse
Affiliation(s)
- Kevin Cm Hermans
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Maastricht University, 50 Universiteitssingel, 6229ER Maastricht, PO Box 616 6200MD, Maastricht, The Netherlands.
| | | | | |
Collapse
|
39
|
The role of cardiac fibroblasts in the transition from inflammation to fibrosis following myocardial infarction. Vascul Pharmacol 2012; 58:182-8. [PMID: 22885638 DOI: 10.1016/j.vph.2012.07.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/03/2012] [Accepted: 07/07/2012] [Indexed: 12/19/2022]
Abstract
Cardiac fibroblasts (CF) play a pivotal role in the repair and remodeling of the heart that occur following myocardial infarction (MI). The transition through the inflammatory, granulation and maturation phases of infarct healing is driven by cellular responses to local levels of cytokines, chemokines and growth factors that fluctuate in a temporal and spatial manner. In the acute inflammatory phase early after MI, CF contribute to the inflammatory milieu through increased secretion of proinflammatory cytokines and chemokines, and they promote extracellular matrix (ECM) degradation by increasing matrix metalloproteinase (MMP) expression and activity. In the granulation phase, CF migrate into the infarct zone, proliferate and produce MMPs and pro-angiogenic molecules to facilitate revascularization. Fibroblasts also undergo a phenotypic change to become myofibroblasts. In the maturation phase, inflammation is reduced by anti-inflammatory cytokines, and increased levels of profibrotic stimuli induce myofibroblasts to synthesize new ECM to form a scar. The scar is contracted through the mechanical force generated by myofibroblasts, preventing cardiac dilation. In this review we discuss the transition from myocardial inflammation to fibrosis with particular focus on how CF respond to alterations in proinflammatory and profibrotic signals. By furthering our understanding of these events, it is hoped that new therapeutic interventions will be developed that selectively reduce adverse myocardial remodeling post-MI, while sparing essential repair mechanisms.
Collapse
|
40
|
Snead AN, Insel PA. Defining the cellular repertoire of GPCRs identifies a profibrotic role for the most highly expressed receptor, protease-activated receptor 1, in cardiac fibroblasts. FASEB J 2012; 26:4540-7. [PMID: 22859370 DOI: 10.1096/fj.12-213496] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
G-protein-coupled receptors (GPCRs) have many roles in cell regulation and are commonly used as drug targets, but the repertoire of GPCRs expressed by individual cell types has not been defined. Here we use an unbiased approach, GPCR RT-PCR array, to define the expression of nonchemosensory GPCRs by cardiac fibroblasts (CFs) isolated from Rattus norvegicus. CFs were selected because of their importance for cardiac structure and function and their contribution to cardiac fibrosis, which occurs with advanced age, after acute injury (e.g., myocardial infarction), and in disease states (e.g., diabetes mellitus, hypertension). We discovered that adult rat CFs express 190 GPCRs and that activation of protease-activated receptor 1 (PAR1), the most highly expressed receptor, raises the expression of profibrotic markers in rat CFs, resulting in a 60% increase in collagen synthesis and conversion to a profibrogenic myofibroblast phenotype. We use siRNA knockdown of PAR1 (90% decrease in mRNA) to show that the profibrotic effects of thrombin are PAR1-dependent. These findings, which define the expression of GPCRs in CFs, provide a proof of principle of an approach to discover previously unappreciated, functionally relevant GPCRs and reveal a potential role for thrombin and PAR1 in wound repair and pathophysiology of the adult heart.
Collapse
Affiliation(s)
- Aaron N Snead
- Department of Pharmacology, University of California at San Diego, La Jolla, California 92093, USA
| | | |
Collapse
|
41
|
van der Velden JLJ, Guala AS, Leggett SE, Sluimer J, Badura ECHL, Janssen-Heininger YMW. Induction of a mesenchymal expression program in lung epithelial cells by wingless protein (Wnt)/β-catenin requires the presence of c-Jun N-terminal kinase-1 (JNK1). Am J Respir Cell Mol Biol 2012; 47:306-14. [PMID: 22461429 DOI: 10.1165/rcmb.2011-0297oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent studies suggest the importance of the transition of airway epithelial cells (EMT) in pulmonary fibrosis, and also indicate a role for Wingless protein (Wnt)/β-catenin signaling in idiopathic pulmonary fibrosis. We investigated the possible role of the Wnt signaling pathway in inducing EMT in lung epithelial cells, and sought to unravel the role of c-Jun-N-terminal-kinase-1 (JNK1). The exposure of C10 lung epithelial cells or primary mouse tracheal epithelial cells (MTECs) to Wnt3a resulted in increases in JNK phosphorylation and nuclear β-catenin content. Because the role of β-catenin as a transcriptional coactivator is well established, we investigated T-cell factor/lymphocyte-enhancement factor (TCF/LEF) transcriptional activity in C10 lung epithelial cells after the activation of Wnt. TCF/LEF transcriptional activity was enhanced after the activation of Wnt, and this increase in TCF/LEF transcriptional activity was diminished after the small interfering (si)RNA-mediated ablation of JNK. The activation of the Wnt pathway by Wnt3a, or the expression of either wild-type or constitutively active β-catenin (S37A), led to the activation of an EMT transcriptome, manifested by the increased mRNA expression of CArG box-binding factor-A, fibroblast-specific protein (FSP)-1, α-smooth muscle actin (α-SMA), and vimentin, increases in the content of α-SMA and FSP1, and the concomitant loss of zona occludens-1. The siRNA-mediated ablation of β-catenin substantially decreased Wnt3a-induced EMT. The siRNA ablation of JNK1 largely abolished Wnt3a, β-catenin, and β-catenin S37a-induced EMT. In MTECs lacking Jnk1, Wnt3a-induced increases in nuclear β-catenin, EMT transcriptome, and the content of α-SMA or FSP1 were substantially diminished. These data show that the activation of the Wnt signaling pathway is capable of inducing an EMT program in lung epithelial cells through β-catenin, and that this process is controlled by JNK1.
Collapse
Affiliation(s)
- Jos L J van der Velden
- Department of Pathology, Health Sciences Research Facility, Room 216A, University of Vermont, Burlington, VT 05405, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Fraccarollo D, Galuppo P, Bauersachs J. Novel therapeutic approaches to post-infarction remodelling. Cardiovasc Res 2012; 94:293-303. [PMID: 22387461 DOI: 10.1093/cvr/cvs109] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Adverse cardiac remodelling is a major cause of morbidity and mortality following acute myocardial infarction (MI). Mechanical and neurohumoral factors involved in structural and molecular post-infarction remodelling were important targets in research and treatment for years. More recently, therapeutic strategies that address myocardial regeneration and pathophysiological mechanisms of infarct wound healing appear to be useful novel tools to prevent progressive ventricular dilation, functional deterioration, life-threatening arrhythmia, and heart failure. This review provides an overview of future and emerging therapies for cardiac wound healing and remodelling after MI.
Collapse
Affiliation(s)
- Daniela Fraccarollo
- Klinik fuer Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | | | | |
Collapse
|
43
|
Daskalopoulos EP, Janssen BJA, Blankesteijn WM. Myofibroblasts in the infarct area: concepts and challenges. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2012; 18:35-49. [PMID: 22214878 DOI: 10.1017/s143192761101227x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Myofibroblasts are differentiated fibroblasts that hold a key role in wound healing and remodeling following myocardial infarction (MI). A large repertoire of stimuli, such as mechanical stretch, growth factors, cytokines, and vasoactive peptides, induces myofibroblast differentiation. Myofibroblasts are responsible for the production and deposition of collagen, leading to the establishment of a dense extracellular matrix that strengthens the infarcted tissue and minimizes dilatation of the infarct area. In addition, cells contributing to fibrosis act on sites distal from the infarct area and promote collagen deposition in noninfarcted tissue, thus contributing to adverse remodeling and consequently to the development of congestive heart failure (CHF). Current drugs that are used to treat post-MI CHF do influence fibroblasts and myofibroblasts; however, their therapeutic efficacy is far from being regarded as ideal. Novel therapeutic agents targeting (myo)fibroblasts are being developed to successfully prevent the cardiac remodeling of sites remote from the infarct area and therefore hinder the establishment of CHF. The purpose of this review article is to discuss the basic concepts of the myofibroblasts' actions in cardiac wound healing processes, factors that influence them, currently available pharmacological agents, and future challenges in this area.
Collapse
Affiliation(s)
- Evangelos P Daskalopoulos
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Maastricht University, 50 Universiteitssingel, 6229ER Maastricht, P.O. Box 616, 6200MD Maastricht, The Netherlands
| | | | | |
Collapse
|
44
|
ter Horst P, Smits JFM, Blankesteijn WM. The Wnt/Frizzled pathway as a therapeutic target for cardiac hypertrophy: where do we stand? Acta Physiol (Oxf) 2012; 204:110-7. [PMID: 21624093 DOI: 10.1111/j.1748-1716.2011.02309.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiac hypertrophy is an enlargement of the heart muscle in response to wall stress. This hypertrophic response often leads to heart failure. In recent years, several studies have shown the involvement of Wnt signalling in hypertrophic growth. In this review, the role of Wnt signalling and the possibilities for therapeutic interventions are discussed. In healthy adult heart tissue, Wnt signalling is very low. However, under pathological condition such as hypertension, Wnt signalling is activated. In recent years, it has become clear that both β-catenin-dependent signalling and β-catenin-independent signalling are involved in hypertrophic growth. Several studies, both in vitro and in vivo, have shown that genetic interventions in Wnt signalling at different levels resulted in an attenuated or diminished hypertrophic response. Therefore, inhibition of Wnt signalling could provide a new therapeutic strategy for cardiac hypertrophy, but further research on the Wnts and Frizzleds involved in the different forms of cardiac hypertrophy will be needed to achieve this goal.
Collapse
Affiliation(s)
- P ter Horst
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | | | | |
Collapse
|
45
|
Abstract
Cellular thiols including GSH (glutathione) and L-Cys (L-cysteine) are essential for cell signalling, growth and differentiation. L-Cys is derived from the extracellular thiol pool and is the rate-limiting compound for intracellular GSH biosynthesis. The present study investigated the effect of thiol-supplemented medium on cell growth, phenotype and total GSH of cultured hPMCs (human peritoneal mesothelial cells). Cells were cultured in medium M199 supplemented with 2% serum, with 'plus' or without 'minus' L-Cys and compared with medium supplemented with either β-ME (β-mercaptoethanol) (0.25 mmol/l) or the receptor tyrosine kinase ligand EGF (epidermal growth factor, 100 ng/ml). β-ME produced a disproportionate increase in total GSH compared with L-Cys and other thiols tested [(procysteine (2-oxothiazolidine-4-carboxylic acid) or NAC (N-acetyl-L-cysteine)], while growth and morphology were identical. Cell behaviour of primary hPMCs is characterized by the transition of fibroblastoid to cobblestone morphology during early passage. L-Cys and β-ME promoted a rapid MET (mesenchymal-to-epithelial transition) within 3 days of culture, confirmed by the presence of cobblestone cells, intact organelles, abundant microvilli, primary cilia and cortical actin. In contrast, EGF produced a biphasic response consisting of delayed growth and retention of a fibroblastoid morphology. During a rapid log phase of growth, MET was accompanied by rapid catch-up growth. Thiols may stabilize the epithelial phenotype by engaging redox-sensitive receptors and transcription factors that modulate differentiation. These data may benefit researchers working on thiol-mediated cell differentiation and strategies to regenerate damage to serosal membranes.
Collapse
|
46
|
Activation of WNT/β-catenin signaling in pulmonary fibroblasts by TGF-β₁ is increased in chronic obstructive pulmonary disease. PLoS One 2011; 6:e25450. [PMID: 21980461 PMCID: PMC3184127 DOI: 10.1371/journal.pone.0025450] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 09/05/2011] [Indexed: 02/07/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is characterized by abnormal extracellular matrix (ECM) turnover. Recently, activation of the WNT/β-catenin pathway has been associated with abnormal ECM turnover in various chronic diseases. We determined WNT-pathway gene expression in pulmonary fibroblasts of individuals with and without COPD and disentangled the role of β-catenin in fibroblast phenotype and function. Methods We assessed the expression of WNT-pathway genes and the functional role of β-catenin, using MRC-5 human lung fibroblasts and primary pulmonary fibroblasts of individuals with and without COPD. Results Pulmonary fibroblasts expressed mRNA of genes required for WNT signaling. Stimulation of fibroblasts with TGF-β1, a growth factor important in COPD pathogenesis, induced WNT-5B, FZD8, DVL3 and β-catenin mRNA expression. The induction of WNT-5B, FZD6, FZD8 and DVL3 mRNA by TGF-β1 was higher in fibroblasts of individuals with COPD than without COPD, whilst basal expression was similar. Accordingly, TGF-β1 activated β-catenin signaling, as shown by an increase in transcriptionally active and total β-catenin protein expression. Furthermore, TGF-β1 induced the expression of collagen1α1, α-sm-actin and fibronectin, which was attenuated by β-catenin specific siRNA and by pharmacological inhibition of β-catenin, whereas the TGF-β1-induced expression of PAI-1 was not affected. The induction of transcriptionally active β-catenin and subsequent fibronectin deposition induced by TGF-β1 were enhanced in pulmonary fibroblasts from individuals with COPD. Conclusions β-catenin signaling contributes to ECM production by pulmonary fibroblasts and contributes to myofibroblasts differentiation. WNT/β-catenin pathway expression and activation by TGF-β1 is enhanced in pulmonary fibroblasts from individuals with COPD. This suggests an important role of the WNT/β-catenin pathway in regulating fibroblast phenotype and function in COPD.
Collapse
|
47
|
Laeremans H, Hackeng TM, van Zandvoort MAMJ, Thijssen VLJL, Janssen BJA, Ottenheijm HCJ, Smits JFM, Blankesteijn WM. Blocking of frizzled signaling with a homologous peptide fragment of wnt3a/wnt5a reduces infarct expansion and prevents the development of heart failure after myocardial infarction. Circulation 2011; 124:1626-35. [PMID: 21931076 DOI: 10.1161/circulationaha.110.976969] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The molecular pathways that control the wound healing after myocardial infarction (MI) are not completely elucidated. One of these pathways is the Wnt/Frizzled pathway. In this study, we evaluated Frizzled as a novel therapeutic target for MI. These Frizzled proteins act as receptors for Wnt proteins and were previously shown to be expressed in the healing infarct. METHODS AND RESULTS Wnt/Frizzled signaling has been studied for decades, but synthetic ligands that interfere with the interaction between Wnts and Frizzled have not been described to date. Here we report the selection of 3 peptides derived from regions of high homology between Wnt3a and Wnt5a that act as antagonists for Frizzled proteins. UM206, the peptide with the highest affinity, antagonized the effect of Wnt3a and Wnt5a in different in vitro assays. Administration of UM206 to mice for 5 weeks, starting immediately after the induction of MI, reduced infarct expansion and increased the numbers of capillaries and myofibroblasts in the infarct area. Moreover, heart failure development was inhibited by this therapy. CONCLUSIONS Blocking of Frizzled signaling reduces infarct expansion and preserves cardiac function after MI. Our findings underscore the potential of Frizzled receptors as a target for pharmacotherapy of cardiac remodeling after MI.
Collapse
Affiliation(s)
- Hilde Laeremans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University, 50 Universiteitssingel, 6229ER Maastricht/PO Box 616, 6200MD Maastricht, Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Carthy JM, Garmaroudi FS, Luo Z, McManus BM. Wnt3a induces myofibroblast differentiation by upregulating TGF-β signaling through SMAD2 in a β-catenin-dependent manner. PLoS One 2011; 6:e19809. [PMID: 21611174 PMCID: PMC3097192 DOI: 10.1371/journal.pone.0019809] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 04/17/2011] [Indexed: 01/08/2023] Open
Abstract
Growing evidence suggests the Wnt family of secreted glycoproteins and their associated signaling pathways, linked to development, are recapitulated during wound repair and regeneration events. However, the role of the Wnt pathway in such settings remains unclear. In the current study, we treated mouse fibroblasts with 250 ng/mL of recombinant Wnt3a for 72 hours and examined its affect on cell morphology and function. Wnt3a induced a spindle-like morphology in fibroblasts characterized by the increased formation of stress fibres. Wnt3a decreased the proliferation of fibroblasts, but significantly increased cell migration as well as fibroblast-mediated contraction of a collagen lattice. Wnt3a significantly increased the expression of TGF-β and its associated signaling through SMAD2. Consistent with this, we observed significantly increased smooth muscle α-actin expression and incorporation of this contractile protein into stress fibres following Wnt3a treatment. Knockdown of β-catenin using siRNA reversed the Wnt3a-induced smooth muscle α-actin expression, suggesting these changes were dependent on canonical Wnt signaling through β-catenin. Neutralization of TGF-β with a blocking antibody significantly inhibited the Wnt3a-induced smooth muscle α-actin expression, indicating these changes were dependent on the increased TGF-β signaling. Collectively, this data strongly suggests Wnt3a promotes the formation of a myofibroblast-like phenotype in cultured fibroblasts, in part, by upregulating TGF-β signaling through SMAD2 in a β-catenin-dependent mechanism. As myofibroblasts are critical regulators of wound healing responses, these findings may have important implications for our understanding of normal and aberrant injury and repair events.
Collapse
Affiliation(s)
- Jon M. Carthy
- UBC James Hogg Research Centre, Institute for Heart+Lung Health, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Farshid S. Garmaroudi
- UBC James Hogg Research Centre, Institute for Heart+Lung Health, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zongshu Luo
- UBC James Hogg Research Centre, Institute for Heart+Lung Health, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce M. McManus
- UBC James Hogg Research Centre, Institute for Heart+Lung Health, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
49
|
Bergmann MW. WNT signaling in adult cardiac hypertrophy and remodeling: lessons learned from cardiac development. Circ Res 2010; 107:1198-208. [PMID: 21071717 DOI: 10.1161/circresaha.110.223768] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
On pathological stress, the heart reactivates several signaling pathways that traditionally were thought to be operational only in the developing heart. One of these pathways is the WNT signaling pathway. WNT controls heart development but is also modulated during adult heart remodeling. This review summarizes the currently available data regarding WNT signaling during left ventricular (LV) remodeling. Upstream, soluble frizzled-related proteins (sFRPs) block WNT-dependent activation of the canonical WNT pathway. By inhibition of WNT activation, these factors also reduce β-catenin-dependent transcription by altering the ratio of cytoplasmic/nuclear β-catenin. In experimental settings, sFRPs injected into the heart attenuated LV remodeling. sFRPs are secreted from autologous bone marrow-derived mononuclear cells. Disheveled is a signaling intermediate of both the canonical and noncanonical WNT pathway. Similarly to the effect of sFRP, depletion of a disheveled isoform attenuated LV remodeling. In contrast, disheveled activation led to progressive dilated cardiomyopathy. Inhibition of nuclear β-catenin signaling downstream of the canonical WNT pathway significantly reduced postinfarct mortality and functional decline of LV function following chronic left anterior descending coronary artery ligation. WNT signaling also affects mobilization and homing of bone marrow-derived vasculogenic progenitor cells. Finally, heart-specific WNT/β-catenin interaction partners have been identified that will possibly allow targeting this pathway in a tissue-specific manner. In summary, the WNT pathway plays a pivotal role in adult cardiac remodeling and may be suitable for therapeutic interventions. Currently, several molecular and cellular mechanisms whereby WNT inhibition attenuates LV remodeling are proposed. Reactivation of the developmental program to restore functional LV myocardium from resident precursor cells may significantly contribute to this process.
Collapse
Affiliation(s)
- Martin W Bergmann
- Experimental and Clinical Research Center, Charité Campus Buch & Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|