1
|
Fiorino E, Rossin D, Vanni R, Aubry M, Giachino C, Rastaldo R. Recent Insights into Endogenous Mammalian Cardiac Regeneration Post-Myocardial Infarction. Int J Mol Sci 2024; 25:11747. [PMID: 39519298 PMCID: PMC11546116 DOI: 10.3390/ijms252111747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Myocardial infarction (MI) is a critical global health issue and a leading cause of heart failure. Indeed, while neonatal mammals can regenerate cardiac tissue mainly through cardiomyocyte proliferation, this ability is lost shortly after birth, resulting in the adult heart's inability to regenerate after injury effectively. In adult mammals, the adverse cardiac remodelling, which compensates for the loss of cardiac cells, impairs cardiac function due to the non-contractile nature of fibrotic tissue. Moreover, the neovascularisation after MI is inadequate to restore blood flow to the infarcted myocardium. This review aims to synthesise the most recent insights into the molecular and cellular players involved in endogenous myocardial and vascular regeneration, facilitating the identification of mechanisms that could be targeted to trigger cardiac regeneration, reduce fibrosis, and improve functional recovery post-MI. Reprogramming adult cardiomyocytes to regain their proliferative potential, along with the modulation of target cells responsible for neovascularisation, represents promising therapeutic strategies. An updated overview of endogenous mechanisms that regulate both myocardial and coronary vasculature regeneration-including stem and progenitor cells, growth factors, cell cycle regulators, and key signalling pathways-could help identify new critical intervention points for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (E.F.); (D.R.); (R.V.); (M.A.); (C.G.)
| |
Collapse
|
2
|
Ergui I, Griffith N, Salama J, Ebner B, Dangl M, Vincent L, Razuk V, Marzouka G, Colombo R. In-Hospital Outcomes in Patients With Non-ST Segment Elevation Myocardial Infarction and Concomitant Neurodevelopmental Disorders in the United States: Insights From the National Inpatient Sample 2011-2020. Cureus 2024; 16:e60289. [PMID: 38746481 PMCID: PMC11093150 DOI: 10.7759/cureus.60289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 05/16/2024] Open
Abstract
Patients with neurodevelopmental disorders (NDDs) encounter significant barriers to receiving quality health care, particularly for acute conditions such as non-ST segment elevation myocardial infarction (NSTEMI). This study addresses the critical gap in knowledge regarding in-hospital outcomes and the use of invasive therapies in this demographic. By analyzing data from the National Inpatient Sample database from 2011 to 2020 using the International Classification of Diseases, Ninth Edition (ICD-9) and Tenth Edition (ICD-10) codes, we identified patients with NSTEMI, both with and without NDDs, and compared baseline characteristics, in-hospital outcomes, and the application of invasive treatments. The analysis involved a weighted sample of 7,482,216 NSTEMI hospitalizations, of which 30,168 (0.40%) patients had NDDs. There were significantly higher comorbidity-adjusted odds of in-hospital mortality, cardiac arrest, endotracheal intubation, infectious complications, ventricular arrhythmias, and restraint use among the NDD cohort. Conversely, this group exhibited lower adjusted odds of undergoing left heart catheterization, percutaneous coronary intervention, or coronary artery bypass graft surgery. These findings underscore the disparities faced by patients with NDDs in accessing invasive cardiac interventions, highlighting the need for further research to address these barriers and improve care quality for this vulnerable population.
Collapse
Affiliation(s)
- Ian Ergui
- Internal Medicine, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| | - Nayrana Griffith
- Cardiology, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| | - Joshua Salama
- Internal Medicine, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| | - Bertrand Ebner
- Cardiology, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| | - Michael Dangl
- Internal Medicine, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| | - Louis Vincent
- Cardiology, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| | - Victor Razuk
- Cardiology, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| | - George Marzouka
- Cardiology, Miami Department of Veterans Affairs, Miami, USA
| | - Rosario Colombo
- Cardiology, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, USA
| |
Collapse
|
3
|
Nam K, Dos Santos HT, Maslow F, Small T, Samuel RZ, Lei P, Andreadis ST, Baker OJ. Fibrin hydrogels fortified with FGF-7/10 and laminin-1 peptides promote regeneration of irradiated salivary glands. Acta Biomater 2023; 172:147-158. [PMID: 37844750 PMCID: PMC10908308 DOI: 10.1016/j.actbio.2023.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Ionizing radiation, commonly used for head and neck cancer treatment, typically damages the salivary glands, resulting in hyposalivation. The development of treatments to restore this lost function is crucial for improving the quality of life for patients suffering from this condition. To address this clinical need, we have developed an innovative hydrogel by chemically conjugating laminin-1 peptides (A99 and YIGSR) and growth factors, FGF-7 and FGF-10, to fibrin hydrogels. Our results demonstrate that FGF-7/10 and laminin-1 peptides fortified fibrin hydrogel [enhanced laminin-1 peptides fibrin hydrogel (Ep-FH)] promotes salivary gland regeneration and functionality by improving epithelial tissue organization, establishing a healthy network of blood vessels and nerves, while reducing fibrosis in a head and neck irradiated mouse model. These results indicate that fibrin hydrogel-based implantable scaffolds containing pro-regenerative signals promote sustained secretory function of irradiated salivary glands, offering a potential alternative treatment for hyposalivation in head and neck cancer patients undergoing radiation treatment. These unique findings emphasize the potential of fibrin hydrogel-based implantable scaffolds enriched with pro-regenerative signals in sustaining the secretory function of irradiated salivary glands and offer a promising alternative treatment for addressing hyposalivation in head and neck cancer patients undergoing radiation therapy. STATEMENT OF SIGNIFICANCE: Radiation therapies used to treat head and neck cancers often result in damaged salivary gland, leading to severe dryness of the oral cavity. In this study, we engineered FGF-7 and FGF-10 and immobilized them into L1p-FH. The resulting hydrogel, Ep-FH, restored irradiated salivary gland functionality by enhancing epithelial tissue organization, promoting the development of a healthy network of blood vessels and nerves as well as reduction of fibrosis.
Collapse
Affiliation(s)
- Kihoon Nam
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States; Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Harim T Dos Santos
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States; Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Frank Maslow
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States; Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Travis Small
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States; Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Ronel Z Samuel
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Pedro Lei
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States; Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States; Center of Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States; Center of Cell, Gene and Tissue Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Olga J Baker
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States; Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States; Department of Biochemistry, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
4
|
Zhou DP, Deng LC, Feng X, Xu HJ, Tian Y, Yang WW, Zeng PP, Zou LH, Yan XH, Zhu XY, Shu DH, Guo Q, Huang XY, Bellusci S, Lou Z, Li XK, Zhang JS. FGF10 mitigates doxorubicin-induced myocardial toxicity in mice via activation of FGFR2b/PHLDA1/AKT axis. Acta Pharmacol Sin 2023; 44:2004-2018. [PMID: 37225844 PMCID: PMC10545682 DOI: 10.1038/s41401-023-01101-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/26/2023] [Indexed: 05/26/2023] Open
Abstract
Doxorubicin is a common chemotherapeutic agent in clinic, but myocardial toxicity limits its use. Fibroblast growth factor (FGF) 10, a multifunctional paracrine growth factor, plays diverse roles in embryonic and postnatal heart development as well as in cardiac regeneration and repair. In this study we investigated the role of FGF10 as a potential modulator of doxorubicin-induced cardiac cytotoxicity and the underlying molecular mechanisms. Fgf10+/- mice and an inducible dominant negative FGFR2b transgenic mouse model (Rosa26rtTA; tet(O)sFgfr2b) were used to determine the effect of Fgf10 hypomorph or blocking of endogenous FGFR2b ligands activity on doxorubicin-induced myocardial injury. Acute myocardial injury was induced by a single injection of doxorubicin (25 mg/kg, i.p.). Then cardiac function was evaluated using echocardiography, and DNA damage, oxidative stress and apoptosis in cardiac tissue were assessed. We showed that doxorubicin treatment markedly decreased the expression of FGFR2b ligands including FGF10 in cardiac tissue of wild type mice, whereas Fgf10+/- mice exhibited a greater degree of oxidative stress, DNA damage and apoptosis as compared with the Fgf10+/+ control. Pre-treatment with recombinant FGF10 protein significantly attenuated doxorubicin-induced oxidative stress, DNA damage and apoptosis both in doxorubicin-treated mice and in doxorubicin-treated HL-1 cells and NRCMs. We demonstrated that FGF10 protected against doxorubicin-induced myocardial toxicity via activation of FGFR2/Pleckstrin homology-like domain family A member 1 (PHLDA1)/Akt axis. Overall, our results unveil a potent protective effect of FGF10 against doxorubicin-induced myocardial injury and identify FGFR2b/PHLDA1/Akt axis as a potential therapeutic target for patients receiving doxorubicin treatment.
Collapse
Affiliation(s)
- De-Pu Zhou
- Medical Research Center and the Department of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Lian-Cheng Deng
- Medical Research Center and the Department of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiao Feng
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Hui-Jing Xu
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ye Tian
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei-Wei Yang
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ping-Ping Zeng
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Li-Hui Zou
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xi-Hua Yan
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xia-Yan Zhu
- Medical Research Center and the Department of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Dan-Hua Shu
- Medical Research Center and the Department of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qiang Guo
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiao-Ying Huang
- Medical Research Center and the Department of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Saverio Bellusci
- Cardio-Pulmonary Institute and Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, 35392, Germany
| | - Zhenkun Lou
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xiao-Kun Li
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Jin-San Zhang
- Medical Research Center and the Department of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- International Collaborative Center on Growth Factor Research, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
5
|
Gafranek JT, D'Aniello E, Ravisankar P, Thakkar K, Vagnozzi RJ, Lim HW, Salomonis N, Waxman JS. Sinus venosus adaptation models prolonged cardiovascular disease and reveals insights into evolutionary transitions of the vertebrate heart. Nat Commun 2023; 14:5509. [PMID: 37679366 PMCID: PMC10485058 DOI: 10.1038/s41467-023-41184-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
How two-chambered hearts in basal vertebrates have evolved from single-chamber hearts found in ancestral chordates remains unclear. Here, we show that the teleost sinus venosus (SV) is a chamber-like vessel comprised of an outer layer of smooth muscle cells. We find that in adult zebrafish nr2f1a mutants, which lack atria, the SV comes to physically resemble the thicker bulbus arteriosus (BA) at the arterial pole of the heart through an adaptive, hypertensive response involving smooth muscle proliferation due to aberrant hemodynamic flow. Single cell transcriptomics show that smooth muscle and endothelial cell populations within the adapting SV also take on arterial signatures. Bulk transcriptomics of the blood sinuses flanking the tunicate heart reinforce a model of greater equivalency in ancestral chordate BA and SV precursors. Our data simultaneously reveal that secondary complications from congenital heart defects can develop in adult zebrafish similar to those in humans and that the foundation of equivalency between flanking auxiliary vessels may remain latent within basal vertebrate hearts.
Collapse
Affiliation(s)
- Jacob T Gafranek
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Division of Molecular Cardiovascular Biology and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Enrico D'Aniello
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121, Napoli, Italy
| | - Padmapriyadarshini Ravisankar
- Division of Molecular Cardiovascular Biology and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Kairavee Thakkar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Ronald J Vagnozzi
- Division of Cardiology, Gates Center for Regenerative Medicine, Consortium for Fibrosis Research and Translation (CFReT), University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Joshua S Waxman
- Division of Molecular Cardiovascular Biology and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
6
|
Wang H, Lin X, Wang X, Liu X, He S, Lyu G. Maternal body fluid lncRNAs serve as biomarkers to diagnose ventricular septal defect: from amniotic fluid to plasma. Front Genet 2023; 14:1254829. [PMID: 37745849 PMCID: PMC10516564 DOI: 10.3389/fgene.2023.1254829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Maternal body fluids contain abundant cell-free fetal RNAs which have the potential to serve as indicators of fetal development and pathophysiological conditions. In this context, this study aimed to explore the potential diagnostic value of maternal circulating long non-coding RNAs (lncRNAs) in ventricular septal defect (VSD). Methods: The potential of lncRNAs as non-invasive prenatal biomarkers for VSD was evaluated using quantitative polymerase chain reaction (qPCR) and receiver operating characteristic (ROC) curve analysis. The biological processes and regulatory network of these lncRNAs were elucidated through bioinformatics analysis. Results: Three lncRNAs (LINC00598, LINC01551, and GATA3-AS1) were found to be consistent in both maternal plasma and amniotic fluid. These lncRNAs exhibited strong diagnostic performance for VSD, with AUC values of 0.852, 0.957, and 0.864, respectively. The bioinformatics analysis revealed the involvement of these lncRNAs in heart morphogenesis, actin cytoskeleton organization, cell cycle regulation, and protein binding through a competitive endogenous RNA (ceRNA) network at the post-transcriptional level. Conclusion: The cell-free lncRNAs present in the amniotic fluid have the potential to be released into the maternal circulation, making them promising candidates for investigating epigenetic regulation in VSD.
Collapse
Affiliation(s)
- Huaming Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xi Lin
- Department of Diagnostic Radiology, Fujian Cancer Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xinda Wang
- Department of Radiology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Xinxiu Liu
- Department of Medical Ultrasound, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Shaozheng He
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Guorong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
7
|
Bragança J, Pinto R, Silva B, Marques N, Leitão HS, Fernandes MT. Charting the Path: Navigating Embryonic Development to Potentially Safeguard against Congenital Heart Defects. J Pers Med 2023; 13:1263. [PMID: 37623513 PMCID: PMC10455635 DOI: 10.3390/jpm13081263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Congenital heart diseases (CHDs) are structural or functional defects present at birth due to improper heart development. Current therapeutic approaches to treating severe CHDs are primarily palliative surgical interventions during the peri- or prenatal stages, when the heart has fully developed from faulty embryogenesis. However, earlier interventions during embryonic development have the potential for better outcomes, as demonstrated by fetal cardiac interventions performed in utero, which have shown improved neonatal and prenatal survival rates, as well as reduced lifelong morbidity. Extensive research on heart development has identified key steps, cellular players, and the intricate network of signaling pathways and transcription factors governing cardiogenesis. Additionally, some reports have indicated that certain adverse genetic and environmental conditions leading to heart malformations and embryonic death may be amendable through the activation of alternative mechanisms. This review first highlights key molecular and cellular processes involved in heart development. Subsequently, it explores the potential for future therapeutic strategies, targeting early embryonic stages, to prevent CHDs, through the delivery of biomolecules or exosomes to compensate for faulty cardiogenic mechanisms. Implementing such non-surgical interventions during early gestation may offer a prophylactic approach toward reducing the occurrence and severity of CHDs.
Collapse
Affiliation(s)
- José Bragança
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Rute Pinto
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Bárbara Silva
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Nuno Marques
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- School of Health, University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
8
|
Gene Therapy for Cardiomyocyte Renewal: Cell Cycle, a Potential Therapeutic Target. Mol Diagn Ther 2023; 27:129-140. [PMID: 36512179 PMCID: PMC10123801 DOI: 10.1007/s40291-022-00625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/14/2022]
Abstract
Heart disease is the primary cause of death worldwide. Even though extensive research has been done, and many pharmacological and surgical treatments have been introduced to treat heart disease, the mortality rate still remains high. Gene therapy is widely used to understand molecular mechanisms of myocardial infarction and to treat cardiomyocyte loss. It was reported that adult cardiomyocytes proliferate at a very low rate; thus, targeting their proliferation has become a new regenerative therapeutic approach. Currently, re-activating cardiomyocyte proliferation appears to be one of the most promising methods to promote adult cardiomyocyte renewal. In this article, we highlight gene therapeutic targets of cell proliferation presently being pursued to re-activate the cell cycle of cardiomyocytes, including cell cycle regulators, transcription factors, microRNAs, signal transduction, and other contributing factors. We also summarize gene delivery vectors that have been used in cardiac research and major challenges to be overcome in the translation to the clinical approach and future directions.
Collapse
|
9
|
Yu L, Yi X, Yu C, Wang F, Tan X. Fibroblast growth factor 10 ameliorates renal ischaemia-reperfusion injury by attenuating mitochondrial damage. Clin Exp Pharmacol Physiol 2023; 50:59-67. [PMID: 36111374 DOI: 10.1111/1440-1681.13724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 12/13/2022]
Abstract
Ischaemia-reperfusion (I/R) injury is one of the leading causes of acute kidney injury (AKI). Its pathologic mechanism is quite complex, involving oxidative stress, inflammatory response, autophagy, and apoptosis. Fibroblast growth factor 10 (FGF10) and 5-hydroxydecanoate (5-HD) play essential roles in kidney injury. Rats were divided into four groups: (i) sham group, sham-operated animals with an unconstructed renal artery; (ii) I/R group, kidneys were subjected to 50 min of ischaemia followed by reperfusion for 2 days; (iii) I/R + FGF10 group, animals treated with 0.5 mg/kg FGF10 (i.p.) 1 h before ischaemia; and (iv) 5-HD group, animals treated with 5 mg/kg 5-HD (i.m.) 30 min before FGF10 treatment. Renal injury, apoptosis damage, mitochondrial oxidative damage, mitochondrial membrane potential (MMP), and expression of the ATP-sensitive K+ (KATP) channel subunit Kir6.2 were evaluated. FGF10 treatment significantly alleviated I/R-induced elevation in the serum creatinine level and the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling-positive tubular cells in the kidney. In addition, FGF10 dramatically ameliorated renal mitochondrial-related damage, including reducing mitochondrial-dependent apoptosis, alleviating oxidative stress, maintaining the mitochondrial membrane potential, and opening the mitochondrial KATP channels. The protective effect of FGF10 was significantly compromised by the ATP-dependent potassium channel blocker 5-HD. Our data suggest that FGF10 offers effective protection against I/R and improves animal survival by attenuating mitochondrial damage.
Collapse
Affiliation(s)
- Lixia Yu
- Department of Pharmacy, Xixi Hospital of Hangzhou, Zhejiang, China
| | - Xiaojiao Yi
- Department of Pharmacy, Xixi Hospital of Hangzhou, Zhejiang, China
| | - Cailong Yu
- Department of Pharmacy, Xixi Hospital of Hangzhou, Zhejiang, China
| | - Fugen Wang
- Department of Pharmacy, Xixi Hospital of Hangzhou, Zhejiang, China
| | - Xiaohua Tan
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
10
|
Chang S, Fulmer D, Hur SK, Thorvaldsen JL, Li L, Lan Y, Rhon-Calderon EA, Leu NA, Chen X, Epstein JA, Bartolomei MS. Dysregulated H19/Igf2 expression disrupts cardiac-placental axis during development of Silver-Russell syndrome-like mouse models. eLife 2022; 11:e78754. [PMID: 36441651 PMCID: PMC9704805 DOI: 10.7554/elife.78754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Dysregulation of the imprinted H19/IGF2 locus can lead to Silver-Russell syndrome (SRS) in humans. However, the mechanism of how abnormal H19/IGF2 expression contributes to various SRS phenotypes remains unclear, largely due to incomplete understanding of the developmental functions of these two genes. We previously generated a mouse model with humanized H19/IGF2 imprinting control region (hIC1) on the paternal allele that exhibited H19/Igf2 dysregulation together with SRS-like growth restriction and perinatal lethality. Here, we dissect the role of H19 and Igf2 in cardiac and placental development utilizing multiple mouse models with varying levels of H19 and Igf2. We report severe cardiac defects such as ventricular septal defects and thinned myocardium, placental anomalies including thrombosis and vascular malformations, together with growth restriction in mouse embryos that correlated with the extent of H19/Igf2 dysregulation. Transcriptomic analysis using cardiac endothelial cells of these mouse models shows that H19/Igf2 dysregulation disrupts pathways related to extracellular matrix and proliferation of endothelial cells. Our work links the heart and placenta through regulation by H19 and Igf2, demonstrating that accurate dosage of both H19 and Igf2 is critical for normal embryonic development, especially related to the cardiac-placental axis.
Collapse
Affiliation(s)
- Suhee Chang
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Diana Fulmer
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Stella K Hur
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Joanne L Thorvaldsen
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Li Li
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yemin Lan
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Eric A Rhon-Calderon
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Nicolae Adrian Leu
- Department of Biomedical Sciences, School of Veterinary Medicine, Institute for Regenerative Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Xiaowen Chen
- Penn Cardiovascular Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Marisa S Bartolomei
- Department of Cell and Developmental Biology, Epigenetics Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
11
|
Li T, Wu Y, Chen WC, Xue X, Suo MJ, Li P, Sheng W, Huang GY. Functional analysis of HECA variants identified in congenital heart disease in the Chinese population. J Clin Lab Anal 2022; 36:e24649. [PMID: 35949005 PMCID: PMC9459261 DOI: 10.1002/jcla.24649] [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: 06/01/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/07/2022] Open
Abstract
Background Congenital heart disease (CHD) is a class of cardiovascular defects that includes septal defects, outflow tract abnormalities, and valve defects. Human homolog of Drosophila headcase (HECA) is a novel cell cycle regulator whose role in CHD has not been elucidated. This is the first study to determine the frequency of HECA mutations in patients with CHD and the association between HECA variants and CHD. Methods In this study, we identified a candidate gene, HECA, by whole‐exome sequencing of an atrial septal defect family. To investigate the association between HECA variants and CHD risk, targeted exon sequencing was conducted in 689 individuals with sporadic CHD. We further analyzed the effect of HECA gene abnormalities on cardiomyocyte phenotype behavior and related signaling pathways by Western blotting, reverse transcription‐quantitative polymerase chain reaction, and scratch assay. Results We found a novel de novo mutation, c.409_410insA (p. W137fs), in the HECA gene and identified five rare deleterious variants that met the filtering criteria in 689 individuals with sporadic CHD. Fisher's exact test revealed a significant association between HECA variations and CHD compared with those in gnomADv2‐East Asians(p = 0.0027). Further functional analysis suggested that the variant p. W137fs resulted in a deficiency of the normal HECA protein, and HECA deficiency altered AC16 cell cycle progression, increased cell proliferation, and migration, and promoted the activation of the PDGF‐BB/PDGFRB/AKT pathway. Conclusions Our study identified HECA and its six rare variants, expanding the spectrum of genes associated with CHD pathogenesis in the Chinese population.
Collapse
Affiliation(s)
- Ting Li
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Yao Wu
- Children's Hospital of Fudan University, Shanghai, China
| | - Wei-Cheng Chen
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Xing Xue
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Mei-Jiao Suo
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Ping Li
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Wei Sheng
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Guo-Ying Huang
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases(2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
12
|
Hasan M, Oster M, Reyer H, Ponsuksili S, Murani E, Wolf P, Fischer DC, Wimmers K. Tissue-Wide Expression of Genes Related to Vitamin D Metabolism and FGF23 Signaling following Variable Phosphorus Intake in Pigs. Metabolites 2022; 12:metabo12080729. [PMID: 36005601 PMCID: PMC9413461 DOI: 10.3390/metabo12080729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Calcium (Ca) and phosphorus (P) homeostasis is maintained by several regulators, including vitamin D and fibroblast growth factor 23 (FGF23), and their tissue-specific activation and signaling cascades. In this study, the tissue-wide expression of key genes linked to vitamin D metabolism (CYP2R1, CYP27A1, CYP27B1, CYP24A1, GC, VDR) and FGF23 signaling (FGF23, FGFR1-4, KL) were investigated in pigs fed conventional (trial 1) and divergent P diets (trial 2). The tissue set comprised kidney, liver, bone, lung, aorta, and gastrointestinal tract sections. Expression patterns revealed that non-renal tissues and cells (NRTC) express genes to form active vitamin D [1,25(OH)2D3] according to site-specific requirements. A low P diet resulted in higher serum calcitriol and increased CYP24A1 expression in the small intestine, indicating local suppression of vitamin D signaling. A high P diet prompted increased mRNA abundances of CYP27B1 for local vitamin D synthesis, specifically in bone. For FGF23 signaling, analyses revealed ubiquitous expression of FGFR1-4, whereas KL was expressed in a tissue-specific manner. Dietary P supply did not affect skeletal FGF23; however, FGFR4 and KL showed increased expression in bone at high P supply, suggesting regulation to balance mineralization. Specific NRTC responses influence vitamin D metabolism and P homeostasis, which should be considered for a thrifty but healthy P supply.
Collapse
Affiliation(s)
- Maruf Hasan
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Michael Oster
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Henry Reyer
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Eduard Murani
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Petra Wolf
- Faculty of Agricultural and Environmental Sciences, University of Rostock, Justus-von-Liebig-Weg 6b, 18059 Rostock, Germany
| | - Dagmar-Christiane Fischer
- Department of Pediatrics, Rostock University Hospital, Ernst-Heydemann-Str. 8, 18057 Rostock, Germany
| | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
- Faculty of Agricultural and Environmental Sciences, University of Rostock, Justus-von-Liebig-Weg 6b, 18059 Rostock, Germany
- Correspondence: ; Tel.: +49-38208-68600
| |
Collapse
|
13
|
Wang N, Dong Y, Xu X, Shen Y, Huang Z, Yu Y, Liu Z, Gong W, Zhang S, Zheng Y, Song Y, Zhu Z, Jin L, Cong W. Fibroblast growth factor 10 protects against UVB‐induced skin injury by activating the ERK/YAP signalling pathway. Cell Prolif 2022; 55:e13315. [DOI: 10.1111/cpr.13315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 11/27/2022] Open
Affiliation(s)
- Nan Wang
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Yetong Dong
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Xiejun Xu
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Yingjie Shen
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
- College of Pharmacy and Research Institute for Drug Development Chonnam National University Gwangju Republic of Korea
| | - Zhiyuan Huang
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Yin Yu
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Zhili Liu
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Wenjie Gong
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Siyi Zhang
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Yeyi Zheng
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Yonghuan Song
- Department of Orthopaedics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou People's Republic of China
| | - Zhongxin Zhu
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Litai Jin
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| | - Weitao Cong
- School of Pharmaceutical Science Wenzhou Medical University Wenzhou People's Republic of China
| |
Collapse
|
14
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
15
|
Assimopoulos S, Hammill C, Fernandes DJ, Spencer Noakes TL, Zhou YQ, Nutter LMJ, Ellegood J, Anagnostou E, Sled JG, Lerch JP. Genetic mouse models of autism spectrum disorder present subtle heterogenous cardiac abnormalities. Autism Res 2022; 15:1189-1208. [PMID: 35445787 PMCID: PMC9325472 DOI: 10.1002/aur.2728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) and congenital heart disease (CHD) are linked on a functional and genetic level. Most work has investigated CHD‐related neurodevelopmental abnormalities. Cardiac abnormalities in ASD have been less studied. We investigated the prevalence of cardiac comorbidities relative to ASD genetic contributors. Using high frequency ultrasound imaging, we screened 9 ASD‐related genetic mouse models (Arid1b(+/−), Chd8(+/−), 16p11.2 (deletion), Sgsh(+/−), Sgsh(−/−), Shank3 Δexon 4–9(+/−), Shank3 Δexon 4–9(−/−), Fmr1(−/−), Vps13b(+/−)), and pooled wild‐type littermates (WTs). We measured heart rate (HR), aorta diameter (AoD), thickness and thickening of the left‐ventricular (LV) anterior and posterior walls, LV chamber diameter, fractional shortening, stroke volume and cardiac output, mitral inflow Peak E and A velocity ratio, ascending aorta velocity time integral (VTI). Mutant groups presented small‐scale alterations in cardiac structure and function compared to WTs (LV anterior wall thickness and thickening, chamber diameter and fractional shortening, HR). A greater number of significant differences was observed among mutant groups than between mutant groups and WTs. Mutant groups differed primarily in structural measures (LV chamber diameter and anterior wall thickness, HR, AoD). The mutant groups with most differences to WTs were 16p11.2 (deletion), Fmr1(−/−), Arid1b(+/−). The mutant groups with most differences from other mutant groups were 16p11.2 (deletion), Sgsh(+/−), Fmr1(−/−). Our results recapitulate the associated clinical findings. The characteristic ASD heterogeneity was recapitulated in the cardiac phenotype. The type of abnormal measures (morphological, functional) can highlight common underlying mechanisms. Clinically, knowledge of cardiac abnormalities in ASD can be essential as even non‐lethal abnormalities impact normal development.
Collapse
Affiliation(s)
- Stephania Assimopoulos
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Hammill
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Darren J Fernandes
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Tara Leigh Spencer Noakes
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Yu-Qing Zhou
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lauryl M J Nutter
- Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Evdokia Anagnostou
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada.,Sickkids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Garcia-Padilla C, Hernandez-Torres F, Lozano-Velasco E, Dueñas A, Muñoz-Gallardo MDM, Garcia-Valencia IS, Palencia-Vincent L, Aranega A, Franco D. The Role of Bmp- and Fgf Signaling Modulating Mouse Proepicardium Cell Fate. Front Cell Dev Biol 2022; 9:757781. [PMID: 35059396 PMCID: PMC8763981 DOI: 10.3389/fcell.2021.757781] [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: 08/12/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Bmp and Fgf signaling are widely involved in multiple aspects of embryonic development. More recently non coding RNAs, such as microRNAs have also been reported to play essential roles during embryonic development. We have previously demonstrated that microRNAs, i.e., miR-130, play an essential role modulating Bmp and Fgf signaling during early stages of cardiomyogenesis. More recently, we have also demonstrated that microRNAs are capable of modulating cell fate decision during proepicardial/septum transversum (PE/ST) development, since over-expression of miR-23 blocked while miR-125, miR-146, miR-223 and miR-195 enhanced PE/ST-derived cardiomyogenesis, respectively. Importantly, regulation of these microRNAs is distinct modulated by Bmp2 and Fgf2 administration in chicken. In this study, we aim to dissect the functional role of Bmp and Fgf signaling during mouse PE/ST development, their implication regulating post-transcriptional modulators such as microRNAs and their impact on lineage determination. Mouse PE/ST explants and epicardial/endocardial cell cultures were distinctly administrated Bmp and Fgf family members. qPCR analyses of distinct microRNAs, cardiomyogenic, fibrogenic differentiation markers as well as key elements directly epithelial to mesenchymal transition were evaluated. Our data demonstrate that neither Bmp2/Bmp4 nor Fgf2/Fgf8 signaling is capable of inducing cardiomyogenesis, fibrogenesis or inducing EMT in mouse PE/ST explants, yet deregulation of several microRNAs is observed, in contrast to previous findings in chicken PE/ST. RNAseq analyses in mouse PE/ST and embryonic epicardium identified novel Bmp and Fgf family members that might be involved in such cell fate differences, however, their implication on EMT induction and cardiomyogenic and/or fibrogenic differentiation is limited. Thus our data support the notion of species-specific differences regulating PE/ST cardiomyogenic lineage commitment.
Collapse
Affiliation(s)
- Carlos Garcia-Padilla
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain.,Department of Anatomy, Embryology and Zoology, School of Medicine, University of Extremadura, Badajoz, Spain
| | - Francisco Hernandez-Torres
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain.,Fundación Medina, Granada, Spain.,Department of Biochemistry and Molecular Biology, School of Medicine, University of Granada, Granada, Spain
| | - Estefania Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain.,Fundación Medina, Granada, Spain
| | - Angel Dueñas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | | | - Isabel S Garcia-Valencia
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | - Lledó Palencia-Vincent
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain
| | - Amelia Aranega
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain.,Fundación Medina, Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, Jaen, Spain.,Fundación Medina, Granada, Spain
| |
Collapse
|
17
|
Meier AB, Raj Murthi S, Rawat H, Toepfer CN, Santamaria G, Schmid M, Mastantuono E, Schwarzmayr T, Berutti R, Cleuziou J, Ewert P, Görlach A, Klingel K, Laugwitz KL, Seidman CE, Seidman JG, Moretti A, Wolf CM. Cell cycle defects underlie childhood-onset cardiomyopathy associated with Noonan syndrome. iScience 2022; 25:103596. [PMID: 34988410 PMCID: PMC8704485 DOI: 10.1016/j.isci.2021.103596] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/10/2021] [Accepted: 12/04/2021] [Indexed: 11/06/2022] Open
Abstract
Childhood-onset myocardial hypertrophy and cardiomyopathic changes are associated with significant morbidity and mortality in early life, particularly in patients with Noonan syndrome, a multisystemic genetic disorder caused by autosomal dominant mutations in genes of the Ras-MAPK pathway. Although the cardiomyopathy associated with Noonan syndrome (NS-CM) shares certain cardiac features with the hypertrophic cardiomyopathy caused by mutations in sarcomeric proteins (HCM), such as pathological myocardial remodeling, ventricular dysfunction, and increased risk for malignant arrhythmias, the clinical course of NS-CM significantly differs from HCM. This suggests a distinct pathophysiology that remains to be elucidated. Here, through analysis of sarcomeric myosin conformational states, histopathology, and gene expression in left ventricular myocardial tissue from NS-CM, HCM, and normal hearts complemented with disease modeling in cardiomyocytes differentiated from patient-derived PTPN11 N308S/+ induced pluripotent stem cells, we demonstrate distinct disease phenotypes between NS-CM and HCM and uncover cell cycle defects as a potential driver of NS-CM.
Collapse
Affiliation(s)
- Anna B. Meier
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Sarala Raj Murthi
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine and Health, Munich 80636, Germany
| | - Hilansi Rawat
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Christopher N. Toepfer
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Gianluca Santamaria
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Manuel Schmid
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Elisa Mastantuono
- Institute of Human Genetics, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg 85764, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Thomas Schwarzmayr
- Institute of Human Genetics, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg 85764, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Riccardo Berutti
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Institute of Neurogenomics, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Julie Cleuziou
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, Technical University of Munich, Munich 80636, Germany
- INSURE (Institute for Translational Cardiac Surgery), Department of Cardiovascular Surgery, German Heart Center Munich, Technical University of Munich, Munich 80636, Germany
| | - Peter Ewert
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine and Health, Munich 80636, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Agnes Görlach
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine and Health, Munich 80636, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Karin Klingel
- Institute for Pathology and Neuropathology, Department of Cardiopathology, University Hospital Tuebingen, Tuebingen 72076, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | | | | | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| | - Cordula M. Wolf
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine and Health, Munich 80636, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich Germany
| |
Collapse
|
18
|
Hubert F, Payan SM, Pelce E, Bouchard L, Sturny R, Lenfant N, Mottola G, Collart F, Kelly RG, Rochais F. FGF10 promotes cardiac repair through a dual cellular mechanism increasing cardiomyocyte renewal and inhibiting fibrosis. Cardiovasc Res 2021; 118:2625-2637. [PMID: 34755840 DOI: 10.1093/cvr/cvab340] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS Promoting cardiomyocyte renewal represents a major therapeutic approach for heart regeneration and repair. Our study aims to investigate the relevance of FGF10 as a potential target for heart regeneration. METHODS AND RESULTS Our results first reveal that Fgf10 levels are upregulated in the injured ventricle after MI. Adult mice with reduced Fgf10 expression subjected to MI display impaired cardiomyocyte proliferation and enhanced cardiac fibrosis, leading to a worsened cardiac function and remodeling post-MI. In contrast, conditional Fgf10 overexpression post-MI revealed that, by enhancing cardiomyocyte proliferation and preventing scar-promoting myofibroblast activation, FGF10 preserves cardiac remodeling and function. Moreover, FGF10 activates major regenerative pathways including the regulation of Meis1 expression levels, the Hippo signaling pathway and a pro-glycolytic metabolic switch. Finally, we demonstrate that elevated FGF10 levels in failing human hearts correlate with reduced fibrosis and enhanced cardiomyocyte proliferation. CONCLUSIONS Altogether, our study shows that FGF10 promotes cardiac regeneration and repair through two cellular mechanisms: elevating cardiomyocyte renewal and limiting fibrosis. This study thus identifies FGF10 as a clinically relevant target for heart regeneration and repair in man.
Collapse
Affiliation(s)
- Fabien Hubert
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Sandy M Payan
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Edeline Pelce
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France.,Department of Cardiac Surgery, Timone Hospital, AP-HM, Marseille, France
| | | | - Rachel Sturny
- Aix Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | | - Giovanna Mottola
- Aix-Marseille Univ, C2VN, INSERM 1263, INRAE 1260, Marseille, France.,Laboratory of Biochemistry, Timone Hospital, Marseille, France
| | - Frédéric Collart
- Department of Cardiac Surgery, Timone Hospital, AP-HM, Marseille, France
| | - Robert G Kelly
- Aix Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | |
Collapse
|
19
|
Advanced maternal age perturbs mouse embryo development and alters the phenotype of derived embryonic stem cells. J Dev Orig Health Dis 2021; 13:395-405. [PMID: 34193331 DOI: 10.1017/s2040174421000325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Advanced maternal age (AMA) is known to reduce fertility, increases aneuploidy in oocytes and early embryos and leads to adverse developmental consequences which may associate with offspring lifetime health risks. However, investigating underlying effects of AMA on embryo developmental potential is confounded by the inherent senescence present in maternal body systems further affecting reproductive success. Here, we describe a new model for the analysis of early developmental mechanisms underlying AMA by the derivation and characterisation of mouse embryonic stem cell (mESC-like) lines from naturally conceived embryos. Young (7-8 weeks) and Old (7-8 months) C57BL/6 female mice were mated with young males. Preimplantation embryos from Old dams displayed developmental retardation in blastocyst morphogenesis. mESC lines established from these blastocysts using conventional techniques revealed differences in genetic, cellular and molecular criteria conserved over several passages in the standardised medium. mESCs from embryos from AMA dams displayed increased incidence of aneuploidy following Giemsa karyotyping compared with those from Young dams. Moreover, AMA caused an altered pattern of expression of pluripotency markers (Sox2, OCT4) in mESCs. AMA further diminished mESC survival and proliferation and reduced the expression of cell proliferation marker, Ki-67. These changes coincided with altered expression of the epigenetic marker, Dnmt3a and other developmental regulators in a sex-dependent manner. Collectively, our data demonstrate the feasibility to utilise mESCs to reveal developmental mechanisms underlying AMA in the absence of maternal senescence and with reduced animal use.
Collapse
|
20
|
Khosravi F, Ahmadvand N, Bellusci S, Sauer H. The Multifunctional Contribution of FGF Signaling to Cardiac Development, Homeostasis, Disease and Repair. Front Cell Dev Biol 2021; 9:672935. [PMID: 34095143 PMCID: PMC8169986 DOI: 10.3389/fcell.2021.672935] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The current focus on cardiovascular research reflects society’s concerns regarding the alarming incidence of cardiac-related diseases and mortality in the industrialized world and, notably, an urgent need to combat them by more efficient therapies. To pursue these therapeutic approaches, a comprehensive understanding of the mechanism of action for multifunctional fibroblast growth factor (FGF) signaling in the biology of the heart is a matter of high importance. The roles of FGFs in heart development range from outflow tract formation to the proliferation of cardiomyocytes and the formation of heart chambers. In the context of cardiac regeneration, FGFs 1, 2, 9, 16, 19, and 21 mediate adaptive responses including restoration of cardiac contracting rate after myocardial infarction and reduction of myocardial infarct size. However, cardiac complications in human diseases are correlated with pathogenic effects of FGF ligands and/or FGF signaling impairment. FGFs 2 and 23 are involved in maladaptive responses such as cardiac hypertrophic, fibrotic responses and heart failure. Among FGFs with known causative (FGFs 2, 21, and 23) or protective (FGFs 2, 15/19, 16, and 21) roles in cardiac diseases, FGFs 15/19, 21, and 23 display diagnostic potential. The effective role of FGFs on the induction of progenitor stem cells to cardiac cells during development has been employed to boost the limited capacity of postnatal cardiac repair. To renew or replenish damaged cardiomyocytes, FGFs 1, 2, 10, and 16 were tested in (induced-) pluripotent stem cell-based approaches and for stimulation of cell cycle re-entry in adult cardiomyocytes. This review will shed light on the wide range of beneficiary and detrimental actions mediated by FGF ligands and their receptors in the heart, which may open new therapeutic avenues for ameliorating cardiac complications.
Collapse
Affiliation(s)
- Farhad Khosravi
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Negah Ahmadvand
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
21
|
Abstract
Branching morphogenesis generates epithelial trees which facilitate gas exchange, filtering, as well as secretion processes with their large surface to volume ratio. In this review, we focus on the developmental mechanisms that control the early stages of lung branching morphogenesis. Lung branching morphogenesis involves the stereotypic, recurrent definition of new branch points, subsequent epithelial budding, and lung tube elongation. We discuss current models and experimental evidence for each of these steps. Finally, we discuss the role of the mesenchyme in determining the organ-specific shape.
Collapse
Affiliation(s)
- Dagmar Iber
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Basel, Switzerland; Swiss Institute of Bioinformatics (SIB), Basel, Switzerland.
| |
Collapse
|
22
|
Peng J, Wang Q, Meng Z, Wang J, Zhou Y, Zhou S, Song W, Chen S, Chen AF, Sun K. A loss-of-function mutation p.T256M in NDRG4 is implicated in the pathogenesis of pulmonary atresia with ventricular septal defect (PA/VSD) and tetralogy of Fallot (TOF). FEBS Open Bio 2021; 11:375-385. [PMID: 33211401 PMCID: PMC7876499 DOI: 10.1002/2211-5463.13044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/10/2020] [Accepted: 11/17/2020] [Indexed: 11/10/2022] Open
Abstract
Pulmonary atresia with ventricular septal defect (PA/VSD) is a rare congenital heart disease (CHD) characterized by a lack of luminal continuity and blood flow from either the right ventricle or the pulmonary artery, together with VSDs. The prevalence of PA/VSD is about 0.2% of live births and approximately 2% of CHDs. PA/VSD is similar to tetralogy of Fallot (TOF) in terms of structural and pathological characteristics. The pathogenesis of these two CHDs remains incompletely understood. It was previously reported that N‐myc downstream‐regulated gene (NDRG)4 is required for myocyte proliferation during early cardiac development. In the present study, we enrolled 80 unrelated patients with PA/VSD or TOF and identified a probably damaging variant p.T256M of NDRG4. The p.T256M variant impaired the proliferation ability of human cardiac myocytes (hCM). Furthermore, the p.T256M variant resulted in G1 and G2 arrest of hCM, followed by an increase in p27 and caspase‐9 expression. Our results provide evidence that the p.T256M variant in NDRG4 is a pathogenic variant associated with impaired hCM proliferation and cell‐cycle arrest and likely contributes towards the pathogenesis of PA/VSD and TOF.
Collapse
Affiliation(s)
- Jiayu Peng
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingjie Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuo Meng
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Zhou
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuang Zhou
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenting Song
- Department of Pediatric Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alex F Chen
- Institute of Cardiovascular Development and Regeneration, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Yan Y, Liu F, Hou X, Wan J, Xiong Q, Zhou R, Liao B. [Joint regulation of Wnt and bone morphogenetic protein signaling pathways to promote differentiation of human induced pluripotent stem cells into cardiomyocytes]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:1313-1321. [PMID: 33063499 DOI: 10.7507/1002-1892.201912087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To explore the role of joint regulation of Wnt and bone morphogenetic protein (BMP) signaling pathways in the differentiation of human induced pluripotent stem cells (hiPSCs) into cardiomyocytes. Methods HiPSCs were cultured and observed under inverted phase contrast microscope. Immunofluorescence staining was used to observe the expressions of hiPSCs pluripotent markers (OCT3/4, NANOG, and TRA-1-60). HiPSCs were passaged which were taken for subsequent experiments within the 35th passage. When the fusion degree of hiPSCs was close to 100%, the CHIR99021 (Wnt pathway activator) was added on the 0th day of differentiation. Different concentrations of IWP4 (inhibitor of Wnt production) were added on the 3rd day of differentiation, and the best concentration of IWP4 was added at different time points. The optimal concentration and the best effective period of IWP4 were obtained by detecting the expression of troponin T (TNNT2) mRNA by real-time fluorescence quantitative PCR. Then, on the basis of adding CHIR99021 and IWP4, different concentrations of BMP-4 were added on the 5th day of differentiation, and the best concentration of BMP-4 was added at different time points. The optimal concentration and best effective period of BMP-4 were obtained by detecting the expression of TNNT2 mRNA. Finally, hiPSCs were divided into three groups: Wnt group, BMP group, and Wnt+BMP group. On the basis of adding CHIR99021 on the 0th day of differentiation, IWP4, BMP-4, and IWP4+BMP-4 were added into Wnt group, BMP group, and Wnt+BMP group respectively according to the screening results. Cells were collected on the 7th and the 15th days of differentiation. The expressions of myocardial precursor cell markers [ISL LIM homeobox 1 (ISL1), NK2 homeobox 5 (NKX2-5)] and cardiomyocyte specific markers [myocyte enhancer factor 2C (MEF2C), myosin light chain 2 (MYL2), MYL7, and TNNT2] were detected by real-time fluorescent quantitative PCR. Cells were collected on the 28th day of differentiation, and the expression of cardiac troponin T (cTnT) was detected by flow cytometry and immunofluorescence staining. Results The results of cell mophology and immunoflurescence staining showed that the OCT3/4, NANOG, and TRA-1-60 were highly expressed in hiPSCs, which suggested that hiPSCs had characteristics of pluripotency. The optimal concentration of IWP4 was 10.0 μmol/L ( P<0.05) and the best effective period was the 3rd day ( P<0.05) in inducing hiPSCs to differentiate into cardiomyocytes. The optimal concentration of BMP-4 was 20.0 ng/mL ( P<0.05) and the best effective period was the 3rd day ( P<0.05). The relative expressions of ISL1, NKX2-5, MEF2C, MYL2, MYL7, and TNNT2 mRNAs, the positive expression ratio of cTnT detected by flow cytometry, and sarcomere structure detected by immunofluorescence staining of Wnt+BMP group were superior to those of Wnt group ( P<0.05). Conclusion Joint regulation of Wnt and BMP signaling pathways can improve the differentiation efficiency of hiPSCs into cardiomyocytes.
Collapse
Affiliation(s)
- Ying Yan
- Clinical Medicine College of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China;College of Integrative Chinese and Western Medicine, Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Feng Liu
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Xiaojie Hou
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Juyi Wan
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Qi Xiong
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Rui Zhou
- Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Bin Liao
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| |
Collapse
|
24
|
Liu L, Song C, Li J, Wang Q, Zhu M, Hu Y, Chen J, Chen C, Zhang JS, Dong N, Chen C. Fibroblast growth factor 10 alleviates particulate matter-induced lung injury by inhibiting the HMGB1-TLR4 pathway. Aging (Albany NY) 2020; 12:1186-1200. [PMID: 31958320 PMCID: PMC7053597 DOI: 10.18632/aging.102676] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022]
Abstract
Exposure to particulate matter (PM) is associated with increased incidence of respiratory diseases. The present study aimed to investigate the roles of fibroblast growth factor 10 (FGF10) in PM-induced lung injury. Mice were intratracheally instilled with FGF10 or phosphate-buffered saline at one hour before instillation of PM for two consecutive days. In addition, the anti-inflammatory impact of FGF10 in vitro and its effect on the high-mobility group box 1 (HMGB1)-toll-like receptor 4 (TLR4) pathway was investigated. It was found that PM exposure is associated with increased inflammatory cell infiltration into the lung and increased vascular protein leakage, while FGF10 pretreatment attenuated both of these effects. FGF10 also decreased the PM-induced expression of interleukin (IL)-6, IL-8, tumor necrosis factor-α and HMGB1 in murine bronchoalveolar lavage fluid and in the supernatants of human bronchial epithelial cells exposed to PM. FGF10 exerted anti-inflammatory and cytoprotective effects by inhibiting the HMGB1-TLR4 pathway. These results indicate that FGF10 may have therapeutic values for PM-induced lung injury.
Collapse
Affiliation(s)
- Lingjing Liu
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Chenjian Song
- Department of Pulmonary Medicine, Yiwu Central Hospital, Yiwu 322000, China
| | - Jingli Li
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Qiang Wang
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Mingyang Zhu
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Yiran Hu
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Junjie Chen
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Chaolei Chen
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Jin-San Zhang
- Department of Pharmacy, Wenzhou Medical University Pharmacy School, Wenzhou 325006, China
| | - Nian Dong
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| | - Chengshui Chen
- Department of Pulmonary Medicine, Wenzhou Medical University First Affiliated Hospital, Wenzhou 325006, China
| |
Collapse
|
25
|
Liu L, Xia Z, Li J, Hu Y, Wang Q, Chen J, Fan S, Wu J, Dong N, Chen C. Fibroblast growth factor 10 protects against particulate matter-induced airway inflammatory response through regulating inflammatory signaling and apoptosis. Am J Transl Res 2019; 11:6977-6988. [PMID: 31814901 PMCID: PMC6895507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
Chronic respiratory disorders are some of the most frequent and severe chronic diseases in China. Epidemiological research has shown that particulate matter (PM) is a risk factor and is closely correlated to the progression of numerous respiratory diseases. Fibroblast growth factor 10 (FGF10) is a mesenchymal-epithelial signaling messenger essential for the development and environmental stability of several tissues. Nevertheless, its role in PM-induced airway inflammation remains unclear. The present study aimed to explore the mechanisms underlying the FGF10-related slowing of lung injury and inflammation in vivo and in vitro, as well as the therapeutic potential of these phenomena. Mice were intraperitoneally injected with a vehicle (PBS) or FGF10 (0.5 mg/kg) at one hour before intratracheal treatment with vehicle (PBS) or PM (4 mg/kg) for two consecutive days. Human airway epithelial BEAS-2B cells were exposed to a vehicle (PBS) or FGF10 (10 ng/ml) in vitro at one hour prior to incubation with a vehicle or PM (200 ug/ml) for 24 hours. Then, the impact on inflammatory molecules was investigated. In vivo, it was found that FGF10 diminished the inflammatory cell aggregation and reduced the apoptosis. Interestingly, in the PM group, the level of FGF10 increased in the bronchoalveolar lavage fluid (BALF). However, the pre-treatment with FGF10 markedly impaired the PM-induced increase in IL-6, IL-8, TNF-α and PGE2 levels in BALF and the cell supernatant. In conclusion, the present findings indicate that FGF10 attenuates PM-induced airway inflammation by inhibiting apoptosis and inflammation. This may be exploited for the prevention and management of PM-induced airway inflammation.
Collapse
Affiliation(s)
- Lingjing Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Ziqiang Xia
- Department of Gastroenterology, First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Jingli Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Yiran Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Qiang Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Junjie Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Shiqian Fan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Jinming Wu
- Department of Gastroenterology, First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Nian Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| | - Chengshui Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325006, China
| |
Collapse
|