1
|
Read DF, Booth GT, Daza RM, Jackson DL, Gladden RG, Srivatsan SR, Ewing B, Franks JM, Spurrell CH, Gomes AR, O'Day D, Gogate AA, Martin BK, Larson H, Pfleger C, Starita L, Lin Y, Shendure J, Lin S, Trapnell C. Single-cell analysis of chromatin and expression reveals age- and sex-associated alterations in the human heart. Commun Biol 2024; 7:1052. [PMID: 39187646 PMCID: PMC11347658 DOI: 10.1038/s42003-024-06582-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: 05/30/2022] [Accepted: 07/11/2024] [Indexed: 08/28/2024] Open
Abstract
Sex differences and age-related changes in the human heart at the tissue, cell, and molecular level have been well-documented and many may be relevant for cardiovascular disease. However, how molecular programs within individual cell types vary across individuals by age and sex remains poorly characterized. To better understand this variation, we performed single-nucleus combinatorial indexing (sci) ATAC- and RNA-Seq in human heart samples from nine donors. We identify hundreds of differentially expressed genes by age and sex and find epigenetic signatures of variation in ATAC-Seq data in this discovery cohort. We then scale up our single-cell RNA-Seq analysis by combining our data with five recently published single nucleus RNA-Seq datasets of healthy adult hearts. We find variation such as metabolic alterations by sex and immune changes by age in differential expression tests, as well as alterations in abundance of cardiomyocytes by sex and neurons with age. In addition, we compare our adult-derived ATAC-Seq profiles to analogous fetal cell types to identify putative developmental-stage-specific regulatory factors. Finally, we train predictive models of cell-type-specific RNA expression levels utilizing ATAC-Seq profiles to link distal regulatory sequences to promoters, quantifying the predictive value of a simple TF-to-expression regulatory grammar and identifying cell-type-specific TFs. Our analysis represents the largest single-cell analysis of cardiac variation by age and sex to date and provides a resource for further study of healthy cardiac variation and transcriptional regulation at single-cell resolution.
Collapse
Affiliation(s)
- David F Read
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Gregory T Booth
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Rula Green Gladden
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sanjay R Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Brent Ewing
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Jennifer M Franks
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | | | - Diana O'Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Aishwarya A Gogate
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Haleigh Larson
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Christian Pfleger
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA, USA
| | - Lea Starita
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Yiing Lin
- Department of Surgery, Washington University, St Louis, MO, USA
| | - Jay Shendure
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Children's Research Institute, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| | - Shin Lin
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA, USA.
| | - Cole Trapnell
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
2
|
Niu Y, Guo D, Wei Y, Li J, Bai Y, Liu Z, Jia X, Chen Z, Li L, Shi B, Zhang X, Zhao Z, Hu J, Wang J, Liu X, Li S. Comparative Transcriptome Analysis of mRNA and miRNA during the Development of Longissimus Dorsi Muscle of Gannan Yak and Tianzhu White Yak. Animals (Basel) 2024; 14:2278. [PMID: 39123804 PMCID: PMC11311108 DOI: 10.3390/ani14152278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The Gannan yak, a superior livestock breed found on the Tibetan Plateau, exhibits significantly enhanced body size, weight, and growth performance in comparison to the Tianzhu white yak. MiRNAs play a pivotal role in regulating muscle growth by negatively modulating target genes. In this study, we found the average diameter, area, and length of myofibers in Gannan yaks were significantly higher than those of Tianzhu white yaks. Further, we focused on analyzing the longissimus dorsi muscle from both Gannan yaks and Tianzhu white yaks through transcriptome sequencing to identify differentially expressed (DE)miRNAs that influence skeletal muscle development. A total of 254 DE miRNAs were identified, of which 126 miRNAs were up-regulated and 128 miRNAs were down-regulated. GO and KEGG enrichment analysis showed that the target genes of these DE miRNAs were significantly enriched in signaling pathways associated with muscle growth and development. By constructing a DE miRNA- DE mRNA interaction network, we screened 18 key miRNAs, and notably, four of the candidates (novel-m0143-3p, novel-m0024-3p, novel-m0128-5p, and novel-m0026-3p) targeted six genes associated with muscle growth and development (DDIT4, ADAMTS1, CRY2, AKIRIN2, SIX1, and FOXO1). These findings may provide theoretical references for further studies on the role of miRNAs in muscle growth and development in Gannan yaks.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.N.); (D.G.); (Y.W.); (J.L.); (Y.B.); (Z.L.); (X.J.); (Z.C.); (L.L.); (B.S.); (X.Z.); (J.H.); (J.W.); (X.L.); (S.L.)
| | | | | | | | | |
Collapse
|
3
|
Rebboah E, Rezaie N, Williams BA, Weimer AK, Shi M, Yang X, Liang HY, Dionne LA, Reese F, Trout D, Jou J, Youngworth I, Reinholdt L, Morabito S, Snyder MP, Wold BJ, Mortazavi A. The ENCODE mouse postnatal developmental time course identifies regulatory programs of cell types and cell states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598567. [PMID: 38915583 PMCID: PMC11195270 DOI: 10.1101/2024.06.12.598567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Postnatal genomic regulation significantly influences tissue and organ maturation but is under-studied relative to existing genomic catalogs of adult tissues or prenatal development in mouse. The ENCODE4 consortium generated the first comprehensive single-nucleus resource of postnatal regulatory events across a diverse set of mouse tissues. The collection spans seven postnatal time points, mirroring human development from childhood to adulthood, and encompasses five core tissues. We identified 30 cell types, further subdivided into 69 subtypes and cell states across adrenal gland, left cerebral cortex, hippocampus, heart, and gastrocnemius muscle. Our annotations cover both known and novel cell differentiation dynamics ranging from early hippocampal neurogenesis to a new sex-specific adrenal gland population during puberty. We used an ensemble Latent Dirichlet Allocation strategy with a curated vocabulary of 2,701 regulatory genes to identify regulatory "topics," each of which is a gene vector, linked to cell type differentiation, subtype specialization, and transitions between cell states. We find recurrent regulatory topics in tissue-resident macrophages, neural cell types, endothelial cells across multiple tissues, and cycling cells of the adrenal gland and heart. Cell-type-specific topics are enriched in transcription factors and microRNA host genes, while chromatin regulators dominate mitosis topics. Corresponding chromatin accessibility data reveal dynamic and sex-specific regulatory elements, with enriched motifs matching transcription factors in regulatory topics. Together, these analyses identify both tissue-specific and common regulatory programs in postnatal development across multiple tissues through the lens of the factors regulating transcription.
Collapse
Affiliation(s)
- Elisabeth Rebboah
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Narges Rezaie
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Brian A. Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Annika K. Weimer
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Minyi Shi
- Department of Next Generation Sequencing and Microchemistry, Proteomics and Lipidomics, Genentech, San Francisco, USA
| | - Xinqiong Yang
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Heidi Yahan Liang
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | | | - Fairlie Reese
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | - Diane Trout
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Jennifer Jou
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Ingrid Youngworth
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | | | - Samuel Morabito
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Barbara J. Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Ali Mortazavi
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| |
Collapse
|
4
|
Alhusaini AM, Alghibiwi HK, Sarawi WS, Alsaab JS, Alshehri SM, Alqahtani QH, Alshanwani AR, Aljassas EA, Alsultan EN, Hasan IH. Resveratrol-Based Liposomes Improve Cardiac Remodeling Induced by Isoproterenol Partially by Modulating MEF2, Cytochrome C and S100A1 Expression. Dose Response 2024; 22:15593258241247980. [PMID: 38645382 PMCID: PMC11027597 DOI: 10.1177/15593258241247980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/28/2024] [Indexed: 04/23/2024] Open
Abstract
Isoproterenol (ISO), a chemically synthesized catecholamine, belongs to β-adrenoceptor agonist used to treat bradycardia. The β-adrenergic agonist is an essential regulator of myocardial metabolism and contractility; however, excessive exposure to ISO can initiate oxidative stress and inflammation. This study aims to investigate the molecular mechanisms underlying ISO-induced cardiac remodeling, the protective efficacy of resveratrol (RSVR), and its liposomal formulation (L-RSVR) against such cardiac change. Wistar albino rats were evenly divided into 4 groups. Control group, ISO group received ISO (50 mg/kg, s.c.) twice a week for 2 weeks, and RSVR- and L-RSVR-treated groups in which rats received either RSVR or L-RSVR (20 mg/kg/day, p.o.) along with ISO for 2 weeks. ISO caused a significant elevation of the expression levels of BAX and MEF2 mRNA, S100A1 and cytochrome C proteins, as well as DNA fragmentation in cardiac tissue compared to the control group. Treatment with either RSVR or L-RSVR for 14 days significantly ameliorated the damage induced by ISO, as evidenced by the improvement of all measured parameters. The present study shows that L-RSVR provides better cardio-protection against ISO-induced cardiac injury in rats, most likely through modulation of cardiac S100A1 protein expression and inhibition of inflammation and apoptosis.
Collapse
Affiliation(s)
- Ahlam M. Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hanan K. Alghibiwi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wedad S. Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Juman S. Alsaab
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Samiyah M. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Qamraa H. Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aliah R. Alshanwani
- Department of Physiology, College of Medicine and King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia
| | - Ebtesam A. Aljassas
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ebtesam N. Alsultan
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Iman H. Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Chea S, Kreger J, Lopez-Burks ME, MacLean AL, Lander AD, Calof AL. Gastrulation-stage gene expression in Nipbl+/- mouse embryos foreshadows the development of syndromic birth defects. SCIENCE ADVANCES 2024; 10:eadl4239. [PMID: 38507484 PMCID: PMC10954218 DOI: 10.1126/sciadv.adl4239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024]
Abstract
In animal models, Nipbl deficiency phenocopies gene expression changes and birth defects seen in Cornelia de Lange syndrome, the most common cause of which is Nipbl haploinsufficiency. Previous studies in Nipbl+/- mice suggested that heart development is abnormal as soon as cardiogenic tissue is formed. To investigate this, we performed single-cell RNA sequencing on wild-type and Nipbl+/- mouse embryos at gastrulation and early cardiac crescent stages. Nipbl+/- embryos had fewer mesoderm cells than wild-type and altered proportions of mesodermal cell subpopulations. These findings were associated with underexpression of genes implicated in driving specific mesodermal lineages. In addition, Nanog was found to be overexpressed in all germ layers, and many gene expression changes observed in Nipbl+/- embryos could be attributed to Nanog overexpression. These findings establish a link between Nipbl deficiency, Nanog overexpression, and gene expression dysregulation/lineage misallocation, which ultimately manifest as birth defects in Nipbl+/- animals and Cornelia de Lange syndrome.
Collapse
Affiliation(s)
- Stephenson Chea
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Jesse Kreger
- Department of Quantitative and Computational Biology, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Martha E. Lopez-Burks
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Arthur D. Lander
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
| | - Anne L. Calof
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA 92697, USA
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
6
|
Wei Y, Guo D, Bai Y, Liu Z, Li J, Chen Z, Shi B, Zhao Z, Hu J, Han X, Wang J, Liu X, Li S, Zhao F. Transcriptome Analysis of mRNA and lncRNA Related to Muscle Growth and Development in Gannan Yak and Jeryak. Int J Mol Sci 2023; 24:16991. [PMID: 38069312 PMCID: PMC10707067 DOI: 10.3390/ijms242316991] [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: 10/23/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
The production performance of Jeryak, resulting from the F1 generation of the cross between Gannan yak and Jersey cattle, exhibits a significantly superior outcome compared with that of Gannan yak. Therefore, we used an RNA-seq approach to identify differentially expressed mRNAs (DEMs) and differentially expressed lncRNAs (DELs) influencing muscle growth and development in Gannan yaks and Jeryaks. A total of 304 differentially expressed lncRNAs and 1819 differentially expressed mRNAs were identified based on the screening criteria of |log 2 FC| > 1 and FDR < 0.05. Among these, 132 lncRNAs and 1081 mRNAs were found to be down-regulated, while 172 lncRNAs and 738 mRNAs were up-regulated. GO and KEGG analyses showed that the identified DELs and DEMs were enriched in the entries of pathways associated with muscle growth and development. On this basis, we constructed an lncRNA-mRNA interaction network. Interestingly, two candidate DELs (MSTRG.16260.9 and MSTRG.22127.1) had targeting relationships with 16 (MYC, IGFBP5, IGFBP2, MYH4, FGF6, etc.) genes related to muscle growth and development. These results could provide a basis for further studies on the roles of lncRNAs and mRNAs in muscle growth in Gannan yaks and Jeryak breeds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.W.); (D.G.); (B.S.)
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.W.); (D.G.); (B.S.)
| | | | | | | | | | | |
Collapse
|
7
|
Kukshal P, Joshi RO, Kumar A, Ahamad S, Murthy PR, Sathe Y, Manohar K, Guhathakurta S, Chellappan S. Case-control association study of congenital heart disease from a tertiary paediatric cardiac centre from North India. BMC Pediatr 2023; 23:290. [PMID: 37322441 PMCID: PMC10268439 DOI: 10.1186/s12887-023-04095-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/27/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Congenital Heart diseases (CHDs) account for 1/3rd of all congenital birth defects. Etiopathogenesis of CHDs remain elusive despite extensive investigations globally. Phenotypic heterogeneity witnessed in this developmental disorder reiterate gene-environment interactions with periconceptional factors as risk conferring; and genetic analysis of both sporadic and familial forms of CHD suggest its multigenic basis. Significant association of de novo and inherited variants have been observed. Approximately 1/5th of CHDs are documented in the ethnically distinct Indian population but genetic insights have been very limited. This pilot case-control based association study was undertaken to investigate the status of Caucasian SNPs in a north Indian cohort. METHOD A total of 306 CHD cases sub-classified into n = 198 acyanotic and n = 108 cyanotic types were recruited from a dedicated tertiary paediatric cardiac centre in Palwal, Haryana. 23 SNPs primarily prioritized from Genome-wide association studies (GWAS) on Caucasians were genotyped using Agena MassARRAY Technology and test of association was performed with adequately numbered controls. RESULTS Fifty percent of the studied SNPs were substantially associated in either allelic, genotypic or sub-phenotype categories validating their strong correlation with disease manifestation. Of note, strongest allelic association was observed for rs73118372 in CRELD1 (p < 0.0001) on Chr3, rs28711516 in MYH6 (p = 0.00083) and rs735712 in MYH7 (p = 0.0009) both on Chr 14 and were also significantly associated with acyanotic, and cyanotic categories separately. rs28711516 (p = 0.003) and rs735712 (p = 0.002) also showed genotypic association. Strongest association was observed with rs735712(p = 0.003) in VSD and maximum association was observed for ASD sub-phenotypes. CONCLUSIONS Caucasian findings were partly replicated in the north Indian population. The findings suggest the contribution of genetic, environmental and sociodemographic factors, warranting continued investigations in this study population.
Collapse
Affiliation(s)
- Prachi Kukshal
- Sri Sathya Sai Sanjeevani Research Foundation, NH-2, Delhi-Mathura Highway, Baghola, Haryana, District Palwal, Pin- 121102, India.
| | - Radha O Joshi
- Present address Sri Sathya Sai Sanjeevani Research Foundation, Kharghar, Navi Mumbai- 410210, Maharashtra, India
| | - Ajay Kumar
- Sri Sathya Sai Sanjeevani Research Foundation, NH-2, Delhi-Mathura Highway, Baghola, Haryana, District Palwal, Pin- 121102, India
| | - Shadab Ahamad
- Sri Sathya Sai Sanjeevani Research Foundation, NH-2, Delhi-Mathura Highway, Baghola, Haryana, District Palwal, Pin- 121102, India
| | - Prabhatha Rashmi Murthy
- Sri Sathya Sai Sanjeevani Centre for Child Heart Care and Training in Paediatric Cardiac Skills, Navi Mumbai Maharashtra, India
| | - Yogesh Sathe
- Sri Sathya Sai Sanjeevani International Centre for Child Heart Care & Research, NH-2, Delhi-Mathura Highway, Baghola, District Palwal, Haryana, Pin 121102, India
| | - Krishna Manohar
- Sri Sathya Sai Sanjeevani International Centre for Child Heart Care & Research, NH-2, Delhi-Mathura Highway, Baghola, District Palwal, Haryana, Pin 121102, India
| | - Soma Guhathakurta
- Sri Sathya Sai Sanjeevani Research Foundation, NH-2, Delhi-Mathura Highway, Baghola, Haryana, District Palwal, Pin- 121102, India
| | - Subramanian Chellappan
- Sri Sathya Sai Sanjeevani International Centre for Child Heart Care & Research, NH-2, Delhi-Mathura Highway, Baghola, District Palwal, Haryana, Pin 121102, India.
| |
Collapse
|
8
|
DiLorenzo MP, Grosse-Wortmann L. Myocardial Fibrosis in Congenital Heart Disease and the Role of MRI. Radiol Cardiothorac Imaging 2023; 5:e220255. [PMID: 37404787 PMCID: PMC10316299 DOI: 10.1148/ryct.220255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 07/06/2023]
Abstract
Progress in the field of congenital heart surgery over the last century can only be described as revolutionary. Recent improvements in patient outcomes have been achieved through refinements in perioperative care. In the current and future eras, the preservation and restoration of myocardial health, beginning with the monitoring of tissue remodeling, will be central to improving cardiac outcomes. Visualization and quantification of fibrotic myocardial remodeling is one of the greatest assets that cardiac MRI brings to the field of cardiology, and its clinical use within the field of congenital heart disease (CHD) has been an area of particular interest in the last few decades. This review summarizes the physical underpinnings of myocardial tissue characterization in CHD, with an emphasis on T1 parametric mapping and late gadolinium enhancement. It describes methods and suggestions for obtaining images, extracting quantitative and qualitative data, and interpreting the results for children and adults with CHD. The tissue characterization observed in different lesions is used to examine the causes and pathomechanisms of fibrotic remodeling in this population. Similarly, the clinical consequences of elevated imaging biomarkers of fibrosis on patient health and outcomes are explored. Keywords: Pediatrics, MR Imaging, Cardiac, Heart, Congenital, Tissue Characterization, Congenital Heart Disease, Cardiac MRI, Parametric Mapping, Fibrosis, Late Gadolinium Enhancement © RSNA, 2023.
Collapse
|
9
|
Pereira CH, Bare DJ, Rosas PC, Dias FAL, Banach K. The role of P21-activated kinase (Pak1) in sinus node function. J Mol Cell Cardiol 2023; 179:90-101. [PMID: 37086972 PMCID: PMC10294268 DOI: 10.1016/j.yjmcc.2023.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/24/2023]
Abstract
Sinoatrial node (SAN) dysfunction (SND) and atrial arrhythmia frequently occur simultaneously with a hazard ratio of 4.2 for new onset atrial fibrillation (AF) in SND patients. In the atrial muscle attenuated activity of p21-activated kinase 1 (Pak1) increases the risk for AF by enhancing NADPH oxidase 2 dependent production of reactive oxygen species (ROS). However, the role of Pak1 dependent ROS regulation in SAN function has not yet been determined. We hypothesize that Pak1 activity maintains SAN activity by regulating the expression of the hyperpolarization activated cyclic nucleotide gated cation channel (HCN). To determine Pak1 dependent changes in heart rate (HR) regulation we quantified the intrinsic sinus rhythm in wild type (WT) and Pak1 deficient (Pak1-/-) mice of both sexes in vivo and in isolated Langendorff perfused hearts. Pak1-/- hearts displayed an attenuated HR in vivo after autonomic blockage and in isolated hearts. The contribution of the Ca2+ clock to pacemaker activity remained unchanged, but Ivabradine (3 μM), a blocker of HCN channels that are a membrane clock component, eliminated the differences in SAN activity between WT and Pak1-/- hearts. Reduced HCN4 expression was confirmed in Pak1-/- right atria. The reduced HCN activity in Pak1-/- could be rescued by class II HDAC inhibition (LMK235), ROS scavenging (TEMPOL) or attenuation of Extracellular Signal-Regulated Kinase (ERK) 1/2 activity (SCH772984). No sex specific differences in Pak1 dependent SAN regulation were determined. Our results establish Pak1 as a class II HDAC regulator and a potential therapeutic target to attenuate SAN bradycardia and AF susceptibility.
Collapse
Affiliation(s)
- Carlos H Pereira
- Dept. of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA; Biological Science Center, Department of Physiology, Av. Cel Francisco H. dos Santos 100, 19031 Centro Politécnico-Curitiba, Brazil.
| | - Dan J Bare
- Dept. of Physiology & Biophysics, The Ohio State University, 5018 Graves Hall, 333 W.10th Ave., Columbus, OH 4321, USA.
| | - Paola C Rosas
- Dept. of Pharmacy Practice, College of Pharmacy, 833 S Wood St., Chicago, IL 60612, USA.
| | - Fernando A L Dias
- Biological Science Center, Department of Physiology, Av. Cel Francisco H. dos Santos 100, 19031 Centro Politécnico-Curitiba, Brazil.
| | - Kathrin Banach
- Dept. of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA.
| |
Collapse
|
10
|
Mohl W, Kiseleva Z, Jusic A, Bruckner M, Mader RM. Signs and signals limiting myocardial damage using PICSO: a scoping review decoding paradigm shifts toward a new encounter. Front Cardiovasc Med 2023; 10:1030842. [PMID: 37229230 PMCID: PMC10204926 DOI: 10.3389/fcvm.2023.1030842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 04/14/2023] [Indexed: 05/27/2023] Open
Abstract
Background Inducing recovery in myocardial ischemia is limited to a timely reopening of infarct vessels and clearing the cardiac microcirculation, but additional molecular factors may impact recovery. Objective In this scoping review, we identify the paradigm shifts decoding the branching points of experimental and clinical evidence of pressure-controlled intermittent coronary sinus occlusion (PICSO), focusing on myocardial salvage and molecular implications on infarct healing and repair. Design The reporting of evidence was structured chronologically, describing the evolution of the concept from mainstream research to core findings dictating a paradigm change. All data reported in this scoping review are based on published data, but new evaluations are also included. Results Previous findings relate hemodynamic PICSO effects clearing reperfused microcirculation to myocardial salvage. The activation of venous endothelium opened a new avenue for understanding PICSO. A flow-sensitive signaling molecule, miR-145-5p, showed a five-fold increase in porcine myocardium subjected to PICSO.Verifying our theory of "embryonic recall," an upregulation of miR-19b and miR-101 significantly correlates to the time of pressure increase in cardiac veins during PICSO (r2 = 0.90, p < 0.05; r2 = 0.98, p < 0.03), suggesting a flow- and pressure-dependent secretion of signaling molecules into the coronary circulation. Furthermore, cardiomyocyte proliferation by miR-19b and the protective role of miR-101 against remodeling show another potential interaction of PICSO in myocardial healing. Conclusion Molecular signaling during PICSO may contribute to retroperfusion toward deprived myocardium and clearing the reperfused cardiac microcirculation. A burst of specific miRNA reiterating embryonic molecular pathways may play a role in targeting myocardial jeopardy and will be an essential therapeutic contribution in limiting infarcts in recovering patients.
Collapse
Affiliation(s)
- Werner Mohl
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Zlata Kiseleva
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Alem Jusic
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Matthäus Bruckner
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Robert M. Mader
- Department of Medicine I, Comprehensive Cancer Center of the Medical University of Vienna, Vienna,Austria
| |
Collapse
|
11
|
Agarwal M, Sharma A, Kagoo R A, Bhargava A. Interactions between genes altered during cardiotoxicity and neurotoxicity in zebrafish revealed using induced network modules analysis. Sci Rep 2023; 13:6257. [PMID: 37069190 PMCID: PMC10110561 DOI: 10.1038/s41598-023-33145-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
As the manufacturing and development of new synthetic compounds increase to keep pace with the expanding global demand, adverse health effects due to these compounds are emerging as critical public health concerns. Zebrafish have become a prominent model organism to study toxicology due to their genomic similarity to humans, optical clarity, well-defined developmental stages, short generation time, and cost-effective maintenance. It also provides a shorter time frame for in vivo toxicology evaluation compared to the mammalian experimental systems. Here, we used meta-analysis to examine the alteration in genes during cardiotoxicity and neurotoxicity in zebrafish, caused by chemical exposure of any kind. First, we searched the literature comprehensively for genes that are altered during neurotoxicity and cardiotoxicity followed by meta-analysis using ConsensusPathDB. Since constant communication between the heart and the brain is an important physiological phenomenon, we also analyzed interactions among genes altered simultaneously during cardiotoxicity and neurotoxicity using induced network modules analysis in ConsensusPathDB. We observed inflammation and regeneration as the major pathways involved in cardiotoxicity and neurotoxicity. A large number of intermediate genes and input genes anchored in these pathways are molecular regulators of cell cycle progression and cell death and are implicated in tumor manifestation. We propose potential predictive biomarkers for neurotoxicity and cardiotoxicity and the major pathways potentially implicated in the manifestation of a particular toxicity phenotype.
Collapse
Affiliation(s)
- Manusmriti Agarwal
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India
| | - Ankush Sharma
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India
| | - Andrea Kagoo R
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India
| | - Anamika Bhargava
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India.
| |
Collapse
|
12
|
Zhang J, Ouyang Z, Xia L, Wang Q, Zheng F, Xu K, Xing Y, Wei K, Shi S, Li C, Yang J. Dynamic chromatin landscape encodes programs for perinatal transition of cardiomyocytes. Cell Death Dis 2023; 9:11. [PMID: 36653336 PMCID: PMC9849264 DOI: 10.1038/s41420-023-01322-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
The perinatal period occurring immediately before and after birth is critical for cardiomyocytes because they must change rapidly to accommodate the switch from fetal to neonatal circulation after birth. This transition is a well-orchestrated process, and any perturbation leads to unhealthy cardiomyocytes and heart disease. Despite its importance, little is known about how this transition is regulated and controlled. Here, by mapping the genome-wide chromatin accessibility, transcription-centered long-range chromatin interactions and gene expression in cardiomyocytes undergoing perinatal transition, we discovered two key transcription factors, MEF2 and AP1, that are crucial for driving the phenotypic changes within the perinatal window. Thousands of dynamic regulatory elements were found in perinatal cardiomyocytes and we show these elements mediated the transcriptional reprogramming through an elegant chromatin high-order architecture. We recompiled transcriptional program of induced stem cell-derived cardiomyocytes according to our discovered network, and they showed adult cardiomyocyte-like electrophysiological expression. Our work provides a comprehensive regulatory resource of cardiomyocytes perinatal reprogramming, and aids the gap-filling of cardiac translational research.
Collapse
Affiliation(s)
- Jing Zhang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Zhaohui Ouyang
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 200092 Shanghai, China
| | - Limei Xia
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Qi Wang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Feng Zheng
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Kun Xu
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Yuexian Xing
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Ke Wei
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 200092 Shanghai, China
| | - Shaolin Shi
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Chaojun Li
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine and China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Gusu School, Nanjing Medical University, 211166 Nanjing, China
| | - Jingping Yang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| |
Collapse
|
13
|
Morita T, Hayashi K. Actin-related protein 5 suppresses the cooperative activation of cardiac gene transcription by myocardin and MEF2. FEBS Open Bio 2023; 13:363-379. [PMID: 36610028 PMCID: PMC9900090 DOI: 10.1002/2211-5463.13549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/15/2022] [Accepted: 01/05/2023] [Indexed: 01/08/2023] Open
Abstract
MYOCD is a transcription factor important for cardiac and smooth muscle development. We previously identified that actin-related protein 5 (ARP5) binds to the N-terminus of MYOCD. Here, we demonstrate that ARP5 inhibits the cooperative action of the cardiac-specific isoform of MYOCD with MEF2. ARP5 overexpression in murine hearts induced cardiac hypertrophy and fibrosis, whereas ARP5 knockdown in P19CL6 cells significantly increased cardiac gene expression. ARP5 was found to bind to a MEF2-binding motif of cardiac MYOCD and inhibit MEF2-mediated transactivation by MYOCD. RNA-seq analysis revealed 849 genes that are upregulated by MYOCD-MEF2 and 650 genes that are repressed by ARP5. ARP5 expression increased with cardiomyopathy and was negatively correlated with the expression of Tnnt2 and Ttn, which were regulated by cardiac MYOCD-MEF2. Overall, our data suggest that ARP5 is a potential suppressor of cardiac MYOCD during physiological and pathological processes.
Collapse
Affiliation(s)
| | - Ken'ichiro Hayashi
- Department of OphthalmologyYamaguchi University Graduate School of MedicineJapan,Department of RNA Biology and NeuroscienceOsaka University Graduate School of MedicineJapan
| |
Collapse
|
14
|
Marzoog BA. Transcription Factors - the Essence of Heart Regeneration: A Potential Novel Therapeutic Strategy. Curr Mol Med 2023; 23:232-238. [PMID: 35170408 DOI: 10.2174/1566524022666220216123650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 02/08/2023]
Abstract
Myocardial cell injury and following sequelae are the primary reasons for death globally. Unfortunately, myocardiocytes in adults have limited regeneration capacity. Therefore, the generation of neo myocardiocytes from non-myocardial cells is a surrogate strategy. Transcription factors (TFs) can be recruited to achieve this tremendous goal. Transcriptomic analyses have suggested that GATA, Mef2c, and Tbx5 (GMT cocktail) are master TFs to transdifferentiate/reprogram cell linage of fibroblasts, somatic cells, mesodermal cells into myocardiocytes. However, adding MESP1, MYOCD, ESRRG, and ZFPM2 TFs induces the generation of more efficient and physiomorphological features for induced myocardiocytes. Moreover, the same cocktail of transcription factors can induce the proliferation and differentiation of induced/pluripotent stem cells into myocardial cells. Amelioration of impaired myocardial cells involves the activation of healing transcription factors, which are induced by inflammation mediators; IL6, tumor growth factor β, and IL22. Transcription factors regulate the cellular and subcellular physiology of myocardiocytes to include mitotic cell cycling regulation, karyokinesis and cytokinesis, hypertrophic growth, adult sarcomeric contractile protein gene expression, fatty acid metabolism, and mitochondrial biogenesis and maturation. Cell therapy by transcription factors can be applied to cardiogenesis and ameliorating impaired cardiocytes. Transcription factors are the cornerstone in cell differentiation.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- Department of Normal and Pathological Physiology, National Research Mordovia State University, Bolshevitskaya Street, 68, Saransk, Rep. Mordovia, 430005, Russia
| |
Collapse
|
15
|
Single-cell transcriptomic analysis identifies murine heart molecular features at embryonic and neonatal stages. Nat Commun 2022; 13:7960. [PMID: 36575170 PMCID: PMC9794824 DOI: 10.1038/s41467-022-35691-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Heart development is a continuous process involving significant remodeling during embryogenesis and neonatal stages. To date, several groups have used single-cell sequencing to characterize the heart transcriptomes but failed to capture the progression of heart development at most stages. This has left gaps in understanding the contribution of each cell type across cardiac development. Here, we report the transcriptional profile of the murine heart from early embryogenesis to late neonatal stages. Through further analysis of this dataset, we identify several transcriptional features. We identify gene expression modules enriched at early embryonic and neonatal stages; multiple cell types in the left and right atriums are transcriptionally distinct at neonatal stages; many congenital heart defect-associated genes have cell type-specific expression; stage-unique ligand-receptor interactions are mostly between epicardial cells and other cell types at neonatal stages; and mutants of epicardium-expressed genes Wt1 and Tbx18 have different heart defects. Assessment of this dataset serves as an invaluable source of information for studies of heart development.
Collapse
|
16
|
Hong JH, Zhang HG. Transcription Factors Involved in the Development and Prognosis of Cardiac Remodeling. Front Pharmacol 2022; 13:828549. [PMID: 35185581 PMCID: PMC8849252 DOI: 10.3389/fphar.2022.828549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/14/2022] [Indexed: 01/09/2023] Open
Abstract
To compensate increasing workload, heart must work harder with structural changes, indicated by increasing size and changing shape, causing cardiac remodeling. However, pathological and unlimited compensated cardiac remodeling will ultimately lead to decompensation and heart failure. In the past decade, numerous studies have explored many signaling pathways involved in cardiac remodeling, but the complete mechanism of cardiac remodeling is still unrecognized, which hinders effective treatment and drug development. As gene transcriptional regulators, transcription factors control multiple cellular activities and play a critical role in cardiac remodeling. This review summarizes the regulation of fetal gene reprogramming, energy metabolism, apoptosis, autophagy in cardiomyocytes and myofibroblast activation of cardiac fibroblasts by transcription factors, with an emphasis on their potential roles in the development and prognosis of cardiac remodeling.
Collapse
|
17
|
Thottakara T, Lund N, Krämer E, Kirchhof P, Carrier L, Patten M. A Novel miRNA Screen Identifies miRNA-4454 as a Candidate Biomarker for Ventricular Fibrosis in Patients with Hypertrophic Cardiomyopathy. Biomolecules 2021; 11:1718. [PMID: 34827715 PMCID: PMC8615621 DOI: 10.3390/biom11111718] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/14/2021] [Indexed: 12/21/2022] Open
Abstract
(1) Background: Left ventricular hypertrophy, myocardial disarray and interstitial fibrosis are the hallmarks of hypertrophic cardiomyopathy (HCM). Access to the myocardium for diagnostic purposes is limited. Circulating biomolecules reflecting the myocardial disease processes could improve the early detection of HCM. Circulating miRNAs have been found to reflect disease processes in several cardiovascular diseases. (2) Methods: We quantified circulating miRNA molecules in the plasma of 24 HCM and 11 healthy controls using the Human v3 miRNA Expression Assay Kit Code set (Nanostring Tech., Seattle, WA, USA) and validated differentially expressed miRNAs using RT-PCR. (3) Results: In comparison to healthy controls, the levels of six miRNAs (miR-1, miR-3144, miR-4454, miR-495-3p, miR-499a-5p and miR-627-3p) were higher in the plasma of HCM patients than healthy individuals (p < 0.05). Of these, higher levels of miR-1, miR-495 and miR-4454 could be validated by real-time PCR. In addition, elevated miR-4454 levels were significantly correlated with cardiac fibrosis, detected by magnetic resonance imaging in HCM patients. (4) Conclusions: Circulating miR-1, miR-495-3p and miR-4454 levels are elevated in the plasma of HCM patients. To the best of our knowledge, this is the first report showing a correlation between miR-4454 levels and cardiac fibrosis in HCM. This suggests miR-4454 as a potential biomarker for fibrosis in these patients.
Collapse
Affiliation(s)
- Tilo Thottakara
- Department of Cardiology, University Heart and Vascular Center Hamburg, 20253 Hamburg, Germany; (T.T.); (N.L.); (P.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany; (E.K.); (L.C.)
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, CA 94158, USA
| | - Natalie Lund
- Department of Cardiology, University Heart and Vascular Center Hamburg, 20253 Hamburg, Germany; (T.T.); (N.L.); (P.K.)
| | - Elisabeth Krämer
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany; (E.K.); (L.C.)
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Paulus Kirchhof
- Department of Cardiology, University Heart and Vascular Center Hamburg, 20253 Hamburg, Germany; (T.T.); (N.L.); (P.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany; (E.K.); (L.C.)
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Lucie Carrier
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany; (E.K.); (L.C.)
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Monica Patten
- Department of Cardiology, University Heart and Vascular Center Hamburg, 20253 Hamburg, Germany; (T.T.); (N.L.); (P.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany; (E.K.); (L.C.)
| |
Collapse
|
18
|
Benner P, Vingron M. Quantifying the tissue-specific regulatory information within enhancer DNA sequences. NAR Genom Bioinform 2021; 3:lqab095. [PMID: 34729474 PMCID: PMC8557370 DOI: 10.1093/nargab/lqab095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 12/04/2022] Open
Abstract
Recent efforts to measure epigenetic marks across a wide variety of different cell types and tissues provide insights into the cell type-specific regulatory landscape. We use these data to study whether there exists a correlate of epigenetic signals in the DNA sequence of enhancers and explore with computational methods to what degree such sequence patterns can be used to predict cell type-specific regulatory activity. By constructing classifiers that predict in which tissues enhancers are active, we are able to identify sequence features that might be recognized by the cell in order to regulate gene expression. While classification performances vary greatly between tissues, we show examples where our classifiers correctly predict tissue-specific regulation from sequence alone. We also show that many of the informative patterns indeed harbor transcription factor footprints.
Collapse
Affiliation(s)
- Philipp Benner
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestraße 73, 14195 Berlin, Germany
| | - Martin Vingron
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestraße 73, 14195 Berlin, Germany
| |
Collapse
|
19
|
Aminu AJ, Petkova M, Atkinson AJ, Yanni J, Morris AD, Simms RT, Chen W, Yin Z, Kuniewicz M, Holda MK, Kuzmin VS, Perde F, Molenaar P, Dobrzynski H. Further insights into the molecular complexity of the human sinus node - The role of 'novel' transcription factors and microRNAs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:86-104. [PMID: 34004232 DOI: 10.1016/j.pbiomolbio.2021.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
RESEARCH PURPOSE The sinus node (SN) is the heart's primary pacemaker. Key ion channels (mainly the funny channel, HCN4) and Ca2+-handling proteins in the SN are responsible for its function. Transcription factors (TFs) regulate gene expression through inhibition or activation and microRNAs (miRs) do this through inhibition. There is high expression of macrophages and mast cells within the SN connective tissue. 'Novel'/unexplored TFs and miRs in the regulation of ion channels and immune cells in the SN are not well understood. Using RNAseq and bioinformatics, the expression profile and predicted interaction of key TFs and cell markers with key miRs in the adult human SN vs. right atrial tissue (RA) were determined. PRINCIPAL RESULTS 68 and 60 TFs significantly more or less expressed in the SN vs. RA respectively. Among those more expressed were ISL1 and TBX3 (involved in embryonic development of the SN) and 'novel' RUNX1-2, CEBPA, GLI1-2 and SOX2. These TFs were predicted to regulate HCN4 expression in the SN. Markers for different cells: fibroblasts (COL1A1), fat (FABP4), macrophages (CSF1R and CD209), natural killer (GZMA) and mast (TPSAB1) were significantly more expressed in the SN vs. RA. Interestingly, RUNX1-3, CEBPA and GLI1 also regulate expression of these cells. MiR-486-3p inhibits HCN4 and markers involved in immune response. MAJOR CONCLUSIONS In conclusion, RUNX1-2, CSF1R, TPSAB1, COL1A1 and HCN4 are highly expressed in the SN but not miR-486-3p. Their complex interactions can be used to treat SN dysfunction such as bradycardia. Interestingly, another research group recently reported miR-486-3p is upregulated in blood samples from severe COVID-19 patients who suffer from bradycardia.
Collapse
Affiliation(s)
- Abimbola J Aminu
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Maria Petkova
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Andrew J Atkinson
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Joseph Yanni
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Alex D Morris
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Robert T Simms
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Weixuan Chen
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Zeyuan Yin
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Marcin Kuniewicz
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom; Department of Anatomy, Jagiellonian University Medical College, Krakow, Poland
| | - Mateusz K Holda
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom; Department of Anatomy, Jagiellonian University Medical College, Krakow, Poland
| | - Vladislav S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russia
| | - Filip Perde
- National Institute of Legal Medicine, Bucharest, Romania
| | - Peter Molenaar
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia; Cardiovascular Molecular & Therapeutics Translational Research Group, University of Queensland, The Prince Charles Hospital, Brisbane, Australia
| | - Halina Dobrzynski
- The Division of Cardiovascular Sciences, University of Manchester, United Kingdom; Department of Anatomy, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
20
|
Meeks KAC, Bentley AR, Gouveia MH, Chen G, Zhou J, Lei L, Adeyemo AA, Doumatey AP, Rotimi CN. Genome-wide analyses of multiple obesity-related cytokines and hormones informs biology of cardiometabolic traits. Genome Med 2021; 13:156. [PMID: 34620218 PMCID: PMC8499470 DOI: 10.1186/s13073-021-00971-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/16/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A complex set of perturbations occur in cytokines and hormones in the etiopathogenesis of obesity and related cardiometabolic conditions such as type 2 diabetes (T2D). Evidence for the genetic regulation of these cytokines and hormones is limited, particularly in African-ancestry populations. In order to improve our understanding of the biology of cardiometabolic traits, we investigated the genetic architecture of a large panel of obesity- related cytokines and hormones among Africans with replication analyses in African Americans. METHODS We performed genome-wide association studies (GWAS) in 4432 continental Africans, enrolled from Ghana, Kenya, and Nigeria as part of the Africa America Diabetes Mellitus (AADM) study, for 13 obesity-related cytokines and hormones, including adipsin, glucose-dependent insulinotropic peptide (GIP), glucagon-like peptide-1 (GLP-1), interleukin-1 receptor antagonist (IL1-RA), interleukin-6 (IL-6), interleukin-10 (IL-10), leptin, plasminogen activator inhibitor-1 (PAI-1), resistin, visfatin, insulin, glucagon, and ghrelin. Exact and local replication analyses were conducted in African Americans (n = 7990). The effects of sex, body mass index (BMI), and T2D on results were investigated through stratified analyses. RESULTS GWAS identified 39 significant (P value < 5 × 10-8) loci across all 13 traits. Notably, 14 loci were African-ancestry specific. In this first GWAS for adipsin and ghrelin, we detected 13 and 4 genome-wide significant loci respectively. Stratified analyses by sex, BMI, and T2D showed a strong effect of these variables on detected loci. Eight novel loci were successfully replicated: adipsin (3), GIP (1), GLP-1 (1), and insulin (3). Annotation of these loci revealed promising links between these adipocytokines and cardiometabolic outcomes as illustrated by rs201751833 for adipsin and blood pressure and locus rs759790 for insulin level and T2D in lean individuals. CONCLUSIONS Our study identified genetic variants underlying variation in multiple adipocytokines, including the first loci for adipsin and ghrelin. We identified population differences in variants associated with adipocytokines and highlight the importance of stratification for discovery of loci. The high number of African-specific loci detected emphasizes the need for GWAS in African-ancestry populations, as these loci could not have been detected in other populations. Overall, our work contributes to the understanding of the biology linking adipocytokines to cardiometabolic traits.
Collapse
Affiliation(s)
- Karlijn A C Meeks
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Mateus H Gouveia
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Lin Lei
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Adebowale A Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA.
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive Bldg 12A rm 4047, Bethesda, MD, 20814, USA.
| |
Collapse
|
21
|
Gupta S, Adhikary S, Hui SP. Decoding the proregenerative competence of regulatory T cells through complex tissue regeneration in zebrafish. Clin Exp Immunol 2021; 206:346-353. [PMID: 34529822 DOI: 10.1111/cei.13661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs ) are specific subtype of T cells that play a central role in sustaining self-antigen tolerance and restricting inflammatory tissue damage. More recently, additional direct functions of Tregs in mammalian tissue repair have emerged, but the regenerative potential of Tregs in non-mammalian vertebrates has not been explored despite the latter possessing a highly developed adaptive immune system. Why complex organs such as the caudal fin, heart, brain, spinal cord and retina regenerate in certain non-mammalian vertebrates, but not in mammals, is an interesting but unresolved question in the field of regenerative biology. Inflammation has traditionally been thought to be an impediment to regeneration due to the formation of scars. Regenerative decline in higher organisms has been speculated to be the evolutionary advent of adaptive immunity. Recent studies, however, have shown that the innate inflammatory response in non-mammalian organisms is required for organ regeneration. It has also been found that highly advanced adaptive immunity is no longer incompatible with regeneration and for that, Tregs are important. Zebrafish regulatory T cells (zTregs ) migrate rapidly to the injury site in damaged organs, where they facilitate the proliferation of regeneration precursor cells by generating tissue-specific regenerative factors by a process distinct from the canonical anti-inflammatory pathway. We review both reparative and proregenerative roles of Tregs in mammals and zebrafish, respectively, and also give an overview of the forkhead box protein 3 (FoxP3) -dependent immunosuppressive function of Tregs in zebrafish, which makes it a useful model organism for future Treg biology and research.
Collapse
Affiliation(s)
- Samudra Gupta
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, India
| | - Satadal Adhikary
- Post Graduate Department of Zoology, ABN Seal College, Cooch Behar, India
| | - Subhra Prakash Hui
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, India
| |
Collapse
|
22
|
The Key Lnc (RNA)s in Cardiac and Skeletal Muscle Development, Regeneration, and Disease. J Cardiovasc Dev Dis 2021; 8:jcdd8080084. [PMID: 34436226 PMCID: PMC8397000 DOI: 10.3390/jcdd8080084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/29/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
Non-coding RNAs (ncRNAs) play a key role in the regulation of transcriptional and epigenetic activity in mammalian cells. Comprehensive analysis of these ncRNAs has revealed sophisticated gene regulatory mechanisms which finely tune the proper gene output required for cellular homeostasis, proliferation, and differentiation. However, this elaborate circuitry has also made it vulnerable to perturbations that often result in disease. Among the many types of ncRNAs, long non-coding RNAs (lncRNAs) appear to have the most diverse mechanisms of action including competitive binding to miRNA targets, direct binding to mRNA, interactions with transcription factors, and facilitation of epigenetic modifications. Moreover, many lncRNAs display tissue-specific expression patterns suggesting an important regulatory role in organogenesis, yet the molecular mechanisms through which these molecules regulate cardiac and skeletal muscle development remains surprisingly limited. Given the structural and metabolic similarities of cardiac and skeletal muscle, it is likely that several lncRNAs expressed in both of these tissues have conserved functions in establishing the striated muscle phenotype. As many aspects of regeneration recapitulate development, understanding the role lncRNAs play in these processes may provide novel insights to improve regenerative therapeutic interventions in cardiac and skeletal muscle diseases. This review highlights key lncRNAs that function as regulators of development, regeneration, and disease in cardiac and skeletal muscle. Finally, we highlight lncRNAs encoded by imprinted genes in striated muscle and the contributions of these loci on the regulation of gene expression.
Collapse
|
23
|
Bosada FM, Rivaud MR, Uhm JS, Verheule S, van Duijvenboden K, Verkerk AO, Christoffels VM, Boukens BJ. A Variant Noncoding Region Regulates Prrx1 and Predisposes to Atrial Arrhythmias. Circ Res 2021; 129:420-434. [PMID: 34092116 DOI: 10.1161/circresaha.121.319146] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Fernanda M Bosada
- Department of Medical Biology (F.M.B., J.-S.U., K.v.D., A.O.V., V.M.C., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Mathilde R Rivaud
- Department of Experimental Cardiology (M.R.R., A.O.V., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Jae-Sun Uhm
- Department of Medical Biology (F.M.B., J.-S.U., K.v.D., A.O.V., V.M.C., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands.,Department of Cardiology, Severance Hospital, College of Medicine, Yonsei University, Seoul, South Korea (J.-S.U.)
| | - Sander Verheule
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (S.V.)
| | - Karel van Duijvenboden
- Department of Medical Biology (F.M.B., J.-S.U., K.v.D., A.O.V., V.M.C., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Arie O Verkerk
- Department of Medical Biology (F.M.B., J.-S.U., K.v.D., A.O.V., V.M.C., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands.,Department of Experimental Cardiology (M.R.R., A.O.V., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology (F.M.B., J.-S.U., K.v.D., A.O.V., V.M.C., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Bastiaan J Boukens
- Department of Medical Biology (F.M.B., J.-S.U., K.v.D., A.O.V., V.M.C., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands.,Department of Experimental Cardiology (M.R.R., A.O.V., B.J.B.), Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| |
Collapse
|
24
|
Herbal Extract from Codonopsis pilosula (Franch.) Nannf. Enhances Cardiogenic Differentiation and Improves the Function of Infarcted Rat Hearts. Life (Basel) 2021; 11:life11050422. [PMID: 34063127 PMCID: PMC8148170 DOI: 10.3390/life11050422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 11/25/2022] Open
Abstract
Background: The roots of Codonopsis pilosula (Franch.) Nannf. have been used in traditional Chinese medicine for treating cardiovascular disease. In the current study, we aimed to discover herbal extracts from C. pilosula that are capable of improving cardiac function of infarcted hearts to develop a potential therapeutic approach. Methods: A mouse embryonic stem (ES) cell-based model with an enhanced green fluorescent protein (eGFP) reporter driven by a cardiomyocyte-specific promoter, the α-myosin heavy chain, was constructed to evaluate the cardiogenic activity of herbal extracts. Then, herbal extracts from C. pilosula with cardiogenic activity based on an increase in eGFP expression during ES cell differentiation were further tested in a rat myocardial infarction model with left anterior descending artery (LAD) ligation. Cardiac function assessments were performed using echocardiography, 1, 3, and 6 weeks post LAD ligation. Results: The herbal extract 417W from C. pilosula was capable of enhancing cardiogenic differentiation in mouse ES cells in vitro. Echocardiography results in the LAD-ligated rat model revealed significant improvements in the infarcted hearts at least 6 weeks after 417W treatment that were determined based on left ventricle fractional shortening (FS), fractional area contraction (FAC), and ejection fraction (EF). Conclusions: The herbal extract 417W can enhance the cardiogenic differentiation of ES cells and improve the cardiac function of infarcted hearts.
Collapse
|
25
|
Miyamoto M, Gangrade H, Tampakakis E. Understanding Heart Field Progenitor Cells for Modeling Congenital Heart Diseases. Curr Cardiol Rep 2021; 23:38. [PMID: 33694131 DOI: 10.1007/s11886-021-01468-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Heart development is a meticulously coordinated process that involves the specification of two distinct populations of cardiac progenitor cells, namely the first and the second heart field. Disruption of heart field progenitors can result in congenital heart defects. In this review, we aim to describe the signaling pathways and transcription factors that link heart field development and congenital heart disease. RECENT FINDINGS Single-cell transcriptomics, lineage-tracing mouse models, and stem cell-based in vitro modeling of cardiogenesis have significantly improved the spatiotemporal characterization of cardiac progenitors. Additionally, novel functional genomic studies have now linked more genetic variants with congenital heart disease. Dysregulation of cardiac progenitor cells causes malformations that can be lethal. Ongoing research will continue to shed light on cardiac morphogenesis and help us better understand and treat patients with congenital heart disease.
Collapse
Affiliation(s)
- Matthew Miyamoto
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 835, Baltimore, MD, 21205, USA
| | - Harshi Gangrade
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 835, Baltimore, MD, 21205, USA
| | - Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 835, Baltimore, MD, 21205, USA.
| |
Collapse
|
26
|
Kyryachenko S, Georges A, Yu M, Barrandou T, Guo L, Bruneval P, Rubio T, Gronwald J, Baraki H, Kutschka I, Aras KK, Efimov IR, Norris RA, Voigt N, Bouatia-Naji N. Chromatin Accessibility of Human Mitral Valves and Functional Assessment of MVP Risk Loci. Circ Res 2021; 128:e84-e101. [PMID: 33508947 PMCID: PMC8316483 DOI: 10.1161/circresaha.120.317581] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/28/2021] [Indexed: 12/07/2022]
Abstract
RATIONALE Mitral valve prolapse (MVP) is a common valvopathy that leads to mitral insufficiency, heart failure, and sudden death. Functional genomic studies in mitral valves are needed to better characterize MVP-associated variants and target genes. OBJECTIVE To establish the chromatin accessibility profiles and assess functionality of variants and narrow down target genes at MVP loci. METHODS AND RESULTS We mapped the open chromatin regions in nuclei from 11 human pathogenic and 7 nonpathogenic mitral valves by an assay for transposase-accessible chromatin with high-throughput sequencing. Open chromatin peaks were globally similar between pathogenic and nonpathogenic valves. Compared with the heart tissue and cardiac fibroblasts, we found that MV-specific assay for transposase-accessible chromatin with high-throughput sequencing peaks are enriched near genes involved in extracellular matrix organization, chondrocyte differentiation, and connective tissue development. One of the most enriched motifs in MV-specific open chromatin peaks was for the nuclear factor of activated T cells family of TFs (transcription factors) involved in valve endocardial and interstitial cell formation. We also found that MVP-associated variants were significantly enriched (P<0.05) in mitral valve open chromatin peaks. Integration of the assay for transposase-accessible chromatin with high-throughput sequencing data with risk loci, extensive functional annotation, and gene reporter assay suggest plausible causal variants for rs2641440 at the SMG6/SRR locus and rs6723013 at the IGFBP2/IGFBP5/TNS1 locus. CRISPR-Cas9 deletion of the sequence including rs6723013 in human fibroblasts correlated with increased expression only for TNS1. Circular chromatin conformation capture followed by high-throughput sequencing experiments provided evidence for several target genes, including SRR, HIC1, and DPH1 at the SMG6/SRR locus and further supported TNS1 as the most likely target gene on chromosome 2. CONCLUSIONS Here, we describe unprecedented genome-wide open chromatin profiles from human pathogenic and nonpathogenic MVs and report specific gene regulation profiles, compared with the heart. We also report in vitro functional evidence for potential causal variants and target genes at MVP risk loci involving established and new biological mechanisms. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
| | | | - Mengyao Yu
- Université de Paris, PARCC, Inserm, Paris,
France
| | | | - Lilong Guo
- Department of Regenerative Medicine and Cell Biology,
Medical University of South Carolina, Charleston, SC, USA
- Department of Medicine, Medical University of South
Carolina, Charleston, SC, USA
| | | | - Tony Rubio
- Institute of Pharmacology and Toxicology, University
Medical Center Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner
Site Göttingen, Germany
| | - Judith Gronwald
- Institute of Pharmacology and Toxicology, University
Medical Center Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner
Site Göttingen, Germany
| | - Hassina Baraki
- DZHK (German Center for Cardiovascular Research), Partner
Site Göttingen, Germany
- Department of Thoracic and Cardiovascular Surgery,
University Medical Center, Göttingen, Germany
| | - Ingo Kutschka
- DZHK (German Center for Cardiovascular Research), Partner
Site Göttingen, Germany
- Department of Thoracic and Cardiovascular Surgery,
University Medical Center, Göttingen, Germany
| | - Kedar K. Aras
- Department of Biomedical Engineering, George Washington
University, Washington, DC, USA
| | - Igor R. Efimov
- Department of Biomedical Engineering, George Washington
University, Washington, DC, USA
| | - Russel A. Norris
- Department of Regenerative Medicine and Cell Biology,
Medical University of South Carolina, Charleston, SC, USA
- Department of Medicine, Medical University of South
Carolina, Charleston, SC, USA
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University
Medical Center Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner
Site Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from
Molecular Machines to Networks of Excitable Cells (MBExC), University of
Göttingen, Germany
| | | |
Collapse
|
27
|
Mef2c factors are required for early but not late addition of cardiomyocytes to the ventricle. Dev Biol 2020; 470:95-107. [PMID: 33245870 PMCID: PMC7819464 DOI: 10.1016/j.ydbio.2020.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
During heart formation, the heart grows and undergoes dramatic morphogenesis to achieve efficient embryonic function. Both in fish and amniotes, much of the growth occurring after initial heart tube formation arises from second heart field (SHF)-derived progenitor cell addition to the arterial pole, allowing chamber formation. In zebrafish, this process has been extensively studied during embryonic life, but it is unclear how larval cardiac growth occurs beyond 3 days post-fertilisation (dpf). By quantifying zebrafish myocardial growth using live imaging of GFP-labelled myocardium we show that the heart grows extensively between 3 and 5 dpf. Using methods to assess cell division, cellular development timing assay and Kaede photoconversion, we demonstrate that proliferation, CM addition, and hypertrophy contribute to ventricle growth. Mechanistically, we show that reduction in Mef2c activity (mef2ca+/-;mef2cb-/-), downstream or in parallel with Nkx2.5 and upstream of Ltbp3, prevents some CM addition and differentiation, resulting in a significantly smaller ventricle by 3 dpf. After 3 dpf, however, CM addition in mef2ca+/-;mef2cb-/- mutants recovers to a normal pace, and the heart size gap between mutants and their siblings diminishes into adulthood. Thus, as in mice, there is an early time window when SHF contribution to the myocardium is particularly sensitive to loss of Mef2c activity.
Collapse
|
28
|
Li J, Hua Y, Miyagawa S, Zhang J, Li L, Liu L, Sawa Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E8893. [PMID: 33255277 PMCID: PMC7727666 DOI: 10.3390/ijms21238893] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Cell Design for Tissue Construction, Faculty of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| |
Collapse
|
29
|
The Future of Direct Cardiac Reprogramming: Any GMT Cocktail Variety? Int J Mol Sci 2020; 21:ijms21217950. [PMID: 33114756 PMCID: PMC7663133 DOI: 10.3390/ijms21217950] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which this technology was described, showed that at least a combination of three transcription factors, GATA4, MEF2C and TBX5 (GMT cocktail), was required for the reprogramming into iCMs in vitro using mouse cells. However, this was later demonstrated to be insufficient for the reprogramming of human cells and additional factors were required. Thereafter, most studies have focused on implementing reprogramming efficiency and obtaining fully reprogrammed and functional iCMs, by the incorporation of other transcription factors, microRNAs or small molecules to the original GMT cocktail. In this respect, great advances have been made in recent years. However, there is still no consensus on which of these GMT-based varieties is best, and robust and highly reproducible protocols are still urgently required, especially in the case of human cells. On the other hand, apart from CMs, other cells such as endothelial and smooth muscle cells to form new blood vessels will be fundamental for the correct reconstruction of damaged cardiac tissue. With this aim, several studies have centered on the direct reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs) able to give rise to all myocardial cell lineages. Especially interesting are reports in which multipotent and highly expandable mouse iCPCs have been obtained, suggesting that clinically relevant amounts of these cells could be created. However, as of yet, this has not been achieved with human iCPCs, and exactly what stage of maturity is appropriate for a cell therapy product remains an open question. Nonetheless, the major concern in regenerative medicine is the poor retention, survival, and engraftment of transplanted cells in the cardiac tissue. To circumvent this issue, several cell pre-conditioning approaches are currently being explored. As an alternative to cell injection, in vivo reprogramming may face fewer barriers for its translation to the clinic. This approach has achieved better results in terms of efficiency and iCMs maturity in mouse models, indicating that the heart environment can favor this process. In this context, in recent years some studies have focused on the development of safer delivery systems such as Sendai virus, Adenovirus, chemical cocktails or nanoparticles. This article provides an in-depth review of the in vitro and in vivo cardiac reprograming technology used in mouse and human cells to obtain iCMs and iCPCs, and discusses what challenges still lie ahead and what hurdles are to be overcome before results from this field can be transferred to the clinical settings.
Collapse
|
30
|
Duddu S, Chakrabarti R, Ghosh A, Shukla PC. Hematopoietic Stem Cell Transcription Factors in Cardiovascular Pathology. Front Genet 2020; 11:588602. [PMID: 33193725 PMCID: PMC7596349 DOI: 10.3389/fgene.2020.588602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Transcription factors as multifaceted modulators of gene expression that play a central role in cell proliferation, differentiation, lineage commitment, and disease progression. They interact among themselves and create complex spatiotemporal gene regulatory networks that modulate hematopoiesis, cardiogenesis, and conditional differentiation of hematopoietic stem cells into cells of cardiovascular lineage. Additionally, bone marrow-derived stem cells potentially contribute to the cardiovascular cell population and have shown potential as a therapeutic approach to treat cardiovascular diseases. However, the underlying regulatory mechanisms are currently debatable. This review focuses on some key transcription factors and associated epigenetic modifications that modulate the maintenance and differentiation of hematopoietic stem cells and cardiac progenitor cells. In addition to this, we aim to summarize different potential clinical therapeutic approaches in cardiac regeneration therapy and recent discoveries in stem cell-based transplantation.
Collapse
Affiliation(s)
| | | | | | - Praphulla Chandra Shukla
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
31
|
Kumar P, Ghosh A, Sundaresan L, Kathirvel P, Sankaranarayanan K, Chatterjee S. Ectopic release of nitric oxide modulates the onset of cardiac development in avian model. In Vitro Cell Dev Biol Anim 2020; 56:593-603. [PMID: 32959218 DOI: 10.1007/s11626-020-00495-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/10/2020] [Indexed: 01/01/2023]
Abstract
Heart development is one of the earliest developmental events, and its pumping action is directly linked to the intensity of development of other organs. Heart contractions mediate the circulation of the nutrients and signalling molecules to the focal points of developing embryos. In the present study, we used in vivo, ex vivo, in vitro, and in silico methods for chick embryo model to characterize and identify molecular targets under the influence of ectopic nitric oxide in reference to cardiogenesis. Spermine NONOate (SpNO) treatment of 10 μM increased the percentage of chick embryos having beating heart at 40th h of incubation by 2.2-fold (p < 0.001). In an ex vivo chick embryo culture, SpNO increased the percentage of embryos having beats by 1.56-fold (p < 0.05) compared with control after 2 h of treatment. Total body weight of SpNO-treated chick embryos at the Hamburger and Hamilton (HH) stage 29 was increased by 1.22-fold (p < 0.005). Cardiac field potential (FP) recordings of chick embryo at HH29 showed 2.5-fold (p < 0.001) increased in the amplitude, 3.2-fold (p < 0.001) increased in frequency of SpNO-treated embryos over that of the control group, whereas FP duration was unaffected. In cultured cardiac progenitors cells (CPCs), SpNO treatment decreased apoptosis and cell death by twofold (p < 0.001) and 1.7-fold (p < 0.001), respectively. Transcriptome analysis of chick embryonic heart isolated from HH15 stage pre-treated with SpNO at HH8 stage showed upregulation of genes involved in heart morphogenesis, heart contraction, cardiac cell development, calcium signalling, structure, and development whereas downregulated genes were enriched under the terms extracellular matrix, wnt pathway, and BMP pathway. The key upstream molecules predicted to be activated were p38 MAPK, MEF2C, TBX5, and GATA4 while KDM5α, DNMT3A, and HNF1α were predicted to be inhibited. This study suggests that the ectopic nitric oxide modulates the onset of cardiac development.
Collapse
Affiliation(s)
- Pavitra Kumar
- Vascular Biology Laboratory, AU-KBC Research Centre, M.I.T Campus of Anna University, Chromepet, Chennai, Tamil Nadu, 600044, India
| | - Anuran Ghosh
- Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Lakshmikirupa Sundaresan
- Vascular Biology Laboratory, AU-KBC Research Centre, M.I.T Campus of Anna University, Chromepet, Chennai, Tamil Nadu, 600044, India.,Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | | | | | - Suvro Chatterjee
- Vascular Biology Laboratory, AU-KBC Research Centre, M.I.T Campus of Anna University, Chromepet, Chennai, Tamil Nadu, 600044, India. .,Department of Biotechnology, Anna University, Chennai, Tamil Nadu, India.
| |
Collapse
|
32
|
Cloning and promoter analysis of palladin 90-kDa, 140-kDa, and 200-kDa isoforms involved in skeletal muscle cell maturation. BMC Res Notes 2020; 13:321. [PMID: 32620172 PMCID: PMC7333403 DOI: 10.1186/s13104-020-05152-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/24/2020] [Indexed: 11/10/2022] Open
Abstract
Objective Palladin is a ubiquitous phosphoprotein expressed in vertebrate cells that works as a scaffolding protein. Several isoforms deriving from alternative splicing are originated from the palladin gene and involved in mesenchymal and muscle cells formation, maturation, migration, and contraction. Recent studies have linked palladin to the invasive spread of cancer and myogenesis. However, since its discovery, the promoter region of the palladin gene has never been studied. The objective of this study was to predict, identify, and measure the activity of the promoter regions of palladin gene. Results By using promoter prediction programs, we successfully identified the transcription start sites for the Palld isoforms and revealed the presence of a variety of transcriptional regulatory elements including TATA box, GATA, MyoD, myogenin, MEF, Nkx2-5, and Tcf3 upstream promoter regions. The transcriptome profiling approach confirmed the active role of predicted transcription factors in the mouse genome. This study complements the missing piece in the characterization of palladin gene and certainly contributes to understanding the complexity and enrollment of palladin regulatory factors in gene transcription.
Collapse
|
33
|
Zhang S, Song Z, An L, Liu X, Hu XW, Naz A, Zhou R, Guo X, He L, Zhu H. WD40 repeat and FYVE domain containing 3 is essential for cardiac development. Cardiovasc Res 2020; 115:1320-1331. [PMID: 30428088 DOI: 10.1093/cvr/cvy285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/22/2018] [Accepted: 11/13/2018] [Indexed: 01/02/2023] Open
Abstract
AIMS WD40 repeat and FYVE domain containing 3 (WDFY3) is an adaptor protein involved in selective degradation of protein aggregates by autophagy. Recent studies have revealed that Wdfy3 is critical in the regulation of brain development and osteoclastogenesis in vivo. However, the function of Wdfy3 in cardiac development remains completely unknown. In this study, we explore the role of Wdfy3 in cardiac morphogenesis using Wdfy3-deficient mice. METHODS AND RESULTS Wdfy3 was expressed in the developing heart in mice and peaked at embryonic day 12.5 (E12.5). Loss of Wdfy3 in mice led to embryonic and neonatal lethality. Wdfy3-deficient mice displayed various congenital heart defects including membranous ventricular septal defect (VSD), aortic overriding (AO), double outlet right ventricle (DORV), thinning of ventricular wall, ventricular dilation, and disorganized ventricular trabeculation at E14.5. Cell proliferation was reduced in the hearts from Wdfy3-deficient mice at E12.5 and E14.5, which was associated with enhanced p21 expression. Cardiomyocyte differentiation was diminished as demonstrated by reduced Myh6 and MLC2v in Wdfy3-deficient mice at E14.5. In addition, Nkx2-5 and Mef2c, two cardiac transcription factors regulating cardiomyocyte differentiation, were decreased in Wdfy3-deficient mice at E14.5. Apoptotic cell death remained unaltered. These data suggest that reduced cell proliferation and cardiomyocyte differentiation contribute to cardiac defects in Wdfy3-deficient mice. Mechanistically, loss of Wdfy3 led to a reduction in protein levels of Notch 1 intracellular domain and its downstream targets Hes1 and Hey1, which was accompanied with enhanced full-length Notch1 protein levels. In vitro luciferase assay showed that Wdfy3 deficiency induced activity of p21 promoter, while diminished activity of Hes1 promoter through modulation of Notch1 signalling. Moreover, Wdfy3 was co-localized with Notch1 in primary embryonic cardiomyocytes. Endogenous Wdfy3 physically interacted with full-length Notch1 in the developing heart. These results suggest that Notch1 signalling is perturbed in the hearts from Wdfy3-deficient mice. No alteration of autophagy was detected in the hearts from Wdfy3-deficient mice. CONCLUSION Taken together, our data suggest that Wdfy3 plays an essential role in cardiac development, which may be mediated by modulation of Notch1 signalling.
Collapse
Affiliation(s)
- Shasha Zhang
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Zongpei Song
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Lin An
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyun Liu
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Wen Hu
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Amber Naz
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Rujiang Zhou
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xizhi Guo
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Lin He
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Hongxin Zhu
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
34
|
Cheng J, Cao XK, Peng SJ, Wang XG, Li Z, Elnour IE, Huang YZ, Lan XY, Chen H. Transcriptional regulation of the bovine FGFR1 gene facilitates myoblast proliferation under hypomethylation of the promoter. J Cell Physiol 2020; 235:8667-8678. [PMID: 32324257 DOI: 10.1002/jcp.29711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
DNA methylation, which can affect the expression level of genes, is one of the most vital epigenetic modifications in mammals. Fibroblast growth factor receptor 1 (FGFR1) plays an important role in muscle development; however, DNA methylation of the FGFR1 promoter has not been studied to date in cattle. Our study focused on methylation of the FGFR1 promoter and its effect on bovine myoblast proliferation and differentiation. We identified the FGFR1 core promoter by using luciferase reporter assays; we then studied FGFR1 expression by reverse transcription quantitative polymerase chain reaction, and the methylation pattern in the FGFR1 core promoter by bisulfite sequencing polymerase chain reaction in bovine muscle tissue at three different developmental stages. We used RNAi strategy to investigate the function of FGFR1 in myoblast proliferation and differentiation. Results showed that the FGFR1 core promoters were located at the R2 (-509 to ~-202 bp) and R4 (-1295 to ~-794 bp) regions upstream of the FGFR1 gene. FGFR1 expression level was negatively associated with the degree of methylation of the FGFR1 core promoter during the developmental process. In addition, we found that FGFR1 can promote myoblast proliferation, but had no effect on myoblast differentiation. In conclusion, our results suggest that FGFR1 can promote myoblast proliferation and its transcription can be regulated by the methylation level of the core promoter. Our findings provide a mechanistic basis for the improvement of animal breeding.
Collapse
Affiliation(s)
- Jie Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiu-Kai Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Shu-Jun Peng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao-Gang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhuang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ibrahim-Elsaeid Elnour
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.,Faculty of Veterinary Science, University of Nyala, Nyala, Sudan
| | - Yong-Zhen Huang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xian-Yong Lan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
35
|
Abstract
Maturation is the last phase of heart development that prepares the organ for strong, efficient, and persistent pumping throughout the mammal's lifespan. This process is characterized by structural, gene expression, metabolic, and functional specializations in cardiomyocytes as the heart transits from fetal to adult states. Cardiomyocyte maturation gained increased attention recently due to the maturation defects in pluripotent stem cell-derived cardiomyocyte, its antagonistic effect on myocardial regeneration, and its potential contribution to cardiac disease. Here, we review the major hallmarks of ventricular cardiomyocyte maturation and summarize key regulatory mechanisms that promote and coordinate these cellular events. With advances in the technical platforms used for cardiomyocyte maturation research, we expect significant progress in the future that will deepen our understanding of this process and lead to better maturation of pluripotent stem cell-derived cardiomyocyte and novel therapeutic strategies for heart disease.
Collapse
Affiliation(s)
- Yuxuan Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - William Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
36
|
van Ouwerkerk AF, Bosada FM, Liu J, Zhang J, van Duijvenboden K, Chaffin M, Tucker NR, Pijnappels D, Ellinor PT, Barnett P, de Vries AAF, Christoffels VM. Identification of Functional Variant Enhancers Associated With Atrial Fibrillation. Circ Res 2020; 127:229-243. [PMID: 32248749 DOI: 10.1161/circresaha.119.316006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
RATIONALE Genome-wide association studies have identified a large number of common variants (single-nucleotide polymorphisms) associated with atrial fibrillation (AF). These variants are located mainly in noncoding regions of the genome and likely include variants that modulate the function of transcriptional regulatory elements (REs) such as enhancers. However, the actual REs modulated by variants and the target genes of such REs remain to be identified. Thus, the biological mechanisms by which genetic variation promotes AF has thus far remained largely unexplored. OBJECTIVE To identify REs in genome-wide association study loci that are influenced by AF-associated variants. METHODS AND RESULTS We screened 2.45 Mbp of human genomic DNA containing 12 strongly AF-associated loci for RE activity using self-transcribing active regulatory region sequencing and a recently generated monoclonal line of conditionally immortalized rat atrial myocytes. We identified 444 potential REs, 55 of which contain AF-associated variants (P<10-8). Subsequently, using an adaptation of the self-transcribing active regulatory region sequencing approach, we identified 24 variant REs with allele-specific regulatory activity. By mining available chromatin conformation data, the possible target genes of these REs were mapped. To define the physiological function and target genes of such REs, we deleted the orthologue of an RE containing noncoding variants in the Hcn4 (potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4) locus of the mouse genome. Mice heterozygous for the RE deletion showed bradycardia, sinus node dysfunction, and selective loss of Hcn4 expression. CONCLUSIONS We have identified REs at multiple genetic loci for AF and found that loss of an RE at the HCN4 locus results in sinus node dysfunction and reduced gene expression. Our approach can be broadly applied to facilitate the identification of human disease-relevant REs and target genes at cardiovascular genome-wide association studies loci.
Collapse
Affiliation(s)
- Antoinette F van Ouwerkerk
- From the Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, the Netherlands (A.F.v.O., F.M.B., K.v.D., P.B., V.M.C.)
| | - Fernanda M Bosada
- From the Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, the Netherlands (A.F.v.O., F.M.B., K.v.D., P.B., V.M.C.)
| | - Jia Liu
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands (J.L., J.Z., D.P., A.A.F.d.V.).,Netherlands Heart Institute, Holland Heart House, Utrecht (J.L., J.Z., D.P., A.A.F.d.V.)
| | - Juan Zhang
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands (J.L., J.Z., D.P., A.A.F.d.V.).,Netherlands Heart Institute, Holland Heart House, Utrecht (J.L., J.Z., D.P., A.A.F.d.V.)
| | - Karel van Duijvenboden
- From the Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, the Netherlands (A.F.v.O., F.M.B., K.v.D., P.B., V.M.C.)
| | - Mark Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (M.C., N.R.T., P.T.E.)
| | - Nathan R Tucker
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (M.C., N.R.T., P.T.E.).,Cardiovascular Research Center, Massachusetts General Hospital, Boston (N.R.T., P.T.E.)
| | - Daniel Pijnappels
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands (J.L., J.Z., D.P., A.A.F.d.V.).,Netherlands Heart Institute, Holland Heart House, Utrecht (J.L., J.Z., D.P., A.A.F.d.V.)
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (M.C., N.R.T., P.T.E.).,Cardiovascular Research Center, Massachusetts General Hospital, Boston (N.R.T., P.T.E.)
| | - Phil Barnett
- From the Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, the Netherlands (A.F.v.O., F.M.B., K.v.D., P.B., V.M.C.)
| | - Antoine A F de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands (J.L., J.Z., D.P., A.A.F.d.V.).,Netherlands Heart Institute, Holland Heart House, Utrecht (J.L., J.Z., D.P., A.A.F.d.V.)
| | - Vincent M Christoffels
- From the Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, the Netherlands (A.F.v.O., F.M.B., K.v.D., P.B., V.M.C.)
| |
Collapse
|
37
|
Pijuan-Sala B, Wilson NK, Xia J, Hou X, Hannah RL, Kinston S, Calero-Nieto FJ, Poirion O, Preissl S, Liu F, Göttgens B. Single-cell chromatin accessibility maps reveal regulatory programs driving early mouse organogenesis. Nat Cell Biol 2020; 22:487-497. [PMID: 32231307 PMCID: PMC7145456 DOI: 10.1038/s41556-020-0489-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/20/2020] [Indexed: 11/29/2022]
Abstract
During mouse embryonic development, pluripotent cells rapidly divide and diversify, yet the regulatory programs that define the cell repertoire for each organ remain ill-defined. To delineate comprehensive chromatin landscapes during early organogenesis, we mapped chromatin accessibility in 19,453 single nuclei from mouse embryos at 8.25 days post-fertilization. Identification of cell-type-specific regions of open chromatin pinpointed two TAL1-bound endothelial enhancers, which we validated using transgenic mouse assays. Integrated gene expression and transcription factor motif enrichment analyses highlighted cell-type-specific transcriptional regulators. Subsequent in vivo experiments in zebrafish revealed a role for the ETS factor FEV in endothelial identity downstream of ETV2 (Etsrp in zebrafish). Concerted in vivo validation experiments in mouse and zebrafish thus illustrate how single-cell open chromatin maps, representative of a mammalian embryo, provide access to the regulatory blueprint for mammalian organogenesis.
Collapse
Affiliation(s)
- Blanca Pijuan-Sala
- Department of Haematology, University of Cambridge, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Nicola K Wilson
- Department of Haematology, University of Cambridge, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Jun Xia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaomeng Hou
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Rebecca L Hannah
- Department of Haematology, University of Cambridge, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Sarah Kinston
- Department of Haematology, University of Cambridge, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Fernando J Calero-Nieto
- Department of Haematology, University of Cambridge, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Olivier Poirion
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Sebastian Preissl
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Berthold Göttgens
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
38
|
Chen P, Li Z, Nie J, Wang H, Yu B, Wen Z, Sun Y, Shi X, Jin L, Wang DW. MYH7B variants cause hypertrophic cardiomyopathy by activating the CaMK-signaling pathway. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1347-1362. [DOI: 10.1007/s11427-019-1627-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 02/27/2020] [Indexed: 12/12/2022]
|
39
|
Wan X, Belanger K, Widen SG, Kuyumcu-Martinez MN, Garg NJ. Genes of the cGMP-PKG-Ca 2+ signaling pathway are alternatively spliced in cardiomyopathy: Role of RBFOX2. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165620. [PMID: 31778749 PMCID: PMC6954967 DOI: 10.1016/j.bbadis.2019.165620] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022]
Abstract
Aberrations in the cGMP-PKG-Ca2+ pathway are implicated in cardiovascular complications of diverse etiologies, though involved molecular mechanisms are not understood. We performed RNA-Seq analysis to profile global changes in gene expression and exon splicing in Chagas disease (ChD) murine myocardium. Ingenuity-Pathway-Analysis of transcriptome dataset identified 26 differentially expressed genes associated with increased mobilization and cellular levels of Ca2+ in ChD hearts. Mixture-of-isoforms and Enrichr KEGG pathway analyses of the RNA-Seq datasets from ChD (this study) and diabetic (previous study) murine hearts identified alternative splicing (AS) in eleven genes (Arhgef10, Atp2b1, Atp2a3, Cacna1c, Itpr1, Mef2a, Mef2d, Pde2a, Plcb1, Plcb4, and Ppp1r12a) of the cGMP-PKG-Ca2+ pathway in diseased hearts. AS of these genes was validated by an exon exclusion-inclusion assay. Further, Arhgef10, Atp2b1, Mef2a, Mef2d, Plcb1, and Ppp1r12a genes consisted RBFOX2 (RNA-binding protein) binding-site clusters, determined by analyzing the RBFOX2 CLIP-Seq dataset. H9c2 rat heart cells transfected with Rbfox2 (vs. scrambled) siRNA confirmed that expression of Rbfox2 is essential for proper exon splicing of genes of the cGMP-PKG-Ca2+ pathway. We conclude that changes in gene expression may influence the Ca2+ mobilization pathway in ChD, and AS impacts the genes involved in cGMP/PKG/Ca2+ signaling pathway in ChD and diabetes. Our findings suggest that ChD patients with diabetes may be at increased risk of cardiomyopathy and heart failure and provide novel ways to restore cGMP-PKG regulated signaling networks via correcting splicing patterns of key factors using oligonucleotide-based therapies for the treatment of cardiovascular complications.
Collapse
Affiliation(s)
- Xianxiu Wan
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, 77555-1070, TX, United States of America
| | - KarryAnne Belanger
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 77555, TX, United States of America
| | - Steven G Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 77555, TX, United States of America
| | - Muge N Kuyumcu-Martinez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 77555, TX, United States of America.
| | - Nisha J Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, 77555-1070, TX, United States of America; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, 77555, TX, United States of America.
| |
Collapse
|
40
|
Programming Skeletal Muscle Metabolic Flexibility in Offspring of Male Rats in Response to Maternal Consumption of Slow Digesting Carbohydrates during Pregnancy. Nutrients 2020; 12:nu12020528. [PMID: 32092940 PMCID: PMC7071425 DOI: 10.3390/nu12020528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 01/20/2023] Open
Abstract
Skeletal muscle plays a relevant role in metabolic flexibility and fuel usage and the associated muscle metabolic inflexibility due to high-fat diets contributing to obesity and type 2 diabetes. Previous research from our group indicates that a high-fat and rapid-digesting carbohydrate diet during pregnancy promotes an excessive adipogenesis and also increases the risk of non-alcoholic fatty liver disease in the offspring. This effect can be counteracted by diets containing carbohydrates with similar glycemic load but lower digestion rates. To address the role of the skeletal muscle in these experimental settings, pregnant rats were fed high-fat diets containing carbohydrates with similar glycemic load but different digestion rates, a high fat containing rapid-digesting carbohydrates diet (HF/RD diet) or a high fat containing slow-digesting carbohydrates diet (HF/SD diet). After weaning, male offspring were fed a standard diet for 3 weeks (weaning) or 10 weeks (adolescence) and the impact of the maternal HF/RD and HF/SD diets on the metabolism, signaling pathways and muscle transcriptome was analyzed. The HF/SD offspring displayed better muscle features compared with the HF/RD group, showing a higher muscle mass, myosin content and differentiation markers that translated into a greater grip strength. In the HF/SD group, metabolic changes such as a higher expression of fatty acids (FAT/CD36) and glucose (GLUT4) transporters, an enhanced glycogen content, as well as changes in regulatory enzymes such as muscle pyruvate kinase and pyruvate dehydrogenase kinase 4 were found, supporting an increased muscle metabolic flexibility and improved muscle performance. The analysis of signaling pathways was consistent with a better insulin sensitivity in the muscle of the HF/SD group. Furthermore, increased expression of genes involved in pathways leading to muscle differentiation, muscle mass regulation, extracellular matrix content and insulin sensitivity were detected in the HF/SD group when compared with HF/RD animals. In the HF/SD group, the upregulation of the ElaV1/HuR gene could be one of the main regulators in the positive effects of the diet in early programming on the offspring. The long-lasting programming effects of the HF/SD diet during pregnancy may depend on a coordinated gene regulation, modulation of signaling pathways and metabolic flexibility that lead to an improved muscle functionality. The dietary early programming associated to HF/SD diet has synergic and positive crosstalk effects in several tissues, mainly muscle, liver and adipose tissue, contributing to maintain the whole body homeostasis in the offspring.
Collapse
|
41
|
Ouyang H, Yu J, Chen X, Wang Z, Nie Q. A novel transcript of MEF2D promotes myoblast differentiation and its variations associated with growth traits in chicken. PeerJ 2020; 8:e8351. [PMID: 32117604 PMCID: PMC7006513 DOI: 10.7717/peerj.8351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/04/2019] [Indexed: 12/28/2022] Open
Abstract
Background Development of skeletal muscle is closely related to broiler production traits. The myocyte-specific enhancer binding factor (MEF) 2D gene (MEF2D) and its variant transcripts play important parts in myogenesis. Methods To identify the transcript variants of chicken MEF2D gene and their function, this study cloned chicken MEF2D gene and identified its transcript variants from different tissue samples. The expression levels of different transcripts of MEF2D gene in different tissues and different periods were measured, and their effects on myoblast proliferation and differentiation were investigated. Variations in MEF2D were identified and association analysis with chicken production traits carried out. Results Four novel transcript variants of MEF2D were obtained, all of which contained highly conserved sequences, including MADS-Box and MEF2-Domain functional regions. Transcript MEF2D-V4 was expressed specifically in muscle, and its expression was increased during embryonic muscle development. The MEF2D-V4 could promote differentiation of chicken myoblasts and its expression was regulated by RBFOX2. The single nucleotide polymorphism g.36186C > T generated a TAG stop codon, caused MEF2D-V4 to terminate translation early, and was associated with several growth traits, especially on early body weight. Conclusion We cloned the muscle-specific transcript of MEF2D and preliminarily revealed its role in embryonic muscle development.
Collapse
Affiliation(s)
- Hongjia Ouyang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.,Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, China
| | - Jiao Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiaolan Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhijun Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qinghua Nie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
42
|
Chen R, Lei S, Jiang T, Zeng J, Zhou S, She Y. Roles of lncRNAs and circRNAs in regulating skeletal muscle development. Acta Physiol (Oxf) 2020; 228:e13356. [PMID: 31365949 DOI: 10.1111/apha.13356] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/26/2019] [Accepted: 07/27/2019] [Indexed: 02/06/2023]
Abstract
The multistep biological process of myogenesis is regulated by a variety of myoblast regulators, such as myogenic differentiation antigen, myogenin, myogenic regulatory factor, myocyte enhancer factor2A-D and myosin heavy chain. Proliferation and differentiation during skeletal muscle myogenesis contribute to the physiological function of muscles. Certain non-coding RNAs, including long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are involved in the regulation of muscle development, and the aberrant expressions of lncRNAs and circRNAs are associated with muscular diseases. In this review, we summarize the recent advances concerning the roles of lncRNAs and circRNAs in regulating the developmental aspects of myogenesis. These findings have remarkably broadened our understanding of the gene regulation mechanisms governing muscle proliferation and differentiation, which makes it more feasible to design novel preventive, diagnostic and therapeutic strategies for muscle disorders.
Collapse
Affiliation(s)
- Rui Chen
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute Guangdong Second Provincial General Hospital Guangzhou China
| | - Si Lei
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute Guangdong Second Provincial General Hospital Guangzhou China
| | - Ting Jiang
- Department of Radiology, The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Jie Zeng
- Department of Medical Ultrasonics, The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Shanyao Zhou
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute Guangdong Second Provincial General Hospital Guangzhou China
| | - Yanling She
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute Guangdong Second Provincial General Hospital Guangzhou China
| |
Collapse
|
43
|
Barrientos G, Llanos P, Basualto-Alarcón C, Estrada M. Androgen-Regulated Cardiac Metabolism in Aging Men. Front Endocrinol (Lausanne) 2020; 11:316. [PMID: 32499759 PMCID: PMC7243157 DOI: 10.3389/fendo.2020.00316] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
The prevalence of cardiovascular mortality is higher in men than in age-matched premenopausal women. Gender differences are linked to circulating sex-related steroid hormone levels and their cardio-specific actions, which are critical factors involved in the prevalence and features of age-associated cardiovascular disease. In women, estrogens have been described as cardioprotective agents, while in men, testosterone is the main sex steroid hormone. The effects of testosterone as a metabolic regulator and cardioprotective agent in aging men are poorly understood. With advancing age, testosterone levels gradually decrease in men, an effect associated with increasing fat mass, decrease in lean body mass, dyslipidemia, insulin resistance and adjustment in energy substrate metabolism. Aging is associated with a decline in metabolism, characterized by modifications in cardiac function, excitation-contraction coupling, and lower efficacy to generate energy. Testosterone deficiency -as found in elderly men- rapidly becomes an epidemic condition, associated with prominent cardiometabolic disorders. Therefore, it is highly probable that senior men showing low testosterone levels will display symptoms of androgen deficiency, presenting an unfavorable metabolic profile and increased cardiovascular risk. Moreover, recent reports establish that testosterone replacement improves cardiomyocyte bioenergetics, increases glucose metabolism and reduces insulin resistance in elderly men. Thus, testosterone-related metabolic signaling and gene expression may constitute relevant therapeutic target for preventing, or treating, age- and gender-related cardiometabolic diseases in men. Here, we will discuss the impact of current evidence showing how cardiac metabolism is regulated by androgen levels in aging men.
Collapse
Affiliation(s)
- Genaro Barrientos
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Universidad de Chile, Santiago, Chile
| | - Paola Llanos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Universidad de Chile, Santiago, Chile
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas (ICOD), Universidad de Chile, Santiago, Chile
| | - Carla Basualto-Alarcón
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile
- Departamento de Anatomía y Medicina Legal, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
- *Correspondence: Manuel Estrada
| |
Collapse
|
44
|
Racioppi C, Wiechecki KA, Christiaen L. Combinatorial chromatin dynamics foster accurate cardiopharyngeal fate choices. eLife 2019; 8:49921. [PMID: 31746740 PMCID: PMC6952182 DOI: 10.7554/elife.49921] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022] Open
Abstract
During embryogenesis, chromatin accessibility profiles control lineage-specific gene expression by modulating transcription, thus impacting multipotent progenitor states and subsequent fate choices. Subsets of cardiac and pharyngeal/head muscles share a common origin in the cardiopharyngeal mesoderm, but the chromatin landscapes that govern multipotent progenitors competence and early fate choices remain largely elusive. Here, we leveraged the simplicity of the chordate model Ciona to profile chromatin accessibility through stereotyped transitions from naive Mesp+ mesoderm to distinct fate-restricted heart and pharyngeal muscle precursors. An FGF-Foxf pathway acts in multipotent progenitors to establish cardiopharyngeal-specific patterns of accessibility, which govern later heart vs. pharyngeal muscle-specific expression profiles, demonstrating extensive spatiotemporal decoupling between early cardiopharyngeal enhancer accessibility and late cell-type-specific activity. We found that multiple cis-regulatory elements, with distinct chromatin accessibility profiles and motif compositions, are required to activate Ebf and Tbx1/10, two key determinants of cardiopharyngeal fate choices. We propose that these 'combined enhancers' foster spatially and temporally accurate fate choices, by increasing the repertoire of regulatory inputs that control gene expression, through either accessibility and/or activity.
Collapse
Affiliation(s)
- Claudia Racioppi
- Center for Developmental Genetics, Department of Biology, New York University, New York, United States
| | - Keira A Wiechecki
- Center for Developmental Genetics, Department of Biology, New York University, New York, United States
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of Biology, New York University, New York, United States
| |
Collapse
|
45
|
Li C, Sun XN, Chen BY, Zeng MR, Du LJ, Liu T, Gu HH, Liu Y, Li YL, Zhou LJ, Zheng XJ, Zhang YY, Zhang WC, Liu Y, Shi C, Shao S, Shi XR, Yi Y, Liu X, Wang J, Auwerx J, Wang ZV, Jia F, Li RG, Duan SZ. Nuclear receptor corepressor 1 represses cardiac hypertrophy. EMBO Mol Med 2019; 11:e9127. [PMID: 31532577 PMCID: PMC6835202 DOI: 10.15252/emmm.201809127] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 01/24/2023] Open
Abstract
The function of nuclear receptor corepressor 1 (NCoR1) in cardiomyocytes is unclear, and its physiological and pathological implications are unknown. Here, we found that cardiomyocyte‐specific NCoR1 knockout (CMNKO) mice manifested cardiac hypertrophy at baseline and had more severe cardiac hypertrophy and dysfunction after pressure overload. Knockdown of NCoR1 exacerbated whereas overexpression mitigated phenylephrine‐induced cardiomyocyte hypertrophy. Mechanistic studies revealed that myocyte enhancer factor 2a (MEF2a) and MEF2d mediated the effects of NCoR1 on cardiomyocyte hypertrophy. The receptor interaction domains (RIDs) of NCoR1 interacted with MEF2a to repress its transcriptional activity. Furthermore, NCoR1 formed a complex with MEF2a and class IIa histone deacetylases (HDACs) to suppress hypertrophy‐related genes. Finally, overexpression of RIDs of NCoR1 in the heart attenuated cardiac hypertrophy and dysfunction induced by pressure overload. In conclusion, NCoR1 cooperates with MEF2 and HDACs to repress cardiac hypertrophy. Targeting NCoR1 and the MEF2/HDACs complex may be an attractive therapeutic strategy to tackle pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Chao Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xue-Nan Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Meng-Ru Zeng
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Hui-Hui Gu
- Shanghai Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Lin Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Lu-Jun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xiao-Jun Zheng
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Yao Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wu-Chang Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chaoji Shi
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shuai Shao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Rui Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Yi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Wang
- Shanghai Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Feng Jia
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
46
|
Mucenski ML, Mahoney R, Adam M, Potter AS, Potter SS. Single cell RNA-seq study of wild type and Hox9,10,11 mutant developing uterus. Sci Rep 2019; 9:4557. [PMID: 30872674 PMCID: PMC6418183 DOI: 10.1038/s41598-019-40923-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
The uterus is a remarkable organ that must guard against infections while maintaining the ability to support growth of a fetus without rejection. The Hoxa10 and Hoxa11 genes have previously been shown to play essential roles in uterus development and function. In this report we show that the Hoxa9,10,11, Hoxc9,10,11, Hoxd9,10,11 genes play a redundant role in the formation of uterine glands. In addition, we use single cell RNA-seq to create a high resolution gene expression atlas of the developing wild type mouse uterus. Cell types and subtypes are defined, for example dividing endothelial cells into arterial, venous, capillary, and lymphatic, while epithelial cells separate into luminal and glandular subtypes. Further, a surprising heterogeneity of stromal and myocyte cell types are identified. Transcription factor codes and ligand/receptor interactions are characterized. We also used single cell RNA-seq to globally define the altered gene expression patterns in all developing uterus cell types for two Hox mutants, with 8 or 9 mutant Hox genes. The mutants show a striking disruption of Wnt signaling as well as the Cxcl12/Cxcr4 ligand/receptor axis.
Collapse
Affiliation(s)
- Michael L Mucenski
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Robert Mahoney
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Andrew S Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
47
|
Prünster MM, Ricci L, Brown FD, Tiozzo S. Modular co-option of cardiopharyngeal genes during non-embryonic myogenesis. EvoDevo 2019; 10:3. [PMID: 30867897 PMCID: PMC6399929 DOI: 10.1186/s13227-019-0116-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/15/2019] [Indexed: 01/03/2023] Open
Abstract
Background In chordates, cardiac and body muscles arise from different embryonic origins. In addition, myogenesis can be triggered in adult organisms, during asexual development or regeneration. In non-vertebrate chordates like ascidians, muscles originate from embryonic precursors regulated by a conserved set of genes that orchestrate cell behavior and dynamics during development. In colonial ascidians, besides embryogenesis and metamorphosis, an adult can propagate asexually via blastogenesis, skipping embryo and larval stages, and form anew the adult body, including the complete body musculature. Results To investigate the cellular origin and mechanisms that trigger non-embryonic myogenesis, we followed the expression of ascidian myogenic genes during Botryllus schlosseri blastogenesis and reconstructed the dynamics of muscle precursors. Based on the expression dynamics of Tbx1/10, Ebf, Mrf, Myh3 for body wall and of FoxF, Tbx1/10, Nk4, Myh2 for heart development, we show that the embryonic factors regulating myogenesis are only partially co-opted in blastogenesis, and that markers for muscle precursors are expressed in two separate domains: the dorsal tube and the ventral mesenchyma. Conclusions Regardless of the developmental pathway, non-embryonic myogenesis shares a similar molecular and anatomical setup as embryonic myogenesis, but implements a co-option and loss of molecular modules. We then propose that the cellular precursors contributing to heart and body muscles may have different origins and may be coordinated by different developmental pathways. Electronic supplementary material The online version of this article (10.1186/s13227-019-0116-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria Mandela Prünster
- 1Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), CNRS, Sorbonne Université, 06230 Villefranche sur Mer, France
| | - Lorenzo Ricci
- 1Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), CNRS, Sorbonne Université, 06230 Villefranche sur Mer, France.,2Department of Organismic and Evolutionary Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138 USA
| | - Federico D Brown
- 3Departamento de Zoologia, Instituto Biociências, Universidade de São Paulo, São Paulo, SP CEP 05508-090 Brazil.,4Centro de Biologia Marinha (CEBIMar), Universidade de São Paulo, São Sebastião, SP CEP 11612-109 Brazil
| | - Stefano Tiozzo
- 1Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), CNRS, Sorbonne Université, 06230 Villefranche sur Mer, France
| |
Collapse
|
48
|
Kannan S, Kwon C. Regulation of cardiomyocyte maturation during critical perinatal window. J Physiol 2019; 598:2941-2956. [PMID: 30571853 DOI: 10.1113/jp276754] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022] Open
Abstract
A primary limitation in the use of pluripotent stem cell-derived cardiomyocytes (PSC-CMs) for both patient health and scientific investigation is the failure of these cells to achieve full functional maturity. In vivo, cardiomyocytes undergo numerous adaptive structural, functional and metabolic changes during maturation. By contrast, PSC-CMs fail to fully undergo these developmental processes, instead remaining arrested at an embryonic stage of maturation. There is thus a significant need to understand the biological processes underlying proper CM maturation in vivo. Here, we discuss what is known regarding the initiation and coordination of CM maturation. We postulate that there is a critical perinatal window, ranging from embryonic day 18.5 to postnatal day 14 in mice, in which the maturation process is exquisitely sensitive to perturbation. While the initiation mechanisms of this process are unknown, it is increasingly clear that maturation proceeds through interconnected regulatory circuits that feed into one another to coordinate concomitant structural, functional and metabolic CM maturation. We highlight PGC1α, SRF and the MEF2 family as transcription factors that may potentially mediate this cross-talk. We lastly discuss several emerging technologies that will facilitate future studies into the mechanisms of CM maturation. Further study will not only produce a better understanding of its key processes, but provide practical insights into developing a robust strategy to produce mature PSC-CMs.
Collapse
Affiliation(s)
- Suraj Kannan
- Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| | - Chulan Kwon
- Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| |
Collapse
|
49
|
Materna SC, Sinha T, Barnes RM, Lammerts van Bueren K, Black BL. Cardiovascular development and survival require Mef2c function in the myocardial but not the endothelial lineage. Dev Biol 2018; 445:170-177. [PMID: 30521808 DOI: 10.1016/j.ydbio.2018.12.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 01/27/2023]
Abstract
MEF2C is a member of the highly conserved MEF2 family of transcription factors and is a key regulator of cardiovascular development. In mice, Mef2c is expressed in the developing heart and vasculature, including the endothelium. Loss of Mef2c function in germline knockout mice leads to early embryonic demise and profound developmental abnormalities in the cardiovascular system. Previous attempts to uncover the cause of embryonic lethality by specifically disrupting Mef2c function in the heart or vasculature failed to recapitulate the global Mef2c knockout phenotype and instead resulted in relatively minor defects that did not compromise viability or result in significant cardiovascular defects. However, previous studies examined the requirement of Mef2c in the myocardial and endothelial lineages using Cre lines that begin to be expressed after the expression of Mef2c has already commenced. Here, we tested the requirement of Mef2c in the myocardial and endothelial lineages using conditional knockout approaches in mice with Cre lines that deleted Mef2c prior to onset of its expression in embryonic development. We found that deletion of Mef2c in the early myocardial lineage using Nkx2-5Cre resulted in cardiac and vascular abnormalities that were indistinguishable from the defects in the global Mef2c knockout. In contrast, early deletion of Mef2c in the vascular endothelium using an Etv2::Cre line active prior to the onset of Mef2c expression resulted in viable offspring that were indistinguishable from wild type controls with no overt defects in vascular development, despite nearly complete early deletion of Mef2c in the vascular endothelium. Thus, these studies support the idea that the requirement of MEF2C for vascular development is secondary to its requirement in the heart and suggest that the observed failure in vascular remodeling in Mef2c knockout mice results from defective heart function.
Collapse
Affiliation(s)
- Stefan C Materna
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Molecular Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA 95343, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ralston M Barnes
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly Lammerts van Bueren
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
50
|
MicroRNA-499a-5p Promotes Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells to Cardiomyocytes. Appl Biochem Biotechnol 2018; 186:245-255. [PMID: 29574510 DOI: 10.1007/s12010-018-2734-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 03/12/2018] [Indexed: 01/09/2023]
Abstract
Since the adult mammalian heart has limited regenerative capacity, cardiac trauma, disease, and aging cause permanent loss of contractile tissue. This has fueled the development of stem cell-based strategies to provide the damaged heart with new cardiomyocytes. Bone marrow-derived mesenchymal stem cells (BM-MSCs) are capable of self-renewal and differentiation into cardiomyocytes, albeit inefficiently. MicroRNAs (miRNAs, miRs) are non-coding RNAs that have the potential to control stem cell fate decisions and are employed in cardiac regeneration and repair. In this study, we tested the hypothesis that overexpression of miR-499a induces cardiomyogenic differentiation in BM-MSCs. Human BM-MSCs (hBM-MSCs) were transduced with lentiviral vectors encoding miR-499a-3p or miR-499a-5p and analyzed by immunostaining and western blotting methods 14 days post-transduction. MiR-499a-5p-transduced cells adopted a polygonal/rod-shaped (myocyte-like) phenotype and showed an increase in the expression of the cardiomyocyte markers α-actinin and cTnI, as cardiogenic differentiation markers. These results indicate that miR-499a-5p overexpression promotes the cardiomyogenic differentiation of hBM-MSCs and may thereby increase their therapeutic efficiency in cardiac regeneration.
Collapse
|