1
|
Leerberg DM, Avillion GB, Priya R, Stainier DY, Yelon D. Regionalized regulation of actomyosin organization influences cardiomyocyte cell shape changes during chamber curvature formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631779. [PMID: 39829878 PMCID: PMC11741281 DOI: 10.1101/2025.01.07.631779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Cardiac chambers emerge from a heart tube that balloons and bends to create expanded ventricular and atrial structures, each containing a convex outer curvature (OC) and a recessed inner curvature (IC). A comprehensive understanding of the cellular and molecular mechanisms underlying the formation of these characteristic curvatures remains lacking. Here, we demonstrate in zebrafish that the initially similar populations of OC and IC ventricular cardiomyocytes diverge in the organization of their actomyosin cytoskeleton and subsequently acquire distinct OC and IC cell shapes. Altering actomyosin dynamics hinders cell shape changes in the OC, and mosaic analyses indicate that actomyosin regulates cardiomyocyte shape in a cell-autonomous manner. Additionally, both blood flow and the transcription factor Tbx5a influence the basal enrichment of actomyosin and squamous cell morphologies in the OC. Together, our findings demonstrate that intrinsic and extrinsic factors intersect to control actomyosin organization in OC cardiomyocytes, which in turn promotes the cell shape changes that drive curvature morphogenesis.
Collapse
Affiliation(s)
- Dena M. Leerberg
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gabriel B. Avillion
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Rashmi Priya
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Didier Y.R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Deborah Yelon
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
2
|
Shireen H, Batool F, Khatoon H, Parveen N, Sehar NU, Hussain I, Ali S, Abbasi AA. Predicting genome-wide tissue-specific enhancers via combinatorial transcription factor genomic occupancy analysis. FEBS Lett 2025; 599:100-119. [PMID: 39367524 DOI: 10.1002/1873-3468.15030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 10/06/2024]
Abstract
Enhancers are non-coding cis-regulatory elements crucial for transcriptional regulation. Mutations in enhancers can disrupt gene regulation, leading to disease phenotypes. Identifying enhancers and their tissue-specific activity is challenging due to their lack of stereotyped sequences. This study presents a sequence-based computational model that uses combinatorial transcription factor (TF) genomic occupancy to predict tissue-specific enhancers. Trained on diverse datasets, including ENCODE and Vista enhancer browser data, the model predicted 25 000 forebrain-specific cis-regulatory modules (CRMs) in the human genome. Validation using biochemical features, disease-associated SNPs, and in vivo zebrafish analysis confirmed its effectiveness. This model aids in predicting enhancers lacking well-characterized chromatin features, complementing experimental approaches in tissue-specific enhancer discovery.
Collapse
Affiliation(s)
- Huma Shireen
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fatima Batool
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hizran Khatoon
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Nazia Parveen
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Noor Us Sehar
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Irfan Hussain
- Centre for Regenerative Medicine and Stem Cells Research, Agha Khan University hospital, Karachi, Pakistan
| | - Shahid Ali
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL, USA
| | - Amir Ali Abbasi
- National Center for Bioinformatics, Program of Comparative and Evolutionary Genomics, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
3
|
Wu T, Chen Z, Zhang Z, Zhou X, Gu Y, Dinenno FA, Chen J. RBPMS and RBPMS2 Cooperate to Safeguard Cardiac Splicing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622565. [PMID: 39574760 PMCID: PMC11581027 DOI: 10.1101/2024.11.07.622565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Background Mutations in cardiac splicing factors (SFs) cause cardiomyopathy and congenital heart disease, underscoring the critical role of SFs in cardiac development and disease. Cardiac SFs are implicated to cooperatively regulate the splicing of essential cardiac genes, but the functional importance of their collaboration remains unclear. RNA Binding Protein with Multiple Splicing (RBPMS) and RBPMS2 are SFs involved in heart development and exhibit similar splicing regulatory activities in vitro , but it is unknown whether they cooperate to regulate splicing in vivo . Methods Rbpms and Rbpms2 single or double cardiomyocyte (CM)-specific knockout (KO) mice were generated and analyzed for cardiac phenotypes. RNA sequencing was performed to assess gene expression and splicing changes in single and double KOs. In silico analyses were used to dissect the mechanisms underlying distinct and overlapping roles of RBPMS and RBPMS2 in heart development. Results Mice lacking both RBPMS and RBPMS2 in CMs died before embryonic day 13.5 and developed sarcomere disarray, whereas Rbpms or Rbpms2 single CM-specific KO mice had normal sarcomere assembly and survived to adulthood. Defective sarcomere assembly is likely owing to the widespread mis-splicing of genes essential for cardiac contraction in double KO mice, underscoring the overlapping role of RBPMS and RBPMS2 in splicing regulation. Mechanistically, we found RBPMS and RBPMS collectively promote cardiac splicing program while repressing non-cardiac splicing programs. Moreover, RNA splicing maps suggested that the binding location of RBPMS and RBPMS2 on pre-mRNA dictates whether they function as splicing activators or repressors. Lastly, the requirement for RBPMS and/or RBPMS2 for splicing regulation arises from intrinsic features of the target exons. Conclusions Our results demonstrate that RBPMS and RBPMS2 work in concert to safeguard the splicing of genes essential for cardiac contraction, highlighting the importance of SF collaboration in maintaining cardiac splicing signature, which should be taken into consideration when devising future therapeutic approaches through modulating the activity of SFs. Novelty and Significance What Is Known?: Mutations in cardiac splicing factors (SFs) cause cardiomyopathy and congenital heart disease, and the splicing of cardiac genes is regulated by multiple SFs. However, the functional importance of the collaboration among specific cardiac SFs is unknown.RBPMS has emerged as a cardiac SF for sarcomere genes but is not required for sarcomere assembly. RBPMS2 can substitute RBPMS in in vitro splicing assays, yet its role in mammalian cardiomyocytes (CMs) remains unclear. What New Information Does This Article Contribute?: RBPMS and RBPMS2 have both distinct and overlapping roles in CMs.RBPMS and RBPMS2 collectively contribute to the maintenance of cardiac splicing program, which is essential for sarcomere assembly and embryonic survival.RNA splicing map of RBPMS and RBPMS2 reveals that they can function either as splicing activators or repressors, depending on their binding locations on pre-mRNA. This study provides compelling evidence of cooperation between cardiac splicing factors during heart development, which, to our knowledge, has not been demonstrated in vivo . Rbpms and Rbpms2 CM-specific double KO mice die in utero and exhibit sarcomere disarray, whereas single KO mice survive to adulthood with normal sarcomere structure but manifest distinct cardiac phenotypes, suggesting RBPMS and RBPMS2 possess both distinct and overlapping functions in CMs. Although mis-splicing in cardiac genes can be seen in all three KOs, the splicing signature of double KO hearts drastically shifts towards non-cardiac tissues, including more prominent mis-splicing in genes related to cardiac contractile function. Our study further reveals that the splicing regulation of RBPMS and RBPMS2 has the characteristics of "positional effects", i.e., the binding location on pre-mRNA dictates whether they function as splicing activators or repressors; and the intrinsic features of the target exon determine the requirement for one or two RBPMS proteins for splicing regulation. Our study sheds light on the functional importance of cardiac SF cooperation in maintaining cardiac splicing signature during heart development.
Collapse
|
4
|
Kliesmete Z, Orchard P, Lee VYK, Geuder J, Krauß SM, Ohnuki M, Jocher J, Vieth B, Enard W, Hellmann I. Evidence for compensatory evolution within pleiotropic regulatory elements. Genome Res 2024; 34:1528-1539. [PMID: 39255977 PMCID: PMC11534155 DOI: 10.1101/gr.279001.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
Pleiotropy, measured as expression breadth across tissues, is one of the best predictors for protein sequence and expression conservation. In this study, we investigated its effect on the evolution of cis-regulatory elements (CREs). To this end, we carefully reanalyzed the Epigenomics Roadmap data for nine fetal tissues, assigning a measure of pleiotropic degree to nearly half a million CREs. To assess the functional conservation of CREs, we generated ATAC-seq and RNA-seq data from humans and macaques. We found that more pleiotropic CREs exhibit greater conservation in accessibility, and the mRNA expression levels of the associated genes are more conserved. This trend of higher conservation for higher degrees of pleiotropy persists when analyzing the transcription factor binding repertoire. In contrast, simple DNA sequence conservation of orthologous sites between species tends to be even lower for pleiotropic CREs than for species-specific CREs. Combining various lines of evidence, we propose that the lack of sequence conservation in functionally conserved pleiotropic CREs is owing to within-element compensatory evolution. In summary, our findings suggest that pleiotropy is also a good predictor for the functional conservation of CREs, even though this is not reflected in the sequence conservation of pleiotropic CREs.
Collapse
Affiliation(s)
- Zane Kliesmete
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Peter Orchard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109-2218, USA
| | - Victor Yan Kin Lee
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Section for Molecular Ecology and Evolution, Globe Institute, University of Copenhagen, 1350 Copenhagen, Denmark
| | - Johanna Geuder
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Simon M Krauß
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Department of Hematology, Cell Therapy, Hemostaseology and Infectious Diseases, University Leipzig Medical Center, 04103 Leipzig, Germany
| | - Mari Ohnuki
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Faculty of Medicine, Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8501, Japan
| | - Jessica Jocher
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Beate Vieth
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Ines Hellmann
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany;
| |
Collapse
|
5
|
Mahiny D, Hauck L, Premsingh B, Grothe D, Billia F. Cdk1 Deficiency Extends the Postnatal Window of Cardiomyocyte Proliferation and Restores Cardiac Function after Myocardial Infarction. Int J Mol Sci 2024; 25:10824. [PMID: 39409153 DOI: 10.3390/ijms251910824] [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: 08/28/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Cyclin-dependent kinase 1 (Cdk1) is a master regulator of the G2-M transition between DNA replication and cell division. This study investigates the regulation of cardiomyocyte (CM) proliferation during the early neonatal period and following ischemic injury in adult mice. We analyzed cell cycle dynamics with the assessment of DNA synthesis, and cytokinesis in murine hearts during the first 15 days after birth. A distinct proliferative block was observed at 1 day, followed by a second wave of DNA synthesis at 4 days, leading to CM binucleation (CMBN) by day 5. Genome-wide mRNA profiling revealed the differential expression of cell cycle regulatory genes during this period, with a downregulation of factors involved in cell division and mitosis. The loss of Cdk1 impaired CMBN but extended the neonatal CM proliferation window until day 10 post-birth. In adult hearts, the cardiac-specific ablation of Cdk1 triggered CM proliferation post-myocardial-infarction (MI) in specific zones, driven by the activation of EGFR1 signaling and suppression of the anti-proliferative p38 and p53 signaling. This was accompanied by restoration of fractional shortening, mitochondrial function, and decreased reactive oxygen species. Additionally, cardiac hypertrophy was mitigated, and survival rates post-MI were increased in Cdk1-knockout mice. These findings reveal a novel role of Cdk1 in regulating cell cycle exit and re-entry in differentiated CMs and offer insights into potential strategies for cardiac repair.
Collapse
Affiliation(s)
- Donya Mahiny
- Toronto General Hospital Research Institute, 100 College St., Toronto, ON M5G 1L7, Canada
| | - Ludger Hauck
- Toronto General Hospital Research Institute, 100 College St., Toronto, ON M5G 1L7, Canada
| | - Benny Premsingh
- Toronto General Hospital Research Institute, 100 College St., Toronto, ON M5G 1L7, Canada
| | - Daniela Grothe
- Toronto General Hospital Research Institute, 100 College St., Toronto, ON M5G 1L7, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute, 100 College St., Toronto, ON M5G 1L7, Canada
- Division of Cardiology, University Health Network (UHN), 200 Elizabeth St., Toronto, ON M5G 2C4, Canada
| |
Collapse
|
6
|
Wang X, Zhang J, Su J, Huang T, Lian L, Nie Q, Zhang X, Li J, Wang Y. Genome-wide mapping of the binding sites of myocyte enhancer factor 2A in chicken primary myoblasts. Poult Sci 2024; 103:104097. [PMID: 39094502 PMCID: PMC11345569 DOI: 10.1016/j.psj.2024.104097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
Myocyte enhancer factor 2A (MEF2A) is a transcription factor that plays a critical role in cell proliferation, differentiation and apoptosis. In contrast to the wide characterization of its regulation mechanism in mammalian skeletal muscle, its role in chickens is limited. Especially, its wide target genes remain to be identified. Therefore, we utilized Cleavage Under Targets and Tagmentation (CUT&Tag) technology to reveal the genome-wide binding profile of MEF2A in chicken primary myoblasts thus gaining insights into its potential role in muscle development. Our results revealed that MEF2A binding sites were primarily distributed in intergenic and intronic regions. Within the promoter region, although only 8.87% of MEF2A binding sites were found, these binding sites were concentrated around the transcription start site (TSS). Following peak annotation, a total of 1903 genes were identified as potential targets of MEF2A. Gene Ontology (GO) enrichment analysis further revealed that MEF2A target genes may be involved in the regulation of embryonic development in multiple organ systems, including muscle development, gland development, and visual system development. Moreover, a comparison of the MEF2A target genes identified in chicken primary myoblasts with those in mouse C2C12 cells revealed 388 target genes are conserved across species, 1515 target genes are chicken specific. Among these conserved genes, ankyrin repeat and SOCS box containing 5 (ASB5), transmembrane protein 182 (TMEM182), myomesin 2 (MYOM2), leucyl and cystinyl aminopeptidase (LNPEP), actinin alpha 2 (ACTN2), sorbin and SH3 domain containing 1 (SORBS1), ankyrin 3 (ANK3), sarcoglycan delta (SGCD), and ORAI calcium release-activated calcium modulator 1 (ORAI1) exhibited consistent expression patterns with MEF2A during embryonic muscle development. Finally, TMEM182, as an important negative regulator of muscle development, has been validated to be regulated by MEF2A by dual-luciferase and quantitative real-time PCR (qPCR) assays. In summary, our study for the first time provides a wide landscape of MEF2A target genes in chicken primary myoblasts, which supports the active role of MEF2A in chicken muscle development.
Collapse
Affiliation(s)
- Xinglong Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Jiannan Zhang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Jiancheng Su
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Tianjiao Huang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Ling Lian
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, PR China
| | - Qinghua Nie
- Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou, PR China
| | - Xin Zhang
- Joint Nutrition Center for Animal Feeding of Sichuan University-Shengliyuan Group
| | - Juan Li
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China; Joint Nutrition Center for Animal Feeding of Sichuan University-Shengliyuan Group
| | - Yajun Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China; Joint Nutrition Center for Animal Feeding of Sichuan University-Shengliyuan Group.
| |
Collapse
|
7
|
Zubrzycki M, Schramm R, Costard-Jäckle A, Grohmann J, Gummert JF, Zubrzycka M. Cardiac Development and Factors Influencing the Development of Congenital Heart Defects (CHDs): Part I. Int J Mol Sci 2024; 25:7117. [PMID: 39000221 PMCID: PMC11241401 DOI: 10.3390/ijms25137117] [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: 06/03/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The traditional description of cardiac development involves progression from a cardiac crescent to a linear heart tube, which in the phase of transformation into a mature heart forms a cardiac loop and is divided with the septa into individual cavities. Cardiac morphogenesis involves numerous types of cells originating outside the initial cardiac crescent, including neural crest cells, cells of the second heart field origin, and epicardial progenitor cells. The development of the fetal heart and circulatory system is subject to regulatation by both genetic and environmental processes. The etiology for cases with congenital heart defects (CHDs) is largely unknown, but several genetic anomalies, some maternal illnesses, and prenatal exposures to specific therapeutic and non-therapeutic drugs are generally accepted as risk factors. New techniques for studying heart development have revealed many aspects of cardiac morphogenesis that are important in the development of CHDs, in particular transposition of the great arteries.
Collapse
Affiliation(s)
- Marek Zubrzycki
- Department of Surgery for Congenital Heart Defects, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany;
| | - Rene Schramm
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Angelika Costard-Jäckle
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Jochen Grohmann
- Department of Congenital Heart Disease/Pediatric Cardiology, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany;
| | - Jan F. Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Maria Zubrzycka
- Department of Clinical Physiology, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
8
|
Jang J, Accornero F, Li D. Epigenetic determinants and non-myocardial signaling pathways contributing to heart growth and regeneration. Pharmacol Ther 2024; 257:108638. [PMID: 38548089 DOI: 10.1016/j.pharmthera.2024.108638] [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: 01/02/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Congenital heart disease is the most common birth defect worldwide. Defective cardiac myogenesis is either a major presentation or associated with many types of congenital heart disease. Non-myocardial tissues, including endocardium and epicardium, function as a supporting hub for myocardial growth and maturation during heart development. Recent research findings suggest an emerging role of epigenetics in nonmyocytes supporting myocardial development. Understanding how growth signaling pathways in non-myocardial tissues are regulated by epigenetic factors will likely identify new disease mechanisms for congenital heart diseases and shed lights for novel therapeutic strategies for heart regeneration.
Collapse
Affiliation(s)
- Jihyun Jang
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA.
| | - Federica Accornero
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Deqiang Li
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA.
| |
Collapse
|
9
|
Broman MT, Nadadur RD, Perez-Cervantes C, Burnicka-Turek O, Lazarevic S, Gams A, Laforest B, Steimle JD, Iddir S, Wang Z, Smith L, Mazurek SR, Olivey HE, Zhou P, Gadek M, Shen KM, Khan Z, Theisen JW, Yang XH, Ikegami K, Efimov IR, Pu WT, Weber CR, McNally EM, Svensson EC, Moskowitz IP. A Genomic Link From Heart Failure to Atrial Fibrillation Risk: FOG2 Modulates a TBX5/GATA4-Dependent Atrial Gene Regulatory Network. Circulation 2024; 149:1205-1230. [PMID: 38189150 PMCID: PMC11152454 DOI: 10.1161/circulationaha.123.066804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND The relationship between heart failure (HF) and atrial fibrillation (AF) is clear, with up to half of patients with HF progressing to AF. The pathophysiological basis of AF in the context of HF is presumed to result from atrial remodeling. Upregulation of the transcription factor FOG2 (friend of GATA2; encoded by ZFPM2) is observed in human ventricles during HF and causes HF in mice. METHODS FOG2 expression was assessed in human atria. The effect of adult-specific FOG2 overexpression in the mouse heart was evaluated by whole animal electrophysiology, in vivo organ electrophysiology, cellular electrophysiology, calcium flux, mouse genetic interactions, gene expression, and genomic function, including a novel approach for defining functional transcription factor interactions based on overlapping effects on enhancer noncoding transcription. RESULTS FOG2 is significantly upregulated in the human atria during HF. Adult cardiomyocyte-specific FOG2 overexpression in mice caused primary spontaneous AF before the development of HF or atrial remodeling. FOG2 overexpression generated arrhythmia substrate and trigger in cardiomyocytes, including calcium cycling defects. We found that FOG2 repressed atrial gene expression promoted by TBX5. FOG2 bound a subset of GATA4 and TBX5 co-bound genomic locations, defining a shared atrial gene regulatory network. FOG2 repressed TBX5-dependent transcription from a subset of co-bound enhancers, including a conserved enhancer at the Atp2a2 locus. Atrial rhythm abnormalities in mice caused by Tbx5 haploinsufficiency were rescued by Zfpm2 haploinsufficiency. CONCLUSIONS Transcriptional changes in the atria observed in human HF directly antagonize the atrial rhythm gene regulatory network, providing a genomic link between HF and AF risk independent of atrial remodeling.
Collapse
Affiliation(s)
- Michael T. Broman
- Department of Medicine, Section of Cardiology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637
| | - Rangarajan D. Nadadur
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Carlos Perez-Cervantes
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Ozanna Burnicka-Turek
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Sonja Lazarevic
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Anna Gams
- Department of Biomedical Engineering, George Washington University
| | - Brigitte Laforest
- Department of Medicine, Section of Cardiology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637
| | - Jeffrey D. Steimle
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Sabrina Iddir
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Zhezhen Wang
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Linsin Smith
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Stefan R. Mazurek
- Department of Medicine, Section of Cardiology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637
| | - Harold E. Olivey
- Department of Biology, Indiana University Northwest, Gary, IN 46408
| | | | - Margaret Gadek
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Kaitlyn M. Shen
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Zoheb Khan
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Joshua W.M. Theisen
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Xinan H. Yang
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | - Kohta Ikegami
- Division of Molecular and Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Igor R. Efimov
- Department of Biomedical Engineering, George Washington University
| | - William T. Pu
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138
- Department of Cardiology, Boston Children’s Hospital, Boston, MA, 02115
| | | | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University, 303 E. Superior, SQ5-516, Chicago, IL 60611
| | | | - Ivan P. Moskowitz
- Department of Pediatrics, University of Chicago, Chicago, IL 60637
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Department of Human Genetics, University of Chicago, Chicago, IL 60637
| |
Collapse
|
10
|
Shaw T, Barr FG, Üren A. The PAX Genes: Roles in Development, Cancer, and Other Diseases. Cancers (Basel) 2024; 16:1022. [PMID: 38473380 PMCID: PMC10931086 DOI: 10.3390/cancers16051022] [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: 02/12/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Since their 1986 discovery in Drosophila, Paired box (PAX) genes have been shown to play major roles in the early development of the eye, muscle, skeleton, kidney, and other organs. Consistent with their roles as master regulators of tissue formation, the PAX family members are evolutionarily conserved, regulate large transcriptional networks, and in turn can be regulated by a variety of mechanisms. Losses or mutations in these genes can result in developmental disorders or cancers. The precise mechanisms by which PAX genes control disease pathogenesis are well understood in some cases, but much remains to be explored. A deeper understanding of the biology of these genes, therefore, has the potential to aid in the improvement of disease diagnosis and the development of new treatments.
Collapse
Affiliation(s)
- Taryn Shaw
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20001, USA
| | - Frederic G Barr
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD 20892, USA
| | - Aykut Üren
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20001, USA
| |
Collapse
|
11
|
Grunert M, Dorn C, Rickert-Sperling S. Cardiac Transcription Factors and Regulatory Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:295-311. [PMID: 38884718 DOI: 10.1007/978-3-031-44087-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiac development is a fine-tuned process governed by complex transcriptional networks, in which transcription factors (TFs) interact with other regulatory layers. In this chapter, we introduce the core cardiac TFs including Gata, Hand, Nkx2, Mef2, Srf, and Tbx. These factors regulate each other's expression and can also act in a combinatorial manner on their downstream targets. Their disruption leads to various cardiac phenotypes in mice, and mutations in humans have been associated with congenital heart defects. In the second part of the chapter, we discuss different levels of regulation including cis-regulatory elements, chromatin structure, and microRNAs, which can interact with transcription factors, modulate their function, or are downstream targets. Finally, examples of disturbances of the cardiac regulatory network leading to congenital heart diseases in human are provided.
Collapse
Affiliation(s)
- Marcel Grunert
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cornelia Dorn
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
12
|
Gao F, Liang T, Lu YW, Pu L, Fu X, Dong X, Hong T, Zhang F, Liu N, Zhou Y, Wang H, Liang P, Guo Y, Yu H, Zhu W, Hu X, Chen H, Zhou B, Pu WT, Mably JD, Wang J, Wang DZ, Chen J. Reduced Mitochondrial Protein Translation Promotes Cardiomyocyte Proliferation and Heart Regeneration. Circulation 2023; 148:1887-1906. [PMID: 37905452 PMCID: PMC10841688 DOI: 10.1161/circulationaha.122.061192] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND The importance of mitochondria in normal heart function are well recognized and recent studies have implicated changes in mitochondrial metabolism with some forms of heart disease. Previous studies demonstrated that knockdown of the mitochondrial ribosomal protein S5 (MRPS5) by small interfering RNA (siRNA) inhibits mitochondrial translation and thereby causes a mitonuclear protein imbalance. Therefore, we decided to examine the effects of MRPS5 loss and the role of these processes on cardiomyocyte proliferation. METHODS We deleted a single allele of MRPS5 in mice and used left anterior descending coronary artery ligation surgery to induce myocardial damage in these animals. We examined cardiomyocyte proliferation and cardiac regeneration both in vivo and in vitro. Doxycycline treatment was used to inhibit protein translation. Heart function in mice was assessed by echocardiography. Quantitative real-time polymerase chain reaction and RNA sequencing were used to assess changes in transcription and chromatin immunoprecipitation (ChIP) and BioChIP were used to assess chromatin effects. Protein levels were assessed by Western blotting and cell proliferation or death by histology and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assays. Adeno-associated virus was used to overexpress genes. The luciferase reporter assay was used to assess promoter activity. Mitochondrial oxygen consumption rate, ATP levels, and reactive oxygen species were also analyzed. RESULTS We determined that deletion of a single allele of MRPS5 in mice results in elevated cardiomyocyte proliferation and cardiac regeneration; this observation correlates with improved cardiac function after induction of myocardial infarction. We identified ATF4 (activating transcription factor 4) as a key regulator of the mitochondrial stress response in cardiomyocytes from Mrps5+/- mice; furthermore, ATF4 regulates Knl1 (kinetochore scaffold 1) leading to an increase in cytokinesis during cardiomyocyte proliferation. The increased cardiomyocyte proliferation observed in Mrps5+/- mice was attenuated when one allele of Atf4 was deleted genetically (Mrps5+/-/Atf4+/-), resulting in the loss in the capacity for cardiac regeneration. Either MRPS5 inhibition (or as we also demonstrate, doxycycline treatment) activate a conserved regulatory mechanism that increases the proliferation of human induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS These data highlight a critical role for MRPS5/ATF4 in cardiomyocytes and an exciting new avenue of study for therapies to treat myocardial injury.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Tian Liang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Yao Wei Lu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Linbin Pu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xuyang Fu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Tingting Hong
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Feng Zhang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Ning Liu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Yuxia Zhou
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Hongkun Wang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Ping Liang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Yuxuan Guo
- Institute of Cardiovascular Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100092 China
| | - Hong Yu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wei Zhu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xinyang Hu
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - William T Pu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - John D. Mably
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Departments of Internal Medicine and Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Jian’an Wang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Departments of Internal Medicine and Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Jinghai Chen
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| |
Collapse
|
13
|
Weisman CM. The permissive binding theory of cancer. Front Oncol 2023; 13:1272981. [PMID: 38023252 PMCID: PMC10666763 DOI: 10.3389/fonc.2023.1272981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
The later stages of cancer, including the invasion and colonization of new tissues, are actively mysterious compared to earlier stages like primary tumor formation. While we lack many details about both, we do have an apparently successful explanatory framework for the earlier stages: one in which genetic mutations hold ultimate causal and explanatory power. By contrast, on both empirical and conceptual grounds, it is not currently clear that mutations alone can explain the later stages of cancer. Can a different type of molecular change do better? Here, I introduce the "permissive binding theory" of cancer, which proposes that novel protein binding interactions are the key causal and explanatory entity in invasion and metastasis. It posits that binding is more abundant at baseline than we observe because it is restricted in normal physiology; that any large perturbation to physiological state revives this baseline abundance, unleashing many new binding interactions; and that a subset of these cause the cellular functions at the heart of oncogenesis, especially invasion and metastasis. Significant physiological perturbations occur in cancer cells in very early stages, and generally become more extreme with progression, providing interactions that continually fuel invasion and metastasis. The theory is compatible with, but not limited to, causal roles for the diverse molecular changes observed in cancer (e.g. gene expression or epigenetic changes), as these generally act causally upstream of proteins, and so may exert their effects by changing the protein binding interactions that occur in the cell. This admits the possibility that molecular changes that appear quite different may actually converge in creating the same few protein complexes, simplifying our picture of invasion and metastasis. If correct, the theory offers a concrete therapeutic strategy: targeting the key novel complexes. The theory is straightforwardly testable by large-scale identification of protein interactions in different cancers.
Collapse
Affiliation(s)
- Caroline M. Weisman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States
| |
Collapse
|
14
|
Armendariz DA, Sundarrajan A, Hon GC. Breaking enhancers to gain insights into developmental defects. eLife 2023; 12:e88187. [PMID: 37497775 PMCID: PMC10374278 DOI: 10.7554/elife.88187] [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: 03/29/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Despite ground-breaking genetic studies that have identified thousands of risk variants for developmental diseases, how these variants lead to molecular and cellular phenotypes remains a gap in knowledge. Many of these variants are non-coding and occur at enhancers, which orchestrate key regulatory programs during development. The prevailing paradigm is that non-coding variants alter the activity of enhancers, impacting gene expression programs, and ultimately contributing to disease risk. A key obstacle to progress is the systematic functional characterization of non-coding variants at scale, especially since enhancer activity is highly specific to cell type and developmental stage. Here, we review the foundational studies of enhancers in developmental disease and current genomic approaches to functionally characterize developmental enhancers and their variants at scale. In the coming decade, we anticipate systematic enhancer perturbation studies to link non-coding variants to molecular mechanisms, changes in cell state, and disease phenotypes.
Collapse
Affiliation(s)
- Daniel A Armendariz
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, United States
| | - Anjana Sundarrajan
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, United States
| | - Gary C Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, United States
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
- Lyda Hill Department of Bioinformatics, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
15
|
Tao Q, Zhu Y, Wang T, Deng Y, Liu H, Wu J. Identification and analysis of lipid metabolism-related genes in allergic rhinitis. Lipids Health Dis 2023; 22:105. [PMID: 37480069 PMCID: PMC10362667 DOI: 10.1186/s12944-023-01825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/28/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Studies have shown that the lipid metabolism mediator leukotriene and prostaglandins are associated with the pathogenesis of allergic rhinitis (AR). The aim of this study was to identify key lipid metabolism-related genes (LMRGs) related to the diagnosis and treatment of AR. MATERIALS AND METHODS AR-related expression datasets (GSE75011, GSE46171) were downloaded through the Gene Expression Omnibus (GEO) database. First, weighted gene co-expression network analysis (WGCNA) was used to get AR-related genes (ARRGs). Next, between control and AR groups in GSE75011, differentially expressed genes (DEGs) were screened, and DEGs were intersected with LMRGs to obtain lipid metabolism-related differentially expressed genes (LMR DEGs). Protein-protein interaction (PPI) networks were constructed for these LMR DEGs. Hub genes were then identified through stress, radiality, closeness and edge percolated component (EPC) analysis and intersected with the ARRGs to obtain candidate genes. Biomarkers with diagnostic value were screened via receiver operating characteristic (ROC) curves. Differential immune cells screened between control and AR groups were then assessed for correlation with the diagnostic genes, and clinical correlation analysis and enrichment analysis were performed. Finally, real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) was made on blood samples from control and AR patients to validate these identified diagnostic genes. RESULTS 73 LMR DEGs were obtained, which were involved in biological processes such as metabolism of lipids and lipid biosynthetic processes. 66 ARRGs and 22 hub genes were intersected to obtain four candidate genes. Three diagnostic genes (LPCAT1, SGPP1, SMARCD3) with diagnostic value were screened according to the AUC > 0.7, with markedly variant between control and AR groups. In addition, two immune cells, regulatory T cells (Treg) and T follicular helper cells (TFH), were marked variations between control and AR groups, and SMARCD3 was significantly associated with TFH. Moreover, SMARCD3 was relevant to immune-related pathways, and correlated significantly with clinical characteristics (age and sex). Finally, RT-qPCR results indicated that changes in the expression of LPCAT1 and SMARCD3 between control and AR groups were consistent with the GSE75011 and GSE46171. CONCLUSION LPCAT1, SGPP1 and SMARCD3 might be used as biomarkers for AR.
Collapse
Affiliation(s)
- Qilei Tao
- Department of Otolaryngology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Yajing Zhu
- Department of Otolaryngology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Tianyu Wang
- Department of Otolaryngology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Yue Deng
- Department of Otolaryngology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Huanhai Liu
- Department of Otolaryngology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| | - Jian Wu
- Department of Otolaryngology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| |
Collapse
|
16
|
Nakhaei-Rad S, Haghighi F, Bazgir F, Dahlmann J, Busley AV, Buchholzer M, Kleemann K, Schänzer A, Borchardt A, Hahn A, Kötter S, Schanze D, Anand R, Funk F, Kronenbitter AV, Scheller J, Piekorz RP, Reichert AS, Volleth M, Wolf MJ, Cirstea IC, Gelb BD, Tartaglia M, Schmitt JP, Krüger M, Kutschka I, Cyganek L, Zenker M, Kensah G, Ahmadian MR. Molecular and cellular evidence for the impact of a hypertrophic cardiomyopathy-associated RAF1 variant on the structure and function of contractile machinery in bioartificial cardiac tissues. Commun Biol 2023; 6:657. [PMID: 37344639 PMCID: PMC10284840 DOI: 10.1038/s42003-023-05013-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/02/2023] [Indexed: 06/23/2023] Open
Abstract
Noonan syndrome (NS), the most common among RASopathies, is caused by germline variants in genes encoding components of the RAS-MAPK pathway. Distinct variants, including the recurrent Ser257Leu substitution in RAF1, are associated with severe hypertrophic cardiomyopathy (HCM). Here, we investigated the elusive mechanistic link between NS-associated RAF1S257L and HCM using three-dimensional cardiac bodies and bioartificial cardiac tissues generated from patient-derived induced pluripotent stem cells (iPSCs) harboring the pathogenic RAF1 c.770 C > T missense change. We characterize the molecular, structural, and functional consequences of aberrant RAF1-associated signaling on the cardiac models. Ultrastructural assessment of the sarcomere revealed a shortening of the I-bands along the Z disc area in both iPSC-derived RAF1S257L cardiomyocytes and myocardial tissue biopsies. The aforementioned changes correlated with the isoform shift of titin from a longer (N2BA) to a shorter isoform (N2B) that also affected the active force generation and contractile tensions. The genotype-phenotype correlation was confirmed using cardiomyocyte progeny of an isogenic gene-corrected RAF1S257L-iPSC line and was mainly reversed by MEK inhibition. Collectively, our findings uncovered a direct link between a RASopathy gene variant and the abnormal sarcomere structure resulting in a cardiac dysfunction that remarkably recapitulates the human disease.
Collapse
Affiliation(s)
- Saeideh Nakhaei-Rad
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Stem Cell Biology and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fereshteh Haghighi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Farhad Bazgir
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Dahlmann
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany
| | - Alexandra Viktoria Busley
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| | - Marcel Buchholzer
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karolin Kleemann
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus Liebig University Giessen, Giessen, Germany
| | - Andrea Borchardt
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas Hahn
- Department of Child Neurology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Sebastian Kötter
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Denny Schanze
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Florian Funk
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annette Vera Kronenbitter
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Roland P Piekorz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marianne Volleth
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany
| | - Matthew J Wolf
- Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Ion Cristian Cirstea
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | - Joachim P Schmitt
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Martina Krüger
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ingo Kutschka
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany.
| | - George Kensah
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany.
| | - Mohammad R Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
17
|
Zhou P, VanDusen NJ, Zhang Y, Cao Y, Sethi I, Hu R, Zhang S, Wang G, Ye L, Mazumdar N, Chen J, Zhang X, Guo Y, Li B, Ma Q, Lee JY, Gu W, Yuan GC, Ren B, Chen K, Pu WT. Dynamic changes in P300 enhancers and enhancer-promoter contacts control mouse cardiomyocyte maturation. Dev Cell 2023; 58:898-914.e7. [PMID: 37071996 PMCID: PMC10231645 DOI: 10.1016/j.devcel.2023.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 04/20/2023]
Abstract
Cardiomyocyte differentiation continues throughout murine gestation and into the postnatal period, driven by temporally regulated expression changes in the transcriptome. The mechanisms that regulate these developmental changes remain incompletely defined. Here, we used cardiomyocyte-specific ChIP-seq of the activate enhancer marker P300 to identify 54,920 cardiomyocyte enhancers at seven stages of murine heart development. These data were matched to cardiomyocyte gene expression profiles at the same stages and to Hi-C and H3K27ac HiChIP chromatin conformation data at fetal, neonatal, and adult stages. Regions with dynamic P300 occupancy exhibited developmentally regulated enhancer activity, as measured by massively parallel reporter assays in cardiomyocytes in vivo, and identified key transcription factor-binding motifs. These dynamic enhancers interacted with temporal changes of the 3D genome architecture to specify developmentally regulated cardiomyocyte gene expressions. Our work provides a 3D genome-mediated enhancer activity landscape of murine cardiomyocyte development.
Collapse
Affiliation(s)
- Pingzhu Zhou
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Nathan J VanDusen
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yanchun Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Yangpo Cao
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Isha Sethi
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Rong Hu
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Shuo Zhang
- Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Guangyu Wang
- Cardiovascular Department, Houston Methodist, Weill Cornell Medical College, Houston, TX, USA
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Neil Mazumdar
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Jian Chen
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Yuxuan Guo
- Peking University Health Science Center, Beijing, China
| | - Bin Li
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Qing Ma
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Julianna Y Lee
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Weiliang Gu
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA; Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bing Ren
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kaifu Chen
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA.
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
18
|
Htet M, Lei S, Bajpayi S, Zoitou A, Chamakioti M, Tampakakis E. The role of noncoding genetic variants in cardiomyopathy. Front Cardiovasc Med 2023; 10:1116925. [PMID: 37283586 PMCID: PMC10239966 DOI: 10.3389/fcvm.2023.1116925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
Cardiomyopathies remain one of the leading causes of morbidity and mortality worldwide. Environmental risk factors and genetic predisposition account for most cardiomyopathy cases. As with all complex diseases, there are significant challenges in the interpretation of the molecular mechanisms underlying cardiomyopathy-associated genetic variants. Given the technical improvements and reduced costs of DNA sequence technologies, an increasing number of patients are now undergoing genetic testing, resulting in a continuously expanding list of novel mutations. However, many patients carry noncoding genetic variants, and although emerging evidence supports their contribution to cardiac disease, their role in cardiomyopathies remains largely understudied. In this review, we summarize published studies reporting on the association of different types of noncoding variants with various types of cardiomyopathies. We focus on variants within transcriptional enhancers, promoters, intronic sites, and untranslated regions that are likely associated with cardiac disease. Given the broad nature of this topic, we provide an overview of studies that are relatively recent and have sufficient evidence to support a significant degree of causality. We believe that more research with additional validation of noncoding genetic variants will provide further mechanistic insights on the development of cardiac disease, and noncoding variants will be increasingly incorporated in future genetic screening tests.
Collapse
Affiliation(s)
- Myo Htet
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Shunyao Lei
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Sheetal Bajpayi
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Asimina Zoitou
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | | | - Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
19
|
Guo Y, Cao Y, Jardin BD, Zhang X, Zhou P, Guatimosim S, Lin J, Chen Z, Zhang Y, Mazumdar N, Lu F, Ma Q, Lu YW, Zhao M, Wang DZ, Dong E, Pu WT. Ryanodine receptor 2 (RYR2) dysfunction activates the unfolded protein response and perturbs cardiomyocyte maturation. Cardiovasc Res 2023; 119:221-235. [PMID: 35576474 PMCID: PMC10233305 DOI: 10.1093/cvr/cvac077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 04/03/2022] [Accepted: 05/05/2022] [Indexed: 11/15/2022] Open
Abstract
AIMS Calcium-handling capacity is a major gauge of cardiomyocyte maturity. Ryanodine receptor 2 (RYR2) is the pre-dominant calcium channel that releases calcium from the sarcoplasmic reticulum/endoplasmic reticulum (SR/ER) to activate cardiomyocyte contraction. Although RYR2 was previously implied as a key regulator of cardiomyocyte maturation, the mechanisms remain unclear. The aim of this study is to solve this problem. METHODS AND RESULTS We performed Cas9/AAV9-mediated somatic mutagenesis to knockout RYR2 specifically in cardiomyocytes in mice. We conducted a genetic mosaic analysis to dissect the cell-autonomous function of RYR2 during cardiomyocyte maturation. We found that RYR2 depletion triggered ultrastructural and transcriptomic defects relevant to cardiomyocyte maturation. These phenotypes were associated with the drastic activation of ER stress pathways. The ER stress alleviator tauroursodeoxycholic acid partially rescued the defects in RYR2-depleted cardiomyocytes. Overexpression of ATF4, a key ER stress transcription factor, recapitulated defects in RYR2-depleted cells. Integrative analysis of RNA-Seq and bioChIP-Seq data revealed that protein biosynthesis-related genes are the major direct downstream targets of ATF4. CONCLUSION RYR2-regulated ER homeostasis is essential for cardiomyocyte maturation. Severe ER stress perturbs cardiomyocyte maturation primarily through ATF4 activation. The major downstream effector genes of ATF4 are related to protein biosynthesis.
Collapse
Affiliation(s)
- Yuxuan Guo
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Yangpo Cao
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Blake D Jardin
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte MG - CEP 31270-901, Brazil
| | - Junsen Lin
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Zhan Chen
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Yueyang Zhang
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Neil Mazumdar
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Fujian Lu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Yao-Wei Lu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Mingming Zhao
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing 100191, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Erdan Dong
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing 100191, China
| | - William T Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| |
Collapse
|
20
|
Cao Y, Zhang X, Akerberg BN, Yuan H, Sakamoto T, Xiao F, VanDusen NJ, Zhou P, Sweat ME, Wang Y, Prondzynski M, Chen J, Zhang Y, Wang P, Kelly DP, Pu WT. In Vivo Dissection of Chamber-Selective Enhancers Reveals Estrogen-Related Receptor as a Regulator of Ventricular Cardiomyocyte Identity. Circulation 2023; 147:881-896. [PMID: 36705030 PMCID: PMC10010668 DOI: 10.1161/circulationaha.122.061955] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiac chamber-selective transcriptional programs underpin the structural and functional differences between atrial and ventricular cardiomyocytes (aCMs and vCMs). The mechanisms responsible for these chamber-selective transcriptional programs remain largely undefined. METHODS We nominated candidate chamber-selective enhancers (CSEs) by determining the genome-wide occupancy of 7 key cardiac transcription factors (GATA4, MEF2A, MEF2C, NKX2-5, SRF, TBX5, TEAD1) and transcriptional coactivator P300 in atria and ventricles. Candidate enhancers were tested using an adeno-associated virus-mediated massively parallel reporter assay. Chromatin features of CSEs were evaluated by performing assay of transposase accessible chromatin sequencing and acetylation of histone H3 at lysine 27-HiChIP on aCMs and vCMs. CSE sequence requirements were determined by systematic tiling mutagenesis of 29 CSEs at 5 bp resolution. Estrogen-related receptor (ERR) function in cardiomyocytes was evaluated by Cre-loxP-mediated inactivation of ERRα and ERRγ in cardiomyocytes. RESULTS We identified 134 066 and 97 506 regions reproducibly occupied by at least 1 transcription factor or P300, in atria or ventricles, respectively. Enhancer activities of 2639 regions bound by transcription factors or P300 were tested in aCMs and vCMs by adeno-associated virus-mediated massively parallel reporter assay. This identified 1092 active enhancers in aCMs or vCMs. Several overlapped loci associated with cardiovascular disease through genome-wide association studies, and 229 exhibited chamber-selective activity in aCMs or vCMs. Many CSEs exhibited differential chromatin accessibility between aCMs and vCMs, and CSEs were enriched for aCM- or vCM-selective acetylation of histone H3 at lysine 27-anchored loops. Tiling mutagenesis of 29 CSEs identified the binding motif of ERRα/γ as important for ventricular enhancer activity. The requirement of ERRα/γ to activate ventricular CSEs and promote vCM identity was confirmed by loss of the vCM gene profile in ERRα/γ knockout vCMs. CONCLUSIONS We identified 229 CSEs that could be useful research tools or direct therapeutic gene expression. We showed that chamber-selective multi-transcription factor, P300 occupancy, open chromatin, and chromatin looping are predictive features of CSEs. We found that ERRα/γ are essential for maintenance of ventricular identity. Finally, our gene expression, epigenetic, 3-dimensional genome, and enhancer activity atlas provide key resources for future studies of chamber-selective gene regulation.
Collapse
Affiliation(s)
- Yangpo Cao
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Brynn N Akerberg
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Haiyun Yuan
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, China (H.Y.)
| | - Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (T.S., D.P.K.)
| | - Feng Xiao
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Nathan J VanDusen
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis (N.J.V.)
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Mason E Sweat
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Yi Wang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Maksymilian Prondzynski
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Jian Chen
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Yan Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Peizhe Wang
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.)
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (T.S., D.P.K.)
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA (Y.C., X.Z., B.N.A., F.X., P.Z., M.E.S., Y.W., M.P., J.C., Y.Z., P.W., W.T.P.).,Harvard Stem Cell Institute, Cambridge, MA (W.T.P.)
| |
Collapse
|
21
|
Aballo TJ, Roberts DS, Bayne EF, Zhu W, Walcott G, Mahmoud AI, Zhang J, Ge Y. Integrated proteomics reveals alterations in sarcomere composition and developmental processes during postnatal swine heart development. J Mol Cell Cardiol 2023; 176:33-40. [PMID: 36657638 PMCID: PMC10006350 DOI: 10.1016/j.yjmcc.2023.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/20/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023]
Abstract
The neonatal swine heart possesses an endogenous ability to regenerate injured myocardium through the proliferation of pre-existing cardiomyocyte (CM) populations. However, this regenerative capacity is lost shortly after birth. Normal postnatal developmental processes and the regenerative capacity of mammalian hearts are tightly linked, but not much is known about how the swine cardiac proteome changes throughout postnatal development. Herein, we integrated robust and quantitative targeted "top-down" and global "bottom-up" proteomic workflows to comprehensively define the dynamic landscape of the swine cardiac proteome throughout postnatal maturation. Using targeted top-down proteomics, we were able to identify significant alterations in sarcomere composition, providing new insight into the proteoform landscape of sarcomeres that can disassemble, a process necessary for productive CM proliferation. Furthermore, we quantified global changes in protein abundance using bottom-up proteomics, identified over 700 differentially expressed proteins throughout postnatal development, and mapped these proteins to changes in developmental and metabolic processes. We envision these results will help guide future investigations to comprehensively understand endogenous cardiac regeneration toward the development of novel therapeutic strategies for heart failure.
Collapse
Affiliation(s)
- Timothy J Aballo
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David S Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Elizabeth F Bayne
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Gregory Walcott
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Ahmed I Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
22
|
He X, Liang J, Paul C, Huang W, Dutta S, Wang Y. Advances in Cellular Reprogramming-Based Approaches for Heart Regenerative Repair. Cells 2022; 11:3914. [PMID: 36497171 PMCID: PMC9740402 DOI: 10.3390/cells11233914] [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: 10/18/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Continuous loss of cardiomyocytes (CMs) is one of the fundamental characteristics of many heart diseases, which eventually can lead to heart failure. Due to the limited proliferation ability of human adult CMs, treatment efficacy has been limited in terms of fully repairing damaged hearts. It has been shown that cell lineage conversion can be achieved by using cell reprogramming approaches, including human induced pluripotent stem cells (hiPSCs), providing a promising therapeutic for regenerative heart medicine. Recent studies using advanced cellular reprogramming-based techniques have also contributed some new strategies for regenerative heart repair. In this review, hiPSC-derived cell therapeutic methods are introduced, and the clinical setting challenges (maturation, engraftment, immune response, scalability, and tumorigenicity), with potential solutions, are discussed. Inspired by the iPSC reprogramming, the approaches of direct cell lineage conversion are merging, such as induced cardiomyocyte-like cells (iCMs) and induced cardiac progenitor cells (iCPCs) derived from fibroblasts, without induction of pluripotency. The studies of cellular and molecular pathways also reveal that epigenetic resetting is the essential mechanism of reprogramming and lineage conversion. Therefore, CRISPR techniques that can be repurposed for genomic or epigenetic editing become attractive approaches for cellular reprogramming. In addition, viral and non-viral delivery strategies that are utilized to achieve CM reprogramming will be introduced, and the therapeutic effects of iCMs or iCPCs on myocardial infarction will be compared. After the improvement of reprogramming efficiency by developing new techniques, reprogrammed iCPCs or iCMs will provide an alternative to hiPSC-based approaches for regenerative heart therapies, heart disease modeling, and new drug screening.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jialiang Liang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Christian Paul
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Wei Huang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Yigang Wang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
23
|
Dinh TT, Xiang M, Rajaraman A, Wang Y, Salazar N, Zhu Y, Roper W, Rhee S, Brulois K, O'Hara E, Kiefel H, Dinh TM, Bi Y, Gonzalez D, Bao EP, Red-Horse K, Balogh P, Gábris F, Gaszner B, Berta G, Pan J, Butcher EC. An NKX-COUP-TFII morphogenetic code directs mucosal endothelial addressin expression. Nat Commun 2022; 13:7448. [PMID: 36460642 PMCID: PMC9718832 DOI: 10.1038/s41467-022-34991-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022] Open
Abstract
Immunoglobulin family and carbohydrate vascular addressins encoded by Madcam1 and St6gal1 control lymphocyte homing into intestinal tissues, regulating immunity and inflammation. The addressins are developmentally programmed to decorate endothelial cells lining gut post-capillary and high endothelial venules (HEV), providing a prototypical example of organ- and segment-specific endothelial specialization. We identify conserved NKX-COUP-TFII composite elements (NCCE) in regulatory regions of Madcam1 and St6gal1 that bind intestinal homeodomain protein NKX2-3 cooperatively with venous nuclear receptor COUP-TFII to activate transcription. The Madcam1 element also integrates repressive signals from arterial/capillary Notch effectors. Pan-endothelial COUP-TFII overexpression induces ectopic addressin expression in NKX2-3+ capillaries, while NKX2-3 deficiency abrogates expression by HEV. Phylogenetically conserved NCCE are enriched in genes involved in neuron migration and morphogenesis of the heart, kidney, pancreas and other organs. Our results define an NKX-COUP-TFII morphogenetic code that targets expression of mucosal vascular addressins.
Collapse
Affiliation(s)
- Thanh Theresa Dinh
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Menglan Xiang
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Anusha Rajaraman
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Yongzhi Wang
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Clinical Science Malmo, Section of Surgery, Lund University, Malmo, Sweden
| | - Nicole Salazar
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yu Zhu
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Walter Roper
- Columbia University Vagelos College of Physicians and Surgeons, New York City, NY, USA
| | - Siyeon Rhee
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Kevin Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Ed O'Hara
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Helena Kiefel
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Truc M Dinh
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Yuhan Bi
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | | | - Evan P Bao
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Peter Balogh
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
- Lymphoid Organogenesis Research Team, Szentágothai Research Center, Pécs, Hungary
| | - Fanni Gábris
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
- Lymphoid Organogenesis Research Team, Szentágothai Research Center, Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscopy Laboratory, University of Pécs Medical School, Pécs, Hungary
| | - Junliang Pan
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA.
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA.
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
24
|
Tang Y, Aryal S, Geng X, Zhou X, Fast VG, Zhang J, Lu R, Zhou Y. TBX20 Improves Contractility and Mitochondrial Function During Direct Human Cardiac Reprogramming. Circulation 2022; 146:1518-1536. [PMID: 36102189 PMCID: PMC9662826 DOI: 10.1161/circulationaha.122.059713] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Direct cardiac reprogramming of fibroblasts into cardiomyocytes has emerged as a promising strategy to remuscularize injured myocardium. However, it is insufficient to generate functional induced cardiomyocytes from human fibroblasts using conventional reprogramming cocktails, and the underlying molecular mechanisms are not well studied. METHODS To discover potential missing factors for human direct reprogramming, we performed transcriptomic comparison between human induced cardiomyocytes and functional cardiomyocytes. RESULTS We identified TBX20 (T-box transcription factor 20) as the top cardiac gene that is unable to be activated by the MGT133 reprogramming cocktail (MEF2C, GATA4, TBX5, and miR-133). TBX20 is required for normal heart development and cardiac function in adult cardiomyocytes, yet its role in cardiac reprogramming remains undefined. We show that the addition of TBX20 to the MGT133 cocktail (MGT+TBX20) promotes cardiac reprogramming and activates genes associated with cardiac contractility, maturation, and ventricular heart. Human induced cardiomyocytes produced with MGT+TBX20 demonstrated more frequent beating, calcium oscillation, and higher energy metabolism as evidenced by increased mitochondria numbers and mitochondrial respiration. Mechanistically, comprehensive transcriptomic, chromatin occupancy, and epigenomic studies revealed that TBX20 colocalizes with MGT reprogramming factors at cardiac gene enhancers associated with heart contraction, promotes chromatin binding and co-occupancy of MGT factors at these loci, and synergizes with MGT for more robust activation of target gene transcription. CONCLUSIONS TBX20 consolidates MGT cardiac reprogramming factors to activate cardiac enhancers to promote cardiac cell fate conversion. Human induced cardiomyocytes generated with TBX20 showed enhanced cardiac function in contractility and mitochondrial respiration.
Collapse
Affiliation(s)
- Yawen Tang
- Department of Biomedical Engineering (Y.T., X.G., V.G.F., J.Z., Y.Z.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Sajesan Aryal
- Department of Medicine, Division of Hematology and Oncology (S.A., X.Z., R.L.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham.,O’Neal Comprehensive Cancer Center (S.A., X.Z., R.L.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Xiaoxiao Geng
- Department of Biomedical Engineering (Y.T., X.G., V.G.F., J.Z., Y.Z.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Xinyue Zhou
- Department of Medicine, Division of Hematology and Oncology (S.A., X.Z., R.L.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham.,O’Neal Comprehensive Cancer Center (S.A., X.Z., R.L.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Vladimir G. Fast
- Department of Biomedical Engineering (Y.T., X.G., V.G.F., J.Z., Y.Z.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Jianyi Zhang
- Department of Biomedical Engineering (Y.T., X.G., V.G.F., J.Z., Y.Z.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Rui Lu
- Department of Medicine, Division of Hematology and Oncology (S.A., X.Z., R.L.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham.,O’Neal Comprehensive Cancer Center (S.A., X.Z., R.L.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| | - Yang Zhou
- Department of Biomedical Engineering (Y.T., X.G., V.G.F., J.Z., Y.Z.), Heersink School of Medicine, School of Engineering, University of Alabama at Birmingham
| |
Collapse
|
25
|
Zhou P, Zhang Y, Sethi I, Ye L, Trembley MA, Cao Y, Akerberg BN, Xiao F, Zhang X, Li K, Jardin BD, Mazumdar N, Ma Q, He A, Zhou B, Pu WT. GATA4 Regulates Developing Endocardium Through Interaction With ETS1. Circ Res 2022; 131:e152-e168. [PMID: 36263775 PMCID: PMC9669226 DOI: 10.1161/circresaha.120.318102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/07/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND The pioneer transcription factor (TF) GATA4 (GATA Binding Protein 4) is expressed in multiple cardiovascular lineages and is essential for heart development. GATA4 lineage-specific occupancy in the developing heart underlies its lineage specific activities. Here, we characterized GATA4 chromatin occupancy in cardiomyocyte and endocardial lineages, dissected mechanisms that control lineage specific occupancy, and analyzed GATA4 regulation of endocardial gene expression. METHODS We mapped GATA4 chromatin occupancy in cardiomyocyte and endocardial cells of embryonic day 12.5 (E12.5) mouse heart using lineage specific, Cre-activated biotinylation of GATA4. Regulation of GATA4 pioneering activity was studied in cell lines stably overexpressing GATA4. GATA4 regulation of endocardial gene expression was analyzed using single cell RNA sequencing and luciferase reporter assays. RESULTS Cardiomyocyte-selective and endothelial-selective GATA4 occupied genomic regions had features of lineage specific enhancers. Footprints within cardiomyocyte- and endothelial-selective GATA4 regions were enriched for NKX2-5 (NK2 homeobox 5) and ETS1 (ETS Proto-Oncogene 1) motifs, respectively, and both of these TFs interacted with GATA4 in co-immunoprecipitation assays. In stable NIH3T3 cell lines expressing GATA4 with or without NKX2-5 or ETS1, the partner TFs re-directed GATA4 pioneer binding and augmented its ability to open previously inaccessible regions, with ETS1 displaying greater potency as a pioneer partner than NKX2-5. Single-cell RNA sequencing of embryonic hearts with endothelial cell-specific Gata4 inactivation identified Gata4-regulated endocardial genes, which were adjacent to GATA4-bound, endothelial regions enriched for both GATA4 and ETS1 motifs. In reporter assays, GATA4 and ETS1 cooperatively stimulated endothelial cell enhancer activity. CONCLUSIONS Lineage selective non-pioneer TFs NKX2-5 and ETS1 guide the activity of pioneer TF GATA4 to bind and open chromatin and create active enhancers and mechanistically link ETS1 interaction to GATA4 regulation of endocardial development.
Collapse
Affiliation(s)
- Pingzhu Zhou
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Yan Zhang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Isha Sethi
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Michael A. Trembley
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Yangpo Cao
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Brynn N. Akerberg
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Feng Xiao
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Kai Li
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Blake D. Jardin
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Neil Mazumdar
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
| | - Aibin He
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138
| |
Collapse
|
26
|
Al-Khindi T, Sherman MB, Kodama T, Gopal P, Pan Z, Kiraly JK, Zhang H, Goff LA, du Lac S, Kolodkin AL. The transcription factor Tbx5 regulates direction-selective retinal ganglion cell development and image stabilization. Curr Biol 2022; 32:4286-4298.e5. [PMID: 35998637 PMCID: PMC9560999 DOI: 10.1016/j.cub.2022.07.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/05/2022] [Accepted: 07/21/2022] [Indexed: 12/14/2022]
Abstract
The diversity of visual input processed by the mammalian visual system requires the generation of many distinct retinal ganglion cell (RGC) types, each tuned to a particular feature. The molecular code needed to generate this cell-type diversity is poorly understood. Here, we focus on the molecules needed to specify one type of retinal cell: the upward-preferring ON direction-selective ganglion cell (up-oDSGC) of the mouse visual system. Single-cell transcriptomic profiling of up- and down-oDSGCs shows that the transcription factor Tbx5 is selectively expressed in up-oDSGCs. The loss of Tbx5 in up-oDSGCs results in a selective defect in the formation of up-oDSGCs and a corresponding inability to detect vertical motion. A downstream effector of Tbx5, Sfrp1, is also critical for vertical motion detection but not up-oDSGC formation. These results advance our understanding of the molecular mechanisms that specify a rare retinal cell type and show how disrupting this specification leads to a corresponding defect in neural circuitry and behavior.
Collapse
Affiliation(s)
- Timour Al-Khindi
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael B Sherman
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Takashi Kodama
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Otolaryngology & Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Preethi Gopal
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhiwei Pan
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James K Kiraly
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hao Zhang
- Department of Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Loyal A Goff
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sascha du Lac
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Otolaryngology & Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alex L Kolodkin
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Hayat R. Dynamics of metabolism and regulation of epigenetics during cardiomyocytes maturation. Cell Biol Int 2022; 47:30-40. [PMID: 36208083 DOI: 10.1002/cbin.11931] [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: 04/22/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 11/09/2022]
Abstract
Maturation is the last step of heart growth that prepares the organ over the lifetime of the mammal for powerful, effective, and sustained pumping. Structural, gene expression, physiological, and functional specialties of cardiomyocytes describe this mechanism as the heart transits from fetus to adult phases. The main cornerstones of maturation of cardiomyocytes are reviewed and primary regulatory mechanisms are summarized to facilitate and organize these cellular activities. During embryonic development, cardiomyocytes proliferate rigorously but leave the cell cycle permanently immediately after the parturition of the child and experience terminal differentiation. The activation of a host of genes specific for the mature heart is correlated with the exit from the cell cycle. Even when exposed to mitogenic stimuli, the bulk of mature cardiomyocytes do not re-join the cell cycle. The reason for this permanent exit from the cell cycle is shown to be linked with stable switching off of the genes of the cell cycle directly involved in the G2/M transition phase and cytokinesis development. Researchers also trying to explain the molecular mechanism involved in stable inhibition of the gene and described structural changes (epigenetic and chromatin) in this mechanism. Substantial developments in the future with advances in the scientific platforms used for cardiomyocyte maturation research will broaden our understanding of this mechanism and result in better maturation of cardiomyocyte-derived pluripotent stem cells and effective treatment approaches for cardiovascular diseases.
Collapse
Affiliation(s)
- Rabia Hayat
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
| |
Collapse
|
28
|
Juan AM, Foong YH, Thorvaldsen JL, Lan Y, Leu NA, Rurik JG, Li L, Krapp C, Rosier CL, Epstein JA, Bartolomei MS. Tissue-specific Grb10/Ddc insulator drives allelic architecture for cardiac development. Mol Cell 2022; 82:3613-3631.e7. [PMID: 36108632 PMCID: PMC9547965 DOI: 10.1016/j.molcel.2022.08.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/12/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
Allele-specific expression of imprinted gene clusters is governed by gametic DNA methylation at master regulators called imprinting control regions (ICRs). Non-gametic or secondary differentially methylated regions (DMRs) at promoters and exonic regions reinforce monoallelic expression but do not control an entire cluster. Here, we unveil an unconventional secondary DMR that is indispensable for tissue-specific imprinting of two previously unlinked genes, Grb10 and Ddc. Using polymorphic mice, we mapped an intronic secondary DMR at Grb10 with paternal-specific CTCF binding (CBR2.3) that forms contacts with Ddc. Deletion of paternal CBR2.3 removed a critical insulator, resulting in substantial shifting of chromatin looping and ectopic enhancer-promoter contacts. Destabilized gene architecture precipitated abnormal Grb10-Ddc expression with developmental consequences in the heart and muscle. Thus, we redefine the Grb10-Ddc imprinting domain by uncovering an unconventional intronic secondary DMR that functions as an insulator to instruct the tissue-specific, monoallelic expression of multiple genes-a feature previously ICR exclusive.
Collapse
Affiliation(s)
- Aimee M Juan
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yee Hoon Foong
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joanne L Thorvaldsen
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yemin Lan
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicolae A Leu
- Department of Biomedical Sciences, Center for Animal Transgenesis and Germ Cell Research, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Joel G Rurik
- Penn Cardiovascular Institute, Department of Medicine, Department Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li Li
- Penn Cardiovascular Institute, Department of Medicine, Department Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Krapp
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Casey L Rosier
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan A Epstein
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, Department of Medicine, Department Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marisa S Bartolomei
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
29
|
Pressler MP, Horvath A, Entcheva E. Sex-dependent transcription of cardiac electrophysiology and links to acetylation modifiers based on the GTEx database. Front Cardiovasc Med 2022; 9:941890. [PMID: 35935618 PMCID: PMC9354462 DOI: 10.3389/fcvm.2022.941890] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
Development of safer drugs based on epigenetic modifiers, e.g., histone deacetylase inhibitors (HDACi), requires better understanding of their effects on cardiac electrophysiology. Using RNAseq data from the genotype-tissue-expression database (GTEx), we created models that link the abundance of acetylation enzymes (HDAC/SIRT/HATs), and the gene expression of ion channels (IC) via select cardiac transcription factors (TFs) in male and female adult human hearts (left ventricle, LV). Gene expression data (transcripts per million, TPM) from GTEx donors (21–70 y.o.) were filtered, normalized and transformed to Euclidian space to allow quantitative comparisons in 84 female and 158 male LVs. Sex-specific partial least-square (PLS) regression models, linking gene expression data for HDAC/SIRT/HATs to TFs and to ICs gene expression, revealed tight co-regulation of cardiac ion channels by HDAC/SIRT/HATs, with stronger clustering in the male LV. Co-regulation of genes encoding excitatory and inhibitory processes in cardiac tissue by the acetylation modifiers may help explain their predominantly net-neutral effects on cardiac electrophysiology. ATP1A1, encoding for the Na/K pump, represented an outlier—with orthogonal regulation by the acetylation modifiers to most of the ICs. The HDAC/SIRT/HAT effects were mediated by strong (+) TF regulators of ICs, e.g., MEF2A and TBX5, in both sexes. Furthermore, for male hearts, PLS models revealed a stronger (+/-) mediatory role on ICs for NKX25 and TGF1B/KLF4, respectively, while RUNX1 exhibited larger (-) TF effects on ICs in females. Male-trained PLS models of HDAC/SIRT/HAT effects on ICs underestimated the effects on some ICs in females. Insights from the GTEx dataset about the co-expression and transcriptional co-regulation of acetylation-modifying enzymes, transcription factors and key cardiac ion channels in a sex-specific manner can help inform safer drug design.
Collapse
Affiliation(s)
- Michael P. Pressler
- Department of Biomedical Engineering, George Washington University, Washington, DC, United States
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, United States
- *Correspondence: Emilia Entcheva,
| |
Collapse
|
30
|
de Sena-Tomás C, Aleman AG, Ford C, Varshney A, Yao D, Harrington JK, Saúde L, Ramialison M, Targoff KL. Activation of Nkx2.5 transcriptional program is required for adult myocardial repair. Nat Commun 2022; 13:2970. [PMID: 35624100 PMCID: PMC9142600 DOI: 10.1038/s41467-022-30468-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
The cardiac developmental network has been associated with myocardial regenerative potential. However, the embryonic signals triggered following injury have yet to be fully elucidated. Nkx2.5 is a key causative transcription factor associated with human congenital heart disease and one of the earliest markers of cardiac progenitors, thus it serves as a promising candidate. Here, we show that cardiac-specific RNA-sequencing studies reveal a disrupted embryonic transcriptional profile in the adult Nkx2.5 loss-of-function myocardium. nkx2.5-/- fish exhibit an impaired ability to recover following ventricular apex amputation with diminished dedifferentiation and proliferation. Complex network analyses illuminate that Nkx2.5 is required to provoke proteolytic pathways necessary for sarcomere disassembly and to mount a proliferative response for cardiomyocyte renewal. Moreover, Nkx2.5 targets embedded in these distinct gene regulatory modules coordinate appropriate, multi-faceted injury responses. Altogether, our findings support a previously unrecognized, Nkx2.5-dependent regenerative circuit that invokes myocardial cell cycle re-entry, proteolysis, and mitochondrial metabolism to ensure effective regeneration in the teleost heart.
Collapse
Affiliation(s)
- Carmen de Sena-Tomás
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Angelika G Aleman
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Caitlin Ford
- Department of Genetics & Development, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Akriti Varshney
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Australian Regenerative Medicine Institute & Systems Biology Institute Australia, Monash University, Clayton, VIC, 3800, Australia
| | - Di Yao
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jamie K Harrington
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Leonor Saúde
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute & Systems Biology Institute Australia, Monash University, Clayton, VIC, 3800, Australia
- Murdoch Children's Research Institute & Department of Peadiatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Kimara L Targoff
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA.
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
31
|
Ma J, Zhan Z, Li N, Huang Y, Li Y, Liu L, Shen Q, Chu Q, Wang X, Wu B, Zhang H. Preliminary Interpretations of Epigenetic Profiling of Cord Blood in Preeclampsia. Genes (Basel) 2022; 13:888. [PMID: 35627272 PMCID: PMC9141867 DOI: 10.3390/genes13050888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/01/2022] [Accepted: 05/14/2022] [Indexed: 12/04/2022] Open
Abstract
Preeclampsia (PE) is characterized by new-onset hypertension after 20 weeks of pregnancy and results in high maternal and fetal mortality worldwide. It has been reported that PE is associated with abnormalities in the umbilical cord and cord blood. However, previous studies were focused primarily on the transcriptomics level, while the underlying gene regulatory landscapes are still unclear. Thus, we performed the Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) using the umbilical cord blood samples collected from a patient with superimposed PE and three healthy donors to uncover the chromatin accessibility changes attributed to PE. We have identified genes associated with immunomodulation and hypoxia response that have higher chromatin accessibility close to their transcription start sites. Motif analysis indicated that the GATA family transcription factor binding was enriched in PE and may play an essential regulatory role in the disease progression. Overall, our findings provide an overview of gene regulatory programs and the corresponding downstream pathways associated with PE that may influence the placenta function and fetal growth.
Collapse
Affiliation(s)
- Junrui Ma
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.M.); (Q.C.); (X.W.)
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhongqun Zhan
- Institute of Translational Medicine, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518106, China; (Z.Z.); (Q.S.)
| | - Ning Li
- Cytotherapy Laboratory, The First Affiliated Hospital (Shenzhen People’s Hospital) Southern University of Science and Technology, Shenzhen 518020, China;
| | - Yanli Huang
- Department of Obstetrics, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518106, China; (Y.H.); (Y.L.); (L.L.)
| | - Yan Li
- Department of Obstetrics, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518106, China; (Y.H.); (Y.L.); (L.L.)
| | - Lu Liu
- Department of Obstetrics, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518106, China; (Y.H.); (Y.L.); (L.L.)
| | - Qi Shen
- Institute of Translational Medicine, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518106, China; (Z.Z.); (Q.S.)
| | - Qiao Chu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.M.); (Q.C.); (X.W.)
| | - Xiaonan Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.M.); (Q.C.); (X.W.)
| | - Benqing Wu
- Institute of Translational Medicine, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518106, China; (Z.Z.); (Q.S.)
- Department of Neonatology, Shenzhen Guangming Maternity & Child Healthcare Hospital, Shenzhen 518107, China
| | - Hui Zhang
- Institute of Translational Medicine, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen 518106, China; (Z.Z.); (Q.S.)
| |
Collapse
|
32
|
Mubeen H, Farooq M, Rehman AU, Zubair M, Haque A. Gene expression and transcriptional regulation driven by transcription factors involved in congenital heart defects. Ir J Med Sci 2022; 192:595-604. [PMID: 35441975 DOI: 10.1007/s11845-022-02974-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/24/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Congenital heart disease (CHD) is one of the most important birth defects caused by more than one mutated gene. Mutations in the genes could cause different types of congenital heart defects including atrial septal defect (ASD), tetralogy of Fallot (TOF), and ventricular septal defect (VSD). OBJECTIVES Cardiac transcription factors are key players for heart development and are actively involved in controlling stress regulation of the heart. Transcription factors are sequence-specific DNA binding proteins that control the process of transcription and work in a synergistic manner. We aim to characterize core cardiac transcription factors including NKX2-5, TBX, SRF, GATA4, and MEF2, which encode homeobox and MADS domain and play a crucial role in heart development. METHODS In this study, we have explored the important transcription factors involved in cardiac development and genes controlling the expression and regulation process by using the bioinformatics approach. RESULTS We have predicted the orthologs and homologs based on their evolutionary history, conserved protein domains, functional sites, and 3D structures for better understanding and presentation of factors responsible for causing CHD. Results showed the importance of these transcription factors for normal heart functioning and development. CONCLUSION Understanding the molecular pathways and genetic basis of CHD will help to open a new door for the treatment of patients with cardiac defects.
Collapse
Affiliation(s)
- Hira Mubeen
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Farooq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan. .,Department of Bioinformatics, Institute of Biochemistry, Biotechnology & Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | | | - Muhammad Zubair
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Asma Haque
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
33
|
Yan Y, Long T, Su Q, Wang Y, Chen K, Yang T, Zhao G, Ma Q, Hu X, Liu C, Liao X, Min W, Li S, Zhang D, Yang Y, Pu WT, Dong Y, Wang DZ, Chen Y, Huang ZP. Cardiac ISL1-Interacting Protein, a Cardioprotective Factor, Inhibits the Transition From Cardiac Hypertrophy to Heart Failure. Front Cardiovasc Med 2022; 9:857049. [PMID: 35369338 PMCID: PMC8970336 DOI: 10.3389/fcvm.2022.857049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
Heart failure is characterized by the inability of the heart to pump effectively and generate proper blood circulation to meet the body’s needs; it is a devastating condition that affects more than 100 million people globally. In spite of this, little is known about the mechanisms regulating the transition from cardiac hypertrophy to heart failure. Previously, we identified a cardiomyocyte-enriched gene, CIP, which regulates cardiac homeostasis under pathological stimulation. Here, we show that the cardiac transcriptional factor GATA4 binds the promotor of CIP gene and regulates its expression. We further determined that both CIP mRNA and protein decrease in diseased human hearts. In a mouse model, induced cardiac-specific overexpression of CIP after the establishment of cardiac hypertrophy protects the heart by inhibiting disease progression toward heart failure. Transcriptome analyses revealed that the IGF, mTORC2 and TGFβ signaling pathways mediate the inhibitory function of CIP on pathologic cardiac remodeling. Our study demonstrates GATA4 as an upstream regulator of CIP gene expression in cardiomyocytes, as well as the clinical significance of CIP expression in human heart disease. More importantly, our investigation suggests CIP is a key regulator of the transition from cardiac hypertrophy to heart failure. The ability of CIP to intervene in the onset of heart failure suggests a novel therapeutic avenue of investigation for the prevention of heart disease progression.
Collapse
Affiliation(s)
- Youchen Yan
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Long
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Qiao Su
- Laboratory Animal Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ken Chen
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Machine Intelligence and Advanced Computing, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Tiqun Yang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Guangyin Zhao
- Laboratory Animal Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Chen Liu
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Wang Min
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shujuan Li
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Dihua Zhang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuedong Yang
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Machine Intelligence and Advanced Computing, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Da-Zhi Wang,
| | - Yili Chen
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Yili Chen,
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
- Zhan-Peng Huang,
| |
Collapse
|
34
|
Jiang L, Liang J, Huang W, Ma J, Park KH, Wu Z, Chen P, Zhu H, Ma JJ, Cai W, Paul C, Niu L, Fan GC, Wang HS, Kanisicak O, Xu M, Wang Y. CRISPR activation of endogenous genes reprograms fibroblasts into cardiovascular progenitor cells for myocardial infarction therapy. Mol Ther 2022; 30:54-74. [PMID: 34678511 PMCID: PMC8753567 DOI: 10.1016/j.ymthe.2021.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/27/2021] [Accepted: 10/18/2021] [Indexed: 01/07/2023] Open
Abstract
Fibroblasts can be reprogrammed into cardiovascular progenitor cells (CPCs) using transgenic approaches, although the underlying mechanism remains unclear. We determined whether activation of endogenous genes such as Gata4, Nkx2.5, and Tbx5 can rapidly establish autoregulatory loops and initiate CPC generation in adult extracardiac fibroblasts using a CRISPR activation system. The induced fibroblasts (>80%) showed phenotypic changes as indicated by an Nkx2.5 cardiac enhancer reporter. The progenitor characteristics were confirmed by colony formation and expression of cardiovascular genes. Cardiac sphere induction segregated the early and late reprogrammed cells that can generate functional cardiomyocytes and vascular cells in vitro. Therefore, they were termed CRISPR-induced CPCs (ciCPCs). Transcriptomic analysis showed that cell cycle and heart development pathways were important to accelerate CPC formation during the early reprogramming stage. The CRISPR system opened the silenced chromatin locus, thereby allowing transcriptional factors to access their own promoters and eventually forming a positive feedback loop. The regenerative potential of ciCPCs was assessed after implantation in mouse myocardial infarction models. The engrafted ciCPCs differentiated into cardiovascular cells in vivo but also significantly improved contractile function and scar formation. In conclusion, multiplex gene activation was sufficient to drive CPC reprogramming, providing a new cell source for regenerative therapeutics.
Collapse
Affiliation(s)
- Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ki Ho Park
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Peng Chen
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jian-Jie Ma
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wenfeng Cai
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Liang Niu
- Division of Biostatistics and Bioinformatics, Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
35
|
Singh VP, Pinnamaneni JP, Pugazenthi A, Sanagasetti D, Mathison M, Martin JF, Yang J, Rosengart TK. Hippo Pathway Effector Tead1 Induces Cardiac Fibroblast to Cardiomyocyte Reprogramming. J Am Heart Assoc 2021; 10:e022659. [PMID: 34889103 PMCID: PMC9075224 DOI: 10.1161/jaha.121.022659] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/09/2021] [Indexed: 01/14/2023]
Abstract
Background The conversion of fibroblasts into induced cardiomyocytes may regenerate myocardial tissue from cardiac scar through in situ cell transdifferentiation. The efficiency transdifferentiation is low, especially for human cells. We explored the leveraging of Hippo pathway intermediates to enhance induced cardiomyocyte generation. Methods and Results We screened Hippo effectors Yap (yes-associated protein), Taz (transcriptional activator binding domain), and Tead1 (TEA domain transcription factor 1; Td) for their reprogramming efficacy with cardio-differentiating factors Gata4, Mef2C, and Tbx5 (GMT). Td induced nearly 3-fold increased expression of cardiomyocyte marker cTnT (cardiac troponin T) by mouse embryonic and adult rat fibroblasts versus GMT administration alone (P<0.0001), while Yap and Taz failed to enhance cTnT expression. Serial substitution demonstrated that Td replacement of TBX5 induced the greatest cTnT expression enhancement and sarcomere organization in rat fibroblasts treated with all GMT substitutions (GMTd versus GMT: 17±1.2% versus 5.4±0.3%, P<0.0001). Cell contractility (beating) was seen in 6% of GMTd-treated cells by 4 weeks after treatment, whereas no beating GMT-treated cells were observed. Human cardiac fibroblasts likewise demonstrated increased cTnT expression with GMTd versus GMT treatment (7.5±0.3% versus 3.0±0.3%, P<0.01). Mechanistically, GMTd administration increased expression of the trimethylated lysine 4 of histone 3 (H3K4me3) mark at the promoter regions of cardio-differentiation genes and mitochondrial biogenesis regulator genes in rat and human fibroblast, compared with GMT. Conclusions These data suggest that the Hippo pathway intermediate Tead1 is an important regulator of cardiac reprogramming that increases the efficiency of maturate induced cardiomyocytes generation and may be a vital component of human cardiodifferentiation strategies.
Collapse
Affiliation(s)
- Vivek P. Singh
- Department of SurgeryBaylor College of MedicineHoustonTX
| | | | | | | | | | - James F. Martin
- Department of Molecular Physiology and BiophysicsBaylor College of MedicineHoustonTX
| | - Jianchang Yang
- Department of SurgeryBaylor College of MedicineHoustonTX
| | | |
Collapse
|
36
|
Maurya SS. Role of Enhancers in Development and Diseases. EPIGENOMES 2021; 5:epigenomes5040021. [PMID: 34968246 PMCID: PMC8715447 DOI: 10.3390/epigenomes5040021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 12/26/2022] Open
Abstract
Enhancers are cis-regulatory elements containing short DNA sequences that serve as binding sites for pioneer/regulatory transcription factors, thus orchestrating the regulation of genes critical for lineage determination. The activity of enhancer elements is believed to be determined by transcription factor binding, thus determining the cell state identity during development. Precise spatio-temporal control of the transcriptome during lineage specification requires the coordinated binding of lineage-specific transcription factors to enhancers. Thus, enhancers are the primary determinants of cell identity. Numerous studies have explored the role and mechanism of enhancers during development and disease, and various basic questions related to the functions and mechanisms of enhancers have not yet been fully answered. In this review, we discuss the recently published literature regarding the roles of enhancers, which are critical for various biological processes governing development. Furthermore, we also highlight that altered enhancer landscapes provide an essential context to understand the etiologies and mechanisms behind numerous complex human diseases, providing new avenues for effective enhancer-based therapeutic interventions.
Collapse
Affiliation(s)
- Shailendra S Maurya
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Department of Developmental Biology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
37
|
Xu H, Xiang M, Qin Y, Cheng H, Chen D, Fu Q, Zhang KK, Xie L. Tbx5 inhibits hedgehog signaling in determination of digit identity. Hum Mol Genet 2021; 29:1405-1416. [PMID: 31373354 DOI: 10.1093/hmg/ddz185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/02/2019] [Accepted: 07/18/2019] [Indexed: 01/27/2023] Open
Abstract
Dominant TBX5 mutation causes Holt-Oram syndrome (HOS), which is characterized by limb defects in humans, but the underlying mechanistic basis is unclear. We used a mouse model with Tbx5 conditional knockdown in Hh-receiving cells (marked by Gli1+) during E8 to E10.5, a previously established model to study atrial septum defects, which displayed polydactyly or hypodactyly. The results suggested that Tbx5 is required for digit identity in a subset of limb mesenchymal cells. Specifically, Tbx5 deletion in this cell population decreased cell apoptosis and increased the proliferation of handplate mesenchymal cells. Furthermore, Tbx5 was found to negatively regulate the Hh-signaling activity through transcriptional regulation of Ptch1, a known Hh-signaling repressor. Repression of Hh-signaling through Smo co-mutation in Tbx5 heterozygotes rescued the limb defects, thus placing Tbx5 upstream of Hh-signaling in limb defects. This work reveals an important missing component necessary for understanding not only limb development but also the molecular and genetic mechanisms underlying HOS.
Collapse
Affiliation(s)
- Huiting Xu
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA.,Hubei Cancer Hospital, Wuhan, Hubei 430079, China
| | - Menglan Xiang
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Yushu Qin
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Henghui Cheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA.,Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Duohua Chen
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA.,Department of Food Science, Changsha University, Changsha, Hunan 410078, China
| | - Qiang Fu
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA.,Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ke K Zhang
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA.,Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Linglin Xie
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA.,Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
38
|
Fan D, Pang S, Chen J, Shan J, Cheng Q, Yan B. Identification and functional study of GATA4 gene regulatory variants in atrial septal defects. BMC Cardiovasc Disord 2021; 21:321. [PMID: 34193080 PMCID: PMC8243876 DOI: 10.1186/s12872-021-02136-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 06/24/2021] [Indexed: 11/29/2022] Open
Abstract
Background Congenital heart disease (CHD) is the leading cause of mortality from birth defects. In adult CHD patients with successful surgical repair, cardiac complications including heart failure develop at late stage, likely due to genetic causes. To date, many mutations in cardiac developmental genes have been associated with CHD. Recently, regulatory variants in genes have been linked to many human diseases. Although mutations and splicing variants in GATA4 gene have been reported in CHD patients, few regulatory variants of GATA4 gene are identified in CHD patients. Methods GATA4 gene regulatory region was investigated in the patients with atrial septal defects (ASD) (n = 332) and ethnic-matched controls (n = 336). Results Five heterozygous regulatory variants including four SNPs [g.31360 T>C (rs372004083), g.31436G>A, g.31437C>A (rs769262495), g.31487C>G (rs1053351749) and g.31856C>T (rs1385460518)] were only identified in ASD patients. Functional analysis indicated that the regulatory variants significantly affected the transcriptional activity of GATA4 gene promoter. Furthermore, two of the five regulatory variants have evidently effected on transcription factor binding sites. Conclusions Our data suggested that GATA4 gene regulatory variants may confer ASD susceptibility by decreasing GATA4 levels. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02136-w.
Collapse
Affiliation(s)
- Dongchen Fan
- Division of Medical Ultrasonics, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China
| | - Shuchao Pang
- Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China.,Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China.,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China
| | - Jing Chen
- Department of Medicine, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Jiping Shan
- Division of Cardiac Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China
| | - Qianjin Cheng
- Division of Cardiac Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China. .,Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China.
| | - Bo Yan
- Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China. .,Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China. .,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, China. .,Center for Molecular Medicine, Yanzhou People's Hospital, Jining, 272100, Shandong, China. .,Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China.
| |
Collapse
|
39
|
Delgado I, Giovinazzo G, Temiño S, Gauthier Y, Balsalobre A, Drouin J, Torres M. Control of mouse limb initiation and antero-posterior patterning by Meis transcription factors. Nat Commun 2021; 12:3086. [PMID: 34035267 PMCID: PMC8149412 DOI: 10.1038/s41467-021-23373-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 04/21/2021] [Indexed: 12/31/2022] Open
Abstract
Meis1 and Meis2 are homeodomain transcription factors that regulate organogenesis through cooperation with Hox proteins. Elimination of Meis genes after limb induction has shown their role in limb proximo-distal patterning; however, limb development in the complete absence of Meis function has not been studied. Here, we report that Meis1/2 inactivation in the lateral plate mesoderm of mouse embryos leads to limb agenesis. Meis and Tbx factors converge in this function, extensively co-binding with Tbx to genomic sites and co-regulating enhancers of Fgf10, a critical factor in limb initiation. Limbs with three deleted Meis alleles show proximal-specific skeletal hypoplasia and agenesis of posterior skeletal elements. This failure in posterior specification results from an early role of Meis factors in establishing the limb antero-posterior prepattern required for Shh activation. Our results demonstrate roles for Meis transcription factors in early limb development and identify their involvement in previously undescribed interaction networks that regulate organogenesis.
Collapse
Affiliation(s)
- Irene Delgado
- Cardiovascular Development Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Giovanna Giovinazzo
- Cardiovascular Development Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Susana Temiño
- Cardiovascular Development Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Yves Gauthier
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
| | - Aurelio Balsalobre
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
| | - Jacques Drouin
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
| | - Miguel Torres
- Cardiovascular Development Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
40
|
Venkatesh I, Mehra V, Wang Z, Simpson MT, Eastwood E, Chakraborty A, Beine Z, Gross D, Cabahug M, Olson G, Blackmore MG. Co-occupancy identifies transcription factor co-operation for axon growth. Nat Commun 2021; 12:2555. [PMID: 33953205 PMCID: PMC8099911 DOI: 10.1038/s41467-021-22828-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 03/29/2021] [Indexed: 12/13/2022] Open
Abstract
Transcription factors (TFs) act as powerful levers to regulate neural physiology and can be targeted to improve cellular responses to injury or disease. Because TFs often depend on cooperative activity, a major challenge is to identify and deploy optimal sets. Here we developed a bioinformatics pipeline, centered on TF co-occupancy of regulatory DNA, and used it to predict factors that potentiate the effects of pro-regenerative Klf6 in vitro. High content screens of neurite outgrowth identified cooperative activity by 12 candidates, and systematic testing in a mouse model of corticospinal tract (CST) damage substantiated three novel instances of pairwise cooperation. Combined Klf6 and Nr5a2 drove the strongest growth, and transcriptional profiling of CST neurons identified Klf6/Nr5a2-responsive gene networks involved in macromolecule biosynthesis and DNA repair. These data identify TF combinations that promote enhanced CST growth, clarify the transcriptional correlates, and provide a bioinformatics approach to detect TF cooperation.
Collapse
Affiliation(s)
- Ishwariya Venkatesh
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA.
| | - Vatsal Mehra
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Zimei Wang
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Matthew T Simpson
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Erik Eastwood
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | | | - Zac Beine
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Derek Gross
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Michael Cabahug
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Greta Olson
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Murray G Blackmore
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA.
| |
Collapse
|
41
|
Garry GA, Bezprozvannaya S, Chen K, Zhou H, Hashimoto H, Morales MG, Liu N, Bassel-Duby R, Olson EN. The histone reader PHF7 cooperates with the SWI/SNF complex at cardiac super enhancers to promote direct reprogramming. Nat Cell Biol 2021; 23:467-475. [PMID: 33941892 DOI: 10.1038/s41556-021-00668-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
Direct cardiac reprogramming of fibroblasts to cardiomyocytes presents an attractive therapeutic strategy to restore cardiac function following injury. Cardiac reprogramming was initially achieved through overexpression of the transcription factors Gata4, Mef2c and Tbx5; later, Hand2 and Akt1 were found to further enhance this process1-5. Yet, staunch epigenetic barriers severely limit the ability of these cocktails to reprogramme adult fibroblasts6,7. We undertook a screen of mammalian gene regulatory factors to discover novel regulators of cardiac reprogramming in adult fibroblasts and identified the histone reader PHF7 as the most potent activating factor8. Mechanistically, PHF7 localizes to cardiac super enhancers in fibroblasts, and through cooperation with the SWI/SNF complex, it increases chromatin accessibility and transcription factor binding at these sites. Furthermore, PHF7 recruits cardiac transcription factors to activate a positive transcriptional autoregulatory circuit in reprogramming. Importantly, PHF7 achieves efficient reprogramming in the absence of Gata4. Here, we highlight the underexplored necessity of cardiac epigenetic readers, such as PHF7, in harnessing chromatin remodelling and transcriptional complexes to overcome critical barriers to direct cardiac reprogramming.
Collapse
Affiliation(s)
- Glynnis A Garry
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Svetlana Bezprozvannaya
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kenian Chen
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huanyu Zhou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hisayuki Hashimoto
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maria Gabriela Morales
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ning Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
42
|
Gacita AM, Fullenkamp DE, Ohiri J, Pottinger T, Puckelwartz MJ, Nobrega MA, McNally EM. Genetic Variation in Enhancers Modifies Cardiomyopathy Gene Expression and Progression. Circulation 2021; 143:1302-1316. [PMID: 33478249 PMCID: PMC8009836 DOI: 10.1161/circulationaha.120.050432] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Inherited cardiomyopathy associates with a range of phenotypes, mediated by genetic and nongenetic factors. Noninherited cardiomyopathy also displays varying progression and outcomes. Expression of cardiomyopathy genes is under the regulatory control of promoters and enhancers, and human genetic variation in promoters and enhancers may contribute to this variability. METHODS We superimposed epigenomic profiling from hearts and cardiomyocytes, including promoter-capture chromatin conformation information, to identify enhancers for 2 cardiomyopathy genes, MYH7 and LMNA. Enhancer function was validated in human cardiomyocytes derived from induced pluripotent stem cells. We also conducted a genome-wide search to ascertain genomic variation in enhancers positioned to alter cardiac expression and correlated one of these variants to cardiomyopathy progression using biobank data. RESULTS Multiple enhancers were identified and validated for LMNA and MYH7, including a key enhancer that regulates the switch from MYH6 expression to MYH7 expression. Deletion of this enhancer resulted in a dose-dependent increase in MYH6 and faster contractile rate in engineered heart tissues. We searched for genomic variation in enhancer sequences across the genome, with a focus on nucleotide changes that create or interrupt transcription factor binding sites. The sequence variant, rs875908, disrupts a T-Box Transcription Factor 5 binding motif and maps to an enhancer region 2 kilobases from the transcriptional start site of MYH7. Gene editing to remove the enhancer that harbors this variant markedly reduced MYH7 expression in human cardiomyocytes. Using biobank-derived data, rs875908 associated with longitudinal echocardiographic features of cardiomyopathy. CONCLUSIONS Enhancers regulate cardiomyopathy gene expression, and genomic variation within these enhancer regions associates with cardiomyopathic progression over time. This integrated approach identified noncoding modifiers of cardiomyopathy and is applicable to other cardiac genes.
Collapse
Affiliation(s)
- Anthony M. Gacita
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Dominic E. Fullenkamp
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Joyce Ohiri
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Tess Pottinger
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Megan J. Puckelwartz
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | | | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| |
Collapse
|
43
|
Vincentz JW, Firulli BA, Toolan KP, Osterwalder M, Pennacchio LA, Firulli AB. HAND transcription factors cooperatively specify the aorta and pulmonary trunk. Dev Biol 2021; 476:1-10. [PMID: 33757801 DOI: 10.1016/j.ydbio.2021.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 01/11/2023]
Abstract
Congenital heart defects (CHDs) affecting the cardiac outflow tract (OFT) constitute a significant cause of morbidity and mortality. The OFT develops from migratory cell populations which include the cardiac neural crest cells (cNCCs) and secondary heart field (SHF) derived myocardium and endocardium. The related transcription factors HAND1 and HAND2 have been implicated in human CHDs involving the OFT. Although Hand1 is expressed within the OFT, Hand1 NCC-specific conditional knockout mice (H1CKOs) are viable. Here we show that these H1CKOs present a low penetrance of OFT phenotypes, whereas SHF-specific Hand1 ablation does not reveal any cardiac phenotypes. Further, HAND1 and HAND2 appear functionally redundant within the cNCCs, as a reduction/ablation of Hand2 on an NCC-specific H1CKO background causes pronounced OFT defects. Double conditional Hand1 and Hand2 NCC knockouts exhibit persistent truncus arteriosus (PTA) with 100% penetrance. NCC lineage-tracing and Sema3c in situ mRNA expression reveal that Sema3c-expressing cells are mis-localized, resulting in a malformed septal bridge within the OFTs of H1CKO;H2CKO embryos. Interestingly, Hand1 and Hand2 also genetically interact within the SHF, as SHF H1CKOs on a heterozygous Hand2 background exhibit Ventricular Septal Defects (VSDs) with incomplete penetrance. Previously, we identified a BMP, HAND2, and GATA-dependent Hand1 OFT enhancer sufficient to drive reporter gene expression within the nascent OFT and aorta. Using these transcription inputs as a probe, we identify a novel Hand2 OFT enhancer, suggesting that a conserved BMP-GATA dependent mechanism transcriptionally regulates both HAND factors. These findings support the hypothesis that HAND factors interpret BMP signaling within the cNCCs to cooperatively coordinate OFT morphogenesis.
Collapse
Affiliation(s)
- Joshua W Vincentz
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA.
| | - Beth A Firulli
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA
| | - Kevin P Toolan
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA; Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008, Bern, Switzerland
| | - Len A Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA; U.S. Department of Energy Joint Genome Institute, Berkeley, CA, 94720, USA; Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, and Medical and Molecular Genetics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
44
|
Feng Y, Xu H, Liu J, Xie N, Gao L, He Y, Yao Y, Lv F, Zhang Y, Lu J, Zhang W, Li CY, Hu X, Yang Z, Xiao RP. Functional and Adaptive Significance of Promoter Mutations That Affect Divergent Myocardial Expressions of TRIM72 in Primates. Mol Biol Evol 2021; 38:2930-2945. [PMID: 33744959 PMCID: PMC8233513 DOI: 10.1093/molbev/msab083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cis-regulatory elements play important roles in tissue-specific gene expression and in the evolution of various phenotypes, and mutations in promoters and enhancers may be responsible for adaptations of species to environments. TRIM72 is a highly conserved protein that is involved in energy metabolism. Its expression in the heart varies considerably in primates, with high levels of expression in Old World monkeys and near absence in hominids. Here, we combine phylogenetic hypothesis testing and experimentation to demonstrate that mutations in promoter are responsible for the differences among primate species in the heart-specific expression of TRIM72. Maximum likelihood estimates of lineage-specific substitution rates under local-clock models show that relative to the evolutionary rate of introns, the rate of promoter was accelerated by 78% in the common ancestor of Old World monkeys, suggesting a role for positive selection in the evolution of the TRIM72 promoter, possibly driven by selective pressure due to changes in cardiac physiology after species divergence. We demonstrate that mutations in the TRIM72 promoter account for the differential myocardial TRIM72 expression of the human and the rhesus macaque. Furthermore, changes in TRIM72 expression alter the expression of genes involved in oxidative phosphorylation, which in turn affects mitochondrial respiration and cardiac energy capacity. On a broader timescale, phylogenetic regression analyses of data from 29 mammalian species show that mammals with high cardiac expression of TRIM72 have high heart rate, suggesting that the expression changes of TRIM72 may be related to differences in the heart physiology of those species.
Collapse
Affiliation(s)
- Yuanqing Feng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Hongzhan Xu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jinghao Liu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Ning Xie
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Lei Gao
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yanyun He
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yuan Yao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Fengxiang Lv
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yan Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jian Lu
- Peking-Tsinghua Center for Life Sciences, Beijing, China.,State Key Laboratory of Protein and Plant Gene Research, Beijing, China.,School of Life Sciences, Peking University, Beijing, China
| | - Wei Zhang
- Peking-Tsinghua Center for Life Sciences, Beijing, China.,State Key Laboratory of Protein and Plant Gene Research, Beijing, China.,School of Life Sciences, Peking University, Beijing, China
| | - Chuan-Yun Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Xinli Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Ziheng Yang
- Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China.,State Key Laboratory of Biomembrane and Membrane Biotechnology, Beijing, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
45
|
Taking Data Science to Heart: Next Scale of Gene Regulation. Curr Cardiol Rep 2021; 23:46. [PMID: 33721129 DOI: 10.1007/s11886-021-01467-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW Technical advances have facilitated high-throughput measurements of the genome in the context of cardiovascular biology. These techniques bring a deluge of gargantuan datasets, which in turn present two fundamentally new opportunities for innovation-data processing and knowledge integration-toward the goal of meaningful basic and translational discoveries. RECENT FINDINGS Big data, integrative analyses, and machine learning have brought cardiac investigations to the cutting edge of chromatin biology, not only to reveal basic principles of gene regulation in the heart, but also to aid in the design of targeted epigenetic therapies. SUMMARY Cardiac studies using big data are only beginning to integrate the millions of recorded data points and the tools of machine learning are aiding this process. Future experimental design should take into consideration insights from existing genomic datasets, thereby focusing on heretofore unexplored epigenomic contributions to disease pathology.
Collapse
|
46
|
Yuan X, Scott IC, Wilson MD. Heart Enhancers: Development and Disease Control at a Distance. Front Genet 2021; 12:642975. [PMID: 33777110 PMCID: PMC7987942 DOI: 10.3389/fgene.2021.642975] [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: 12/17/2020] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
Bound by lineage-determining transcription factors and signaling effectors, enhancers play essential roles in controlling spatiotemporal gene expression profiles during development, homeostasis and disease. Recent synergistic advances in functional genomic technologies, combined with the developmental biology toolbox, have resulted in unprecedented genome-wide annotation of heart enhancers and their target genes. Starting with early studies of vertebrate heart enhancers and ending with state-of-the-art genome-wide enhancer discovery and testing, we will review how studying heart enhancers in metazoan species has helped inform our understanding of cardiac development and disease.
Collapse
Affiliation(s)
- Xuefei Yuan
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ian C. Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Michael D. Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
47
|
Carreras D, Martinez-Moreno R, Pinsach-Abuin M, Santafe MM, Gomà P, Brugada R, Scornik FS, Pérez GJ, Pagans S. Epigenetic Changes Governing Scn5a Expression in Denervated Skeletal Muscle. Int J Mol Sci 2021; 22:ijms22052755. [PMID: 33803193 PMCID: PMC7963191 DOI: 10.3390/ijms22052755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
The SCN5A gene encodes the α-subunit of the voltage-gated cardiac sodium channel (NaV1.5), a key player in cardiac action potential depolarization. Genetic variants in protein-coding regions of the human SCN5A have been largely associated with inherited cardiac arrhythmias. Increasing evidence also suggests that aberrant expression of the SCN5A gene could increase susceptibility to arrhythmogenic diseases, but the mechanisms governing SCN5A expression are not yet well understood. To gain insights into the molecular basis of SCN5A gene regulation, we used rat gastrocnemius muscle four days following denervation, a process well known to stimulate Scn5a expression. Our results show that denervation of rat skeletal muscle induces the expression of the adult cardiac Scn5a isoform. RNA-seq experiments reveal that denervation leads to significant changes in the transcriptome, with Scn5a amongst the fifty top upregulated genes. Consistent with this increase in expression, ChIP-qPCR assays show enrichment of H3K27ac and H3K4me3 and binding of the transcription factor Gata4 near the Scn5a promoter region. Also, Gata4 mRNA levels are significantly induced upon denervation. Genome-wide analysis of H3K27ac by ChIP-seq suggest that a super enhancer recently described to regulate Scn5a in cardiac tissue is activated in response to denervation. Altogether, our experiments reveal that similar mechanisms regulate the expression of Scn5a in denervated muscle and cardiac tissue, suggesting a conserved pathway for SCN5A expression among striated muscles.
Collapse
Affiliation(s)
- David Carreras
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
| | - Rebecca Martinez-Moreno
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
| | - Mel·lina Pinsach-Abuin
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
| | - Manel M. Santafe
- Unit of Histology and Neurobiology, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Rovira i Virgili University, 43003 Reus, Spain;
| | - Pol Gomà
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 21005 Madrid, Spain
- Hospital Josep Trueta, 17007 Girona, Spain
| | - Fabiana S. Scornik
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 21005 Madrid, Spain
- Correspondence: (F.S.S.); (G.J.P.); (S.P.)
| | - Guillermo J. Pérez
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 21005 Madrid, Spain
- Correspondence: (F.S.S.); (G.J.P.); (S.P.)
| | - Sara Pagans
- Cardiovascular Genetics Center, Biomedical Research Institute of Girona, 17190 Salt, Spain; (D.C.); (R.M.-M.); (M.P.-A.); (P.G.); (R.B.)
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 21005 Madrid, Spain
- Correspondence: (F.S.S.); (G.J.P.); (S.P.)
| |
Collapse
|
48
|
Smad4 regulates the nuclear translocation of Nkx2-5 in cardiac differentiation. Sci Rep 2021; 11:3588. [PMID: 33574455 PMCID: PMC7878807 DOI: 10.1038/s41598-021-82954-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/25/2021] [Indexed: 01/16/2023] Open
Abstract
Bmp plays an important role in cardiomyocyte differentiation, but the function of Smad4 in Bmp signaling remains elusive. Here, we show that disruption of the Smad4 gene in cardiac progenitors expressing Sfrp5 led to embryonic lethality with hypoplastic heart formation. Although the expression of Nkx2-5 is regulated by Bmp signaling, expression of Nkx2-5 was weakly detected in the mutant heart. However, the nuclear translocation of Nkx2-5 was impaired. Expression of CK2 or PP1, which could alter the phosphorylation status of the NLS of Nkx2-5, was not affected, but Nkx2-5 was found to bind to Smad4 by co-immunoprecipitation experiments. Introduction of Smad4 into cells derived from Smad4 conditional knockout embryonic hearts restored the nuclear localization of Nkx2-5, and exogenous Nkx2-5 failed to translocate into the nucleus of Smad4-depleted fibroblasts. These results suggest that Smad4 plays an essential role in cardiomyocyte differentiation by controlling not only transcription but also the nuclear localization of Nkx2-5.
Collapse
|
49
|
Xia X, Yu CY, Bian M, Sun CB, Tanasa B, Chang KC, Bruffett DM, Thakur H, Shah SH, Knasel C, Cameron EG, Kapiloff MS, Goldberg JL. MEF2 transcription factors differentially contribute to retinal ganglion cell loss after optic nerve injury. PLoS One 2020; 15:e0242884. [PMID: 33315889 PMCID: PMC7735573 DOI: 10.1371/journal.pone.0242884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Loss of retinal ganglion cells (RGCs) in optic neuropathies results in permanent partial or complete blindness. Myocyte enhancer factor 2 (MEF2) transcription factors have been shown to play a pivotal role in neuronal systems, and in particular MEF2A knockout was shown to enhance RGC survival after optic nerve crush injury. Here we expanded these prior data to study bi-allelic, tri-allelic and heterozygous allele deletion. We observed that deletion of all MEF2A, MEF2C, and MEF2D alleles had no effect on RGC survival during development. Our extended experiments suggest that the majority of the neuroprotective effect was conferred by complete deletion of MEF2A but that MEF2D knockout, although not sufficient to increase RGC survival on its own, increased the positive effect of MEF2A knockout. Conversely, MEF2A over-expression in wildtype mice worsened RGC survival after optic nerve crush. Interestingly, MEF2 transcription factors are regulated by post-translational modification, including by calcineurin-catalyzed dephosphorylation of MEF2A Ser-408 known to increase MEF2A-dependent transactivation in neurons. However, neither phospho-mimetic nor phospho-ablative mutation of MEF2A Ser-408 affected the ability of MEF2A to promote RGC death in vivo after optic nerve injury. Together these findings demonstrate that MEF2 gene expression opposes RGC survival following axon injury in a complex hierarchy, and further support the hypothesis that loss of or interference with MEF2A expression might be beneficial for RGC neuroprotection in diseases such as glaucoma and other optic neuropathies.
Collapse
Affiliation(s)
- Xin Xia
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Caroline Y. Yu
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Minjuan Bian
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Catalina B. Sun
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Bogdan Tanasa
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Kun-Che Chang
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Dawn M. Bruffett
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Hrishikesh Thakur
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Sahil H. Shah
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Cara Knasel
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Evan G. Cameron
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Michael S. Kapiloff
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
- Department of Medicine and Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
- * E-mail: (MSK); (JLG)
| | - Jeffrey L. Goldberg
- Mary M. and Sash A. Spencer Center for Vision Research, Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States of America
- * E-mail: (MSK); (JLG)
| |
Collapse
|
50
|
Wang H, Liu Y, Guan H, Fan GL. The Regulation of Target Genes by Co-occupancy of Transcription Factors, c-Myc and Mxi1 with Max in the Mouse Cell Line. Curr Bioinform 2020. [DOI: 10.2174/1574893614666191106103633] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background:
The regulatory function of transcription factors on genes is not only related
to the location of binding genes and its related functions, but is also related to the methods of
binding.
Objective:
It is necessary to study the regulation effects in different binding methods on target genes.
Methods:
In this study, we provided a reliable theoretical basis for studying gene expression
regulation of co-binding transcription factors and further revealed the specific regulation of
transcription factor co-binding in cancer cells.
Results:
Transcription factors tend to combine with other transcription factors in the regulatory
region to form a competitive or synergistic relationship to regulate target genes accurately.
Conclusion:
We found that up-regulated genes in cancer cells were involved in the regulation of
their own immune system related to the normal cells.
Collapse
Affiliation(s)
- Hui Wang
- Department of Physics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, China
| | - Yuan Liu
- Department of Physics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, China
| | - Hua Guan
- ENT Department, Huhhot First Hospital, Hohhot, China
| | - Guo-Liang Fan
- Department of Physics, School of Physical Science and Technology, Inner Mongolia University, Hohhot, China
| |
Collapse
|