1
|
Núñez-Martínez HN, Tapia-Urzúa G, Cerecedo-Castillo Á, Peralta-Alvarez C, Guerrero G, Huarte M, Recillas-Targa F. The lncRNA DUBR is regulated by CTCF and coordinates chromatin landscape and gene expression in hematopoietic cells. Nucleic Acids Res 2025; 53:gkaf093. [PMID: 39995041 PMCID: PMC11850227 DOI: 10.1093/nar/gkaf093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Master hematopoietic transcription factors (TFs) and long noncoding RNAs (lncRNAs) coordinate shaping lineage-specific gene expression programs during hematopoietic differentiation. The architectural protein CCCTC-binding factor (CTCF) has emerged as a pivotal regulator of gene expression in cell differentiation. However, the relationship and its regulatory effect of CTCF on lncRNA genes in hematopoiesis remain elusive. We demonstrated that CTCF constrains the lncRNA DUBRtranscription throughout erythroid differentiation. DUBR is highly expressed in human hematopoietic stem and progenitor cells (HSPCs) but depleted in erythroblasts. DUBR perturbation dysregulates hematopoietic-erythroid cell differentiation genes and facilitates genome-wide activation of regulatory elements. A genomic map of RNA occupancy revealed that DUBR associates with a set of genes involved in regulating hematopoietic differentiation, including the erythroid repressor HES1, which targets a subset of regulatory elements of DUBR-dysregulated genes. Our results support the role of DUBR as a regulator of a hematopoietic differentiation gene program by coordinating the expression of genes and influencing their chromatin regulatory landscape.
Collapse
Affiliation(s)
- Hober Nelson Núñez-Martínez
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Gustavo Tapia-Urzúa
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Ángel Josué Cerecedo-Castillo
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Carlos Alberto Peralta-Alvarez
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
- Unidad de Bioinformática y Manejo de la Información, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Georgina Guerrero
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Maite Huarte
- Center for Applied Medical Research, Department of Gene Therapy and Regulation of Gene Expression, University of Navarra, Pamplona, 31008, Spain
| | - Félix Recillas-Targa
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| |
Collapse
|
2
|
Guan X, Fan Y, Six R, Van Soom A, Pavani KC, Peelman L. MicroRNAs bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 have adverse effects on blastocyst formation. Theriogenology 2025; 233:88-99. [PMID: 39613498 DOI: 10.1016/j.theriogenology.2024.11.018] [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: 08/09/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
In a previous study we found that the levels of the novel microRNAs (miRNAs) bta-novel-miR-117 bta-novel-miR-234 and bta-novel-miR-417 (P < 0.001) are significantly up-regulated in extracellular vesicles (EVs) in the culture medium of degenerating embryos compared to blastocysts. Because the functions of these novel miRNAs are still unknown, we investigated their regulatory roles during bovine blastocyst development by adding their mimics and inhibitors to the culture medium. The addition of mimics for bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 resulted in a decreased blastocyst rate, and supplementation of bta-novel-miR-234 inhibitors increased the cleavage rate significantly (P < 0.001). Low-input transcriptome analysis and RT-qPCR results revealed that bta-novel-miR-117, bta-novel-miR-234 and bta-novel-miR-417 co-target genes such as ANKEF1, HAND2 and SLC2A2, downregulated their expression significantly (P < 0.001). These genes associated with glucose transmembrane transport and plasma membrane raft metabolism play crucial roles in embryonic development. The results suggest that overexpressing of these three novel miRNAs impairs embryonic development, and they might serve as biomarkers to detect failing bovine embryos.
Collapse
Affiliation(s)
- Xuefeng Guan
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Yuan Fan
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Rani Six
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium
| | - Ann Van Soom
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Krishna Chaitanya Pavani
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, Salisburylaan 133, B-9820, Merelbeke, Belgium; Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Gent, Belgium.
| | - Luc Peelman
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820, Merelbeke, Belgium.
| |
Collapse
|
3
|
DNA-binding-deficient Hand2 dimerizes with Tcf3a to control zebrafish cardiogenesis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1383-1384. [PMID: 39658722 DOI: 10.1038/s44161-024-00589-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
|
4
|
Xu Y, Gehlot R, Capon SJ, Albu M, Gretz J, Bloomekatz J, Mattonet K, Vucicevic D, Talyan S, Kikhi K, Günther S, Looso M, Firulli BA, Sanda M, Firulli AB, Lacadie SA, Yelon D, Stainier DYR. PDGFRA is a conserved HAND2 effector during early cardiac development. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1531-1548. [PMID: 39658721 PMCID: PMC11634778 DOI: 10.1038/s44161-024-00574-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/29/2024] [Indexed: 12/12/2024]
Abstract
The basic helix-loop-helix transcription factor HAND2 has multiple roles during vertebrate organogenesis, including cardiogenesis. However, much remains to be uncovered about its mechanism of action. Here, we show the generation of several hand2 mutant alleles in zebrafish and demonstrate that dimerization-deficient mutants display the null phenotype but DNA-binding-deficient mutants do not. Rescue experiments with Hand2 variants using a newly identified hand2 enhancer confirmed these observations. To identify Hand2 effectors critical for cardiogenesis, we analyzed the transcriptomes of hand2 loss- and gain-of-function embryonic cardiomyocytes and tested the function of eight candidate genes in vivo; pdgfra was most effective in rescuing myocardial migration in hand2 mutants. Accordingly, we identified a putative Hand2-binding region in the zebrafish pdgfra locus that is important for its expression. In addition, Hand2 loss- and gain-of-function experiments in mouse embryonic stem cell-derived cardiac cells decreased and increased Pdgfra expression, respectively. Altogether, these results further our mechanistic understanding of HAND2 function during early cardiogenesis.
Collapse
Affiliation(s)
- Yanli Xu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rupal Gehlot
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Samuel J Capon
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marga Albu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jonas Gretz
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Joshua Bloomekatz
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Biology, University of Mississippi, University, MS, USA
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Dubravka Vucicevic
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Sweta Talyan
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Khrievono Kikhi
- Flow Cytometry Service Group, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Beth A Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN, USA
| | - Miloslav Sanda
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana Medical School, Indianapolis, IN, USA
| | - Scott Allen Lacadie
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| |
Collapse
|
5
|
Siewert A, Hoeland S, Mangold E, Ludwig KU. Combining genetic and single-cell expression data reveals cell types and novel candidate genes for orofacial clefting. Sci Rep 2024; 14:26492. [PMID: 39489835 PMCID: PMC11532359 DOI: 10.1038/s41598-024-77724-9] [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: 08/22/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024] Open
Abstract
Non-syndromic cleft lip with/without cleft palate (nsCL/P) is one of the most common birth defects and has a multifactorial etiology. To date, over 45 loci harboring common risk variants have been identified. However, the effector genes at these loci, and the cell types that are affected by risk alleles, remain largely unknown. To address this, we combined genetic data from an nsCL/P genome-wide association study (GWAS) with single-cell RNA sequencing data obtained from the heads of unaffected human embryos. Using the recently developed single-cell disease relevance score (scDRS) approach, we identified two major cell types involved in nsCL/P development, namely the epithelium and the HAND2+ pharyngeal arches (PA). Combining scDRS with co-expression networks and differential gene expression analysis, we prioritized nsCL/P candidate genes, some of which were additionally supported by GWAS data (e.g., CTNND1, PRTG, RPL35A, RAB11FIP1, KRT19). Our results suggest that specific epithelial and PA sub-cell types are involved in nsCL/P development, and harbor a substantial fraction of the genetic risk for nsCL/P.
Collapse
Affiliation(s)
- Anna Siewert
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany.
| | - Simone Hoeland
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Elisabeth Mangold
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Kerstin U Ludwig
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
6
|
To K, Fei L, Pett JP, Roberts K, Blain R, Polański K, Li T, Yayon N, He P, Xu C, Cranley J, Moy M, Li R, Kanemaru K, Huang N, Megas S, Richardson L, Kapuge R, Perera S, Tuck E, Wilbrey-Clark A, Mulas I, Memi F, Cakir B, Predeus AV, Horsfall D, Murray S, Prete M, Mazin P, He X, Meyer KB, Haniffa M, Barker RA, Bayraktar O, Chédotal A, Buckley CD, Teichmann SA. A multi-omic atlas of human embryonic skeletal development. Nature 2024; 635:657-667. [PMID: 39567793 PMCID: PMC11578895 DOI: 10.1038/s41586-024-08189-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/09/2024] [Indexed: 11/22/2024]
Abstract
Human embryonic bone and joint formation is determined by coordinated differentiation of progenitors in the nascent skeleton. The cell states, epigenetic processes and key regulatory factors that underlie lineage commitment of these cells remain elusive. Here we applied paired transcriptional and epigenetic profiling of approximately 336,000 nucleus droplets and spatial transcriptomics to establish a multi-omic atlas of human embryonic joint and cranium development between 5 and 11 weeks after conception. Using combined modelling of transcriptional and epigenetic data, we characterized regionally distinct limb and cranial osteoprogenitor trajectories across the embryonic skeleton and further described regulatory networks that govern intramembranous and endochondral ossification. Spatial localization of cell clusters in our in situ sequencing data using a new tool, ISS-Patcher, revealed mechanisms of progenitor zonation during bone and joint formation. Through trajectory analysis, we predicted potential non-canonical cellular origins for human chondrocytes from Schwann cells. We also introduce SNP2Cell, a tool to link cell-type-specific regulatory networks to polygenic traits such as osteoarthritis. Using osteolineage trajectories characterized here, we simulated in silico perturbations of genes that cause monogenic craniosynostosis and implicate potential cell states and disease mechanisms. This work forms a detailed and dynamic regulatory atlas of bone and cartilage maturation and advances our fundamental understanding of cell-fate determination in human skeletal development.
Collapse
Affiliation(s)
- Ken To
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Lijiang Fei
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - J Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Kenny Roberts
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Raphael Blain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nadav Yayon
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
| | - Peng He
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - James Cranley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Madelyn Moy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Ruoyan Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Stathis Megas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Centre for AI in Medicine, Department of Applied Mathematics and Theoretical Physics, Cambridge, UK
| | | | - Rakesh Kapuge
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Elizabeth Tuck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Ilaria Mulas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Fani Memi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - David Horsfall
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Simon Murray
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Pavel Mazin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Xiaoling He
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Newcastle University, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Omer Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Institut de Pathologie, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | | | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Centre for AI in Medicine, Department of Applied Mathematics and Theoretical Physics, Cambridge, UK.
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- CIFAR Macmillan Multi-scale Human Programme, CIFAR, Toronto, Canada.
| |
Collapse
|
7
|
Signal B, Phipps AJ, Giles KA, Huskins SN, Mercer TR, Robinson MD, Woodhouse A, Taberlay PC. Ageing-Related Changes to H3K4me3, H3K27ac, and H3K27me3 in Purified Mouse Neurons. Cells 2024; 13:1393. [PMID: 39195281 PMCID: PMC11353134 DOI: 10.3390/cells13161393] [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: 07/09/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
Neurons are central to lifelong learning and memory, but ageing disrupts their morphology and function, leading to cognitive decline. Although epigenetic mechanisms are known to play crucial roles in learning and memory, neuron-specific genome-wide epigenetic maps into old age remain scarce, often being limited to whole-brain homogenates and confounded by glial cells. Here, we mapped H3K4me3, H3K27ac, and H3K27me3 in mouse neurons across their lifespan. This revealed stable H3K4me3 and global losses of H3K27ac and H3K27me3 into old age. We observed patterns of synaptic function gene deactivation, regulated through the loss of the active mark H3K27ac, but not H3K4me3. Alongside this, embryonic development loci lost repressive H3K27me3 in old age. This suggests a loss of a highly refined neuronal cellular identity linked to global chromatin reconfiguration. Collectively, these findings indicate a key role for epigenetic regulation in neurons that is inextricably linked with ageing.
Collapse
Affiliation(s)
- Brandon Signal
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS 7000, Australia; (B.S.); (K.A.G.); (S.N.H.)
| | - Andrew J. Phipps
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS 7000, Australia;
| | - Katherine A. Giles
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS 7000, Australia; (B.S.); (K.A.G.); (S.N.H.)
- Children’s Medical Research Institute, University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Shannon N. Huskins
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS 7000, Australia; (B.S.); (K.A.G.); (S.N.H.)
| | - Timothy R. Mercer
- Australian Institute for Bioengineering and Nanotechnology, Corner College and Cooper Roads, Brisbane, QLD 4072, Australia;
| | - Mark D. Robinson
- SIB Swiss Institute of Bioinformatics, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland;
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS 7000, Australia;
| | - Phillippa C. Taberlay
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS 7000, Australia; (B.S.); (K.A.G.); (S.N.H.)
| |
Collapse
|
8
|
Guo H, Hang C, Lin B, Lin Z, Xiong H, Zhang M, Lu R, Liu J, Shi D, Xie D, Liu Y, Liang D, Yang J, Chen YH. HAND factors regulate cardiac lineage commitment and differentiation from human pluripotent stem cells. Stem Cell Res Ther 2024; 15:31. [PMID: 38317221 PMCID: PMC10845658 DOI: 10.1186/s13287-024-03649-9] [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: 11/06/2023] [Accepted: 01/25/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Transcription factors HAND1 and HAND2 (HAND1/2) play significant roles in cardiac organogenesis. Abnormal expression and deficiency of HAND1/2 result in severe cardiac defects. However, the function and mechanism of HAND1/2 in regulating human early cardiac lineage commitment and differentiation are still unclear. METHODS With NKX2.5eGFP H9 human embryonic stem cells (hESCs), we established single and double knockout cell lines for HAND1 and HAND2, respectively, whose cardiomyocyte differentiation efficiency could be monitored by assessing NKX2.5-eGFP+ cells with flow cytometry. The expression of specific markers for heart fields and cardiomyocyte subtypes was examined by quantitative PCR, western blot and immunofluorescence staining. Microelectrode array and whole-cell patch clamp were performed to determine the electrophysiological characteristics of differentiated cardiomyocytes. The transcriptomic changes of HAND knockout cells were revealed by RNA sequencing. The HAND1/2 target genes were identified and validated experimentally by integrating with HAND1/2 chromatin immunoprecipitation sequencing data. RESULTS Either HAND1 or HAND2 knockout did not affect the cardiomyocyte differentiation kinetics, whereas depletion of HAND1/2 resulted in delayed differentiation onset. HAND1 knockout biased cardiac mesoderm toward second heart field progenitors at the expense of first heart field progenitors, leading to increased expression of atrial and outflow tract cardiomyocyte markers, which was further confirmed by the appearance of atrial-like action potentials. By contrast, HAND2 knockout cardiomyocytes had reduced expression of atrial cardiomyocyte markers and displayed ventricular-like action potentials. HAND1/2-deficient hESCs were more inclined to second heart field lineage and its derived cardiomyocytes with atrial-like action potentials than HAND1 single knockout during differentiation. Further mechanistic investigations suggested TBX5 as one of the downstream targets of HAND1/2, whose overexpression partially restored the abnormal cardiomyocyte differentiation in HAND1/2-deficient hESCs. CONCLUSIONS HAND1/2 have specific and redundant roles in cardiac lineage commitment and differentiation. These findings not only reveal the essential function of HAND1/2 in cardiac organogenesis, but also provide important information on the pathogenesis of HAND1/2 deficiency-related congenital heart diseases, which could potentially lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Huixin Guo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Chengwen Hang
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
| | - Bowen Lin
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
| | - Zheyi Lin
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Hui Xiong
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Cell Biology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Mingshuai Zhang
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Cell Biology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Renhong Lu
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
| | - Junyang Liu
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Cell Biology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Dan Shi
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
| | - Duanyang Xie
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yi Liu
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Dandan Liang
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, 200092, China
| | - Jian Yang
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China.
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China.
- Department of Cell Biology, Tongji University School of Medicine, Shanghai, 200092, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, 200092, China.
| | - Yi-Han Chen
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China.
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China.
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
9
|
Haridhasapavalan KK, Sundaravadivelu PK, Joshi N, Das NJ, Mohapatra A, Voorkara U, Kaveeshwar V, Thummer RP. Generation of a recombinant version of a biologically active cell-permeant human HAND2 transcription factor from E. coli. Sci Rep 2022; 12:16129. [PMID: 36167810 PMCID: PMC9515176 DOI: 10.1038/s41598-022-19745-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Transcription factor HAND2 has a significant role in vascularization, angiogenesis, and cardiac neural crest development. It is one of the key cardiac factors crucial for the enhanced derivation of functional and mature myocytes from non-myocyte cells. Here, we report the generation of the recombinant human HAND2 fusion protein from the heterologous system. First, we cloned the full-length human HAND2 gene (only protein-coding sequence) after codon optimization along with the fusion tags (for cell penetration, nuclear translocation, and affinity purification) into the expression vector. We then transformed and expressed it in Escherichia coli strain, BL21(DE3). Next, the effect (in terms of expression) of tagging fusion tags with this recombinant protein at two different terminals was also investigated. Using affinity chromatography, we established the one-step homogeneous purification of recombinant human HAND2 fusion protein; and through circular dichroism spectroscopy, we established that this purified protein had retained its secondary structure. We then showed that this purified human protein could transduce the human cells and translocate to its nucleus. The generated recombinant HAND2 fusion protein showed angiogenic potential in the ex vivo chicken embryo model. Following transduction in MEF2C overexpressing cardiomyoblast cells, this purified recombinant protein synergistically activated the α-MHC promoter and induced GFP expression in the α-MHC-eGFP reporter assay. Prospectively, the purified bioactive recombinant HAND2 protein can potentially be a safe and effective molecular tool in the direct cardiac reprogramming process and other biological applications.
Collapse
Affiliation(s)
- Krishna Kumar Haridhasapavalan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Pradeep Kumar Sundaravadivelu
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Neha Joshi
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Nayan Jyoti Das
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Anshuman Mohapatra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Udayashree Voorkara
- Department of Obstetrics and Gynaecology, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India
| | - Vishwas Kaveeshwar
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India.
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
10
|
Prazak L, Iwasaki Y, Kim AR, Kozlov K, King K, Gergen JP. A dual role for DNA binding by Runt in activation and repression of sloppy paired transcription. Mol Biol Cell 2021; 32:ar26. [PMID: 34432496 PMCID: PMC8693977 DOI: 10.1091/mbc.e20-08-0509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
This work investigates the role of DNA binding by Runt in regulating the sloppy paired 1 (slp1) gene and in particular two distinct cis-regulatory elements that mediate regulation by Runt and other pair-rule transcription factors during Drosophila segmentation. We find that a DNA-binding-defective form of Runt is ineffective at repressing both the distal (DESE) and proximal (PESE) early stripe elements of slp1 and is also compromised for DESE-dependent activation. The function of Runt-binding sites in DESE is further investigated using site-specific transgenesis and quantitative imaging techniques. When DESE is tested as an autonomous enhancer, mutagenesis of the Runt sites results in a clear loss of Runt-dependent repression but has little to no effect on Runt-dependent activation. Notably, mutagenesis of these same sites in the context of a reporter gene construct that also contains the PESE enhancer results in a significant reduction of DESE-dependent activation as well as the loss of repression observed for the autonomous mutant DESE enhancer. These results provide strong evidence that DNA binding by Runt directly contributes to the regulatory interplay of interactions between these two enhancers in the early embryo.
Collapse
Affiliation(s)
- Lisa Prazak
- Department of Biology, Farmingdale State College, Farmingdale, NY 11735-1021.,Department of Biochemistry and Cell Biology and Center for Developmental Genetics.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Yasuno Iwasaki
- Department of Biochemistry and Cell Biology and Center for Developmental Genetics
| | - Ah-Ram Kim
- Graduate Program in Biochemistry and Structural Biology, and
| | - Konstantin Kozlov
- Department of Applied Mathematics, St. Petersburg State Polytechnical University, St. Petersburg, Russia 195251
| | - Kevin King
- Department of Biochemistry and Cell Biology and Center for Developmental Genetics.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - J Peter Gergen
- Department of Biochemistry and Cell Biology and Center for Developmental Genetics
| |
Collapse
|
11
|
Šućurović S, Nikolić T, Brosens JJ, Mulac-Jeričević B. Analysis of heart and neural crest derivatives-expressed protein 2 (HAND2)-progesterone interactions in peri-implantation endometrium†. Biol Reprod 2021; 102:1111-1121. [PMID: 31982918 DOI: 10.1093/biolre/ioaa013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/21/2019] [Accepted: 01/23/2020] [Indexed: 12/31/2022] Open
Abstract
Implantation is restricted to a narrow window when the local endometrial microenvironment is supportive of the invading embryo. The ovarian steroid hormones estrogen (E) and progesterone (P) are principal regulators of uterine receptivity. Suppression of E-dependent proliferation of luminal epithelium (LE) by P is mandatory for embryo implantation. Here, we report that the balance of E receptor α (ERα) and P receptors (PR) activity controls HAND2 expression, a key transcription factor that determines the fate of the implanting embryo and thereby pregnancy outcome. As a model, we used wild-type mice as well as mice in which either both PR isoforms or the A-isoform was genetically ablated (PRKO and PRAKO, respectively). Detailed spatiotemporal analyses of PR, HAND2, and ERα expression at implantation site demonstrated co-expression of HAND2 and PR but not ERα. Furthermore, in hormonally treated ovariectomized WT, PRAKO and PRKO mice, E suppresses endometrial HAND2 expression. Adding P together with E partially rescues HAND2 expression in WT, but not PRAKO and PRKO animals. Therefore, infertility in PRAKO mice is at least in part associated with the loss of PR-A-regulated HAND2 expression.
Collapse
Affiliation(s)
- Sandra Šućurović
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia and
| | - Tamara Nikolić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia and
| | - Jan J Brosens
- Division of Biomedical Sciences, Warwick Medical School, Coventry, United Kingdom
| | - Biserka Mulac-Jeričević
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia and
| |
Collapse
|
12
|
Correction to: Epigenetic and Transcriptional Networks Underlying Atrial Fibrillation. Circ Res 2020; 127:e143-e146. [DOI: 10.1161/res.0000000000000429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Hand2 Selectively Reorganizes Chromatin Accessibility to Induce Pacemaker-like Transcriptional Reprogramming. Cell Rep 2020; 27:2354-2369.e7. [PMID: 31116981 PMCID: PMC6657359 DOI: 10.1016/j.celrep.2019.04.077] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/25/2019] [Accepted: 04/17/2019] [Indexed: 01/01/2023] Open
Abstract
Gata4, Hand2, Mef2c, and Tbx5 (GHMT) can reprogram transduced fibroblasts into induced pacemaker-like myocytes (iPMs), but the underlying mechanisms remain obscure. Here, we explore the role of Hand2 in iPM formation by using a combination of transcriptome, genome, and biochemical as-says. We found many shared transcriptional signatures between iPMs and the endogenous sinoatrial node (SAN), yet key regulatory networks remain missing. We demonstrate that Hand2 augments chromatin accessibility at loci involved in sarcomere organization, electrical coupling, and membrane depolarization. Focusing on an established cardiac Hand2 cistrome, we observe selective reorganization of chromatin accessibility to promote pacemaker-specific gene expression. Moreover, we identify a Hand2 cardiac subtype diversity (CSD) domain through biochemical analysis of the N terminus. By integrating our RNA-seq and ATAC-seq datasets, we highlight desmosome organization as a hallmark feature of iPM formation. Collectively, our results illuminate Hand2-dependent mechanisms that may guide future efforts to rationally improve iPM formation. Gata4, Hand2, Mef2c, and Tbx5 can reprogram fibroblasts into cardiomyocyte-like cells, including induced pacemakers (iPMs). Fernandez-Perez et al. show that Hand2 coordinates this process by influencing chromatin accessibility and gene expression in fibroblasts undergoing iPM lineage conversion. These insights could eventually inform the production of superior replacement cells.
Collapse
|
14
|
van Ouwerkerk AF, Hall AW, Kadow ZA, Lazarevic S, Reyat JS, Tucker NR, Nadadur RD, Bosada FM, Bianchi V, Ellinor PT, Fabritz L, Martin J, de Laat W, Kirchhof P, Moskowitz I, Christoffels VM. Epigenetic and Transcriptional Networks Underlying Atrial Fibrillation. Circ Res 2020; 127:34-50. [PMID: 32717170 PMCID: PMC8315291 DOI: 10.1161/circresaha.120.316574] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies have uncovered over a 100 genetic loci associated with atrial fibrillation (AF), the most common arrhythmia. Many of the top AF-associated loci harbor key cardiac transcription factors, including PITX2, TBX5, PRRX1, and ZFHX3. Moreover, the vast majority of the AF-associated variants lie within noncoding regions of the genome where causal variants affect gene expression by altering the activity of transcription factors and the epigenetic state of chromatin. In this review, we discuss a transcriptional regulatory network model for AF defined by effector genes in Genome-wide association studies loci. We describe the current state of the field regarding the identification and function of AF-relevant gene regulatory networks, including variant regulatory elements, dose-sensitive transcription factor functionality, target genes, and epigenetic states. We illustrate how altered transcriptional networks may impact cardiomyocyte function and ionic currents that impact AF risk. Last, we identify the need for improved tools to identify and functionally test transcriptional components to define the links between genetic variation, epigenetic gene regulation, and atrial function.
Collapse
Affiliation(s)
- Antoinette F. van Ouwerkerk
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Amelia W. Hall
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zachary A. Kadow
- Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sonja Lazarevic
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Jasmeet S. Reyat
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nathan R. Tucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Masonic Medical Research Institute, Utica, NY, USA
| | - Rangarajan D. Nadadur
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Fernanda M. Bosada
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Valerio Bianchi
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- SWBH and UHB NHS Trusts, Birmingham, UK
| | - Jim Martin
- Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
- Texas Heart Institute, Houston, Texas, 77030
| | - Wouter de Laat
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- SWBH and UHB NHS Trusts, Birmingham, UK
- University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Ivan Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Vincent M. Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
15
|
Progestin-induced heart and neural crest derivatives-expressed transcript 2 inhibits angiopoietin 2 via fibroblast growth factor 9 in human endometrial stromal cells. Reprod Biol 2019; 19:14-21. [DOI: 10.1016/j.repbio.2019.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/14/2019] [Accepted: 02/16/2019] [Indexed: 01/26/2023]
|
16
|
Riedlinger T, Liefke R, Meier-Soelch J, Jurida L, Nist A, Stiewe T, Kracht M, Schmitz ML. NF-κB p65 dimerization and DNA-binding is important for inflammatory gene expression. FASEB J 2018; 33:4188-4202. [PMID: 30526044 PMCID: PMC6404571 DOI: 10.1096/fj.201801638r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Increasing evidence shows that many transcription factors execute important biologic functions independent from their DNA-binding capacity. The NF-κB p65 (RELA) subunit is a central regulator of innate immunity. Here, we investigated the relative functional contribution of p65 DNA-binding and dimerization in p65-deficient human and murine cells reconstituted with single amino acid mutants preventing either DNA-binding (p65 E/I) or dimerization (p65 FL/DD). DNA-binding of p65 was required for RelB-dependent stabilization of the NF-κB p100 protein. The antiapoptotic function of p65 and expression of the majority of TNF-α–induced genes were dependent on p65’s ability to bind DNA and to dimerize. Chromatin immunoprecipitation with massively parallel DNA sequencing experiments revealed that impaired DNA-binding and dimerization strongly diminish the chromatin association of p65. However, there were also p65-independent TNF-α–inducible genes and a subgroup of p65 binding sites still allowed some residual chromatin association of the mutants. These sites were enriched in activator protein 1 (AP-1) binding motifs and showed increased chromatin accessibility and basal transcription. This suggests a mechanism of assisted p65 chromatin association that can be in part facilitated by chromatin priming and cooperativity with other transcription factors such as AP-1.—Riedlinger, T., Liefke, R., Meier-Soelch, J., Jurida, L., Nist, A., Stiewe, T., Kracht, M., Schmitz, M. L. NF-κB p65 dimerization and DNA-binding is important for inflammatory gene expression.
Collapse
Affiliation(s)
- Tabea Riedlinger
- Institute of Biochemistry, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Robert Liefke
- Institute of Biochemistry, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany.,Institute of Molecular Biology and Tumor Research (IMT), Philipps University Marburg, Marburg, Germany
| | - Johanna Meier-Soelch
- Rudolf-Buchheim-Institute of Pharmacology, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany; and
| | - Liane Jurida
- Rudolf-Buchheim-Institute of Pharmacology, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany; and
| | - Andrea Nist
- Genomics Core Facility-Institute of Molecular Oncology, Philipps University Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility-Institute of Molecular Oncology, Philipps University Marburg, Marburg, Germany
| | - Michael Kracht
- Rudolf-Buchheim-Institute of Pharmacology, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany; and
| | - M Lienhard Schmitz
- Institute of Biochemistry, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
17
|
George RM, Firulli AB. Hand Factors in Cardiac Development. Anat Rec (Hoboken) 2018; 302:101-107. [PMID: 30288953 DOI: 10.1002/ar.23910] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/01/2018] [Accepted: 02/15/2018] [Indexed: 12/23/2022]
Abstract
Congenital heart defects account for 1% of infant mortality and 10% of in utero deaths. As the vertebrate embryo develops, multiple tissue types develop in tandem to morphologically pattern the functional heart. Underlying cardiac development is a network of transcription factors known to tightly control these morphological events. Members of the Twist family of basic helix-loop-helix transcription factors, Hand1 and Hand2, are essential to this process. The expression patterns and functional role of Hand factors in neural crest cells, endocardium, myocardium, and epicardium is indicative of their importance during cardiogenesis; however, to date, an extensive understanding of the transcriptional targets of Hand proteins and their overall mechanism of action remain unclear. In this review, we summarize the recent findings that further outline the crucial functions of Hand factors during heart development and in post-natal heart function. Anat Rec, 302:101-107, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rajani M George
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research Department of Pediatrics, Anatomy, Biochemistry, Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
18
|
Kato N, Iwase A, Ishida C, Nagai T, Mori M, Bayasula, Nakamura T, Osuka S, Ganiyeva U, Qin Y, Miki R, Kikkawa F. Upregulation of Fibroblast Growth Factors Caused by Heart and Neural Crest Derivatives Expressed 2 Suppression in Endometriotic Cells: A Possible Therapeutic Target in Endometriosis. Reprod Sci 2018; 26:979-987. [DOI: 10.1177/1933719118802053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several features exist that distinguish endometriotic cells from eutopic endometrial cells. Progesterone resistance is one of the main distinguishing features, although how progesterone resistance affects the phenotype of endometriotic cells is not fully elucidated. Heart and neural crest derivatives expressed 2 (HAND2) is a transcriptional factor that plays an important role in maintaining endometrial function in a progesterone-dependent manner. Therefore, we explored whether progesterone-dependent HAND2 is implicated in the progression of endometriosis. HAND2 was less expressed by endometriotic tissues compared to endometrial tissues. Suppression of HAND2 expression induced fibroblast growth factor 1 (FGF1), FGF2, and FGF9 in endometriotic stromal cells and consequently enhanced migration and invasion capacity. AZD4547, a FGF receptor inhibitor, diminished the migration and invasion of endometriotic cells in vitro. In the murine model of endometriosis, AZD4547 showed suppressive effects on the development of endometriotic lesions at a relatively low concentration. In conclusion, we demonstrated that FGF1, FGF2, and FGF9 are downstream effectors of HAND2 in endometriotic cells. Since HAND2-dependent FGFs play roles in enhancing invasive capacity of endometriotic cells, our results suggest that FGF receptor inhibitors, such as AZD4547, can be promising therapeutic targets for endometriosis.
Collapse
Affiliation(s)
- Nao Kato
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Chiharu Ishida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Nagai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Mori
- Aichi Cancer Center, Department of Gynecologic Oncology, Nagoya, Japan
| | - Bayasula
- Bell Research Center for Reproductive Health and Cancer, Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Maternal and Perinatal Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Umida Ganiyeva
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ying Qin
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rika Miki
- Bell Research Center for Reproductive Health and Cancer, Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
19
|
Jiang X. Silencing of heart and neural crest derivatives expressed transcript 2 attenuates transforming growth factor-β1-enhanced apoptosis of human bronchial epithelial cells. Oncol Lett 2018; 16:4997-5005. [PMID: 30250565 PMCID: PMC6144912 DOI: 10.3892/ol.2018.9299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/22/2018] [Indexed: 01/10/2023] Open
Abstract
Human bronchial epithelial (HBE) cells form the first protective barrier of the airway to protect patients from pulmonary diseases. The present study was performed to illustrate the mechanism underlying the effect of silencing heart and neural crest derivatives expressed transcript 2 (HAND2) on attenuating the transforming growth factor (TGF)-β1-enhanced apoptosis of HBE cells. TGF-β1 (10 µg/ml) was applied to HBE cells, and the HBE cells were transfected with small interfering RNA targeting HAND2 or were transfected with non-specific sequence. Subsequently, cell proliferation was measured using a Cell Counting kit-8 assay, whereas cell cycle and apoptosis status were measured using a flow cytometer. Reverse transcription-quantitative polymerase chain reaction and western blot analyses were performed to detect the expression levels of cell cycle- and apoptosis-related factors. Western blot analysis was also used to detect the phosphorylation levels of extracellular signal-regulated kinase (ERK), P38 and c-Jun-N-terminal kinase (JNK) of mitogen-activated protein kinase (MAPK) pathways. The results showed that TGF-β1 decreased HBE cell proliferation ability, arrested cell cycle at the G2 phase and promoted cell apoptosis with statistical significance. The expression levels of P21 and Cyclin D1 were inhibited, and those of caspase-3, caspase-8 and caspase-9 were promoted by TGF-β1. The phosphorylation levels of ERK, P38 and JNK were increased by TGF-β1. HAND2-silencing significantly alleviated the above functions of TGF-β1 on the HBE cells. In conclusion, the silencing of HAND2 attenuated the TGF-β1-stimulated apoptosis of HBE cells through regulating cell cycle, apoptosis-related factors and ERK/P38/JNK MAPK pathways. This may provide a novel treatment strategy for pulmonary disease, with HAND2 as the novel gene target.
Collapse
Affiliation(s)
- Xiaohui Jiang
- Intensive Care Unit, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
20
|
HAND2 Target Gene Regulatory Networks Control Atrioventricular Canal and Cardiac Valve Development. Cell Rep 2018; 19:1602-1613. [PMID: 28538179 DOI: 10.1016/j.celrep.2017.05.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 03/20/2017] [Accepted: 04/28/2017] [Indexed: 02/08/2023] Open
Abstract
The HAND2 transcriptional regulator controls cardiac development, and we uncover additional essential functions in the endothelial to mesenchymal transition (EMT) underlying cardiac cushion development in the atrioventricular canal (AVC). In Hand2-deficient mouse embryos, the EMT underlying AVC cardiac cushion formation is disrupted, and we combined ChIP-seq of embryonic hearts with transcriptome analysis of wild-type and mutants AVCs to identify the functionally relevant HAND2 target genes. The HAND2 target gene regulatory network (GRN) includes most genes with known functions in EMT processes and AVC cardiac cushion formation. One of these is Snai1, an EMT master regulator whose expression is lost from Hand2-deficient AVCs. Re-expression of Snai1 in mutant AVC explants partially restores this EMT and mesenchymal cell migration. Furthermore, the HAND2-interacting enhancers in the Snai1 genomic landscape are active in embryonic hearts and other Snai1-expressing tissues. These results show that HAND2 directly regulates the molecular cascades initiating AVC cardiac valve development.
Collapse
|
21
|
Perens EA, Garavito-Aguilar ZV, Guio-Vega GP, Peña KT, Schindler YL, Yelon D. Hand2 inhibits kidney specification while promoting vein formation within the posterior mesoderm. eLife 2016; 5:19941. [PMID: 27805568 PMCID: PMC5132343 DOI: 10.7554/elife.19941] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/01/2016] [Indexed: 12/29/2022] Open
Abstract
Proper organogenesis depends upon defining the precise dimensions of organ progenitor territories. Kidney progenitors originate within the intermediate mesoderm (IM), but the pathways that set the boundaries of the IM are poorly understood. Here, we show that the bHLH transcription factor Hand2 limits the size of the embryonic kidney by restricting IM dimensions. The IM is expanded in zebrafish hand2 mutants and is diminished when hand2 is overexpressed. Within the posterior mesoderm, hand2 is expressed laterally adjacent to the IM. Venous progenitors arise between these two territories, and hand2 promotes venous development while inhibiting IM formation at this interface. Furthermore, hand2 and the co-expressed zinc-finger transcription factor osr1 have functionally antagonistic influences on kidney development. Together, our data suggest that hand2 functions in opposition to osr1 to balance the formation of kidney and vein progenitors by regulating cell fate decisions at the lateral boundary of the IM. DOI:http://dx.doi.org/10.7554/eLife.19941.001 The human body is made up of many different types of cells, yet they are all descended from one single fertilized egg cell. The process by which cells specialize into different types is complex and has many stages. At each step of the process, the selection of cell types that a cell can eventually become is increasingly restricted. The entire system is controlled by switching different genes on and off in different groups of cells. Balancing the activity of these genes ensures that enough cells of each type are made in order to build a complete and healthy body. Upsetting this balance can result in organs that are too large, too small or even missing altogether. The cells that form the kidneys and bladder originate within a tissue called the intermediate mesoderm. Controlling the size of this tissue is an important part of building working kidneys. Perens et al. studied how genes control the size of the intermediate mesoderm of zebrafish embryos, which is very similar to the intermediate mesoderm of humans. The experiments revealed that a gene called hand2, which is switched on in cells next to the intermediate mesoderm, restricts the size of this tissue in order to determine the proper size of the kidney. Switching off the hand2 gene resulted in zebrafish with abnormally large kidneys. Loss of hand2 also led to the loss of a different type of cell that forms veins. These findings suggest that cells with an active hand2 gene are unable to become intermediate mesoderm cells and instead go on to become part of the veins. These experiments also demonstrated that a gene called osr1 works in opposition to hand2 to determine the right number of cells that are needed to build the kidneys. Further work will reveal how hand2 prevents cells from joining the intermediate mesoderm and how its role is balanced by the activity of osr1. Understanding how the kidneys form could eventually help to diagnose or treat several genetic diseases and may make it possible to grow replacement kidneys from unspecialized cells. DOI:http://dx.doi.org/10.7554/eLife.19941.002
Collapse
Affiliation(s)
- Elliot A Perens
- Division of Biological Sciences, University of California, San Diego, San Diego, United States.,Department of Pediatrics, School of Medicine, University of California, San Diego, San Diego, United States
| | - Zayra V Garavito-Aguilar
- Division of Biological Sciences, University of California, San Diego, San Diego, United States.,Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Gina P Guio-Vega
- Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Karen T Peña
- Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Yocheved L Schindler
- Division of Biological Sciences, University of California, San Diego, San Diego, United States
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, San Diego, United States
| |
Collapse
|
22
|
Cook JR, Matsumoto M, Banks AS, Kitamura T, Tsuchiya K, Accili D. A mutant allele encoding DNA binding-deficient FoxO1 differentially regulates hepatic glucose and lipid metabolism. Diabetes 2015; 64:1951-65. [PMID: 25576059 PMCID: PMC4439558 DOI: 10.2337/db14-1506] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/07/2015] [Indexed: 12/27/2022]
Abstract
Insulin signaling in the liver blunts glucose production and stimulates triglyceride biosynthesis. FoxO1 is required for cAMP induction of hepatic glucose production and is permissive for the effect of insulin to suppress this process. Moreover, FoxO1 ablation increases lipogenesis. In this study, we investigated the pleiotropic actions of FoxO1 on glucose and lipid metabolism. To this end, we reconstituted FoxO1 function in mice with a liver-specific deletion of Foxo1 using targeted knock-in of an allele encoding a DNA binding-deficient FoxO1 mutant (L-DBD). Chow-reared L-DBD mice showed defects in hepatic glucose production but normal liver triglyceride content despite increased rates of de novo lipogenesis and impaired fatty acid oxidation in isolated hepatocytes. Gene expression studies indicated that FoxO1 regulates the expression of glucokinase via a cell-nonautonomous coregulatory mechanism, while its regulation of glucose-6-phosphatase proceeds via a cell-autonomous action as a direct transcriptional activator. These conclusions support a differential regulation of hepatic glucose and lipid metabolism by FoxO1 based on the mechanism by which it alters the expression of key target genes involved in each process.
Collapse
Affiliation(s)
- Joshua R Cook
- Department of Medicine, Columbia University, New York, NY
| | - Michihiro Matsumoto
- Department of Medicine, Columbia University, New York, NY Department of Molecular Metabolic Regulation, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Alexander S Banks
- Department of Medicine, Columbia University, New York, NY Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Tadahiro Kitamura
- Department of Medicine, Columbia University, New York, NY Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Kyoichiro Tsuchiya
- Department of Medicine, Columbia University, New York, NY Department of Clinical and Molecular Endocrinology, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | | |
Collapse
|
23
|
Katsumura KR, Yang C, Boyer ME, Li L, Bresnick EH. Molecular basis of crosstalk between oncogenic Ras and the master regulator of hematopoiesis GATA-2. EMBO Rep 2014; 15:938-47. [PMID: 25056917 DOI: 10.15252/embr.201438808] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Disease mutations provide unique opportunities to decipher protein and cell function. Mutations in the master regulator of hematopoiesis GATA-2 underlie an immunodeficiency associated with myelodysplastic syndrome and leukemia. We discovered that a GATA-2 disease mutant (T354M) defective in chromatin binding was hyperphosphorylated by p38 mitogen-activated protein kinase. p38 also induced multisite phosphorylation of wild-type GATA-2, which required a single phosphorylated residue (S192). Phosphorylation of GATA-2, but not T354M, stimulated target gene expression. While crosstalk between oncogenic Ras and GATA-2 has been implicated as an important axis in cancer biology, its mechanistic underpinnings are unclear. Oncogenic Ras enhanced S192-dependent GATA-2 phosphorylation, nuclear foci localization, and transcriptional activation. These studies define a mechanism that controls a key regulator of hematopoiesis and a dual mode of impairing GATA-2-dependent genetic networks: mutational disruption of chromatin occupancy yielding insufficient GATA-2, and oncogenic Ras-mediated amplification of GATA-2 activity.
Collapse
Affiliation(s)
- Koichi R Katsumura
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Carbone Cancer Center, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Chenxi Yang
- Department of Chemistry, University of Wisconsin, Madison, WI, USA
| | - Meghan E Boyer
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Carbone Cancer Center, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin, Madison, WI, USA University of Wisconsin School of Pharmacy, Madison, WI, USA
| | - Emery H Bresnick
- UW-Madison Blood Research Program, Department of Cell and Regenerative Biology, Carbone Cancer Center, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
24
|
Schindler YL, Garske KM, Wang J, Firulli BA, Firulli AB, Poss KD, Yelon D. Hand2 elevates cardiomyocyte production during zebrafish heart development and regeneration. Development 2014; 141:3112-22. [PMID: 25038045 DOI: 10.1242/dev.106336] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Embryonic heart formation requires the production of an appropriate number of cardiomyocytes; likewise, cardiac regeneration following injury relies upon the recovery of lost cardiomyocytes. The basic helix-loop-helix (bHLH) transcription factor Hand2 has been implicated in promoting cardiomyocyte formation. It is unclear, however, whether Hand2 plays an instructive or permissive role during this process. Here, we find that overexpression of hand2 in the early zebrafish embryo is able to enhance cardiomyocyte production, resulting in an enlarged heart with a striking increase in the size of the outflow tract. Our evidence indicates that these increases are dependent on the interactions of Hand2 in multimeric complexes and are independent of direct DNA binding by Hand2. Proliferation assays reveal that hand2 can impact cardiomyocyte production by promoting division of late-differentiating cardiac progenitors within the second heart field. Additionally, our data suggest that hand2 can influence cardiomyocyte production by altering the patterning of the anterior lateral plate mesoderm, potentially favoring formation of the first heart field at the expense of hematopoietic and vascular lineages. The potency of hand2 during embryonic cardiogenesis suggested that hand2 could also impact cardiac regeneration in adult zebrafish; indeed, we find that overexpression of hand2 can augment the regenerative proliferation of cardiomyocytes in response to injury. Together, our studies demonstrate that hand2 can drive cardiomyocyte production in multiple contexts and through multiple mechanisms. These results contribute to our understanding of the potential origins of congenital heart disease and inform future strategies in regenerative medicine.
Collapse
Affiliation(s)
- Yocheved L Schindler
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA Developmental Genetics Program and Department of Cell Biology, Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Kristina M Garske
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jinhu Wang
- Department of Cell Biology, Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Beth A Firulli
- Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Departments of Pediatrics and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anthony B Firulli
- Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Departments of Pediatrics and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kenneth D Poss
- Department of Cell Biology, Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA Developmental Genetics Program and Department of Cell Biology, Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
25
|
Anderson E, Hill RE. Long range regulation of the sonic hedgehog gene. Curr Opin Genet Dev 2014; 27:54-9. [PMID: 24859115 DOI: 10.1016/j.gde.2014.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/18/2014] [Accepted: 03/25/2014] [Indexed: 12/22/2022]
Abstract
The regulatory architecture that controls developmental genes is often a collection of enhancers that, in combination, generate a complex spatial and temporal pattern of expression. These enhancers populate domains operating at long distances and, in the case of the sonic hedgehog (Shh) locus, this regulatory domain covers ∼900-1000kb. Within this context each embryonic tissue that expresses Shh has acquired its own regulatory apparatus which may require the activity from several distinct enhancers. Expression of Shh in the developing limb bud is driven by a single enhancer that interprets a myriad of genetic information to initiate expression in the posterior margin of the limb bud, inhibits expression along the anterior margin, defines the level of expression, and sets the tissue boundary of expression.
Collapse
Affiliation(s)
- Eve Anderson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Robert E Hill
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
26
|
Shindoh H, Okada H, Tsuzuki T, Nishigaki A, Kanzaki H. Requirement of heart and neural crest derivatives-expressed transcript 2 during decidualization of human endometrial stromal cells in vitro. Fertil Steril 2014; 101:1781-90.e1-5. [PMID: 24745730 DOI: 10.1016/j.fertnstert.2014.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 03/04/2014] [Accepted: 03/11/2014] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate the role of heart and neural crest derivatives-expressed transcript 2 (HAND2) during decidualization of human endometrial stromal cells (ESCs). DESIGN In vitro experiment. SETTING Research laboratory. PATIENT(S) Twenty-six patients undergoing hysterectomy for benign reasons. INTERVENTION(S) ESCs were cultured for 12 days with HAND2 small interfering RNA (siRNA) or nonsilencing RNA during decidualization by medroxyprogesterone acetate (MPA) and E2. MAIN OUTCOME MEASURE(S) Decidualization was monitored by changes in cellular morphology and the expression of several decidual-specific genes. RESULT(S) HAND2 siRNA effectively suppressed HAND2 levels in ESCs after 12 days of E2 + MPA treatment. ESCs cultured with HAND2 siRNA retained a long fibroblast-like shape, whereas the cells cultured with control siRNA transformed into enlarged polygonal cells. Silencing of HAND2 expression significantly reduced connexin-43 involved in the morphologic changes. HAND2 silencing significantly reduced the mRNA levels of fibulin-1, prolactin, tissue inhibitor of metalloproteinase 3, interleukin-15, and forkhead box O1A (FOXO1A), but had no effect on the mRNA levels of dickkopf-1, serum glucocorticoid kinase 1, and insulin-like growth factor-binding protein 5. HAND2 siRNA effectively suppressed the levels of nuclear FOXO1A protein as a regulator of decidualization. CONCLUSION(S) These results suggest that HAND2 plays a key role in the regulation of progestin-induced decidualization of human ESCs.
Collapse
Affiliation(s)
- Hisayuu Shindoh
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Hidetaka Okada
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan.
| | - Tomoko Tsuzuki
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Akemi Nishigaki
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Hideharu Kanzaki
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| |
Collapse
|
27
|
Tamura M, Amano T, Shiroishi T. The Hand2 Gene Dosage Effect in Developmental Defects and Human Congenital Disorders. Curr Top Dev Biol 2014; 110:129-52. [DOI: 10.1016/b978-0-12-405943-6.00003-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
The research on screening differentially expressed genes in Hirschsprung's disease by using Microarray. J Pediatr Surg 2013; 48:2281-8. [PMID: 24210200 DOI: 10.1016/j.jpedsurg.2013.06.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 06/05/2013] [Accepted: 06/22/2013] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To study the differential expression of genes between Hirschsprung's disease (HSCR) and normal tissue by using microarray for exploring the mechanism of HSCR development and establishing the gene expression profiles of HSCR. METHODS Colon tissues (aganglionic and normal segments) of 4 patients with HSCR were detected by the Agilent SurePrint G3 Human GE 8x60K Microarrays. RT-PCR was used to verify the results of Microarray test. Then, immunohistochemistry was used to demonstrate the expression of HAND2 in the myenteric plexus of the colon from 46 patients with HSCR to further explore the relationship between HAND2 and development of HSCR. RESULTS A total of 12,125 meaningful expressed genes were screened out. 4 pairs of specimens had 622 differentially expressed genes, 584 (93.89%) of which were up-regulated while 38(6.11%) were down-regulated. 6 of the 622 genes were tested by RT-PCR, which were consistent with the results detected by Microarray. The average optical density of positive expression of HAND2 in myenteric plexus was compared between the aganglionic, transitional, dilated, normal segments and control group. The average optical density in the aganglionic segments was obviously reduced. Statistical analyzed data showed that it has significant deviation (P<0.01). CONCLUSION 1. A set of differentially expressed genes between aganglionic and normal segments of HSCR was obtained. Our data may provide significant information to research the pathogenesis of HSCR. 2. Reduced protein expression of HAND2 in the myenteric plexus of the aganglionic would suggest that HAND2 was involved in the pathogenesis of HSCR.
Collapse
|
29
|
Cho H, Okada H, Tsuzuki T, Nishigaki A, Yasuda K, Kanzaki H. Progestin-induced heart and neural crest derivatives expressed transcript 2 is associated with fibulin-1 expression in human endometrial stromal cells. Fertil Steril 2012; 99:248-255.e2. [PMID: 23036802 DOI: 10.1016/j.fertnstert.2012.08.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 08/28/2012] [Accepted: 08/28/2012] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate whether heart and neural crest derivatives expressed transcript 2 (HAND2) regulates fibulin-1 (FBLN1) expression during decidualization of human endometrial stromal cells (ESCs). DESIGN In vitro experiment. SETTING Research laboratory. PATIENT(S) Twenty-four patients undergoing hysterectomy for benign reasons. INTERVENTION(S) ESCs were cultured with various progestins (medroxyprogesterone acetate [MPA], norethisterone, levonorgestrel, dienogest, and P), E(2), dexamethasone, and/or 8-bromoadenosine 3', 5'-cyclic monophosphate (8-Br-cAMP). HAND2 and FBLN1 were silenced by small interfering RNA technology. MAIN OUTCOME MEASURE(S) HAND2 and FBLN1 expression levels were assessed by real-time polymerase chain reaction and Western blot analysis. RESULT(S) MPA or E(2) + MPA increased HAND2 mRNA levels in ESCs in a time- and dose-dependent manner, and this stimulatory effect was blocked by RU-486 (P receptor antagonist). HAND2 was increased by E(2) + MPA earlier than FBLN1. Simultaneous MPA and 8-Br-cAMP treatment synergistically enhanced HAND2 mRNA levels. P and all the progestins significantly increased HAND2 mRNA levels, whereas E(2), 8-Br-cAMP, or dexamethasone alone had no effect. Silencing of HAND2 expression significantly reduced FBLN1 expression, whereas FBLN1 silencing had no effect on HAND2 expression. CONCLUSION(S) These results suggest that progestin-induced HAND2 contributes to FBLN1 expression in human ESCs.
Collapse
Affiliation(s)
- Hisayuu Cho
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Hidetaka Okada
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan.
| | - Tomoko Tsuzuki
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Akemi Nishigaki
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Katsuhiko Yasuda
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Hideharu Kanzaki
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| |
Collapse
|
30
|
Strehle EM, Yu L, Rosenfeld JA, Donkervoort S, Zhou Y, Chen TJ, Martinez JE, Fan YS, Barbouth D, Zhu H, Vaglio A, Smith R, Stevens CA, Curry CJ, Ladda RL, Fan ZJ, Fox JE, Martin JA, Abdel-Hamid HZ, McCracken EA, McGillivray BC, Masser-Frye D, Huang T. Genotype-phenotype analysis of 4q deletion syndrome: proposal of a critical region. Am J Med Genet A 2012; 158A:2139-51. [PMID: 22847869 DOI: 10.1002/ajmg.a.35502] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 05/07/2012] [Indexed: 12/12/2022]
Abstract
Chromosome 4q deletion syndrome (4q- syndrome) is a rare condition, with an estimated incidence of 1 in 100,000. Although variable, the clinical spectrum commonly includes craniofacial, developmental, digital, skeletal, and cardiac involvement. Data on the genotype-phenotype correlation within the 4q arm are limited. We present detailed clinical and genetic information by array CGH on 20 patients with 4q deletions. We identified a patient who has a ∼465 kb deletion (186,770,069-187,234,800, hg18 coordinates) in 4q35.1 with all clinical features for 4q deletion syndrome except for developmental delay, suggesting that this is a critical region for this condition and a specific gene responsible for orofacial clefts and congenital heart defects resides in this region. Since the patients with terminal deletions all had cleft palate, our results provide further evidence that a gene associated with clefts is located on the terminal segment of 4q. By comparing and contrasting our patients' genetic information and clinical features, we found significant genotype-phenotype correlations at a single gene level linking specific phenotypes to individual genes. Based on these data, we constructed a hypothetical partial phenotype-genotype map for chromosome 4q which includes BMP3, SEC31A, MAPK10, SPARCL1, DMP1, IBSP, PKD2, GRID2, PITX2, NEUROG2, ANK2, FGF2, HAND2, and DUX4 genes.
Collapse
|
31
|
Rabinowitz AH, Vokes SA. Integration of the transcriptional networks regulating limb morphogenesis. Dev Biol 2012; 368:165-80. [PMID: 22683377 DOI: 10.1016/j.ydbio.2012.05.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/29/2012] [Accepted: 05/29/2012] [Indexed: 12/29/2022]
Abstract
The developing limb is one of the best described vertebrate systems for understanding how coordinated gene expression during embryogenesis leads to the structures present in the mature organism. This knowledge, derived from decades of research, is largely based upon gain- and loss-of-function experiments. These studies have provided limited information about how the key signaling pathways interact with each other and the downstream effectors of these pathways. We summarize our current understanding of known genetic interactions in the context of three temporally defined gene regulatory networks. These networks crystallize our current knowledge, depicting a dynamic process involving multiple feedback loops between the ectoderm and mesoderm. At the same time, they highlight the fact that many essential processes are still largely undescribed. Much of the dynamic transcriptional activity occurring during development is regulated by distal cis-regulatory elements. Modern genomic tools have provided new approaches for studying the function of cis-regulatory elements and we discuss the results of these studies in regard to understanding limb development. Ultimately, these genomic techniques will allow scientists to understand how multiple signaling pathways are integrated in space and time to drive gene expression and regulate the formation of the limb.
Collapse
Affiliation(s)
- Adam H Rabinowitz
- Section of Molecular Cell & Developmental Biology, Institute for Cellular and Molecular Biology, One University Station A4800, Austin, TX 78712, USA
| | | |
Collapse
|
32
|
Bieganski T, Jamsheer A, Sowinska A, Baranska D, Niedzielski K, Kozlowski K, Czarny-Ratajczak M. Three new patients with FATCO: Fibular agenesis with ectrodactyly. Am J Med Genet A 2012; 158A:1542-50. [DOI: 10.1002/ajmg.a.35369] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Accepted: 02/05/2012] [Indexed: 11/08/2022]
|
33
|
Li Z, Fan J, Zhao W, Jin L, Ma L. The specific binding of peptide ligands to cardiomyocytes derived from mouse embryonic stem cells. J Pept Sci 2011; 17:771-82. [DOI: 10.1002/psc.1401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 07/07/2011] [Accepted: 07/07/2011] [Indexed: 02/05/2023]
Affiliation(s)
- Zhuokun Li
- Department of Biological sciences and Biotechnology; Tsinghua University; Beijing China
- Life Science Division, Graduate School at Shenzhen; Tsinghua University; Shenzhen China
| | - Jiusong Fan
- Department of Biological sciences and Biotechnology; Tsinghua University; Beijing China
- Life Science Division, Graduate School at Shenzhen; Tsinghua University; Shenzhen China
| | - Wenxiu Zhao
- Life Science Division, Graduate School at Shenzhen; Tsinghua University; Shenzhen China
| | - Lei Jin
- Department of Biological sciences and Biotechnology; Tsinghua University; Beijing China
- Life Science Division, Graduate School at Shenzhen; Tsinghua University; Shenzhen China
| | - Lan Ma
- Life Science Division, Graduate School at Shenzhen; Tsinghua University; Shenzhen China
| |
Collapse
|
34
|
Vincentz JW, Barnes RM, Firulli AB. Hand factors as regulators of cardiac morphogenesis and implications for congenital heart defects. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2011; 91:485-94. [PMID: 21462297 PMCID: PMC3119928 DOI: 10.1002/bdra.20796] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/06/2011] [Accepted: 02/02/2011] [Indexed: 11/08/2022]
Abstract
Almost 15 years of careful study have established the related basic Helix-Loop-Helix (bHLH) transcription factors Hand1 and Hand2 as critical for heart development across evolution. Hand factors make broad contributions, revealed through animal models, to the development of multiple cellular lineages that ultimately contribute to the heart. They perform critical roles in ventricular cardiomyocyte growth, differentiation, morphogenesis, and conduction. They are also important for the proper development of the cardiac outflow tract, epicardium, and endocardium. Molecularly, they function both through DNA binding and through protein-protein interactions, which are regulated transcriptionally, posttranscriptionally by microRNAs, and posttranslationally through phosphoregulation. Although direct Hand factor transcriptional targets are progressively being identified, confirmed direct targets of Hand factor transcriptional activity in the heart are limited. Identification of these targets will be critical to model the mechanisms by which Hand factor bHLH interactions affect developmental pathways. Improved understanding of Hand factor-mediated transcriptional cascades will be necessary to determine how Hand factor dysregulation translates to human disease phenotypes. This review summarizes the insight that animal models have provided into the regulation and function of these factors during heart development, in addition to the recent findings that suggest roles for HAND1 and HAND2 in human congenital heart disease.
Collapse
Affiliation(s)
- Joshua W. Vincentz
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Ralston M. Barnes
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Anthony B. Firulli
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| |
Collapse
|
35
|
Lu S, Nie J, Luan Q, Feng Q, Xiao Q, Chang Z, Shan C, Hess D, Hemmings BA, Yang Z. Phosphorylation of the Twist1-family basic helix-loop-helix transcription factors is involved in pathological cardiac remodeling. PLoS One 2011; 6:e19251. [PMID: 21559426 PMCID: PMC3084786 DOI: 10.1371/journal.pone.0019251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 03/24/2011] [Indexed: 01/12/2023] Open
Abstract
Background The Twist1-family basic helix-loop-helix (bHLH) transcription factors including Twist1, Hand1 and Hand2, play an essential role in heart development and are implicated in pathological heart remodeling. Previously, it was reported that these bHLH transcription factors can be regulated by phosphorylation within the basic-helix I domain, which is involved in developmental processes such as limb formation and trophoblast differentiation. However, how phosphorylation of Twist1 family functions in post-natal heart is elusive. Principal Findings Here, we generated transgenic mice with over-expression of Hand1 and Twist1 mutants (to mimic or to abolish phosphorylation) in cardiomyocytes and found pathological cardiac remodeling leading to heart failure and sudden death. Gene expression profile analysis revealed up-regulation of growth-promoting genes and down-regulation of metabolic genes. It is well known that aberrant activation of Akt signaling causes pathological cardiac remodeling and results in heart failure. The basic-helix I domain of Twist1 family members contain Akt substrate consensus motif and may be downstream targets of Akt signaling. Using biochemical analysis, we demonstrated that Hand1 and Twist1 were phosphorylated by Akt in the basic-helix I domain. Phosphorylation of Hand1 regulated its transcriptional activation of luciferase reporter genes and DNA binding ability. Conclusions This study provides novel insights into the regulation of Twist1 family in cardiac remodeling and suggests that the Twist1 family can be regulated by Akt signaling.
Collapse
Affiliation(s)
- Shuangshuang Lu
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Junwei Nie
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Qing Luan
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Qiuting Feng
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Qi Xiao
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Zai Chang
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Congjia Shan
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Daniel Hess
- The Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Brian A. Hemmings
- The Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Zhongzhou Yang
- The Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
- * E-mail:
| |
Collapse
|
36
|
|
37
|
Tsuchihashi T, Maeda J, Shin CH, Ivey KN, Black BL, Olson EN, Yamagishi H, Srivastava D. Hand2 function in second heart field progenitors is essential for cardiogenesis. Dev Biol 2010; 351:62-9. [PMID: 21185281 DOI: 10.1016/j.ydbio.2010.12.023] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 12/06/2010] [Accepted: 12/15/2010] [Indexed: 10/18/2022]
Abstract
Cardiogenesis involves the contributions of multiple progenitor pools, including mesoderm-derived cardiac progenitors known as the first and second heart fields. Disruption of genetic pathways regulating individual subsets of cardiac progenitors likely underlies many forms of human cardiac malformations. Hand2 is a member of the basic helix loop helix (bHLH) family of transcription factors and is expressed in numerous cell lineages that contribute to the developing heart. However, the early embryonic lethality of Hand2-null mice has precluded lineage-specific study of its function in myocardial progenitors. Here, we generated and used a floxed allele of Hand2 to ablate its expression in specific cardiac cell populations at defined developmental points. We found that Hand2 expression within the mesoderm-derived second heart field progenitors was required for their survival and deletion in this domain recapitulated the complete Hand2-null phenotype. Loss of Hand2 at later stages of development and in restricted domains of the second heart field revealed a spectrum of cardiac anomalies resembling forms of human congenital heart disease. Molecular analyses of Hand2 mutant cells revealed several genes by which Hand2 may influence expansion of the cardiac progenitors. These findings demonstrate that Hand2 is essential for survival of second heart field progenitors and that the graded loss of Hand2 function in this cardiac progenitor pool can cause a spectrum of congenital heart malformation.
Collapse
Affiliation(s)
- Takatoshi Tsuchihashi
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Galli A, Robay D, Osterwalder M, Bao X, Bénazet JD, Tariq M, Paro R, Mackem S, Zeller R. Distinct roles of Hand2 in initiating polarity and posterior Shh expression during the onset of mouse limb bud development. PLoS Genet 2010; 6:e1000901. [PMID: 20386744 PMCID: PMC2851570 DOI: 10.1371/journal.pgen.1000901] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 03/09/2010] [Indexed: 01/21/2023] Open
Abstract
The polarization of nascent embryonic fields and the endowment of cells with organizer properties are key to initiation of vertebrate organogenesis. One such event is antero-posterior (AP) polarization of early limb buds and activation of morphogenetic Sonic Hedgehog (SHH) signaling in the posterior mesenchyme, which in turn promotes outgrowth and specifies the pentadactylous autopod. Inactivation of the Hand2 transcriptional regulator from the onset of mouse forelimb bud development disrupts establishment of posterior identity and Shh expression, which results in a skeletal phenotype identical to Shh deficient limb buds. In wild-type limb buds, Hand2 is part of the protein complexes containing Hoxd13, another essential regulator of Shh activation in limb buds. Chromatin immunoprecipitation shows that Hand2-containing chromatin complexes are bound to the far upstream cis-regulatory region (ZRS), which is specifically required for Shh expression in the limb bud. Cell-biochemical studies indicate that Hand2 and Hoxd13 can efficiently transactivate gene expression via the ZRS, while the Gli3 repressor isoform interferes with this positive transcriptional regulation. Indeed, analysis of mouse forelimb buds lacking both Hand2 and Gli3 reveals the complete absence of antero-posterior (AP) polarity along the entire proximo-distal axis and extreme digit polydactyly without AP identities. Our study uncovers essential components of the transcriptional machinery and key interactions that set-up limb bud asymmetry upstream of establishing the SHH signaling limb bud organizer. During early limb bud development, posterior mesenchymal cells are selected to express Sonic Hedgehog (Shh), which controls antero-posterior (AP) limb axis formation (axis from thumb to little finger). We generated a conditional loss-of-function Hand2 allele to inactivate Hand2 specifically in mouse limb buds. This genetic analysis reveals the pivotal role of Hand2 in setting up limb bud asymmetry as initiation of posterior identity and establishment of the Shh expression domain are completely disrupted in Hand2 deficient limb buds. The resulting loss of the ulna and digits mirror the skeletal malformations observed in Shh-deficient limbs. We show that Hand2 is part of the chromatin complexes that are bound to the cis-regulatory region that controls Shh expression specifically in limb buds. In addition, we show that Hand2 is part of a protein complex containing Hoxd13, which also participates in limb bud mesenchymal activation of Shh expression. Indeed, Hand2 and Hoxd13 stimulate ZRS–mediated transactivation in cells, while the Gli3 repressor form (Gli3R) interferes with this up-regulation. Interestingly, limb buds lacking both Hand2 and Gli3 lack AP asymmetry and are severely polydactylous. Molecular analysis reveals some of the key interactions and hierarchies that govern establishment of AP limb asymmetries upstream of SHH.
Collapse
Affiliation(s)
- Antonella Galli
- Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
- * E-mail: (AG); (RZ)
| | - Dimitri Robay
- Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marco Osterwalder
- Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Xiaozhong Bao
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jean-Denis Bénazet
- Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Muhammad Tariq
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Renato Paro
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Faculty of Sciences, University of Basel, Basel, Switzerland
| | - Susan Mackem
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Rolf Zeller
- Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
- * E-mail: (AG); (RZ)
| |
Collapse
|
39
|
Maves L, Tyler A, Moens CB, Tapscott SJ. Pbx acts with Hand2 in early myocardial differentiation. Dev Biol 2009; 333:409-18. [PMID: 19607825 DOI: 10.1016/j.ydbio.2009.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 06/09/2009] [Accepted: 07/06/2009] [Indexed: 12/30/2022]
Abstract
Transcription factors of the basic helix-loop-helix (bHLH) family are critical regulators of muscle cell differentiation. For example, Myod drives skeletal muscle differentiation, and Hand2 potentiates cardiac muscle differentiation. Understanding how these bHLH factors regulate distinct transcriptional targets in a temporally and spatially controlled manner is critical for understanding their activity in cellular differentiation. We previously showed that Pbx homeodomain proteins modulate the activity of Myod to promote the differentiation of fast-twitch skeletal muscle. Here, we test the hypothesis that Pbx proteins are also necessary for cardiac muscle differentiation through interacting with Hand2. We show that Pbx proteins are required for the activation of cardiac muscle differentiation in zebrafish embryos. Loss of Pbx activity leads to delay of myocardial differentiation and subsequent defective cardiac morphogenesis, similar to reduced Hand2 activity. Genetic interaction experiments support the hypothesis that Pbx proteins modulate the activity of Hand2 in myocardial differentiation. Furthermore, we show that Pbx proteins directly bind the promoter of the myocardial differentiation gene myl7 in vitro, supporting a direct role for Pbx proteins in promoting cardiac muscle differentiation. Our findings demonstrate new roles for Pbx proteins in vertebrate cardiac development and also provide new insight into connections between the transcriptional regulation of skeletal and cardiac muscle differentiation programs.
Collapse
Affiliation(s)
- Lisa Maves
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | |
Collapse
|