1
|
Siddiqui HB, Golcez T, Çelik M, Sevgin B, Çoban M, Süder İ, Kaya Ö, Özören N, Pekkan K. Modulation of mechanosensitive genes during embryonic aortic arch development. Dev Dyn 2024. [PMID: 39096177 DOI: 10.1002/dvdy.728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/30/2024] [Accepted: 07/17/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Early embryonic aortic arches (AA) are a dynamic vascular structures that are in the process of shaping into the great arteries of cardiovascular system. Previously, a time-lapsed mechanosensitive gene expression map was established for AA subject to altered mechanical loads in the avian embryo. To validate this map, we investigated effects on vascular microstructure and material properties following the perturbation of key genes using an in-house microvascular gene knockdown system. RESULTS All siRNA vectors show a decrease in the expression intensity of desired genes with no significant differences between vectors. In TGFβ3 knockdowns, we found a reduction in expression intensities of TGFβ3 (≤76%) and its downstream targets such as ELN (≤99.6%), Fbn1 (≤60%), COL1 (≤52%) and COL3 (≤86%) and an increase of diameter in the left AA (23%). MMP2 knockdown also reduced expression levels in MMP2 (≤30%) and a 6-fold increase in its downstream target COL3 with a decrease in stiffness of the AA wall and an increase in the diameter of the AA (55%). These in vivo measurements were confirmed using immunohistochemistry, western blotting and a computational growth model of the vascular extracellular matrix (ECM). CONCLUSIONS Localized spatial genetic modification of the aortic arch region governs the vascular phenotype and ECM composition of the embryo and can be integrated with mechanically-induced congenital heart disease models.
Collapse
Affiliation(s)
| | - Tansu Golcez
- Biomedical Sciences and Engineering Program, Koç University, Istanbul, Turkey
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Merve Çelik
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Börteçine Sevgin
- Biomedical Sciences and Engineering Program, Koç University, Istanbul, Turkey
| | - Mervenur Çoban
- Biomedical Sciences and Engineering Program, Koç University, Istanbul, Turkey
| | - İlke Süder
- Department of Molecular Biology and Genetics, Bogazici University, Turkey
| | - Özen Kaya
- Department of Molecular Biology and Genetics, Bogazici University, Turkey
- Department of Mechanical Engineering, Koç University, Istanbul, Turkey
| | - Nesrin Özören
- Department of Molecular Biology and Genetics, Bogazici University, Turkey
| | - Kerem Pekkan
- Biomedical Sciences and Engineering Program, Koç University, Istanbul, Turkey
- Department of Mechanical Engineering, Koç University, Istanbul, Turkey
| |
Collapse
|
2
|
Ren P, Chen M, Li J, Lin Z, Yang C, Yu C, Zhang D, Liu Y. MYH1F promotes the proliferation and differentiation of chicken skeletal muscle satellite cells into myotubes. Anim Biotechnol 2023; 34:3074-3084. [PMID: 36244007 DOI: 10.1080/10495398.2022.2132953] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
In diploid organisms, interactions between alleles determine phenotypic variation. In previous experiments, only MYH1F was found to show both ASE (spatiotemporal allele-specific expression) and TRD (allelic transmission ratio distortion) characteristics in the pectoral muscle by comparing the genome-wide allele lists of hybrid populations (F1) of meat- and egg- type chickens. In addition, MYH1F is a member of the MYH gene family, which plays an important role in skeletal muscle and non-muscle cells of animals, but the specific expression and function of this gene in chickens are still unknown. Therefore, qRT-PCR was used to detect the expression of MYH1F in different tissues of chicken. Proliferation and differentiation of chicken skeletal muscle satellite cells (SMSCs) have been detected by transfection of MYH1F-specific small interfering RNA (siRNA). The results showed that the expression of MYH1F in chicken skeletal muscle was higher than that in other tissues. Combined with CCK-8 assay, EdU assay, immunofluorescence, and Western blot Assay, it was found that MYH1F knockdown could significantly suppress the proliferation of chicken SMSCs and depress the differentiation and fusion of the cells. These results suggest that MYH1F plays a critical role in myogenesis in poultry, which is of great significance for exploring the regulatory mechanisms of muscle development and improving animal productivity.
Collapse
Affiliation(s)
- Peng Ren
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Meiying Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Jingjing Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhongzhen Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Chunlin Yu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Donghao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Yu Y, Quan J, Zou M, Zhao W, Su Y, Xu Y. Effects of ketamine-induced H3K9 hypoacetylation during pregnancy on cardiogenesis of mouse offspring. Birth Defects Res 2023; 115:770-781. [PMID: 36899481 DOI: 10.1002/bdr2.2168] [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: 11/08/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND Prenatal exposure to adverse factors can cause congenital heart defects. Ketamine, a widely used anesthetic drug, produces several adverse reactions such as tachycardia, hypertension, and laryngospasm, especially in pediatric patients. This study aimed to detect the effects of ketamine exposure during pregnancy on the cardiogenesis of mouse offspring and the potential mechanisms. METHODS In this study, ketamine at an addictive dose (5 mg/kg) was administered to mice during early gestation to explore the epigenetic mechanism of its causing cardiac dysplasia. The cardiac morphology of the mouse offspring was observed through hematoxylin-eosin staining and transmission electron microscopy. The heart function of one-month-old neonates was detected by echocardiography. The expression of cardiomyogenesis-related genes was detected by western blot and RT-qPCR. The acetylation level of histone H3K9 at the Mlc2 promoter and its deacetylase level and activity were detected by CHIP-qPCR, RT-qPCR, and ELISA, respectively. RESULTS Our data revealed that ketamine exposure during pregnancy could cause cardiac enlargement, myocardial sarcomere disorganization, and decreased cardiac contractile function in mouse offspring. Moreover, ketamine reduced the expression of Myh6, Myh7, Mlc2, Mef2c, and cTnI. The histone H3K9 acetylation level at the Mlc2 promoter was down-regulated by increasing the histone deacetylase activity and HDAC3 level upon ketamine administration. CONCLUSIONS Our work indicates that H3K9 acetylation is a vital player in cardiac dysplasia in offspring caused by prenatal ketamine exposure and HDAC3 is a key regulatory factor.
Collapse
Affiliation(s)
- Yujuan Yu
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Department of Anesthesiology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Sichuan, China
| | - Junjun Quan
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Mou Zou
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Wei Zhao
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing, China
| | - Yujuan Su
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ying Xu
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
4
|
Jamalpoor A, Hartvelt S, Dimopoulou M, Zwetsloot T, Brandsma I, Racz PI, Osterlund T, Hendriks G. A novel human stem cell-based biomarker assay for in vitro assessment of developmental toxicity. Birth Defects Res 2022; 114:1210-1228. [PMID: 35289129 DOI: 10.1002/bdr2.2001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Testing for developmental toxicity according to the current regulatory guidelines requires large numbers of animals, making these tests very resource intensive, time-consuming, and ethically debatable. Over the past decades, several alternative in vitro assays have been developed, but these often suffered from low predictability and the inability to provide a mechanistic understanding of developmental toxicity. METHODS To identify embryotoxic compounds, we developed a human induced pluripotent stem cells (hiPSCs)-based biomarker assay. The assay is based on the differentiation of hiPSCs into functional cardiomyocytes and hepatocytes. Proper stem cell differentiation is investigated by morphological profiling and assessment of time-dependent expression patterns of cell-specific biomarkers. In this system, a decrease in the expression of the biomarker genes and morphology disruption of the differentiated cells following compound treatment indicated teratogenicity. RESULTS The hiPSCs-based biomarker assay was validated with 21 well-established in vivo animal teratogenic and non-teratogenic compounds during cardiomyocyte and hepatocyte differentiation. The in vivo teratogenic compounds (e.g., thalidomide and valproic acid) markedly disrupted morphology, functionality, and the expression pattern of the biomarker genes in either one or both cell types. Non-teratogenic chemicals generally had no effect on the morphology of differentiated cells, nor on the expression of the biomarker genes. Compared to the in vivo classification, the assay achieved high accuracy (91%), sensitivity (91%), and specificity (90%). CONCLUSION The assay, which we named ReproTracker®, is a state-of-the-art in vitro method that can identify the teratogenicity potential of new pharmaceuticals and chemicals and signify the outcome of in vivo test systems.
Collapse
Affiliation(s)
- Amer Jamalpoor
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| | - Sabine Hartvelt
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| | - Myrto Dimopoulou
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| | - Tom Zwetsloot
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| | - Inger Brandsma
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| | - Peter I Racz
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| | - Torben Osterlund
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| | - Giel Hendriks
- Toxys B.V., Leiden Bio Science Park, Oegstgeest, The Netherlands
| |
Collapse
|
5
|
Ren P, Deng F, Chen S, Ran J, Li J, Yin L, Wang Y, Yin H, Zhu Q, Liu Y. Whole-genome resequencing reveals loci with allelic transmission ratio distortion in F 1 chicken population. Mol Genet Genomics 2021; 296:331-339. [PMID: 33404883 DOI: 10.1007/s00438-020-01744-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/30/2020] [Indexed: 11/28/2022]
Abstract
Allelic transmission ratio distortion (TRD) is the significant deviation from the expected ratio under Mendelian inheritance theory, which may be resulted from multiple disrupted biological processes, including germline selection, meiotic drive, gametic competition, imprint error, and embryo lethality. However, it is less known that whether or what extent the allelic TRD is present in farm animals. In this study, whole-genome resequencing technology was applied to reveal TRD loci in chicken by constructing a full-sib F1 hybrid population. Through the whole-genome resequencing data of two parents (30 ×) and 38 offspring (5 ×), we detected a total of 2850 TRD SNPs (p-adj < 0.05) located within 400 genes showing TRD, and all of them were unevenly distributed on macrochromosomes and microchromosomes. Our findings suggested that TRD in the chicken chromosome 16 might play an important role in chicken immunity and disease resistance and the MYH1F with significant TRD and allele-specific expression could play a key role in the fast muscle development. In addition, functional enrichment analyses revealed that many genes (e.g., TGFBR2, TGFBR3, NOTCH1, and NCOA1) with TRD were found in the significantly enriched biological process and InterPro terms in relation to embryonic lethality and germline selection. Our results suggested that TRD is considerably prevalent in the chicken genome and has functional implications.
Collapse
Affiliation(s)
- Peng Ren
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Feilong Deng
- Special Key Laboratory of Microbial Resources and Drug Development, Zunyi Medical University, Zunyi, 563000, China
| | - Shiyi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Jinshan Ran
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Jingjing Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Lingqian Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu, 611130, China. .,Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, 211 Huiming Road, Wenjiang, Sichuan, 611130, China.
| |
Collapse
|
6
|
The transcriptome of anterior regeneration in earthworm Eudrilus eugeniae. Mol Biol Rep 2020; 48:259-283. [PMID: 33306150 DOI: 10.1007/s11033-020-06044-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/28/2020] [Indexed: 12/25/2022]
Abstract
The oligochaete earthworm, Eudrilus eugeniae is capable of regenerating both anterior and posterior segments. The present study focuses on the transcriptome analysis of earthworm E. eugeniae to identify and functionally annotate the key genes supporting the anterior blastema formation and regulating the anterior regeneration of the worm. The Illumina sequencing generated a total of 91,593,182 raw reads which were assembled into 105,193 contigs using CLC genomics workbench. In total, 40,946 contigs were annotated against the NCBI nr and SwissProt database and among them, 15,702 contigs were assigned to 14,575 GO terms. Besides a total of 9389 contigs were mapped to 416 KEGG biological pathways. The RNA-Seq comparison study identified 10,868 differentially expressed genes (DEGs) and of them, 3986 genes were significantly upregulated in the anterior regenerated blastema tissue samples of the worm. The GO enrichment analysis showed angiogenesis and unfolded protein binding as the top enriched functions and the pathway enrichment analysis denoted TCA cycle as the most significantly enriched pathway associated with the upregulated gene dataset of the worm. The identified DEGs and their function and pathway information can be effectively utilized further to interpret the key cellular, genetic and molecular events associated with the regeneration of the worm.
Collapse
|
7
|
Maran S, Ee R, Faten SA, Sy Bing C, Khaw KY, Erin Lim SH, Lai KS, Wan Ibrahim WP, Mohd Zain MR, Chan KG, Gan SH, Tan HL. Mutations in the tail domain of MYH3 contributes to atrial septal defect. PLoS One 2020; 15:e0230982. [PMID: 32315303 PMCID: PMC7173802 DOI: 10.1371/journal.pone.0230982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/12/2020] [Indexed: 11/18/2022] Open
Abstract
Atrial septal defect (ASD) is one of the most common congenital heart defects diagnosed in children. Sarcomeric genes has been attributed to ASD and knockdown of MYH3 functionally homologues gene in chick models indicated abnormal atrial septal development. Here, we report for the first time, a case-control study investigating the role of MYH3 among non-syndromic ASD patients in contributing to septal development. Four amplicons which will amplifies the 40 kb MYH3 were designed and amplified using long range-PCR. The amplicons were then sequenced using indexed paired-end libraries on the MiSeq platform. The STREGA guidelines were applied for planning and reporting. The non-synonymous c. 3574G>A (p.Ala1192Thr) [p = 0.001, OR = 2.30 (1.36-3.87)] located within the tail domain indicated a highly conserved protein region. The mutant model of c. 3574G>A (p.Ala1192Thr) showed high root mean square deviation (RMSD) values compared to the wild model. To our knowledge, this is the first study to provide compelling evidence on the pathogenesis of MYH3 variants towards ASD hence, suggesting the crucial role of non-synonymous variants in the tail domain of MYH3 towards atrial septal development. It is hoped that this gene can be used as panel for diagnosis of ASD in future.
Collapse
Affiliation(s)
- Sathiya Maran
- School of Pharmacy, Monash University, Sunway, Selangor, Malaysia
- Human Genome Centre, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- * E-mail:
| | - Robson Ee
- Division of Genetics and Molecular Biology, Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Siti Aisyah Faten
- Human Genome Centre, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Choi Sy Bing
- School of Data Sciences, Perdana University, Selangor, Malaysia
| | - Kooi Yeong Khaw
- School of Pharmacy, Monash University, Sunway, Selangor, Malaysia
| | - Swee-Hua Erin Lim
- Health Sciences Division, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi, United Arab Emirates
| | - Kok-Song Lai
- Health Sciences Division, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi, United Arab Emirates
| | - Wan Pauzi Wan Ibrahim
- Faculty of Medicine and Health Sciences, Universiti Sultan Zainal Abidin, Kuala Terengganu, Terengganu, Malaysia
| | - Mohd Rizal Mohd Zain
- Department of Paediatrics, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Kok Gan Chan
- Division of Genetics and Molecular Biology, Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
- International Genome Centre, Jiangsu University, Zhenjiang, China
| | - Siew Hua Gan
- School of Pharmacy, Monash University, Sunway, Selangor, Malaysia
| | - Huay Lin Tan
- Human Genome Centre, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
8
|
Agarwal M, Sharma A, Kumar P, Kumar A, Bharadwaj A, Saini M, Kardon G, Mathew SJ. Myosin heavy chain-embryonic regulates skeletal muscle differentiation during mammalian development. Development 2020; 147:dev184507. [PMID: 32094117 PMCID: PMC7157585 DOI: 10.1242/dev.184507] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/12/2020] [Indexed: 12/12/2022]
Abstract
Myosin heavy chain-embryonic (MyHC-emb) is a skeletal muscle-specific contractile protein expressed during muscle development. Mutations in MYH3, the gene encoding MyHC-emb, lead to Freeman-Sheldon and Sheldon-Hall congenital contracture syndromes. Here, we characterize the role of MyHC-emb during mammalian development using targeted mouse alleles. Germline loss of MyHC-emb leads to neonatal and postnatal alterations in muscle fiber size, fiber number, fiber type and misregulation of genes involved in muscle differentiation. Deletion of Myh3 during embryonic myogenesis leads to the depletion of the myogenic progenitor cell pool and an increase in the myoblast pool, whereas fetal myogenesis-specific deletion of Myh3 causes the depletion of both myogenic progenitor and myoblast pools. We reveal that the non-cell-autonomous effect of MyHC-emb on myogenic progenitors and myoblasts is mediated by the fibroblast growth factor (FGF) signaling pathway, and exogenous FGF rescues the myogenic differentiation defects upon loss of MyHC-emb function in vitro Adult Myh3 null mice exhibit scoliosis, a characteristic phenotype exhibited by individuals with Freeman-Sheldon and Sheldon-Hall congenital contracture syndrome. Thus, we have identified MyHC-emb as a crucial myogenic regulator during development, performing dual cell-autonomous and non-cell-autonomous functions.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Megha Agarwal
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
- Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Akashi Sharma
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
- KIIT University, Patia, Bhubaneswar, 751024, Odisha, India
| | - Pankaj Kumar
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
- Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Amit Kumar
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
| | - Anushree Bharadwaj
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
| | - Masum Saini
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, 15 N 2030 E, Salt Lake City, UT 84112, USA
| | - Sam J Mathew
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
- Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- KIIT University, Patia, Bhubaneswar, 751024, Odisha, India
| |
Collapse
|
9
|
Li S, Wen H, Du S. Defective sarcomere organization and reduced larval locomotion and fish survival in slow muscle heavy chain 1 (smyhc1) mutants. FASEB J 2020; 34:1378-1397. [PMID: 31914689 PMCID: PMC6956737 DOI: 10.1096/fj.201900935rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 11/11/2022]
Abstract
Zebrafish skeletal muscles are broadly divided into slow-twitch and fast-twitch muscle fibers. The slow fibers, which express a slow fiber-specific myosin heavy chain 1 (Smyhc1), are the first group of muscle fibers formed during myogenesis. To uncover Smyhc1 function in muscle growth, we generated three mutant alleles with reading frame shift mutations in the zebrafish smyhc1 gene using CRISPR. The mutants showed shortened sarcomeres with no thick filaments and M-lines in slow fibers of the mutant embryos. However, the formation of slow muscle precursors and expression of other slow muscle genes were not affected and fast muscles appeared normal. The smyhc1 mutant embryos and larvae showed reduced locomotion and food intake. The mutant larvae exhibited increased lethality of incomplete penetrance. Approximately 2/5 of the homozygous mutants were viable and grew into reproductive adults. These adult mutants displayed a typical pattern of slow and fast muscle fiber distribution, and regained normal slow muscle formation. Together, our studies indicate that Smyhc1 is essential for myogenesis in embryonic slow muscles, and loss of Smyhc1 results in defective sarcomere assembly, reduces larval motility and fish survival, but has no visible impact on muscle growth in juvenile and adult zebrafish that escape the larval lethality.
Collapse
Affiliation(s)
- Siping Li
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
- The Key Laboratory of Mariculture, Ministry of Education, Fishery College of Ocean University of China, Qingdao 266003, China
| | - Haishen Wen
- The Key Laboratory of Mariculture, Ministry of Education, Fishery College of Ocean University of China, Qingdao 266003, China
| | - Shaojun Du
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| |
Collapse
|
10
|
Xu B, Zhang M, Perlingeiro RCR, Shen W. Skeletal Muscle Constructs Engineered from Human Embryonic Stem Cell Derived Myogenic Progenitors Exhibit Enhanced Contractile Forces When Differentiated in a Medium Containing EGM‐2 Supplements. ACTA ACUST UNITED AC 2019; 3:e1900005. [DOI: 10.1002/adbi.201900005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 10/08/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Bin Xu
- Department of Biomedical Engineering University of Minnesota Minneapolis MN 55455 USA
| | - Mengen Zhang
- Department of Biomedical Engineering University of Minnesota Minneapolis MN 55455 USA
| | - Rita C. R. Perlingeiro
- Department of Medicine University of Minnesota Minneapolis MN 55455 USA
- Stem Cell Institute and Institute for Engineering in Medicine University of Minnesota Minneapolis Minnesota 55455 USA
| | - Wei Shen
- Department of Biomedical Engineering University of Minnesota Minneapolis MN 55455 USA
- Stem Cell Institute and Institute for Engineering in Medicine University of Minnesota Minneapolis Minnesota 55455 USA
| |
Collapse
|
11
|
England J, Granados-Riveron J, Polo-Parada L, Kuriakose D, Moore C, Brook JD, Rutland CS, Setchfield K, Gell C, Ghosh TK, Bu'Lock F, Thornborough C, Ehler E, Loughna S. Tropomyosin 1: Multiple roles in the developing heart and in the formation of congenital heart defects. J Mol Cell Cardiol 2017; 106:1-13. [PMID: 28359939 PMCID: PMC5441184 DOI: 10.1016/j.yjmcc.2017.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 12/03/2022]
Abstract
Tropomyosin 1 (TPM1) is an essential sarcomeric component, stabilising the thin filament and facilitating actin's interaction with myosin. A number of sarcomeric proteins, such as alpha myosin heavy chain, play crucial roles in cardiac development. Mutations in these genes have been linked to congenital heart defects (CHDs), occurring in approximately 1 in 145 live births. To date, TPM1 has not been associated with isolated CHDs. Analysis of 380 CHD cases revealed three novel mutations in the TPM1 gene; IVS1 + 2T > C, I130V, S229F and a polyadenylation signal site variant GATAAA/AATAAA. Analysis of IVS1 + 2T > C revealed aberrant pre-mRNA splicing. In addition, abnormal structural properties were found in hearts transfected with TPM1 carrying I130V and S229F mutations. Phenotypic analysis of TPM1 morpholino-treated embryos revealed roles for TPM1 in cardiac looping, atrial septation and ventricular trabeculae formation and increased apoptosis was seen within the heart. In addition, sarcomere assembly was affected and altered action potentials were exhibited. This study demonstrated that sarcomeric TPM1 plays vital roles in cardiogenesis and is a suitable candidate gene for screening individuals with isolated CHDs. Four mutations identified in the TPM1 gene; IVS1 + 2T > C, I130V, S229F and GATAAA/AATAAA. In vitro analysis of IVS1 + 2T > C revealed aberrant pre-mRNA splicing. I130V and S229F mutations caused abnormal structural properties in the sarcomere. Reduced TPM1 expression during early cardiogenesis causes aberrant gross morphology. Apoptosis, sarcomere assembly and cardiac conduction were also affected.
Collapse
Affiliation(s)
| | - Javier Granados-Riveron
- Laboratory of Genomics, Genetics and Bioinformatics, Hospital Infantil de México Federico Gómez, Mexico
| | - Luis Polo-Parada
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, USA
| | | | | | - J David Brook
- School of Life Sciences, University of Nottingham, UK
| | - Catrin S Rutland
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | | | | | | | - Frances Bu'Lock
- East Midlands Congenital Heart Centre, Glenfield Hospital, Leicester, UK
| | | | - Elisabeth Ehler
- Randall Division of Cell and Molecular Biophysics, The Cardiovascular Division, King's College London, UK
| | | |
Collapse
|
12
|
Simpson S, Rutland P, Rutland CS. Genomic Insights into Cardiomyopathies: A Comparative Cross-Species Review. Vet Sci 2017; 4:E19. [PMID: 29056678 PMCID: PMC5606618 DOI: 10.3390/vetsci4010019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/15/2017] [Accepted: 03/15/2017] [Indexed: 12/19/2022] Open
Abstract
In the global human population, the leading cause of non-communicable death is cardiovascular disease. It is predicted that by 2030, deaths attributable to cardiovascular disease will have risen to over 20 million per year. This review compares the cardiomyopathies in both human and non-human animals and identifies the genetic associations for each disorder in each species/taxonomic group. Despite differences between species, advances in human medicine can be gained by utilising animal models of cardiac disease; likewise, gains can be made in animal medicine from human genomic insights. Advances could include undertaking regular clinical checks in individuals susceptible to cardiomyopathy, genetic testing prior to breeding, and careful administration of breeding programmes (in non-human animals), further development of treatment regimes, and drugs and diagnostic techniques.
Collapse
Affiliation(s)
- Siobhan Simpson
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, The University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK.
| | - Paul Rutland
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.
| | - Catrin Sian Rutland
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, The University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK.
| |
Collapse
|
13
|
Inhibition of Gata4 and Tbx5 by Nicotine-Mediated DNA Methylation in Myocardial Differentiation. Stem Cell Reports 2017; 8:290-304. [PMID: 28111280 PMCID: PMC5312513 DOI: 10.1016/j.stemcr.2016.12.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 11/23/2022] Open
Abstract
Maternal nicotine exposure causes alteration of gene expression and cardiovascular programming. The discovery of nicotine-medicated regulation in cardiogenesis is of major importance for the study of cardiac defects. The present study investigated the effect of nicotine on cardiac gene expression and epigenetic regulation during myocardial differentiation. Persistent nicotine exposure selectively inhibited expression of two cardiac genes, Tbx5 and Gata4, by promoter DNA hypermethylation. The nicotine-induced suppression on cardiac differentiation was restored by general nicotinic acetylcholine receptor inhibition. Consistent results of Tbx5 and Gata4 gene suppression and cardiac function impairment with decreased left ventricular ejection fraction were obtained from in vivo studies in offspring. Our results present a direct repressive effect of nicotine on myocardial differentiation by regulating cardiac gene suppression via promoter DNA hypermethylation, contributing to the etiology of smoking-associated cardiac defects. Nicotine downregulates Tbx5 and Gata4 during in vitro and in vivo cardiogenesis Nicotine causes diminished cardiac differentiation and impaired cardiac function Nicotine causes Tbx5 and Gata4 gene suppression via promoter DNA hypermethylation nAChR antagonist restores nicotine-induced gene suppression and DNA methylation
Collapse
|
14
|
England J, Pang KL, Parnall M, Haig MI, Loughna S. Cardiac troponin T is necessary for normal development in the embryonic chick heart. J Anat 2016; 229:436-49. [PMID: 27194630 PMCID: PMC4974548 DOI: 10.1111/joa.12486] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2016] [Indexed: 12/30/2022] Open
Abstract
The heart is the first functioning organ to develop during embryogenesis. The formation of the heart is a tightly regulated and complex process, and alterations to its development can result in congenital heart defects. Mutations in sarcomeric proteins, such as alpha myosin heavy chain and cardiac alpha actin, have now been associated with congenital heart defects in humans, often with atrial septal defects. However, cardiac troponin T (cTNT encoded by gene TNNT2) has not. Using gene‐specific antisense oligonucleotides, we have investigated the role of cTNT in chick cardiogenesis. TNNT2 is expressed throughout heart development and in the postnatal heart. TNNT2‐morpholino treatment resulted in abnormal atrial septal growth and a reduction in the number of trabeculae in the developing primitive ventricular chamber. External analysis revealed the development of diverticula from the ventricular myocardial wall which showed no evidence of fibrosis and still retained a myocardial phenotype. Sarcomeric assembly appeared normal in these treated hearts. In humans, congenital ventricular diverticulum is a rare condition, which has not yet been genetically associated. However, abnormal haemodynamics is known to cause structural defects in the heart. Further, structural defects, including atrial septal defects and congenital diverticula, have previously been associated with conduction anomalies. Therefore, to provide mechanistic insights into the effect that cTNT knockdown has on the developing heart, quantitative PCR was performed to determine the expression of the shear stress responsive gene NOS3 and the conduction gene TBX3. Both genes were differentially expressed compared to controls. Therefore, a reduction in cTNT in the developing heart results in abnormal atrial septal formation and aberrant ventricular morphogenesis. We hypothesize that alterations to the haemodynamics, indicated by differential NOS3 expression, causes these abnormalities in growth in cTNT knockdown hearts. In addition, the muscular diverticula reported here suggest a novel role for mutations of structural sarcomeric proteins in the pathogenesis of congenital cardiac diverticula. From these studies, we suggest TNNT2 is a gene worthy of screening for those with a congenital heart defect, particularly atrial septal defects and ventricular diverticula.
Collapse
Affiliation(s)
- Jennifer England
- School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Kar Lai Pang
- School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Matthew Parnall
- School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Maria Isabel Haig
- School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Siobhan Loughna
- School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| |
Collapse
|
15
|
Wang C, Zhan Y, Wang F, Li H, Xie L, Liu B, Li Y, Mu D, Zheng H, Zhou K, Hua Y. Parental occupational exposures to endocrine disruptors and the risk of simple isolated congenital heart defects. Pediatr Cardiol 2015; 36:1024-37. [PMID: 25628158 DOI: 10.1007/s00246-015-1116-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 01/13/2015] [Indexed: 11/25/2022]
Abstract
This study aims to explore the associations between parental occupational exposures to endocrine disruptors (EDs) and simple isolated congenital heart defects (CHDs). A case-control study with standardized data collection involving 761 children with isolated CHDs and 609 children without any congenital malformations was conducted in Sichuan Province of China from March in 2012 to August in 2013. An adjusted job exposure matrix was used for occupational EDs exposure assessment. Logistic regression analysis was performed to assess the associations between parental occupational EDs exposures and CHDs. Maternal age at births, maternal education level, gravity, parity, induced abortion, folic acid use, medication use, drinking capacity and area of residence periconceptionally were selected as confounding factors for mothers. For fathers, we selected the following confounding factors: paternal education level, smoking, drinking frequencies and drinking capacity periconceptionally. Maternal occupational exposures to phthalates are associated with perimembranous ventricular septal defect (PmVSD) (P = 0.001, adjusted OR 3.7, 95 % CI 1.7-8.0), patent ductus arteriosus (PDA) (P = 0.002, adjusted OR 3.8, 95 % CI 1.6-8.9), secundum atrial septal defect (s-ASD) (P = 0.008, adjusted OR 3.5, 95 % CI 1.4-8.7) and pulmonary valve stenosis (PS) (P = 0.035, adjusted OR 4.2, 95 % CI 1.1-16.0), to alkylphenolic compounds and PmVSD (P = 0.003, adjusted OR 2.2, 95 % CI 1.3-3.6), PDA (P = 0.005, adjusted OR 2.0, 95 % CI 1.1-3.5) and PS (P = 0.004, adjusted OR 3.8, 95 % CI 1.5-9.4), to heavy metals with PmVSD (P = 0.003, adjusted OR 7.3, 95 % CI 2.0-27.6) and s-ASD (P = 0.034, adjusted OR 6.5, 95 % CI 1.1-36.7). Paternal occupational exposures to phthalates are associated with PmVSD (P = 0.035, adjusted OR 1.6, 95 % CI 1.0-2.4) and PS (P = 0.026, adjusted OR 2.4, 95 % CI 1.1-5.2), to alkylphenolic compounds (P = 0.027, adjusted OR 1.5, 95 % CI 1.0-2.2) with PmVSD. In conclusion, parental occupational exposures to some specific EDs, in particular phthalates and alkylphenolic compounds, are associated with an increased risk of some CHD phenotypes. However, the findings need to be considered more circumspectly regarding a crude measure of exposure probabilities and small numbers.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Simmons O, Snider P, Wang J, Schwartz RJ, Chen Y, Conway SJ. Persistent Noggin arrests cardiomyocyte morphogenesis and results in early in utero lethality. Dev Dyn 2014; 244:457-67. [PMID: 25428115 DOI: 10.1002/dvdy.24233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 11/13/2014] [Accepted: 11/16/2014] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Multiple bone morphogenetic protein (BMP) genes are expressed in the developing heart from the initiation to late-differentiation stages, and play pivotal roles in cardiovascular development. In this study, we investigated the requirement of BMP activity in heart development by transgenic over-expression of extracellular BMP antagonist Noggin. RESULTS Using Nkx2.5-Cre to drive lineage-restricted Noggin within cardiomyocyte progenitors, we show persistent Noggin arrests cardiac development at the linear heart stage. This is coupled with a significantly reduced cell proliferation rate, subsequent cardiomyocyte programmed cell death and reduction of downstream intracellular pSMAD1/5/8 expression. Noggin mutants exhibit reduced heartbeat which likely results in subsequent fully penetrant in utero lethality. Significantly, confocal and electron micrographic examination revealed considerably fewer contractile elements, as well as a lack of maturation of actin-myosin microfilaments. Molecular analysis demonstrated that ectopic Noggin-expressing regions in the early heart's pacemaker region, failed to express the potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4 (Hcn4), resulting in an overall decrease in Hcn4 levels. CONCLUSIONS Combined, our results reveal a novel role for BMP signaling in the progression of heart development from the tubular heart stage to the looped stage by means of regulation of proliferation and promotion of maturation of the in utero heart's contractile apparatus and pacemaker.
Collapse
Affiliation(s)
- Olga Simmons
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | | | | | | |
Collapse
|
17
|
Zhang L, Jia S, Yang M, Xu Y, Li C, Sun J, Huang Y, Lan X, Lei C, Zhou Y, Zhang C, Zhao X, Chen H. Detection of copy number variations and their effects in Chinese bulls. BMC Genomics 2014; 15:480. [PMID: 24935859 PMCID: PMC4073501 DOI: 10.1186/1471-2164-15-480] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 06/10/2014] [Indexed: 02/03/2023] Open
Abstract
Background Copy number variations (CNVs) are a main source of genomic structural variations underlying animal evolution and production traits. Here, with one pure-blooded Angus bull as reference, we describe a genome-wide analysis of CNVs based on comparative genomic hybridization arrays in 29 Chinese domesticated bulls and examined their effects on gene expression and cattle growth traits. Results We identified 486 copy number variable regions (CNVRs), covering 2.45% of the bovine genome, in 24 taurine (Bos taurus), together with 161 ones in 2 yaks (Bos grunniens) and 163 ones in 3 buffaloes (Bubalus bubalis). Totally, we discovered 605 integrated CNVRs, with more “loss” events than both “gain” and “both” ones, and clearly clustered them into three cattle groups. Interestingly, we confirmed their uneven distributions across chromosomes, and the differences of mitochondrion DNA copy number (gain: taurine, loss: yak & buffalo). Furthermore, we confirmed approximately 41.8% (253/605) and 70.6% (427/605) CNVRs span cattle genes and quantitative trait loci (QTLs), respectively. Finally, we confirmed 6 CNVRs in 9 chosen ones by using quantitative PCR, and further demonstrated that CNVR22 had significantly negative effects on expression of PLA2G2D gene, and both CNVR22 and CNVR310 were associated with body measurements in Chinese cattle, suggesting their key effects on gene expression and cattle traits. Conclusions The results advanced our understanding of CNV as an important genomic structural variation in taurine, yak and buffalo. This study provides a highly valuable resource for Chinese cattle’s evolution and breeding researches. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-480) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Hong Chen
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi, China.
| |
Collapse
|
18
|
Xu Y, Shi T, Cai H, Zhou Y, Lan X, Zhang C, Lei C, Qi X, Chen H. Associations of MYH3 gene copy number variations with transcriptional expression and growth traits in Chinese cattle. Gene 2013; 535:106-11. [PMID: 24316128 DOI: 10.1016/j.gene.2013.11.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/25/2013] [Accepted: 11/21/2013] [Indexed: 12/22/2022]
Abstract
Copy number variations (CNVs) have been recently recognized as another important genetic variability complementary to single nucleotide polymorphisms (SNPs). Compelling evidence has indicated that CNVs are responsible for phenotypic traits by changing the copy numbers of functional genes. Myosin heavy chain 3 (MYH3) gene is a critical regulatory factor in skeletal muscle development, and has been detected in the CNVs region by comparative genomic hybridization (CGH) array. This study was conducted to validate and detect the distribution of MYH3 copy numbers (relative to Angus cattle) in four Chinese cattle breeds (NY, QC, LX, and CY), and further to investigate the associations of the copy number changes with its transcriptional expression and cattle growth traits. Substantial genetic differences of MYH3 copy numbers were identified between NY and the other three breeds (P<0.01). The copy numbers of MYH3 gene presented the positive correlations with the transcript level of MYH3 gene in both fetal and adult skeletal muscles (P<0.05). Statistical analysis revealed that CNVs of MYH3 gene were significantly associated with growth traits of NY cattle, and the individuals with copy number gain showed better phenotypes than the loss and/or median groups (P<0.05). This study firstly attempted to establish the correlations between CNVs of candidate genes and growth traits, and our results suggested that the CNVs of MYH3 gene may be utilized as the potential markers for economic traits in selection breeding programs of Chinese cattle.
Collapse
Affiliation(s)
- Yao Xu
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Tao Shi
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Hanfang Cai
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Yang Zhou
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Chuzhao Lei
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Xinglei Qi
- Bureau of Animal Husbandry of Biyang County, Biyang, Henan 463700, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A & F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China.
| |
Collapse
|
19
|
Ye F, Yuan F, Li X, Cooper N, Tinney JP, Keller BB. Gene expression profiles in engineered cardiac tissues respond to mechanical loading and inhibition of tyrosine kinases. Physiol Rep 2013; 1:e00078. [PMID: 24303162 PMCID: PMC3841024 DOI: 10.1002/phy2.78] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 08/07/2013] [Indexed: 12/17/2022] Open
Abstract
Engineered cardiac tissues (ECTs) are platforms to investigate cardiomyocyte maturation and functional integration, the feasibility of generating tissues for cardiac repair, and as models for pharmacology and toxicology bioassays. ECTs rapidly mature in vitro to acquire the features of functional cardiac muscle and respond to mechanical load with increased proliferation and maturation. ECTs are now being investigated as platforms for in vitro models for human diseases and for pharmacologic screening for drug toxicities. We tested the hypothesis that global ECT gene expression patterns are complex and sensitive to mechanical loading and tyrosine kinase inhibitors similar to the maturing myocardium. We generated ECTs from day 14.5 rat embryo ventricular cells, as previously published, and then conditioned constructs after 5 days in culture for 48 h with mechanical stretch (5%, 0.5 Hz) and/or the p38 MAPK (p38 mitogen-activated protein kinase) inhibitor BIRB796. RNA was isolated from individual ECTs and assayed using a standard Agilent rat 4 × 44k V3 microarray and Pathway Analysis software for transcript expression fold changes and changes in regulatory molecules and networks. Changes in expression were confirmed by quantitative-polymerase chain reaction (q-PCR) for selected regulatory molecules. At the threshold of a 1.5-fold change in expression, stretch altered 1559 transcripts, versus 1411 for BIRB796, and 1846 for stretch plus BIRB796. As anticipated, top pathways altered in response to these stimuli include cellular development, cellular growth and proliferation; tissue development; cell death, cell signaling, and small molecule biochemistry as well as numerous other pathways. Thus, ECTs display a broad spectrum of altered gene expression in response to mechanical load and/or tyrosine kinase inhibition, reflecting a complex regulation of proliferation, differentiation, and architectural alignment of cardiomyocytes and noncardiomyocytes within ECT.
Collapse
Affiliation(s)
- Fei Ye
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville Louisville, Kentucky ; Affiliated Hospital of Guiyang Medical College Guiyang, China
| | | | | | | | | | | |
Collapse
|
20
|
Niu F, Wang L, Liu X, Wang H, Yang J, Liu Y, Chen L. Genetic diversity of MYH3 gene associated with growth and carcass traits in Chinese Qinchuan cattle. Mol Biol Rep 2013; 40:5635-43. [PMID: 24065521 DOI: 10.1007/s11033-013-2665-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 09/14/2013] [Indexed: 10/26/2022]
Abstract
MYH3, whose function is to convert chemical energy to mechanical energy through ATP hydrolysis, is mainly expressed in skeletal muscle at various stages and is indispensable in the procedure of development of skeletal muscle and heart. In the study, genetic variations and genotypes of MYH 3 gene in a total of 365 Qinchuan cattles were analyzed by polymerase chain reaction-restriction fragment length polymorphism, as well as verified the effect on growth and carcass traits. After PCR products were digested by restriction enzymes, eight SNPs were identified and individuals were genotyped. It showed that the SNPs at nucleotides were all in low linkage disequilibrium, therefore no dominated haplotype was found in the population. The result of statistic analysis indicated seven SNPs were significantly associated with growth and carcass traits (P < 0.05, N = 365) except locus G13791A. To sum up, the result of the study proved that polymorphisms in MYH3 gene are associated with the growth performance of Chinese Qinchuan cattle, so the variations of the gene could be used as possible molecular assisted-makers in the beef cattle breeding program and management.
Collapse
Affiliation(s)
- Fubiao Niu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
21
|
Wang L, Liu X, Niu F, Wang H, He H, Gu Y. Single nucleotide polymorphisms, haplotypes and combined genotypes in MYH₃ gene and their associations with growth and carcass traits in Qinchuan cattle. Mol Biol Rep 2012; 40:417-26. [PMID: 23073773 PMCID: PMC3518803 DOI: 10.1007/s11033-012-2076-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 10/03/2012] [Indexed: 10/27/2022]
Abstract
MYH₃ is a major contractile protein which converts chemical energy into mechanical energy through the ATP hydrolysis. MYH₃ is mainly expressed in the skeletal muscle in different stages especially embryonic period, and it has a role in the development of skeletal muscle and heart. In this study, polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was applied to analyze the genetic variations of the MYH₃ gene and verify the effect on growth and carcass traits in a total of 365 Qinchuan cattles. The PCR product was digested with some restriction enzyme and demonstrated the polymorphism in the population, the single nucleotide polymorphisms (SNPs) at nucleotides g. +1215T>C, g. +3377C>T, and g. +28625C>T were in linkage disequilibrium with each other. The result of haplotype analysis showed that nineteen different haplotypes were identified among the five SNPs. The statistical analyses indicated that the five SNPs were significant association with growth and carcass traits (P < 0.05, N = 365); whereas the five SNPs were no significant association between 18 combined genotypes of MYH₃ gene and growth and carcass traits. Taken together, our results provide the evidence that polymorphisms in MYH₃ are associated with growth and carcass traits in Qinchuan cattle, and may be used as a possible candidate for marker-assisted selection and management in beef cattle breeding program.
Collapse
Affiliation(s)
- Lijun Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, People's Republic of China
| | | | | | | | | | | |
Collapse
|
22
|
Heavy and light roles: myosin in the morphogenesis of the heart. Cell Mol Life Sci 2012; 70:1221-39. [PMID: 22955375 PMCID: PMC3602621 DOI: 10.1007/s00018-012-1131-1] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/08/2012] [Accepted: 08/13/2012] [Indexed: 01/10/2023]
Abstract
Myosin is an essential component of cardiac muscle, from the onset of cardiogenesis through to the adult heart. Although traditionally known for its role in energy transduction and force development, recent studies suggest that both myosin heavy-chain and myosin light-chain proteins are required for a correctly formed heart. Myosins are structural proteins that are not only expressed from early stages of heart development, but when mutated in humans they may give rise to congenital heart defects. This review will discuss the roles of myosin, specifically with regards to the developing heart. The expression of each myosin protein will be described, and the effects that altering expression has on the heart in embryogenesis in different animal models will be discussed. The human molecular genetics of the myosins will also be reviewed.
Collapse
|
23
|
Clause KC, Tchao J, Powell MC, Liu LJ, Huard J, Keller BB, Tobita K. Developing cardiac and skeletal muscle share fast-skeletal myosin heavy chain and cardiac troponin-I expression. PLoS One 2012; 7:e40725. [PMID: 22808244 PMCID: PMC3393685 DOI: 10.1371/journal.pone.0040725] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 06/14/2012] [Indexed: 01/26/2023] Open
Abstract
Skeletal muscle derived stem cells (MDSCs) transplanted into injured myocardium can differentiate into fast skeletal muscle specific myosin heavy chain (sk-fMHC) and cardiac specific troponin-I (cTn-I) positive cells sustaining recipient myocardial function. We have recently found that MDSCs differentiate into a cardiomyocyte phenotype within a three-dimensional gel bioreactor. It is generally accepted that terminally differentiated myocardium or skeletal muscle only express cTn-I or sk-fMHC, respectively. Studies have shown the presence of non-cardiac muscle proteins in the developing myocardium or cardiac proteins in pathological skeletal muscle. In the current study, we tested the hypothesis that normal developing myocardium and skeletal muscle transiently share both sk-fMHC and cTn-I proteins. Immunohistochemistry, western blot, and RT-PCR analyses were carried out in embryonic day 13 (ED13) and 20 (ED20), neonatal day 0 (ND0) and 4 (ND4), postnatal day 10 (PND10), and 8 week-old adult female Lewis rat ventricular myocardium and gastrocnemius muscle. Confocal laser microscopy revealed that sk-fMHC was expressed as a typical striated muscle pattern within ED13 ventricular myocardium, and the striated sk-fMHC expression was lost by ND4 and became negative in adult myocardium. cTn-I was not expressed as a typical striated muscle pattern throughout the myocardium until PND10. Western blot and RT-PCR analyses revealed that gene and protein expression patterns of cardiac and skeletal muscle transcription factors and sk-fMHC within ventricular myocardium and skeletal muscle were similar at ED20, and the expression patterns became cardiac or skeletal muscle specific during postnatal development. These findings provide new insight into cardiac muscle development and highlight previously unknown common developmental features of cardiac and skeletal muscle.
Collapse
Affiliation(s)
- Kelly C. Clause
- Cardiovascular Development Research Program, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jason Tchao
- Cardiovascular Development Research Program, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mary C. Powell
- Cardiovascular Development Research Program, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Li J. Liu
- Cardiovascular Development Research Program, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Johnny Huard
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- McGowan Institutes for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Bradley B. Keller
- Department of Pediatrics, University of Louisville, Louisville, Kentucky, United States of America
| | - Kimimasa Tobita
- Cardiovascular Development Research Program, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- McGowan Institutes for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|