1
|
Henry M, Mertens L. Echocardiographic assessment of Right Ventricular Diastolic Function in Children and Adults: Present State and Future Directions. Can J Cardiol 2025:S0828-282X(25)00234-X. [PMID: 40158654 DOI: 10.1016/j.cjca.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025] Open
Abstract
Diastolic function plays an important but often overlooked role in overall ventricular performance and is comprised of sequence of events which together result in adequate filling at low filling pressures. From a physiologic point of view there are two distinct, yet related periods that drive diastole: active relaxation and late filling. Much of the literature on diastolic function is based on the left ventricle however there are significant differences in morphology, physiology and adaptation between ventricles. Echocardiographic assessment of RV diastolic function is challenging due to our imperfect understanding in RV physiology, suboptimal imaging tools and the use of models that have been built using the left ventricle. Conventional assessment includes IVC size, RA volume and the use of Doppler to quantify tissue and blood velocity (TV E/A, e', a', E/e', IVRT, hepatic vein and pulmonary artery). In adults, TV E/A used in combination with e', IVC size, may be used to classify diastolic impairment however in children this is dependent to a greater degree on age and pathology. Right atrial and ventricular strain suffers from fewer limitations and may show incremental benefit however remains understudied, particularly in children. Novel methods made possible with ultrafast ultrasound provide a means to non-invasively assess intraventricular pressure differences as a measure of active relaxation, and shear wave velocity to directly assess myocardial stiffness, however these remain in pre-clinical stages. This review discusses right ventricular diastolic physiology, the current state and limitations of echocardiographic evaluation and explores promising new methods for its assessment.
Collapse
Affiliation(s)
- Matthew Henry
- Cardiology Program, Division of Pediatric Cardiology, University of Alberta, Alberta, Canada; Centre for Research in Cardiac Imaging and Innovation in Medicine, Edmonton, Alberta, Canada.
| | - Luc Mertens
- Cardiology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
| |
Collapse
|
2
|
Zheng Q, Liu Y, Guo M, Zhang X, Zhang Q, Yu XY, Lin Z. Discovery of therapeutic targets in cardiovascular diseases using high-throughput chromosome conformation capture (Hi-C). Front Genet 2025; 16:1515010. [PMID: 40182924 PMCID: PMC11966399 DOI: 10.3389/fgene.2025.1515010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Epigenetic changes have been associated with several cardiovascular diseases. In recent years, epigenetic inheritance based on spatial changes has gradually attracted attention. Alterations in three-dimensional chromatin structures have been shown to regulate gene expression and influence disease onset and progression. High-throughput Chromosome Conformation Capture (Hi-C) is a powerful method to detect spatial chromatin conformation changes. Since its development, Hi-C technology has been widely adopted for discovering novel therapeutic targets in cardiovascular research. In this review, we summarize key targets identified by Hi-C in cardiovascular diseases and discuss their potential implications for epigenetic therapy.
Collapse
Affiliation(s)
- Quan Zheng
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Ying Liu
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
- Department of Pharmacology, School of Pharmacy, Guangzhou Xinhua University, Guangzhou, China
| | - Minghao Guo
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Xin Zhang
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Qingbin Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xi-Yong Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhongxiao Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Zrinski Topic R, Lenicek Krleza J. Cardiac Markers in Pediatric Laboratory Medicine: Critical Review. Diagnostics (Basel) 2025; 15:165. [PMID: 39857049 PMCID: PMC11763470 DOI: 10.3390/diagnostics15020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Currently, there are no validated guidelines or recommendations for how to interpret cardiac biomarkers in the pediatric population. The most commonly used cardiac biomarkers are cardiac troponins and natriuretic peptides, but the clinical value of common cardiac biomarkers in pediatric laboratory medicine is restricted due to age- and sex-specific interpretations, and there are no standardized cut-off values. The results from the studies on reference values, as well as results from clinical studies, are difficult to compare with identical studies due to the heterogeneity of subject characteristics (gestational and chronological age, sex, pubertal status, menstrual cycle, exercise), assay characteristics (type of assay, generation of assay, analytical platform used), and experimental protocol characteristics (prospective or retrospective studies, reference population selection, patient population selection, inclusion and exclusion criteria, number of subjects). Future studies need to establish evidence-based cut-offs for specific indications to optimize utilization and standardize the interpretation of common cardiac biomarkers in neonates, children, and adolescents. The aim of this article was to summarize the current analytical and clinical limitations of cardiac troponins and natriuretic peptides in the pediatric population, as informed by the existing published literature.
Collapse
Affiliation(s)
- Renata Zrinski Topic
- Department of Laboratory Diagnostics, Children’s Hospital Zagreb, 10000 Zagreb, Croatia;
- Department of Laboratory Medical Diagnostics, University of Applied Health Sciences, 10000 Zagreb, Croatia
| | - Jasna Lenicek Krleza
- Department of Laboratory Diagnostics, Children’s Hospital Zagreb, 10000 Zagreb, Croatia;
- Department of Laboratory Medical Diagnostics, University of Applied Health Sciences, 10000 Zagreb, Croatia
- Department of Nursing, Catholic University of Croatia, 10000 Zagreb, Croatia
| |
Collapse
|
4
|
Wang G, Shen WB, Chen AW, Reece EA, Yang P. Diabetes and Early Development: Epigenetics, Biological Stress, and Aging. Am J Perinatol 2024. [PMID: 39209306 DOI: 10.1055/a-2405-1493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pregestational diabetes, either type 1 or type 2 diabetes, induces structural birth defects including neural tube defects and congenital heart defects in human fetuses. Rodent models of type 1 and type 2 diabetic embryopathy have been established and faithfully mimic human conditions. Hyperglycemia of maternal diabetes triggers oxidative stress in the developing neuroepithelium and the embryonic heart leading to the activation of proapoptotic kinases and excessive cell death. Oxidative stress also activates the unfolded protein response and endoplasmic reticulum stress. Hyperglycemia alters epigenetic landscapes by suppressing histone deacetylation, perturbing microRNA (miRNA) expression, and increasing DNA methylation. At cellular levels, besides the induction of cell apoptosis, hyperglycemia suppresses cell proliferation and induces premature senescence. Stress signaling elicited by maternal diabetes disrupts cellular organelle homeostasis leading to mitochondrial dysfunction, mitochondrial dynamic alteration, and autophagy impairment. Blocking oxidative stress, kinase activation, and cellular senescence ameliorates diabetic embryopathy. Deleting the mir200c gene or restoring mir322 expression abolishes maternal diabetes hyperglycemia-induced senescence and cellular stress, respectively. Both the autophagy activator trehalose and the senomorphic rapamycin can alleviate diabetic embryopathy. Thus, targeting cellular stress, miRNAs, senescence, or restoring autophagy or mitochondrial fusion is a promising approach to prevent poorly controlled maternal diabetes-induced structural birth defects. In this review, we summarize the causal events in diabetic embryopathy and propose preventions for this pathological condition. KEY POINTS: · Maternal diabetes induces structural birth defects.. · Kinase signaling and cellular organelle stress are critically involved in neural tube defects.. · Maternal diabetes increases DNA methylation and suppresses developmental gene expression.. · Cellular apoptosis and senescence are induced by maternal diabetes in the neuroepithelium.. · microRNAs disrupt mitochondrial fusion leading to congenital heart diseases in diabetic pregnancy..
Collapse
Affiliation(s)
- Guanglei Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anna Wu Chen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - E Albert Reece
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
5
|
Yaqoob H, Ahmad H, Ali SI, Patel N, Arif A. Missense mutations in the CITED2 gene may contribute to congenital heart disease. BMC Cardiovasc Disord 2024; 24:516. [PMID: 39333893 PMCID: PMC11429617 DOI: 10.1186/s12872-024-04035-2] [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: 03/15/2024] [Accepted: 07/08/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is a lifelong abnormality present from birth. Multiple studies have shown that mutations in genes involved in heart development could cause congenital heart disease. The CITED2 gene works as a transcription factor in the hypoxic pathway for the development of the heart. Therefore, five CHD types, ventricular septal defect, atrial septal defect, atrioventricular septal defect, tetralogy of fallot, and patent ductus arteriosus, were evaluated by conducting a targeted single nucleotide polymorphism (SNP) analysis of the CITED2 gene variant rs375393125 (T > C). This study aimed to identify the association of CITED2 gene mutations in CHD patients. METHODS Three hundred fifty samples, 250 from patients and 100 from controls, were collected for this genetic analysis. Allele-specific PCR and gel electrophoresis were used to identify the target missense mutations. The genotypic results of the CHDs were further validated through Sanger sequencing. RESULTS The frequency of the homozygous mutant (CC) in CHD patients was 48.4%, and of the heterozygous mutant (TC) genotype was 11.4%; these percentages are higher than controls (1%). The control samples had only one heterozygous TC and no homozygous CC genotype. The chi-square value was obtained at 103.9 with a probability of 0.05, more significant than the significance value of 21.03. The odds ratio was 43.7, which is > 1. The calculated value of ANOVA was 11.6, which was more significant than the F critical value of 3.7. As a result of sequencing, the mutant sample of each selected CHD type was found heterozygous or homozygous, and the results were like those obtained through conventional PCR. CONCLUSION The samples of CHD patients showed mutations. Therefore, the CITED2 gene SNP might be associated with CHD.
Collapse
Affiliation(s)
- Hira Yaqoob
- The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi, Pakistan
| | - Hussain Ahmad
- The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi, Pakistan
| | - Syed Irtiza Ali
- The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi, Pakistan
| | - Najma Patel
- National Institute of Cardiovascular Diseases Pakistan Rafiqui (H.J.), Shaheed Road, Karachi, Pakistan
| | - Afsheen Arif
- The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi, Pakistan.
| |
Collapse
|
6
|
Ling S, Yan L, Mao R, Li J, Xi H, Wang F, Li X, He M. A Coarse-Fine Collaborative Learning Model for Three Vessel Segmentation in Fetal Cardiac Ultrasound Images. IEEE J Biomed Health Inform 2024; 28:4036-4047. [PMID: 38635389 DOI: 10.1109/jbhi.2024.3390688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Congenital heart disease (CHD) is the most frequent birth defect and a leading cause of infant mortality, emphasizing the crucial need for its early diagnosis. Ultrasound is the primary imaging modality for prenatal CHD screening. As a complement to the four-chamber view, the three-vessel view (3VV) plays a vital role in detecting anomalies in the great vessels. However, the interpretation of fetal cardiac ultrasound images is subjective and relies heavily on operator experience, leading to variability in CHD detection rates, particularly in resource-constrained regions. In this study, we propose an automated method for segmenting the pulmonary artery, ascending aorta, and superior vena cava in the 3VV using a novel deep learning network named CoFi-Net. Our network incorporates a coarse-fine collaborative strategy with two parallel branches dedicated to simultaneous global localization and fine segmentation of the vessels. The coarse branch employs a partial decoder to leverage high-level semantic features, enabling global localization of objects and suppression of irrelevant structures. The fine branch utilizes attention-parameterized skip connections to improve feature representations and improve boundary information. The outputs of the two branches are fused to generate accurate vessel segmentations. Extensive experiments conducted on a collected dataset demonstrate the superiority of CoFi-Net compared to state-of-the-art segmentation models for 3VV segmentation, indicating its great potential for enhancing CHD diagnostic efficiency in clinical practice. Furthermore, CoFi-Net outperforms other deep learning models in breast lesion segmentation on a public breast ultrasound dataset, despite not being specifically designed for this task, demonstrating its potential and robustness for various segmentation tasks.
Collapse
|
7
|
Zubrzycki M, Schramm R, Costard-Jäckle A, Grohmann J, Gummert JF, Zubrzycka M. Cardiac Development and Factors Influencing the Development of Congenital Heart Defects (CHDs): Part I. Int J Mol Sci 2024; 25:7117. [PMID: 39000221 PMCID: PMC11241401 DOI: 10.3390/ijms25137117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The traditional description of cardiac development involves progression from a cardiac crescent to a linear heart tube, which in the phase of transformation into a mature heart forms a cardiac loop and is divided with the septa into individual cavities. Cardiac morphogenesis involves numerous types of cells originating outside the initial cardiac crescent, including neural crest cells, cells of the second heart field origin, and epicardial progenitor cells. The development of the fetal heart and circulatory system is subject to regulatation by both genetic and environmental processes. The etiology for cases with congenital heart defects (CHDs) is largely unknown, but several genetic anomalies, some maternal illnesses, and prenatal exposures to specific therapeutic and non-therapeutic drugs are generally accepted as risk factors. New techniques for studying heart development have revealed many aspects of cardiac morphogenesis that are important in the development of CHDs, in particular transposition of the great arteries.
Collapse
Affiliation(s)
- Marek Zubrzycki
- Department of Surgery for Congenital Heart Defects, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany;
| | - Rene Schramm
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Angelika Costard-Jäckle
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Jochen Grohmann
- Department of Congenital Heart Disease/Pediatric Cardiology, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany;
| | - Jan F. Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital, Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany; (R.S.); (A.C.-J.); (J.F.G.)
| | - Maria Zubrzycka
- Department of Clinical Physiology, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
8
|
Blade SP, Falkowski DJ, Bachand SN, Pagano SJ, Chin L. Mechanobiology of Adipocytes. BIOLOGY 2024; 13:434. [PMID: 38927314 PMCID: PMC11200640 DOI: 10.3390/biology13060434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
The growing obesity epidemic necessitates increased research on adipocyte and adipose tissue function and disease mechanisms that progress obesity. Historically, adipocytes were viewed simply as storage for excess energy. However, recent studies have demonstrated that adipocytes play a critical role in whole-body homeostasis, are involved in cell communication, experience forces in vivo, and respond to mechanical stimuli. Changes to the adipocyte mechanical microenvironment can affect function and, in some cases, contribute to disease. The aim of this review is to summarize the current literature on the mechanobiology of adipocytes. We reviewed over 100 papers on how mechanical stress is sensed by the adipocyte, the effects on cell behavior, and the use of cell culture scaffolds, particularly those with tunable stiffness, to study adipocyte behavior, adipose cell and tissue mechanical properties, and computational models. From our review, we conclude that adipocytes are responsive to mechanical stimuli, cell function and adipogenesis can be dictated by the mechanical environment, the measurement of mechanical properties is highly dependent on testing methods, and current modeling practices use many different approaches to recapitulate the complex behavior of adipocytes and adipose tissue. This review is intended to aid future studies by summarizing the current literature on adipocyte mechanobiology.
Collapse
Affiliation(s)
- Sean P. Blade
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Dylan J. Falkowski
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Sarah N. Bachand
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Steven J. Pagano
- Department of Mechanical Engineering, Widener University, Chester, PA 19013, USA;
| | - LiKang Chin
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| |
Collapse
|
9
|
Haybar H, Hadi H, Purrahman D, Mahmoudian-Sani MR, Saki N. Emerging roles of HOTAIR lncRNA in the pathogenesis and prognosis of cardiovascular diseases. Biomark Med 2024; 18:203-219. [PMID: 38411079 DOI: 10.2217/bmm-2023-0368] [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] [Indexed: 02/28/2024] Open
Abstract
Highlights HOTAIR, a long noncoding RNA, plays a role in the regulation of proteins involved in the pathogenesis of cardiovascular disease. Furthermore, it has been identified as a biomarker of this type of disease. Several factors and cells contribute to atherosclerosis, a progressive disease. However, the prognosis of HOTAIR in this disease varies depending on the path in which it plays a role. For this condition, there is no single prognosis to consider.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hakimeh Hadi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Daryush Purrahman
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Reza Mahmoudian-Sani
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
10
|
Liu XX, Zhao DY, Zhao X, Zhang XA, Yu ZL, Sun LH. The effect of China's birth policy changes on birth defects-A large hospital-based cross-sectional study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:1156-1167. [PMID: 37158781 DOI: 10.1080/09603123.2023.2207469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023]
Abstract
A retrospective analysis of birth data hospital-based obtained from 14 monitoring areas in the Huaihe River Basin from 2009 to 2019 was conducted. Trend in the total prevalence of birth defects (BDs) and subgroups were analyzed using the Joinpoint Regression model. The incidence of BDs increased gradually from 118.87 per 10,000 in 2009 to 241.18 per 10,000 in 2019 (AAPC = 5.91, P < 0.001). Congenital heart diseases were the most common subtype of BDs. The proportion of maternal age younger than 25 decreased but the age 25-40 years increased significantly (AAPC<20=-5.58; AAPC20-24=-6.38; AAPC25-29 = 5.15; AAPC30-35 = 7.07; AAPC35-40 = 8.27; All P < 0.05). Compared with the one-child policy period, the risk of BDs was greater for groups among maternal age younger than 40 years during the partial and universal two-child policy period (P < 0.001). The incidence of BDs and the proportion of women with advanced maternal age in Huaihe River Basin is increasing. There was an interaction between changes in birth policy and the mother's age on the risk of BDs.
Collapse
Affiliation(s)
- Xin-Xin Liu
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Dan-Yang Zhao
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Zhao
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-An Zhang
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zeng-Li Yu
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Huan Sun
- The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
11
|
Yuan X, Chen R, Luo G, Sun P, Song X, Ma J, Sun R, Yu T, Jiang Z. Role and mechanism of miR-871-3p/Megf8 in regulating formaldehyde-induced cardiomyocyte inflammation and congenital heart disease. Int Immunopharmacol 2024; 126:111297. [PMID: 38039718 DOI: 10.1016/j.intimp.2023.111297] [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: 08/17/2023] [Revised: 11/02/2023] [Accepted: 11/24/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVE AND DESIGN We aimed to investigate the molecular mechanism underlying formaldehyde (FA)-induced congenital heart disease (CHD) using in vitro and in vivo models. MATERIALS AND SUBJECTS Neonatal rat heart tissues and H9C2 cells were used for in vitro studies, while FA-exposed new-born rats were used for in vivo studies. TREATMENT H9C2 cells were exposed to FA concentrations of 0, 50, 100 and 150 μM/mL for 24 h. METHODS Whole transcriptome gene sequencing identified differentially expressed miRNAs in neonatal rat heart tissues, while Real-time quantitative PCR (RT-qPCR) assessed miR-871-3p and Megf8 expression. RNA pull-down and dual-luciferase reporter assays determined miR-871-3p and Megf8 relationships. Inflammatory cytokine expression was assessed by western blotting. A FA-induced CHD model was used to validate miR-871-3p regulatory effects in vivo. RESULTS We identified 89 differentially expressed miRNAs, with 28 up-regulated and 61 down-regulated (fold change ≥ 2.0, P < 0.05). Inflammation (interleukin) and signalling pathways were found to control FA-induced cardiac dysplasia. miR-871-3p was upregulated in FA-exposed heart tissues, modulated inflammation, and directly targeted Megf8. In vivo experiments showed miR-871-3p knockdown inhibited FA-induced inflammation and CHD. CONCLUSION We demonstrated miR-871-3p's role in FA-induced CHD by targeting Megf8, providing potential targets for CHD intervention and improved diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Xiaoli Yuan
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong Province, People's Republic of China
| | - Rui Chen
- Heart Center, Women and Children's Hospital, Qingdao University, Qingdao 266034, Shandong Province, People's Republic of China
| | - Gang Luo
- Heart Center, Women and Children's Hospital, Qingdao University, Qingdao 266034, Shandong Province, People's Republic of China
| | - Pin Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong Province, People's Republic of China
| | - Xiaoxia Song
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong Province, People's Republic of China
| | - Jianmin Ma
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong Province, People's Republic of China
| | - Ruicong Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong Province, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong Province, People's Republic of China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, Shandong Province, People's Republic of China.
| | - Zhirong Jiang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong Province, People's Republic of China.
| |
Collapse
|
12
|
Lin CY, Chang YM, Tseng HY, Shih YL, Yeh HH, Liao YR, Tang HH, Hsu CL, Chen CC, Yan YT, Kao CF. Epigenetic regulator RNF20 underlies temporal hierarchy of gene expression to regulate postnatal cardiomyocyte polarization. Cell Rep 2023; 42:113416. [PMID: 37967007 DOI: 10.1016/j.celrep.2023.113416] [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: 12/08/2022] [Revised: 09/19/2023] [Accepted: 10/25/2023] [Indexed: 11/17/2023] Open
Abstract
Differentiated cardiomyocytes (CMs) must undergo diverse morphological and functional changes during postnatal development. However, the mechanisms underlying initiation and coordination of these changes remain unclear. Here, we delineate an integrated, time-ordered transcriptional network that begins with expression of genes for cell-cell connections and leads to a sequence of structural, cell-cycle, functional, and metabolic transitions in mouse postnatal hearts. Depletion of histone H2B ubiquitin ligase RNF20 disrupts this gene network and impairs CM polarization. Subsequently, assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis confirmed that RNF20 contributes to chromatin accessibility in this context. As such, RNF20 is likely to facilitate binding of transcription factors at the promoters of genes involved in cell-cell connections and actin organization, which are crucial for CM polarization and functional integration. These results suggest that CM polarization is one of the earliest events during postnatal heart development and provide insights into how RNF20 regulates CM polarity and the postnatal gene program.
Collapse
Affiliation(s)
- Chia-Yeh Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsin-Yi Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yen-Ling Shih
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsiao-Hui Yeh
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - You-Rou Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Han-Hsuan Tang
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chia-Ling Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| |
Collapse
|
13
|
Ashiq S, Sabar MF. Association of maternal hypertension and diabetes with variants of the NKX2-5, LEFTY1 and LEFTY2 genes in children with congenital heart defects: a case-control study from Pakistani Population. Mol Biol Rep 2023; 50:5013-5020. [PMID: 37097539 DOI: 10.1007/s11033-023-08418-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/30/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND Globally, congenital heart defect (CHD) is the most common congenital malformation, responsible for higher morbidity and mortality in the pediatric population. It is a complex multifactorial disease influenced by gene-environment and gene-gene interactions. The current study was the first attempt to study these polymorphisms in common clinical phenotypes of CHD in Pakistan and the association between maternal hypertension and diabetes with single nucleotide polymorphisms (SNPs) in children. METHODS A total of 376 subjects were recruited in this current case-control study. Six variants from three genes were analyzed by cost-effective multiplex PCR and genotyped by minisequencing. Statistical analysis was done by GraphPad prism and Haploview. The association of SNPs and CHD was determined using logistic regression. RESULTS The risk allele frequency was higher in cases as compared to healthy subjects, but the results were not significant for rs703752. However, stratification analysis suggested that rs703752 was significantly associated with the tetralogy of Fallot. The rs2295418 was significantly associated with maternal hypertension (OR = 16.41, p = 0.003), while a weak association was present between maternal diabetes and rs360057 (p = 0.08). CONCLUSION In conclusion, variants in transcriptional and signaling genes were associated with Pakistani pediatric CHD patients that showed varied susceptibility between different clinical phenotypes of CHD. In addition, this study was the first report regarding the significant association between maternal hypertension and the LEFTY2 gene variant.
Collapse
Affiliation(s)
- Sana Ashiq
- Centre for Applied Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Thokar Niaz Baig, Lahore, 53700, Pakistan
| | - Muhammad Farooq Sabar
- Centre for Applied Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Thokar Niaz Baig, Lahore, 53700, Pakistan.
| |
Collapse
|
14
|
Scott AK, Rafuse M, Neu CP. Mechanically induced alterations in chromatin architecture guide the balance between cell plasticity and mechanical memory. Front Cell Dev Biol 2023; 11:1084759. [PMID: 37143893 PMCID: PMC10151697 DOI: 10.3389/fcell.2023.1084759] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 04/07/2023] [Indexed: 05/06/2023] Open
Abstract
Phenotypic plasticity, or adaptability, of a cell determines its ability to survive and function within changing cellular environments. Changes in the mechanical environment, ranging from stiffness of the extracellular matrix (ECM) to physical stress such as tension, compression, and shear, are critical environmental cues that influence phenotypic plasticity and stability. Furthermore, an exposure to a prior mechanical signal has been demonstrated to play a fundamental role in modulating phenotypic changes that persist even after the mechanical stimulus is removed, creating stable mechanical memories. In this mini review, our objective is to highlight how the mechanical environment alters both phenotypic plasticity and stable memories through changes in chromatin architecture, mainly focusing on examples in cardiac tissue. We first explore how cell phenotypic plasticity is modulated in response to changes in the mechanical environment, and then connect the changes in phenotypic plasticity to changes in chromatin architecture that reflect short-term and long-term memories. Finally, we discuss how elucidating the mechanisms behind mechanically induced chromatin architecture that lead to cell adaptations and retention of stable mechanical memories could uncover treatment methods to prevent mal-adaptive permanent disease states.
Collapse
Affiliation(s)
- Adrienne K. Scott
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Michael Rafuse
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Corey P. Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
15
|
Epigenetic Evaluation of the TBX20 Gene and Environmental Risk Factors in Mexican Paediatric Patients with Congenital Septal Defects. Cells 2023; 12:cells12040586. [PMID: 36831251 PMCID: PMC9953838 DOI: 10.3390/cells12040586] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/25/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
The TBX20 gene has a key role during cardiogenesis, and it has been related to epigenetic mechanisms in congenital heart disease (CHD). The purpose of this study was to assess the association between DNA methylation status and congenital septal defects. The DNA methylation of seven CpG sites in the TBX20 gene promoter was analyzed through pyrosequencing as a quantitative method in 48 patients with congenital septal defects and 104 individuals with patent ductus arteriosus (PDA). The average methylation was higher in patients than in PDA (p < 0.001). High methylation levels were associated with a higher risk of congenital septal defects (OR = 4.59, 95% CI = 1.57-13.44, p = 0.005). The ROC curve analysis indicated that methylation of the TBX20 gene could be considered a risk marker for congenital septal defects (AUC = 0.682; 95% CI = 0.58-0.77; p < 0.001). The analysis of environmental risk factors in patients with septal defects and PDA showed an association between the consumption of vitamins (OR = 0.10; 95% CI = 0.01-0.98; p = 0.048) and maternal infections (OR = 3.10; 95% CI = 1.26-7.60; p = 0.013). These results suggest that differences in DNA methylation of the TBX20 gene can be associated with septal defects.
Collapse
|
16
|
Semenovykh D, Benak D, Holzerova K, Cerna B, Telensky P, Vavrikova T, Kolar F, Neckar J, Hlavackova M. Myocardial m6A regulators in postnatal development: effect of sex. Physiol Res 2022. [DOI: 10.33549/physiolres.934970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
N6-methyladenosine (m6A) is an abundant mRNA modification affecting mRNA stability and protein expression. It is a highly dynamic process, and its outcomes during postnatal heart development are poorly understood. Here we studied m6A machinery in the left ventricular myocardium of Fisher344 male and female rats (postnatal days one to ninety; P1-P90) using Western Blot. A downward pattern of target protein levels (demethylases FTO and ALKBH5, methyltransferase METTL3, reader YTHDF2) was revealed in male and female rat during postnatal development. On P1, the FTO protein level was significantly higher in male compared to females.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - M Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic. E-mail:
| |
Collapse
|
17
|
SEMENOVYKH D, BENAK D, HOLZEROVA K, CERNA B, TELENSKY P, VAVRIKOVA T, KOLAR F, NECKAR J, HLAVACKOVA M. Myocardial m6A regulators in postnatal development: effect of sex. Physiol Res 2022; 71:877-882. [PMID: 36426889 PMCID: PMC9814979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
N6-methyladenosine (m6A) is an abundant mRNA modification affecting mRNA stability and protein expression. It is a highly dynamic process, and its outcomes during postnatal heart development are poorly understood. Here we studied m6A machinery in the left ventricular (LV) myocardium of Fisher344 male and female rats (postnatal days one to ninety; P1-P90) using Western Blot. A downward pattern of target protein levels (demethylases FTO and ALKBH5, methyltransferase METTL3, reader YTHDF2) was revealed in male and female rat LVs during postnatal development. On P1, the FTO protein level was significantly higher in male LVs compared to females.
Collapse
Affiliation(s)
- Dmytro SEMENOVYKH
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic,Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Daniel BENAK
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic,Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Kristyna HOLZEROVA
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Barbora CERNA
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr TELENSKY
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic,International Clinical Research Center of St. Anne’s University Hospital Brno, Dementia Research Group, Brno, Czech Republic
| | - Tereza VAVRIKOVA
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Frantisek KOLAR
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan NECKAR
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marketa HLAVACKOVA
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
18
|
Congenital Heart Diseases: Genetic Risk Variants and Their Methylation Status. Genes (Basel) 2022; 13:genes13112115. [DOI: 10.3390/genes13112115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/01/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The interaction between single nucleotide variants (SNVs) associated with congenital heart diseases (CHDs) and their gene methylation status has not been well researched. The aim of the present study was to determine if there is a relationship between the methy lation status (MS) of genes and the allelic variants associated with CHDs. (2) Methods: Seven SNVs of the genes AXIN1, TBX1, TBX20, and MTHFR were selected from the literature. DNA extraction, genotyping, and a methylation analysis were performed on healthy subjects and subjects with CHDs. (3) Results: Twenty-two subjects with CHDs were selected as the case group (15 with ventricular septal defects (VSDs) and 7 with atrial septal defects (ASDs)), and 44 healthy subjects comprised the control group. The MTHFR and AXIN1 genes were hypermethylated in the control group when compared to the case group. When analyzed separately, those with atrial septum defects exhibited greater methylation, except for the gene MTHFR where there were no differences. Only the alternate alleles of MTHFR showed a significantly different methylation status in those without cardiopathy. (4) Conclusions: The MTHFR and AXIN genes were hypermethylated in the control group; however, only the alternate alleles of MTHFR (rs1801133 and rs1801131) showed a significantly different methylation status.
Collapse
|
19
|
Zuela-Sopilniak N, Lammerding J. Can't handle the stress? Mechanobiology and disease. Trends Mol Med 2022; 28:710-725. [PMID: 35717527 PMCID: PMC9420767 DOI: 10.1016/j.molmed.2022.05.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
Mechanobiology is a rapidly growing research area focused on how mechanical forces and properties influence biological systems at the cell, molecular, and tissue level, and how those biological systems, in turn, control mechanical parameters. Recently, it has become apparent that disrupted mechanobiology has a significant role in many diseases, from cardiovascular disease to muscular dystrophy and cancer. An improved understanding of this intricate process could be harnessed toward developing alternative and more targeted treatment strategies, and to advance the fields of regenerative and personalized medicine. Modulating the mechanical properties of the cellular microenvironment has already been used successfully to boost antitumor immune responses and to induce cardiac and spinal regeneration, providing inspiration for further research in this area.
Collapse
Affiliation(s)
- Noam Zuela-Sopilniak
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
20
|
Wang Z, Zhong C, Li H. Histone demethylase KDM5B catalyzed H3K4me3 demethylation to promote differentiation of bone marrow mesenchymal stem cells into cardiomyocytes. Mol Biol Rep 2022; 49:7239-7249. [PMID: 35788877 PMCID: PMC9304058 DOI: 10.1007/s11033-022-07428-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022]
Abstract
Background Studies have shown that histone H3 methylation is involved in regulating the differentiation of Bone Marrow Mesenchymal Stem Cells (BMSCs). KDM5B can specifically reduce the level of histone 3 lysine 4 trimethylation (H3K4me3), thereby activating the expression of related genes and participating in biological processes such as cell differentiation, embryonic development and tumor formation. Whether KDM5B is involved in the regulation of BMSCs differentiation into cardiomyocytes through the above manner has not been reported. Objective To investigate the effect of KDM5B on the induction and differentiation of swine BMSCs into myocardial cells in vitro. Methods Swine bone marrow BMSCs were isolated and cultured, and the overexpression, interference expression and blank vector of KMD5B were constructed and transfected by lentivirus. BMSCs was induced to differentiate into cardiomyocytes by 5-azacytidine (5-AZA) in vitro, and the differentiation efficiency was compared by immunofluorescence, RT-PCR, Western Blot and whole-cell patch clamp detection. Result Compared with the control group, the expression levels of histone H3K4me3 and pluripotency gene Nanog in KDM5B overexpression group were significantly decreased, while the expression level of key myocardial gene HCN4 and myocardial marker gene α-Actin and cTNT were significantly increased, and the Na+ current density on the surface of differentiated myocardial cell membrane was significantly increased. Meanwhile, the corresponding results of the KDM5B silent expression group were just opposite. Conclusions It indicated that enhanced KDM5B expression could promote the differentiation of BMSCs into cardiomyocytes and improve the differentiation efficiency by controlling H3K4 methylation levels.
Collapse
Affiliation(s)
- Zhen Wang
- Medical College of Yangzhou University, Yangzhou, 225001, Jiangsu, China.,Friendliness Hospital Yangzhou, Jiangsu, 225009, China
| | - Chenlu Zhong
- Medical College of Yangzhou University, Yangzhou, 225001, Jiangsu, China.,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China
| | - Hongxiao Li
- Medical College of Yangzhou University, Yangzhou, 225001, Jiangsu, China. .,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
21
|
Molecular Mechanisms Contributing to the Etiology of Congenital Diaphragmatic Hernia: A Review and Novel Cases. J Pediatr 2022; 246:251-265.e2. [PMID: 35314152 DOI: 10.1016/j.jpeds.2022.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 12/25/2022]
|
22
|
Shar JA, Keswani SG, Grande-Allen KJ, Sucosky P. Significance of aortoseptal angle anomalies to left ventricular hemodynamics and subaortic stenosis: A numerical study. Comput Biol Med 2022; 146:105613. [PMID: 35751200 PMCID: PMC10570849 DOI: 10.1016/j.compbiomed.2022.105613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Discrete subaortic stenosis (DSS) is an obstructive cardiac disease caused by a membranous lesion in the left ventricular (LV) outflow tract (LVOT). Although its etiology is unknown, the higher prevalence of DSS in LVOT anatomies featuring a steep aortoseptal angle (AoSA) suggests a potential role for hemodynamics. Therefore, the objective of this study was to quantify the impact of AoSA steepening on the LV three-dimensional (3D) hemodynamic stress environment. METHODS A 3D LV model reconstructed from cardiac cine-magnetic resonance imaging was connected to four LVOT geometrical variations spanning the clinical AoSA range (115°-160°). LV hemodynamic stresses were characterized in terms of cycle-averaged pressure, temporal shear magnitude (TSM), and oscillatory shear index. The wall shear stress (WSS) topological skeleton was further analyzed by computing the scaled divergence of the WSS vector field. RESULTS AoSA steepening caused an increasingly perturbed subaortic flow marked by LVOT flow skewness and complex 3D secondary flow patterns. These disturbances generated WSS overloads (>45% increase in TSM vs. 160° model) on the inferior LVOT wall, and increased WSS contraction (>66% decrease in WSS divergence vs. 160° model) in regions prone to DSS membrane formation. CONCLUSIONS AoSA steepening generated substantial hemodynamic stress abnormalities in LVOT regions prone to DSS formation. Further studies are needed to assess the possible impact of such mechanical abnormalities on the tissue and cellular responses.
Collapse
Affiliation(s)
- Jason A Shar
- Department of Mechanical Engineering, Kennesaw State University, 840 Polytechnic Lane, Marietta, GA, 30060, USA.
| | - Sundeep G Keswani
- Division of Pediatric Surgery, Texas Children's Hospital, Department of Surgery, Baylor College of Medicine, USA.
| | | | - Philippe Sucosky
- Department of Mechanical Engineering, Kennesaw State University, 840 Polytechnic Lane, Marietta, GA, 30060, USA.
| |
Collapse
|
23
|
Analysis of commonly expressed genes between first trimester fetal heart and placenta cell types in the context of congenital heart disease. Sci Rep 2022; 12:10756. [PMID: 35750800 PMCID: PMC9232495 DOI: 10.1038/s41598-022-14955-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/16/2022] [Indexed: 02/08/2023] Open
Abstract
Congenital heart disease (CHD) is often associated with fetal growth abnormalities. During the first trimester of pregnancy, the heart and placenta develop concurrently, and share key developmental pathways. It is hypothesized that defective morphogenesis of either organ is synergistically linked. However, many studies determined to understand the mechanisms behind CHD overlook the contribution of the placenta. In this study, we aimed to identify commonly expressed genes between first trimester heart and placenta cells using two publicly available single cell sequencing databases. Using a systematic computational approach, we identified 328 commonly expressed genes between heart and placenta endothelial cells and enrichment in pathways including Vasculature Development (GO:0001944, FDR 2.90E−30), and Angiogenesis (GO:0001525, FDR 1.18E−27). We also found, in comparison with fetal heart endothelial cells, 197 commonly expressed genes with placenta extravillous trophoblasts, 128 with cytotrophoblasts and 80 with syncytiotrophoblasts, and included genes such as FLT1, GATA2, ENG and CDH5. Finally, comparison of first trimester cardiomyocytes and placenta cytotrophoblasts revealed 53 commonly expressed genes and enrichment in biological processes integral to cellular function including Cellular Respiration (GO:0045333; FDR 5.05E−08), Ion Transport (GO:0006811; FDR 2.08E−02), and Oxidation–Reduction Process (GO:0055114; FDR 1.58E−07). Overall, our results identify specific genes and cellular pathways common between first trimester fetal heart and placenta cells which if disrupted may concurrently contribute to the developmental perturbations resulting in CHD.
Collapse
|
24
|
Linglart L, Bonnet D. Epigenetics and Congenital Heart Diseases. J Cardiovasc Dev Dis 2022; 9:185. [PMID: 35735814 PMCID: PMC9225036 DOI: 10.3390/jcdd9060185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/22/2022] Open
Abstract
Congenital heart disease (CHD) is a frequent occurrence, with a prevalence rate of almost 1% in the general population. However, the pathophysiology of the anomalous heart development is still unclear in most patients screened. A definitive genetic origin, be it single-point mutation or larger chromosomal disruptions, only explains about 35% of identified cases. The precisely choreographed embryology of the heart relies on timed activation of developmental molecular cascades, spatially and temporally regulated through epigenetic regulation: chromatin conformation, DNA priming through methylation patterns, and spatial accessibility to transcription factors. This multi-level regulatory network is eminently susceptible to outside disruption, resulting in faulty cardiac development. Similarly, the heart is unique in its dynamic development: growth is intrinsically related to mechanical stimulation, and disruption of the intrauterine environment will have a direct impact on fetal embryology. These two converging axes offer new areas of research to characterize the cardiac epigenetic regulation and identify points of fragility in order to counteract its teratogenic consequences.
Collapse
Affiliation(s)
- Léa Linglart
- M3C-Necker, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France;
| | - Damien Bonnet
- M3C-Necker, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France;
- School of Medicine, Université de Paris Cité, 75006 Paris, France
| |
Collapse
|
25
|
Deep sequencing unveils altered cardiac miRNome in congenital heart disease. Mol Genet Genomics 2022; 297:1123-1139. [PMID: 35668131 DOI: 10.1007/s00438-022-01908-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
Abstract
Congenital heart disease (CHD) surges from fetal cardiac dysmorphogenesis and chiefly contributes to perinatal morbidity and cardiovascular disease mortality. A continual rise in prevalence and prerequisite postoperative disease management creates need for better understanding and new strategies to control the disease. The interaction between genetic and non-genetic factors roots the multifactorial status of this disease, which remains incompletely explored. The small non-coding microRNAs (miRs, miRNAs) regulate several biological processes via post-transcriptional regulation of gene expression. Abnormal expression of miRs in developing and adult heart is associated with anomalous cardiac cell differentiation, cardiac dysfunction, and cardiovascular diseases. Here, we attempt to discover the changes in cardiac miRNA transcriptome in CHD patients over those without CHD (non-CHD) and find its role in CHD through functional annotation. This study explores the miRNome in three most commonly occurring CHD subtypes, namely atrial septal defect (ASD), ventricular septal defect (VSD), and tetralogy of fallot (TOF). We found 295 dysregulated miRNAs through high-throughput sequencing of the cardiac tissues. The bioinformatically predicted targets of these differentially expressed miRs were functionally annotated to know they were entailed in cell signal regulatory pathways, profoundly responsible for cell proliferation, survival, angiogenesis, migration and cell cycle regulation. Selective miRs (hsa-miR-221-3p, hsa-miR-218-5p, hsa-miR-873-5p) whose expression was validated by qRT-PCR, have been reported for cardiogenesis, cardiomyocyte proliferation, cardioprotection and cardiac dysfunction. These results indicate that the altered miRNome to be responsible for the disease status in CHD patients. Our data expand the existing knowledge on the epigenetic changes in CHD. In future, characterization of these cardiac-specific miRs will add huge potential to understand cardiac development, function, and molecular pathogenesis of heart diseases with a prospect of epigenetic manipulation for cardiac repair.
Collapse
|
26
|
Nana L, Lu L, Zhen L, Ying D, Meixian W, Zhao J, Zeng S, Hong K, Yanping W, Jun Z, Jianxin Z, Ping Y. The effect of maternal polycyclic aromatic hydrocarbons exposure and methylation levels of CHDs-candidate genes on the risk of congenital heart diseases. Prenat Diagn 2022; 42:1142-1154. [PMID: 35556253 DOI: 10.1002/pd.6167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/27/2022] [Accepted: 05/08/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To evaluate the impact of maternal exposure to polycyclic aromatic hydrocarbons (PAHs) and methylation levels of CHDs-candidate genes on the risk of congenital heart diseases (CHDs), and the effect of PAHs exposure on DNA methylation states. METHODS A case-control study involving 60 mother -fetus pairs was performed by measuring 1-OHPG concentration in maternal urine and methylation levels of 20 CHDs-candidate genes in cord bloods. Logistic regression models were applied to determine the effect of maternal PAHs exposure and fetal methylation levels on the risk of CHDs. Spearman correlation was performed to correlate PAHs exposure and methylation levels. RESULTS Maternal higher PAHs exposure was associated with the risk of CHDs (aOR = 3.245, 95% CI: 1.060, 9.937) or some subtypes. The methylation levels of 23 amplicons within 11 genes exhibited significant differences between CHDs and controls. Higher methylation of NKX2-5_M1 was associated with decreased risk of CHDs (aOR=0.182, 95% CI:0.034, 0.983). No significant correlations were found between 1-OHPG concentration and methylation levels of NKX2-5_M1. CONCLUSIONS Maternal PAHs exposure was linked with CHDs. Higher methylation of the upstream sequence of NKX2-5 promoter decreased the risk of CHDs. There was no correlation between maternal PAHs exposure and the methylation level of NKX2-5. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Li Nana
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Li Lu
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Liu Zhen
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Deng Ying
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Wang Meixian
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Jinju Zhao
- Department of Gynecology and Obstetrics, Xichang people's Hospital, Xichang, China
| | - Shengli Zeng
- Department of Gynecology and Obstetrics, Rongchang Maternal and Child care Hospital, Chongqing, China
| | - Kang Hong
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Wang Yanping
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Zhu Jun
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Zhao Jianxin
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Yu Ping
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Abstract
Embryonic heart development is an intricate process that mainly involves morphogens, transcription factors, and cardiac genes. The precise spatiotemporal expression of these genes during different developmental stages underlies normal heart development. Thus, mutation or aberrant expression of these genes may lead to congenital heart disease (CHD). However, evidence demonstrates that the mutation of genes accounts for only a small portion of CHD cases, whereas the aberrant expression regulated by epigenetic modification plays a predominant role in the pathogenesis of CHD. In this review, we provide essential knowledge on the aberrant epigenetic modification involved in the pathogenesis of CHD. Then, we discuss recent advances in the identification of novel epigenetic biomarkers. Last, we highlight the epigenetic roles in some adverse intrauterine environment‐related CHD, which may help the prevention, diagnosis, and treatment of these kinds of CHD.
Collapse
Affiliation(s)
- Guanglei Wang
- Department of Obstetrics, Gynecology, & Reproductive Sciences University of Maryland School of Medicine Baltimore MD
| | - Bingbing Wang
- Department of Obstetrics, Gynecology, & Reproductive Sciences University of Maryland School of Medicine Baltimore MD
| | - Peixin Yang
- Department of Obstetrics, Gynecology, & Reproductive Sciences University of Maryland School of Medicine Baltimore MD
- Department of Biochemistry & Molecular Biology University of Maryland School of Medicine Baltimore MD
| |
Collapse
|
28
|
Salman HE, Kamal RY, Hijazi ZM, Yalcin HC. Hemodynamic and Structural Comparison of Human Fetal Heart Development Between Normally Growing and Hypoplastic Left Heart Syndrome-Diagnosed Hearts. Front Physiol 2022; 13:856879. [PMID: 35399257 PMCID: PMC8984126 DOI: 10.3389/fphys.2022.856879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/02/2022] [Indexed: 11/20/2022] Open
Abstract
Congenital heart defects (CHDs) affect a wide range of societies with an incidence rate of 1.0–1.2%. These defects initiate at the early developmental stage and result in critical health disorders. Although genetic factors play a role in the formation of CHDs, the occurrence of cases in families with no history of CHDs suggests that mechanobiological forces may also play a role in the initiation and progression of CHDs. Hypoplastic left heart syndrome (HLHS) is a critical CHD, which is responsible for 25–40% of all prenatal cardiac deaths. The comparison of healthy and HLHS hearts helps in understanding the main hemodynamic differences related to HLHS. Echocardiography is the most common imaging modality utilized for fetal cardiac assessment. In this study, we utilized echocardiographic images to compare healthy and HLHS human fetal hearts for determining the differences in terms of heart chamber dimensions, valvular flow rates, and hemodynamics. The cross-sectional areas of chamber dimensions are determined from 2D b-mode ultrasound images. Valvular flow rates are measured via Doppler echocardiography, and hemodynamic quantifications are performed with the use of computational fluid dynamics (CFD) simulations. The obtained results indicate that cross-sectional areas of the left and right sides of the heart are similar for healthy fetuses during gestational development. The left side of HLHS heart is underdeveloped, and as a result, the hemodynamic parameters such as flow velocity, pressure, and wall shear stress (WSS) are significantly altered compared to those of healthy hearts.
Collapse
Affiliation(s)
- Huseyin Enes Salman
- Department of Mechanical Engineering, TOBB University of Economics and Technology, Ankara, Turkey
| | - Reema Yousef Kamal
- Pediatric Cardiology Division, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Ziyad M. Hijazi
- Sidra Heart Center, Sidra Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Huseyin Cagatay Yalcin
- Biomedical Research Center, Qatar University, Doha, Qatar
- *Correspondence: Huseyin Cagatay Yalcin,
| |
Collapse
|
29
|
DNA Methylation Levels of the TBX5 Gene Promoter Are Associated with Congenital Septal Defects in Mexican Paediatric Patients. BIOLOGY 2022; 11:biology11010096. [PMID: 35053095 PMCID: PMC8773106 DOI: 10.3390/biology11010096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022]
Abstract
The TBX5 gene regulates morphological changes during heart development, and it has been associated with epigenetic abnormalities observed in congenital heart defects (CHD). The aim of this research was to evaluate the association between DNA methylation levels of the TBX5 gene promoter and congenital septal defects. DNA methylation levels of six CpG sites in the TBX5 gene promoter were evaluated using pyrosequencing analysis in 35 patients with congenital septal defects and 48 controls. Average methylation levels were higher in individuals with congenital septal defects than in the controls (p < 0.004). In five CpG sites, we also found higher methylation levels in patients than in the controls (p < 0.05). High methylation levels were associated with congenital septal defects (OR = 3.91; 95% CI = 1.02–14.8; p = 0.045). The analysis of Receiver Operating Characteristic (ROC) showed that the methylation levels of the TBX5 gene could be used as a risk marker for congenital septal defects (AUC = 0.68, 95% CI = 0.56–0.80; p = 0.004). Finally, an analysis of environmental factors indicated that maternal infections increased the risk (OR = 2.90; 95% CI = 1.01–8.33; p = 0.048) of congenital septal defects. Our data suggest that a high DNA methylation of the TBX5 gene could be associated with congenital septal defects.
Collapse
|
30
|
Yang P, Yang Y, He X, Sun P, Zhang Y, Song X, Tian Y, Zong T, Ma J, Chen X, Lv Q, Yu T, Jiang Z. miR-153-3p Targets βII Spectrin to Regulate Formaldehyde-Induced Cardiomyocyte Apoptosis. Front Cardiovasc Med 2022; 8:764831. [PMID: 34977182 PMCID: PMC8714842 DOI: 10.3389/fcvm.2021.764831] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Formaldehyde (FA) is ubiquitous in the environment and can be transferred to the fetus through placental circulation, causing miscarriage and congenital heart disease (CHD). Studies have shown that βII spectrin is necessary for cardiomyocyte survival and differentiation, and its loss leads to heart development defects and cardiomyocyte apoptosis. Additionally, previous studies have demonstrated that miRNA is essential in heart development and remodeling. However, whether miRNA regulates FA-induced CHD and cardiomyocyte apoptosis remains unclear. Methods: Using commercially available rat embryonic cardiomyocytes and a rat model of fetal cardiomyocyte apoptosis. Real-time quantitative PCR (RT-qPCR) and Western blot were performed to examine the level of miR-153-3p, βII spectrin, caspase 7, cleaved caspase7, Bax, Bcl-2 expression in embryonic cardiomyocytes and a rat model of fetal cardiomyocyte apoptosis. Apoptotic cell populations were evaluated by flow cytometry and Tunel. Luciferase activity assay and RNA pull-down assay were used to detect the interaction between miR-153-3p and βII spectrin. Masson's trichrome staining detects the degree of tissue fibrosis. Fluorescence in situ hybridization (FISH) and Immunohistochemistry were used to detect the expression of miR-153-3p and βII spectrin in tissues. Results: Using commercially available rat embryonic cardiomyocytes and a rat model of fetal cardiomyocyte apoptosis, our studies indicate that miR-153-3p plays a regulatory role by directly targeting βII spectrin to promote cardiomyocyte apoptosis. miR-153-3p mainly regulates cardiomyocyte apoptosis by regulating the expression of caspase7, further elucidating the importance of apoptosis in heart development. Finally, the results with our animal model revealed that targeting the miR-153-3p/βII spectrin pathway effectively regulated FA-induced damage during heart development. Recovery experiments with miR-153-3p antagomir resulted in the reversal of FA-induced cardiomyocyte apoptosis and fetal cardiac fibrosis. Conclusion: This study investigated the molecular mechanism underpinning the role of βII spectrin in FA-induced CHD and the associated upstream miRNA pathway. The study findings suggest that miR-153-3p may provide a potential target for the clinical diagnosis and treatment of CHD.
Collapse
Affiliation(s)
- Panyu Yang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao, China
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pin Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Zhang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoxia Song
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yu Tian
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tingyu Zong
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianmin Ma
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaofei Chen
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qifeng Lv
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhirong Jiang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
31
|
Akiel M. The genetic architecture behind congenital heart disease: A review of genetic and epigenetic factors. JOURNAL OF NATURE AND SCIENCE OF MEDICINE 2022. [DOI: 10.4103/jnsm.jnsm_126_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
32
|
Environmental Alterations during Embryonic Development: Studying the Impact of Stressors on Pluripotent Stem Cell-Derived Cardiomyocytes. Genes (Basel) 2021; 12:genes12101564. [PMID: 34680959 PMCID: PMC8536136 DOI: 10.3390/genes12101564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022] Open
Abstract
Non-communicable diseases (NCDs) sauch as diabetes, obesity and cardiovascular diseases are rising rapidly in all countries world-wide. Environmental maternal factors (e.g., diet, oxidative stress, drugs and many others), maternal illnesses and other stressors can predispose the newborn to develop diseases during different stages of life. The connection between environmental factors and NCDs was formulated by David Barker and colleagues as the Developmental Origins of Health and Disease (DOHaD) hypothesis. In this review, we describe the DOHaD concept and the effects of several environmental stressors on the health of the progeny, providing both animal and human evidence. We focus on cardiovascular diseases which represent the leading cause of death worldwide. The purpose of this review is to discuss how in vitro studies with pluripotent stem cells (PSCs), such as embryonic and induced pluripotent stem cells (ESC, iPSC), can underpin the research on non-genetic heart conditions. The PSCs could provide a tool to recapitulate aspects of embryonic development “in a dish”, studying the effects of environmental exposure during cardiomyocyte (CM) differentiation and maturation, establishing a link to molecular mechanism and epigenetics.
Collapse
|
33
|
Balasubramanian R, Vuppalapati S, Avanthika C, Jhaveri S, Peddi NC, Ahmed S, Reddy A, Kaur J. Epidemiology, Genetics and Epigenetics of Congenital Heart Diseases in Twins. Cureus 2021; 13:e17253. [PMID: 34540478 PMCID: PMC8448266 DOI: 10.7759/cureus.17253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2021] [Indexed: 12/13/2022] Open
Abstract
Congenital heart defects (CHDs) refer to abnormalities in the heart function that arise at the fetal stages. It is the most common birth defect that affects 0.8% of all liveborn infants. There is an increase in the incidence of congenital heart disease in monochorionic twin gestation. A six-fold increase in CHDs exists among monochorionic twins especially in association with twin-twin transfusion syndrome (TTTS) compared to dichorionic twin pregnancy. In this review article, we discussed the epidemiology, the role of genetics like protein-coding genes, epigenetics, placenta, hemodynamics and environmental factors in the etiology of CHD in twins. We conducted a literature search in PubMed indexed journals using the medical terms "twin pregnancy" and "congenital heart defect" to provide an overview of the uptrend in CHD in twin pregnancies, primarily due to assisted reproductive technologies (ARTs) and multiple other factors. Both the heart and placenta are vascular and share a common development window; therefore, CHD can develop secondary to placental pathologies. Among environmental factors, the strongest association of maternal smoking with CHD has been seen. We studied the causative factors to suggest improvement in echocardiographic skills in case of abnormal findings in twin gestations to decrease the CHD-associated morbidity and mortality, as early diagnosis allows doctors to precisely determine the risk of CHD. Systemic ultrasound scanning with five transverse views is very effective in diagnosing fetal CHD in twin pregnancy. In the case of genetics, prenatal counseling allows the expectant to understand the full ramifications of possible events after the pregnancy. The pathological basis of malformations specific to conjoined twinning and twin reversed arterial perfusion sequence is addressed. Also, there is evidence that folate supplementation may be protective against CHD but more research is needed to clarify the mechanisms. We concluded from the literature that monochorionic twins are at high risk of CHD. Chorionicity seems to play a more vital role than zygosity. Even the type of heart defect in monochorial twin pregnancies was unique from single, dizygotic, or dichorionic twin pregnancies. We also emphasize improving echocardiographic skills of technicians in referring ART dichorionic twin fetuses with suspicious findings to fetal cardiologists and performing postnatal scans in the case of TTTS. To understand the role of the placenta, making use of newer technologies and examining the placenta both during pregnancy and beyond delivery will play a vital role in understanding the etiology. Even identifying early signals impacting the heart and placental vasculature and correcting them using advanced technology could downtrend the incidence in coming years. Increased maternal age as well as multiple pregnancies increasing the risk of CHD has also been implicated. For more clarity on the role of genetics, the cost of DNA sequencing needs to decrease. This will enable whole-genome sequencing in the future thus helping to discover the gene responsible for CHD ultimately proving beneficial for future generations. For environmental factors, we have to rely on observational studies to assess the risk to the unborn child. There is difficulty in studying natural factors due to the unreliability of exposure to contaminants like pesticides and air pollution.
Collapse
Affiliation(s)
| | - Sravya Vuppalapati
- Pediatrics, People's Education Society Institute of Medical Sciences and Research, Kuppam, IND
| | | | - Sharan Jhaveri
- Internal Medicine, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Ahmedabad, IND
| | - Nikhil Chowdary Peddi
- Pediatrics, People's Education Society Institute of Medical Sciences and Research, Kuppam, IND
| | - Sana Ahmed
- Internal Medicine, Smt. Kashibai Navale Medical College, Pune, IND
| | - Apeksha Reddy
- Pediatrics, People's Education Society Institute of Medical Sciences and Research, Kuppam, IND
| | | |
Collapse
|
34
|
Yamada D, Nakamura M, Takao T, Takihira S, Yoshida A, Kawai S, Miura A, Ming L, Yoshitomi H, Gozu M, Okamoto K, Hojo H, Kusaka N, Iwai R, Nakata E, Ozaki T, Toguchida J, Takarada T. Induction and expansion of human PRRX1 + limb-bud-like mesenchymal cells from pluripotent stem cells. Nat Biomed Eng 2021; 5:926-940. [PMID: 34373601 DOI: 10.1038/s41551-021-00778-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/01/2021] [Indexed: 02/05/2023]
Abstract
Current protocols for the differentiation of human pluripotent stem cells (hPSCs) into chondrocytes do not allow for the expansion of intermediate progenitors so as to prospectively assess their chondrogenic potential. Here we report a protocol that leverages PRRX1-tdTomato reporter hPSCs for the selective induction of expandable and ontogenetically defined PRRX1+ limb-bud-like mesenchymal cells under defined xeno-free conditions, and the prospective assessment of the cells' chondrogenic potential via the cell-surface markers CD90, CD140B and CD82. The cells, which proliferated stably and exhibited the potential to undergo chondrogenic differentiation, formed hyaline cartilaginous-like tissue commensurate to their PRRX1-expression levels. Moreover, we show that limb-bud-like mesenchymal cells derived from patient-derived induced hPSCs can be used to identify therapeutic candidates for type II collagenopathy and we developed a method to generate uniformly sized hyaline cartilaginous-like particles by plating the cells on culture dishes coated with spots of a zwitterionic polymer. PRRX1+ limb-bud-like mesenchymal cells could facilitate the mass production of chondrocytes and cartilaginous tissues for applications in drug screening and tissue engineering.
Collapse
Affiliation(s)
- Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Nakamura
- Precision Health, Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tomoka Takao
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shota Takihira
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Aki Yoshida
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shunsuke Kawai
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Akihiro Miura
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Lu Ming
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Yoshitomi
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mai Gozu
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kumi Okamoto
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyuki Kusaka
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama, Japan
| | - Ryosuke Iwai
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama, Japan
| | - Eiji Nakata
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Junya Toguchida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
35
|
Assessing chromatin condensation for epigenetics with a DNA-targeting sensor by FRET and FLIM techniques. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.02.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
Zhou J, Xiong Y, Dong X, Wang H, Qian Y, Ma D, Li X. Genome-wide methylation analysis reveals differentially methylated CpG sites and altered expression of heart development-associated genes in fetuses with cardiac defects. Exp Ther Med 2021; 22:1032. [PMID: 34373718 PMCID: PMC8343574 DOI: 10.3892/etm.2021.10464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 05/25/2021] [Indexed: 12/16/2022] Open
Abstract
DNA methylation, as an epigenetic mechanism, has a vital role in heart development. An increasing number of studies have investigated aberrant DNA methylation in pediatric or adult heart samples from patients with congenital heart defects (CHD). Placenta tissue, umbilical cord blood, or newborn blood have also been used to detect DNA methylation biomarkers for CHD. However, few studies have compared the methylation levels in fetal heart tissue with cardiac defects with that in normal controls. The present study conducted an integrative whole-genome and CpG site-specific DNA methylation analysis of fetal heart samples from 17 isolated cardiac defect cases, 14 non-isolated cardiac defect cases, and 22 controls with normal hearts, using methylated DNA immunoprecipitation microarray and MassARRAY EpiTYPER assays. Expression of genes adjacent to differentially methylated regions (DMRs) was measured by RT-qPCR and western blot analysis. The results revealed that fetuses with cardiac defects presented global hypomethylation. Genomic analysis of DMRs revealed that a proportion of DMRs were located in exons (12.4%), distal intergenic regions (11.14%), and introns (8.97%). Only 55.7% of DMRs were observed at promoter regions. Functional enrichment analysis for genes adjacent to these DMRs revealed that hypomethylated genes were involved in embryonic heart tube morphogenesis and immune-related regulation functions. Intergenic hypermethylation of EGFR and solute carrier family 19 member 1 (SLC19A1), and intragenic hypomethylation of NOTCH1 were validated in fetal heart tissues with cardiac defects. Only SLC19A1 expression was significantly decreased at the mRNA level, while EGFR, NOTCH1, and SLC19A1 expression were all significantly decreased at the protein level. In conclusion, the present study demonstrated that fetal cardiac defects may be associated with alterations in regional and single CpG site methylation outside of promoter regions, resulting in differentiated expression of corresponding genes associated with heart development. These results present new insights into the epigenetic mechanisms underlying abnormal heart development.
Collapse
Affiliation(s)
- Jizi Zhou
- Department of Prenatal Diagnosis and Fetal Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, P.R. China
| | - Yu Xiong
- Department of Prenatal Diagnosis and Fetal Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, P.R. China
| | - Xinran Dong
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Huijun Wang
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Yanyan Qian
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China.,Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Xiaotian Li
- Department of Prenatal Diagnosis and Fetal Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, P.R. China.,Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China.,The Shanghai Key Laboratory of Birth Defects, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
37
|
Gu Y, Li Q, Lin R, Jiang W, Wang X, Zhou G, Su J, Fan X, Gao P, Jin M, Wang Y, Du J. Prognostic Model to Predict Postoperative Adverse Events in Pediatric Patients With Aortic Coarctation. Front Cardiovasc Med 2021; 8:672627. [PMID: 34095260 PMCID: PMC8175771 DOI: 10.3389/fcvm.2021.672627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Postoperative adverse events remain excessively high in surgical patients with coarctation of aorta (CoA). Currently, there is no generally accepted strategy to predict these patients' individual outcomes. Objective: This study aimed to develop a risk model for the prediction of postoperative risk in pediatric patients with CoA. Methods: In total, 514 patients with CoA at two centers were enrolled. Using daily clinical practice data, we developed a model to predict 30-day or in-hospital adverse events after the operation. The least absolute shrinkage and selection operator approach was applied to select predictor variables and logistic regression was used to develop the model. Model performance was estimated using the receiver-operating characteristic curve, the Hosmer–Lemeshow test and the calibration plot. Net reclassification improvement (NRI) and integrated discrimination improvement (IDI) compared with existing risk strategies were assessed. Results: Postoperative adverse events occurred in 195 (37.9%) patients in the overall population. Nine predictive variables were identified, including incision of left thoracotomy, preoperative ventilation, concomitant ventricular septal defect, preoperative cardiac dysfunction, severe pulmonary hypertension, height, weight-for-age z-score, left ventricular ejection fraction and left ventricular posterior wall thickness. A multivariable logistic model [area under the curve = 0.8195 (95% CI: 0.7514–0.8876)] with adequate calibration was developed. Model performance was significantly improved compared with the existing Aristotle Basic Complexity (ABC) score (NRI = 47.3%, IDI = 11.5%) and the Risk Adjustment for Congenital Heart Surgery (RACHS-1) (NRI = 75.0%, IDI = 14.9%) in the validation set. Conclusion: Using daily clinical variables, we generated and validated an easy-to-apply postoperative risk model for patients with CoA. This model exhibited a remarkable improvement over the ABC score and the RACHS-1 method.
Collapse
Affiliation(s)
- Yan Gu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China.,Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.,Department of Pediatric Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Pediatric Heart Centre, Beijing, China
| | - Qianqian Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Rui Lin
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China.,Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Wenxi Jiang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China.,Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Xue Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China.,Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Gengxu Zhou
- Department of Pediatric Cardiology, Bayi Children's Hospital Affiliated to the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Junwu Su
- Department of Pediatric Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Pediatric Heart Centre, Beijing, China
| | - Xiangming Fan
- Department of Pediatric Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Pediatric Heart Centre, Beijing, China
| | - Pei Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.,Peking University Clinical Research Institute, Peking University Health Science Center, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Mei Jin
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China.,Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.,Department of Pediatric Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Pediatric Heart Centre, Beijing, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China.,Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China.,Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
38
|
A new era of genetic testing in congenital heart disease: A review. Trends Cardiovasc Med 2021; 32:311-319. [PMID: 33964404 DOI: 10.1016/j.tcm.2021.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/07/2021] [Accepted: 04/29/2021] [Indexed: 11/24/2022]
Abstract
Genetic and genomic testing in pediatric CHD is becoming increasingly routine, and can have important psychosocial, clinical and reproductive implications. In this paper we highlight important challenges and considerations when providing genetics consults and testing in pediatric CHD and illustrate the role of a dedicated CHD genetics clinic. Key lessons include that a) a genetic diagnosis can have clinical utility that justifies testing early in life, b) adequate genetic counselling is crucial to ensure families are supported, understand the range of possible results, and are prepared for new or unexpected health information, and c) further integration of the clinical genetics and cardiology workflows will be required to effectively manage the burgeoning information arising from genetic testing. Our experience demonstrates that a dedicated CHD genetics clinic is a valuable addition to a multidisciplinary team providing care to children with CHD.
Collapse
|
39
|
The role of DNA methylation in syndromic and non-syndromic congenital heart disease. Clin Epigenetics 2021; 13:93. [PMID: 33902696 PMCID: PMC8077695 DOI: 10.1186/s13148-021-01077-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Congenital heart disease (CHD) is a common structural birth defect worldwide, and defects typically occur in the walls and valves of the heart or enlarged blood vessels. Chromosomal abnormalities and genetic mutations only account for a small portion of the pathogenic mechanisms of CHD, and the etiology of most cases remains unknown. The role of epigenetics in various diseases, including CHD, has attracted increased attention. The contributions of DNA methylation, one of the most important epigenetic modifications, to CHD have not been illuminated. Increasing evidence suggests that aberrant DNA methylation is related to CHD. Here, we briefly introduce DNA methylation and CHD and then review the DNA methylation profiles during cardiac development and in CHD, abnormalities in maternal genome-wide DNA methylation patterns are also described. Whole genome methylation profile and important differentially methylated genes identified in recent years are summarized and clustered according to the sample type and methodologies. Finally, we discuss the novel technology for and prospects of CHD-related DNA methylation.
Collapse
|
40
|
Rufaihah AJ, Chen CK, Yap CH, Mattar CNZ. Mending a broken heart: In vitro, in vivo and in silico models of congenital heart disease. Dis Model Mech 2021; 14:dmm047522. [PMID: 33787508 PMCID: PMC8033415 DOI: 10.1242/dmm.047522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Birth defects contribute to ∼0.3% of global infant mortality in the first month of life, and congenital heart disease (CHD) is the most common birth defect among newborns worldwide. Despite the significant impact on human health, most treatments available for this heterogenous group of disorders are palliative at best. For this reason, the complex process of cardiogenesis, governed by multiple interlinked and dose-dependent pathways, is well investigated. Tissue, animal and, more recently, computerized models of the developing heart have facilitated important discoveries that are helping us to understand the genetic, epigenetic and mechanobiological contributors to CHD aetiology. In this Review, we discuss the strengths and limitations of different models of normal and abnormal cardiogenesis, ranging from single-cell systems and 3D cardiac organoids, to small and large animals and organ-level computational models. These investigative tools have revealed a diversity of pathogenic mechanisms that contribute to CHD, including genetic pathways, epigenetic regulators and shear wall stresses, paving the way for new strategies for screening and non-surgical treatment of CHD. As we discuss in this Review, one of the most-valuable advances in recent years has been the creation of highly personalized platforms with which to study individual diseases in clinically relevant settings.
Collapse
Affiliation(s)
- Abdul Jalil Rufaihah
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Ching Kit Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Choon Hwai Yap
- Division of Cardiology, Department of Paediatrics, Khoo Teck Puat -National University Children's Medical Institute, National University Health System, Singapore 119228
- Department of Bioengineering, Imperial College London, London, UK
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore 119228
| |
Collapse
|
41
|
Steinweg JK, Hui GTY, Pietsch M, Ho A, van Poppel MP, Lloyd D, Colford K, Simpson JM, Razavi R, Pushparajah K, Rutherford M, Hutter J. T2* placental MRI in pregnancies complicated with fetal congenital heart disease. Placenta 2021; 108:23-31. [PMID: 33798991 DOI: 10.1016/j.placenta.2021.02.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/05/2021] [Accepted: 02/25/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Congenital heart disease (CHD) is one of the most important and common group of congenital malformations in humans. Concurrent development and close functional links between the fetal heart and placenta emphasise the importance of understanding placental function and its influence in pregnancy outcomes. The aim of this study was to evaluate placental oxygenation by relaxometry (T2*) to assess differences in placental phenotype and function in CHD. METHODS In this prospective cross-sectional observational study, 69 women with a fetus affected with CHD and 37 controls, whole placental T2* was acquired using a 1.5-Tesla MRI scanner. Gaussian Process Regression was used to assess differences in placental phenotype in CHD cohorts compared to our controls. RESULTS Placental T2* maps demonstrated significant differences in CHD compared to controls at equivalent gestational age. Mean T2* values over the entire placental volume were lowest compared to predicted normal in right sided obstructive lesions (RSOL) (Z-Score 2.30). This cohort also showed highest lacunarity indices (Z-score -1.7), as a marker of lobule size. Distribution patterns of T2* values over the entire placental volume were positively skewed in RSOL (Z-score -4.69) and suspected, not confirmed coarctation of the aorta (CoA-) (Z-score -3.83). Deviations were also reflected in positive kurtosis in RSOL (Z-score -3.47) and CoA- (Z-score -2.86). CONCLUSION Placental structure and function appear to deviate from normal development in pregnancies with fetal CHD. Specific patterns of altered placental function assessed by T2* deliver crucial complementary information to antenatal assessments in the presence of fetal CHD.
Collapse
Affiliation(s)
- Johannes K Steinweg
- Department of Cardiovascular Imaging, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom.
| | - Grace Tin Yan Hui
- Centre for the Developing Brain, King's College London, London, United Kingdom
| | - Maximilian Pietsch
- Centre for the Developing Brain, King's College London, London, United Kingdom; Department of Biomedical Engineering, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom
| | - Alison Ho
- Centre for the Developing Brain, King's College London, London, United Kingdom
| | - Milou Pm van Poppel
- Department of Cardiovascular Imaging, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom
| | - David Lloyd
- Department of Cardiovascular Imaging, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom; Department of Congenital Heart Disease, Evelina Children's Hospital, London, United Kingdom
| | - Kathleen Colford
- Centre for the Developing Brain, King's College London, London, United Kingdom
| | - John M Simpson
- Department of Cardiovascular Imaging, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom; Department of Congenital Heart Disease, Evelina Children's Hospital, London, United Kingdom
| | - Reza Razavi
- Department of Cardiovascular Imaging, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom; Department of Congenital Heart Disease, Evelina Children's Hospital, London, United Kingdom
| | - Kuberan Pushparajah
- Department of Cardiovascular Imaging, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom; Department of Congenital Heart Disease, Evelina Children's Hospital, London, United Kingdom
| | - Mary Rutherford
- Centre for the Developing Brain, King's College London, London, United Kingdom
| | - Jana Hutter
- Centre for the Developing Brain, King's College London, London, United Kingdom; Department of Biomedical Engineering, School of Biomedical Engineering & Imaging Science, King's College London, London, United Kingdom
| |
Collapse
|
42
|
Abstract
Congenital heart disease is the most common congenital defect observed in newborns. Within the spectrum of congenital heart disease are left‐sided obstructive lesions (LSOLs), which include hypoplastic left heart syndrome, aortic stenosis, bicuspid aortic valve, coarctation of the aorta, and interrupted aortic arch. These defects can arise in isolation or as a component of a defined syndrome; however, nonsyndromic defects are often observed in multiple family members and associated with high sibling recurrence risk. This clear evidence for a heritable basis has driven a lengthy search for disease‐causing variants that has uncovered both rare and common variants in genes that, when perturbed in cardiac development, can result in LSOLs. Despite advancements in genetic sequencing platforms and broadening use of exome sequencing, the currently accepted LSOL‐associated genes explain only 10% to 20% of patients. Further, the combinatorial effects of common and rare variants as a cause of LSOLs are emerging. In this review, we highlight the genes and variants associated with the different LSOLs and discuss the strengths and weaknesses of the present genetic associations. Furthermore, we discuss the research avenues needed to bridge the gaps in our current understanding of the genetic basis of nonsyndromic congenital heart disease.
Collapse
Affiliation(s)
- Lauren E Parker
- Division of Cardiology Department of Pediatrics Duke University School of Medicine Durham NC
| | - Andrew P Landstrom
- Division of Cardiology Department of Pediatrics Duke University School of Medicine Durham NC.,Department of Cell Biology Duke University School of Medicine Durham NC
| |
Collapse
|
43
|
Mousavi A, Vahdat S, Baheiraei N, Razavi M, Norahan MH, Baharvand H. Multifunctional Conductive Biomaterials as Promising Platforms for Cardiac Tissue Engineering. ACS Biomater Sci Eng 2020; 7:55-82. [PMID: 33320525 DOI: 10.1021/acsbiomaterials.0c01422] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Adult cardiomyocytes are terminally differentiated cells that result in minimal intrinsic potential for the heart to self-regenerate. The introduction of novel approaches in cardiac tissue engineering aims to repair damages from cardiovascular diseases. Recently, conductive biomaterials such as carbon- and gold-based nanomaterials, conductive polymers, and ceramics that have outstanding electrical conductivity, acceptable mechanical properties, and promoted cell-cell signaling transduction have attracted attention for use in cardiac tissue engineering. Nevertheless, comprehensive classification of conductive biomaterials from the perspective of cardiac cell function is a subject for discussion. In the present review, we classify and summarize the unique properties of conductive biomaterials considered beneficial for cardiac tissue engineering. We attempt to cover recent advances in conductive biomaterials with a particular focus on their effects on cardiac cell functions and proposed mechanisms of action. Finally, current problems, limitations, challenges, and suggested solutions for applications of these biomaterials are presented.
Collapse
Affiliation(s)
- Ali Mousavi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Sadaf Vahdat
- Tissue Engineering and Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, 14117-13116 Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, 1665659911 Tehran, Iran
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, 14117-13116 Tehran, Iran
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida 32816, United States
| | - Mohammad Hadi Norahan
- Centro de Biotecnología-FEMSA, Department of Sciences, Tecnologico de Monterrey, Monterrey 64849, NL, México
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, 1665659911 Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
44
|
Abstract
Congenital heart disease (CHD) is one of the most common combined malformations of microtia. There is currently no specific study that investigates the relationship between microtia and CHD. METHODS This study collected microtia inpatients admitted from May 1, 2015 to July 31, 2016. The diagnosis of CHD was based on patient's symptoms, past history, and echocardiography. Pearson χ test was used to analyze the correlation between CHD and microtia. RESULTS A total of 30 cases (3.35%) were documented with CHD, including atrial septal defect (12/40.00%), ventricular septal defect (7/23.30%), patent ductus arteriosus (2/6.70%), complex congenital heart disease (3/10.00%), combined CHD (2/6.70%) and other malformations (4/13.30%). Analysis showed no statistically significant relation between CHD and the side of affected ear or gender. CONCLUSIONS The occurrence of CHD in microtia patients was higher than that in the general population. The relationship between them was explored mainly from the etiological perspective. Microtia and CHD were often combined in syndromes such as Goldenhar syndrome, 22q11 deletion syndrome, and CHARGE syndrome. Absence of genes or abnormal embryo development associated with these syndromes leads to the occurrence of both.
Collapse
|
45
|
Joshi RO, Chellappan S, Kukshal P. Exploring the Role of Maternal Nutritional Epigenetics in Congenital Heart Disease. Curr Dev Nutr 2020; 4:nzaa166. [PMID: 33294766 PMCID: PMC7703391 DOI: 10.1093/cdn/nzaa166] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022] Open
Abstract
Congenital heart disease (CHD) is one of the major debilitating birth defects resulting in significant impact on neonatal and child mortality globally. The etiology of CHD is complex and multifactorial. Many causative genes responsible for CHDs have been identified from the familial forms previously. Still, the non-Mendelian inheritance and predominant sporadic cases have stimulated research to understand the epigenetic basis and environmental impact on the incidence of CHD. The fetal epigenetic programming affecting cardiac development is susceptible to the availability of key dietary factors during the crucial periconceptional period. This article highlights the need and importance of in-depth research in the new emerging area of maternal nutritional epigenetics and CHD. It summarizes the current research and underlines the limitations in these types of studies. This review will benefit the future research on nutrition as a modifiable environmental factor to decrease the incidence of CHD.
Collapse
Affiliation(s)
- Radha O Joshi
- Department of Genomics Research, Sri Sathya Sai Sanjeevani Research Foundation, Palwal, Haryana, India
| | - Subramanian Chellappan
- Department of Anesthesia, Sri Sathya Sai Sanjeevani International Centre for Child Heart Care and Research, Palwal, Haryana, India
| | - Prachi Kukshal
- Department of Genomics Research, Sri Sathya Sai Sanjeevani Research Foundation, Palwal, Haryana, India
| |
Collapse
|
46
|
Jarrell DK, Vanderslice EJ, VeDepo MC, Jacot JG. Engineering Myocardium for Heart Regeneration-Advancements, Considerations, and Future Directions. Front Cardiovasc Med 2020; 7:586261. [PMID: 33195474 PMCID: PMC7588355 DOI: 10.3389/fcvm.2020.586261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 12/28/2022] Open
Abstract
Heart disease is the leading cause of death in the United States among both adults and infants. In adults, 5-year survival after a heart attack is <60%, and congenital heart defects are the top killer of liveborn infants. Problematically, the regenerative capacity of the heart is extremely limited, even in newborns. Furthermore, suitable donor hearts for transplant cannot meet the demand and require recipients to use immunosuppressants for life. Tissue engineered myocardium has the potential to replace dead or fibrotic heart tissue in adults and could also be used to permanently repair congenital heart defects in infants. In addition, engineering functional myocardium could facilitate the development of a whole bioartificial heart. Here, we review and compare in vitro and in situ myocardial tissue engineering strategies. In the context of this comparison, we consider three challenges that must be addressed in the engineering of myocardial tissue: recapitulation of myocardial architecture, vascularization of the tissue, and modulation of the immune system. In addition to reviewing and analyzing current progress, we recommend specific strategies for the generation of tissue engineered myocardial patches for heart regeneration and repair.
Collapse
Affiliation(s)
- Dillon K Jarrell
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ethan J Vanderslice
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mitchell C VeDepo
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jeffrey G Jacot
- Jacot Laboratory for Pediatric Regenerative Medicine, Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
47
|
Kolomenski JE, Delea M, Simonetti L, Fabbro MC, Espeche LD, Taboas M, Nadra AD, Bruque CD, Dain L. An update on genetic variants of the NKX2-5. Hum Mutat 2020; 41:1187-1208. [PMID: 32369864 DOI: 10.1002/humu.24030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 04/03/2020] [Accepted: 04/26/2020] [Indexed: 12/13/2022]
Abstract
NKX2-5 is a homeodomain transcription factor that plays a crucial role in heart development. It is the first gene where a single genetic variant (GV) was found to be associated with congenital heart diseases in humans. In this study, we carried out a comprehensive survey of NKX2-5 GVs to build a unified, curated, and updated compilation of all available GVs. We retrieved a total of 1,380 unique GVs. From these, 970 had information on their frequency in the general population and 143 have been linked to pathogenic phenotypes in humans. In vitro effect was ascertained for 38 GVs. The homeodomain had the biggest cluster of pathogenic variants in the protein: 49 GVs in 60 residues, 23 in its third α-helix, where 11 missense variants may affect protein-DNA interaction or the hydrophobic core. We also pinpointed the likely location of pathogenic GVs in four linear motifs. These analyses allowed us to assign a putative explanation for the effect of 90 GVs. This study pointed to reliable pathogenicity for GVs in helix 3 of the homeodomain and may broaden the scope of functional and structural studies that can be done to better understand the effect of GVs in NKX2-5 function.
Collapse
Affiliation(s)
- Jorge E Kolomenski
- Departamento de Química Biológica Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET, Buenos Aires, Argentina.,Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología Traslacional, iB3, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisol Delea
- Centro Nacional de Genética Médica, ANLIS, Buenos Aires, Argentina
| | - Leandro Simonetti
- Department of Chemistry-Biomedical Centre, Uppsala University, Uppsala, Sweden
| | | | - Lucía D Espeche
- Centro Nacional de Genética Médica, ANLIS, Buenos Aires, Argentina
| | - Melisa Taboas
- Centro Nacional de Genética Médica, ANLIS, Buenos Aires, Argentina
| | - Alejandro D Nadra
- Departamento de Química Biológica Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET, Buenos Aires, Argentina.,Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología Traslacional, iB3, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos D Bruque
- Centro Nacional de Genética Médica, ANLIS, Buenos Aires, Argentina.,Instituto de Biología y Medicina Experimental, (IBYME-CONICET), Buenos Aires, Argentina
| | - Liliana Dain
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología Traslacional, iB3, Universidad de Buenos Aires, Buenos Aires, Argentina.,Centro Nacional de Genética Médica, ANLIS, Buenos Aires, Argentina.,Instituto de Biología y Medicina Experimental, (IBYME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
48
|
Kosugi M, Otani M, Kikkawa Y, Itakura Y, Sakai K, Ito T, Toyoda M, Sekita Y, Kimura T. Mutations of histone demethylase genes encoded by X and Y chromosomes, Kdm5c and Kdm5d, lead to noncompaction cardiomyopathy in mice. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30311-9. [PMID: 32081420 DOI: 10.1016/j.bbrc.2020.02.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/07/2020] [Indexed: 12/11/2022]
Abstract
Mammalian X and Y chromosomes evolved from a pair of autosomes. Although most ancestral genes have been lost from the Y chromosome, a small number of ancestral X-Y gene pairs are still present on the sex chromosomes. The KDM5C and KDM5D genes, which encode H3K4 histone demethylases, are a surviving ancestral gene pair located on the X and Y chromosomes, respectively. Mutations in KDM5C cause X-linked intellectual disability in human males, suggesting functional divergence between KDM5C and KDM5D in the nervous system. In this study, to explore the functional conservation and divergence between these two genes in other organs, we generated female mice lacking Kdm5c (homozygous X5c- X5c- females) and male mice lacking both Kdm5c and Kdm5d (compound hemizygous X5c- Y5d- males). Both X5c- X5c- females and X5c- Y5d- males showed lower body weights and postnatal lethality. Histological examination of the hearts showed prominent trabecular extension and a thin layer of compacted myocardium in the left and right ventricles, indicating noncompaction cardiomyopathy. However, hemizygous males lacking either Kdm5c or Kdm5d showed no signs of noncompaction cardiomyopathy. These results clearly demonstrate that the function of Kdm5c and Kdm5d in heart development is conserved.
Collapse
Affiliation(s)
- Mayuko Kosugi
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Mai Otani
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Yurika Kikkawa
- Research Team for Geriatric Medicine (Vascular Medicine), Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yoko Itakura
- Research Team for Geriatric Medicine (Vascular Medicine), Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kohei Sakai
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Toshiaki Ito
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Masashi Toyoda
- Research Team for Geriatric Medicine (Vascular Medicine), Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yoichi Sekita
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Tohru Kimura
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan.
| |
Collapse
|