1
|
Dolfini D, Imbriano C, Mantovani R. The role(s) of NF-Y in development and differentiation. Cell Death Differ 2025; 32:195-206. [PMID: 39327506 DOI: 10.1038/s41418-024-01388-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
NF-Y is a conserved sequence-specific trimeric Transcription Factor -TF- binding to the CCAAT element. We review here the role(s) in development, from pre-implantation embryo to terminally differentiated tissues, by rationalizing and commenting on genetic, genomic, epigenetic and biochemical studies. This effort brings to light the impact of NF-YA isoforms on stemness and differentiation, as well as binding to distal vs promoter proximal sites and connections with selected TFs.
Collapse
Affiliation(s)
- Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Carol Imbriano
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, Modena, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
2
|
Hirono K, Hata Y, Imamura T, Tsuboi K, Takarada S, Okabe M, Nakaoka H, Ibuki K, Ozawa S, Ichimata S, Nishida N, Iwasaki H, Urata S, Okada S, Hiratsuji T, Sakaguchi H, Takigiku K, Nakazawa M, Nishihara E, Harada M, Matsuo O, Yasuda K, Yoshida Y, Namiki H, Yasuda K, Ifuku T, Urayama K, Oka H, Ogino K, Kato A, Kan N, Seki S, Seki M, Odanaka Y, Iwashima S, Yoshida S, Miyata T, Miyamoto T, Watanabe K, Kuwabara N, Inuzuka R, Takahashi Y, Sakazaki H, Muneuchi J, Kogaki S, Numano F, Kido S, Nii M, Hoshino S, Ishida H, Maeda J, Hayabuchi Y, Otsubo Y, Ikeda K, Tsukano S, Watanabe M, Momoi N, Fujii T, Fujioka T, Fujino M, Uchiyama H, Baba S, Horigome H, Honda T, Suzuki K, Ichida F. Determination of Genotype and Phenotypes in Pediatric Patients With Biventricular Noncompaction. J Am Heart Assoc 2024; 13:e035614. [PMID: 39494597 DOI: 10.1161/jaha.124.035614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Left ventricular noncompaction (LVNC) is a hereditary type of cardiomyopathy characterized by prominent trabeculations. Detailed characteristics of biventricular noncompaction (BiVNC) remain unknown. This study aimed to elucidate the clinical characteristics and genetic landscape of BiVNC. METHODS AND RESULTS We recruited children with left ventricular noncompaction from Japanese multi-institutional centers from 2013 to 2021. Left ventricular noncompaction was classified as BiVNC, congenital heart disease, arrhythmia, dilated cardiomyopathy, or normal function. In these patients, cardiomyopathy-associated genes were screened. A total of 234 patients (127 male; mean age, 4 months [range, 0-6.6 years]) were enrolled in this study, of whom 25 had BiVNC; 55, normal function; 84, dilated cardiomyopathy; 38, congenital heart disease; and 32, arrhythmia. BiVNC was diagnosed during the perinatal period in 10 patients, in whom the prevalence was higher than that in other patients. A total of 14 patients in the group with BiVNC had congenital heart disease, but not necessarily right heart lesions. Left ventricular dyskinesis was frequently observed in the lateral wall (24%) and apex (28%). Eleven pathogenic variants were found in 11 patients with BiVNC (44.0%). The group with BiVNC had a higher ratio of mitochondrial and developmental gene variants than the other groups. Among all groups, the group with BiVNC had the worst survival rate (P=0.0009). CONCLUSIONS Pediatric patients with BiVNC had a high rate of ventricular dyskinesis and poor outcome. A comprehensive and careful screening for disease-causing genes and phenotype may help identify specific patients with left ventricular noncompaction and mortality-related cardiac phenotypes.
Collapse
Affiliation(s)
- Keiichi Hirono
- Department of Pediatrics, Faculty of Medicine University of Toyama Japan
| | - Yukiko Hata
- Legal Medicine, Faculty of Medicine University of Toyama Japan
| | - Teruhiko Imamura
- 2nd Department of Internal Medicine, Faculty of Medicine University of Toyama Japan
| | - Kaori Tsuboi
- Department of Pediatrics, Faculty of Medicine University of Toyama Japan
| | - Shinya Takarada
- Department of Pediatrics, Faculty of Medicine University of Toyama Japan
| | - Mako Okabe
- Department of Pediatrics, Faculty of Medicine University of Toyama Japan
| | - Hideyuki Nakaoka
- Department of Pediatrics, Faculty of Medicine University of Toyama Japan
| | - Keijiro Ibuki
- Department of Pediatrics, Faculty of Medicine University of Toyama Japan
| | - Sayaka Ozawa
- Department of Pediatrics, Faculty of Medicine University of Toyama Japan
| | | | - Naoki Nishida
- Legal Medicine, Faculty of Medicine University of Toyama Japan
| | - Hidenori Iwasaki
- Department of Pediatrics Kanazawa University Hospital Ishikawa Japan
| | - Susumu Urata
- Division of Cardiology National Center for Child Health and Development Tokyo Japan
| | - Seigo Okada
- Department of Pediatrics Yamaguchi University Graduate School of Medicine Yamaguchi Japan
| | - Tomoya Hiratsuji
- Department of Pediatrics Okinawa Prefectural Hokubu Hospital Okinawa Japan
| | - Heima Sakaguchi
- Department of Pediatric Cardiology National Cerebral and Cardiovascular Center Osaka Japan
| | - Kiyohiro Takigiku
- Department of Pediatric Cardiology Nagano Children's Hospital Nagano Japan
| | - Makoto Nakazawa
- Department of Pediatrics Southern Tohoku Research Institute for Neuroscience Fukushima Japan
| | - Eiki Nishihara
- Department of Pediatric Cardiology and Neonatology Ogaki Municipal Hospital Gifu Japan
| | - Masako Harada
- Department of Pediatrics Miyazaki University Hospital Miyazaki Japan
| | - Osamu Matsuo
- Department of Pediatrics Kumamoto University Kumamoto Japan
| | - Kenji Yasuda
- Department of Pediatrics Shimane University Hospital Shimane Japan
| | - Yoko Yoshida
- Decision of Pediatric Electrophysiology Osaka City General Hospital Osaka Japan
| | - Hidemasa Namiki
- Department of Pediatrics and Child Health Nihon University School of Medicine & Itabashi Hospital Tokyo Japan
| | - Kazushi Yasuda
- Department of Pediatric Cardiology Aichi Children's Health and Medical Center Aichi Japan
| | - Toshinobu Ifuku
- Department of Pediatrics Miyazaki Prefectural Miyazaki Hospital Miyazaki Japan
| | - Kotaro Urayama
- Department of Pediatrics Tsuchiya General Hospital Hiroshima Japan
| | - Hideharu Oka
- Department of Pediatrics Asahikawa Medical University Hospital Asahikawa Hokkaido Japan
| | - Kayo Ogino
- Department of Pediatrics Kurashiki Central Hospital Okayama Japan
| | - Akio Kato
- Department of Pediatric Cardiology Okinawa Prefectural Nanbu Medical Center and Children's Medical Center Okinawa Japan
| | - Nobuhiko Kan
- Department of Fetal and Neonatal Cardiology Fukuoka Children's Hospital Fukuoka Japan
| | - Shunji Seki
- Department of Pediatrics Ibusuki Medical Center Kagoshima Japan
| | - Mitsuru Seki
- Department of Pediatrics Jichi Medical University Tochigi Japan
| | - Yutaka Odanaka
- Department of Pediatrics Osaka Medical and Pharmaceutical University Osaka Japan
| | - Satoru Iwashima
- Department of Pediatrics Chutoen General Medical Center Shizuoka Japan
| | - Shuichiro Yoshida
- Department of Pediatrics Cardiology, Chukyo Hospital Japan Community Healthcare Organization Aichi Japan
| | - Toyohisa Miyata
- Department of Pediatrics Ehime University Hospital Ehime Japan
| | | | - Ken Watanabe
- Department of Pediatrics Kitano Hospital Tazuke Kofukai Medical Research Institute Osaka Japan
| | - Naoki Kuwabara
- Department of Pediatric Cardiology Gifu Prefectural General Medical Center Gifu Japan
| | - Ryo Inuzuka
- Department of Pediatrics Tokyo University Hospital Tokyo Japan
| | | | - Hisanori Sakazaki
- Department of Pediatric Cardiology Hyogo Prefectural Amagasaki General Medical Center Hyogo Japan
| | - Jun Muneuchi
- Department of Pediatrics, Kyushu Hospital Japan Community Healthcare Organization Fukuoka Japan
| | - Shigetoyo Kogaki
- Department of Pediatrics and Neonatology Osaka General Medical Center Osaka Japan
| | - Fujito Numano
- Department of Pediatrics Niigata University Medical and Dental Hospital Niigata Japan
| | - Sachiko Kido
- Department of Cardiology Hyogo Prefectural Children's Hospital Hyogo Japan
| | - Masaki Nii
- Department of Cardiology Shizuoka Children's Hospital Shizuoka Japan
| | - Shinsuke Hoshino
- Department of Pediatrics Shiga University of Medical Science Otsu Shiga Japan
| | - Hidekazu Ishida
- Department of Pediatrics Osaka University Graduate School of Medicine Osaka Japan
| | - Jun Maeda
- Division of Cardiology Tokyo Metropolitan Children's Medical Center Tokyo Japan
| | | | - Yoshikazu Otsubo
- Department of Pediatrics Sasebo City General Hospital Nagasaki Japan
| | - Kazuyuki Ikeda
- Department of Pediatrics Kyoto Prefectural University of Medicine Kyoto Japan
| | - Shinya Tsukano
- Department of Pediatrics Niigata City General Hospital Niigata Japan
| | - Makoto Watanabe
- Department of Pediatrics Nippon Medical School Hospital Tokyo Japan
| | - Nobuo Momoi
- Department of Pediatrics Fukushima Medical University Hospital Fukushima Japan
| | - Takanari Fujii
- Pediatric Heart Disease and Adult Congenital Heart Disease Center Showa University Hospital Tokyo Japan
| | - Tao Fujioka
- Department of Pediatrics Japanese Red Cross Medical Center Tokyo Japan
| | - Mitsuhiro Fujino
- Department of Pediatric Cardiology Osaka City General Hospital Osaka Japan
| | - Hiroki Uchiyama
- Department of Pediatrics Hamamatsu University Hospital Shizuoka Japan
| | - Shigehito Baba
- Department of Pediatrics Niigata University Medical and Dental Hospital Niigata Japan
| | - Hitoshi Horigome
- Department of Pediatrics University of Tsukuba Hospital Ibaraki Japan
| | - Takashi Honda
- Department of Pediatrics Kitasato University School of Medicine Kanagawa Japan
| | - Kazutaka Suzuki
- Department of Pediatrics Nagoya City University Hospital Aichi Japan
| | - Fukiko Ichida
- Department of Pediatrics International University of Health and Welfare Tokyo Japan
| |
Collapse
|
3
|
Ma Q, Zhang YH, Guo W, Feng K, Huang T, Cai YD. Machine Learning in Identifying Marker Genes for Congenital Heart Diseases of Different Cardiac Cell Types. Life (Basel) 2024; 14:1032. [PMID: 39202774 PMCID: PMC11355424 DOI: 10.3390/life14081032] [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: 05/13/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Congenital heart disease (CHD) represents a spectrum of inborn heart defects influenced by genetic and environmental factors. This study advances the field by analyzing gene expression profiles in 21,034 cardiac fibroblasts, 73,296 cardiomyocytes, and 35,673 endothelial cells, utilizing single-cell level analysis and machine learning techniques. Six CHD conditions: dilated cardiomyopathy (DCM), donor hearts (used as healthy controls), hypertrophic cardiomyopathy (HCM), heart failure with hypoplastic left heart syndrome (HF_HLHS), Neonatal Hypoplastic Left Heart Syndrome (Neo_HLHS), and Tetralogy of Fallot (TOF), were investigated for each cardiac cell type. Each cell sample was represented by 29,266 gene features. These features were first analyzed by six feature-ranking algorithms, resulting in several feature lists. Then, these lists were fed into incremental feature selection, containing two classification algorithms, to extract essential gene features and classification rules and build efficient classifiers. The identified essential genes can be potential CHD markers in different cardiac cell types. For instance, the LASSO identified key genes specific to various heart cell types in CHD subtypes. FOXO3 was found to be up-regulated in cardiac fibroblasts for both Dilated and hypertrophic cardiomyopathy. In cardiomyocytes, distinct genes such as TMTC1, ART3, ARHGAP24, SHROOM3, and XIST were linked to dilated cardiomyopathy, Neo-Hypoplastic Left Heart Syndrome, hypertrophic cardiomyopathy, HF-Hypoplastic Left Heart Syndrome, and Tetralogy of Fallot, respectively. Endothelial cell analysis further revealed COL25A1, NFIB, and KLF7 as significant genes for dilated cardiomyopathy, hypertrophic cardiomyopathy, and Tetralogy of Fallot. LightGBM, Catboost, MCFS, RF, and XGBoost further delineated key genes for specific CHD subtypes, demonstrating the efficacy of machine learning in identifying CHD-specific genes. Additionally, this study developed quantitative rules for representing the gene expression patterns related to CHDs. This research underscores the potential of machine learning in unraveling the molecular complexities of CHD and establishes a foundation for future mechanism-based studies.
Collapse
Affiliation(s)
- Qinglan Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| | - Yu-Hang Zhang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China;
| | - Kaiyan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China;
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China;
| |
Collapse
|
4
|
Sessa F, Chisari M, Salerno M, Esposito M, Zuccarello P, Capasso E, Scoto E, Cocimano G. Congenital heart diseases (CHDs) and forensic investigations: Searching for the cause of death. Exp Mol Pathol 2024; 137:104907. [PMID: 38820762 DOI: 10.1016/j.yexmp.2024.104907] [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: 04/16/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024]
Abstract
Congenital Heart Diseases (CHDs) are a group of structural abnormalities or defects of the heart that are present at birth. CHDs could be connected to sudden death (SD), defined by the WHO (World Health Organization) as "death occurring within 24 h after the onset of the symptoms" in an apparently "healthy" subject. These conditions can range from relatively mild defects to severe, life-threatening anomalies. The prevalence of CHDs varies across populations, but they affect millions of individuals worldwide. This article aims to discuss the post-mortem investigation of death related to CHDs, exploring the forensic approach, current methodologies, challenges, and potential advancements in this challenging field. A further goal of this article is to provide a guide for understanding these complex diseases, highlighting the pivotal role of autopsy, histopathology, and genetic investigations in defining the cause of death, and providing evidence about the translational use of autopsy reports. Forensic investigations play a crucial role in understanding the complexities of CHDs and determining the cause of death accurately. Through collaboration between medical professionals and forensic experts, meticulous examinations, and analysis of evidence, valuable insights can be gained. These insights not only provide closure to the families affected but also contribute to the prevention of future tragedies.
Collapse
Affiliation(s)
- Francesco Sessa
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, 95121 Catania, Italy.
| | - Mario Chisari
- "Rodolico-San Marco" Hospital, Santa Sofia Street, 87, Catania 95121, Italy.
| | - Monica Salerno
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, 95121 Catania, Italy.
| | | | - Pietro Zuccarello
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, 95121 Catania, Italy.
| | - Emanuele Capasso
- Department of Advanced Biomedical Science-Legal Medicine Section, University of Naples "Federico II", 80131 Naples, Italy.
| | - Edmondo Scoto
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, 95121 Catania, Italy
| | - Giuseppe Cocimano
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Vanvitelli", 80121 Napoli, Italy.
| |
Collapse
|
5
|
Wang Z, Liu XY, Yang CX, Zhou HM, Li YJ, Qiu XB, Huang RT, Cen SS, Wang Y, Xu YJ, Qiu HY, Yang YQ. Discovery and functional investigation of BMP4 as a new causative gene for human congenital heart disease. Am J Transl Res 2024; 16:2034-2048. [PMID: 38883374 PMCID: PMC11170606 DOI: 10.62347/dgcd4269] [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: 02/06/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024]
Abstract
OBJECTIVE Aggregating evidence highlights the strong genetic basis underpinning congenital heart disease (CHD). Here BMP4 was chosen as a prime candidate gene causative of human CHD predominantly because BMP4 was amply expressed in the embryonic hearts and knockout of Bmp4 in mice led to embryonic demise mainly from multiple cardiovascular developmental malformations. The aim of this retrospective investigation was to discover a novel BMP4 mutation underlying human CHD and explore its functional impact. METHODS A sequencing examination of BMP4 was implemented in 212 index patients suffering from CHD and 236 unrelated non-CHD individuals as well as the family members available from the proband carrying a discovered BMP4 mutation. The impacts of the discovered CHD-causing mutation on the expression of NKX2-5 and TBX20 induced by BMP4 were measured by employing a dual-luciferase analysis system. RESULTS A new heterozygous BMP4 mutation, NM_001202.6:c.318T>G;p.(Tyr106*), was found in a female proband affected with familial CHD. Genetic research of the mutation carrier's relatives unveiled that the truncating mutation was in co-segregation with CHD in the pedigree. The nonsense mutation was absent from 236 unrelated non-CHD control persons. Quantitative biologic measurement revealed that Tyr106*-mutant BMP4 failed to induce the expression of NKX2-5 and TBX20, two genes whose expression is lost in CHD. CONCLUSION The current findings indicate BMP4 as a new gene predisposing to human CHD, allowing for improved prenatal genetic counseling along with personalized treatment of CHD patients.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Cardiovascular Medicine, Women and Children's Hospital of Ningbo University Ningbo 315012, Zhejiang, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine Shanghai 200065, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University Shanghai 200240, China
| | - Hui-Min Zhou
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine Shanghai 200065, China
| | - Yan-Jie Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine Shanghai 200030, China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine Shanghai 200030, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine Shanghai 200127, China
| | - Shu-Shu Cen
- Health Science Center, Ningbo University Ningbo 315211, Zhejiang, China
| | - Yuan Wang
- Health Science Center, Ningbo University Ningbo 315211, Zhejiang, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University Shanghai 200240, China
| | - Hai-Yan Qiu
- Department of Cardiovascular Medicine, Women and Children's Hospital of Ningbo University Ningbo 315012, Zhejiang, China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University Shanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University Shanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People's Hospital, Fudan University Shanghai 200240, China
| |
Collapse
|
6
|
Gorini F, Coi A, Pierini A, Assanta N, Bottoni A, Santoro M. Hypothyroidism in Patients with Down Syndrome: Prevalence and Association with Congenital Heart Defects. CHILDREN (BASEL, SWITZERLAND) 2024; 11:513. [PMID: 38790508 PMCID: PMC11119539 DOI: 10.3390/children11050513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
This population-based study aimed to assess the prevalence of congenital hypothyroidism (CH) and overt hypothyroidism (OH) and their association with congenital heart defects (CHDs) in patients with Down syndrome (DS). The population included all live births residing in Tuscany (Italy) diagnosed with DS recorded in the Registry of Congenital Defects and in the Registry of Rare Diseases of Tuscany in the years 2003-2017. The prevalence of CH and OH in DS patients was calculated by sex and by period. The association of CH and OH with CHDs in DS patients was assessed using multivariate logistic regression. The cohort included 228 subjects. The prevalence of CH and OH was 11.4% (95%CI: 7.4-16.7%) and 12.7% (95%CI: 8.5-12.3%), respectively, with no significant difference by sex. A significant increase in the prevalence of CH (p < 0.0001) was found in the years 2010-2017 compared to the previous period, and among preterm infants (p = 0.009). The presence of CH was associated with a higher prevalence of CHDs (adjusted OR = 2.24, p = 0.082). A significant association between ventricular septal defects (VSDs) and the occurrence of OH (adjusted OR = 3.07, p = 0.025) was also observed. This study confirmed the higher prevalence of both CH and OH in DS compared to the general population. Furthermore, the risk of association between DS and CHDs was higher in the presence of CH, while VSDs are associated with OH, providing relevant insights into the epidemiology of hypothyroidism in DS and associated anomalies.
Collapse
Affiliation(s)
- Francesca Gorini
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Alessio Coi
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Anna Pierini
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
- Foundation Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Nadia Assanta
- Foundation Gabriele Monasterio CNR-Regione Toscana, 54100 Massa, Italy
| | - Antonio Bottoni
- Foundation Gabriele Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Michele Santoro
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
7
|
Jin Y, Zhao M, Guo Q, Zhao W, Lei M, Zhang Y, Zhang Y, Shen Y, Lin K, Yang Z, Chu J, Sun H, Luo Z. Association study of FLT4 and HYDIN single nucleotide polymorphisms with atrial septal defect susceptibility in the Han Chinese population of Southwest China. Ital J Pediatr 2024; 50:62. [PMID: 38581027 PMCID: PMC10998412 DOI: 10.1186/s13052-024-01630-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/18/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Atrial septal defect (ASD) is a common form of congenital heart disease. Although several genes related to ASD have been found, the genetic factors of ASD remain unclear. This study aimed to evaluate the correlation between 10 candidate single nucleotide polymorphisms (SNPs) and sporadic atrial septal defects. METHODS Based on the results of 34 individual whole exome sequences, 10 candidate SNPs were selected. In total, 489 ASD samples and 420 normal samples were collected. The 10 SNPs in the case group and the control group were identified through Snapshot genotyping technology. The χ2-test and unconditional regression model were used to evaluate the relationship between ASD and each candidate SNP. Haploview software was used to perform linkage disequilibrium and haplotype analysis. RESULTS The χ2 results showed that the FLT4 rs383985 (P = 0.003, OR = 1.115-1.773), HYDIN rs7198975 (P = 0.04621, OR = 1.003-1.461), and HYDIN rs1774266 (P = 0.04621, OR = 1.003-1.461) alleles were significantly different between the control group and the case group (P < 0.05). Only the association with the FLT4 polymorphism was statistically significant after adjustment for multiple comparisons. CONCLUSION These findings suggest that a possible molecular pathogenesis associated with sporadic ASD is worth exploring in future studies.
Collapse
Affiliation(s)
- Ye Jin
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Miao Zhao
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Qiuzhe Guo
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Wanyu Zhao
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Min Lei
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Yifei Zhang
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Yunhan Zhang
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Yan Shen
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Keqin Lin
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China
| | - Zhaoqing Yang
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China
| | - Jiayou Chu
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China
| | - Hao Sun
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China.
| | - Zhiling Luo
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China.
| |
Collapse
|
8
|
Tabrizi F, Khatami M, Heidari MM, Bragança J, Tatari H, Namnabat M, Hadadzadeh M, Navabi Shirazi MA. Novel and deleterious nucleotide variations in the HAND1 gene probably affect miRNA target sites and protein function in pediatric patients with congenital heart disease. Mol Biol Rep 2024; 51:468. [PMID: 38551686 DOI: 10.1007/s11033-024-09410-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/01/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Congenital heart disease (CHD) is the most prevalent developmental defect and principal cause of infant mortality and affects cardiac and large blood vessel structures in approximately 1% of live births worldwide. To date, numerous studies have related critical genetic dysfunctions to the pathogenesis of CHDs. However, the genetic basis underlying CHD remains largely unknown. In the present study, we investigated the association of nucleotide variations in coding and noncoding regions of the HAND1 gene with the risk of CHD. The HAND1 gene, encoding a helix-loop-helix transcription factor, is particularly relevant for mechanisms underlying CHD since it plays a significant role in heart development. METHODS AND RESULTS The genomic DNA of 150 unrelated pediatric patients with CHD was screened by PCR-SSCP and direct sequencing. Four novel and heterozygous missense mutations were identified in the first exon, with three causing amino acid substitutions (p.Val149Met, p.Tyr142His, and p.Leu146Met). In-silico analysis also indicated their deleterious impact on protein structure and function. In addition, we identified five novel nucleotide variants in the 3'UTR region (c.*461, c.*342, c.*529, c.*448, c.*593), potentially altering the target sites of miRNAs. These changes include the loss of certain target sites and the acquisition of new ones. CONCLUSIONS These findings confirm the phenotypic association between CHDs and HAND1 mutations and can pave the way for developing new preventive and therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - José Bragança
- Faculty of Medicine and Biomedical Sciences, Algarve Biomedical Centre Research Institute (ABC-RI), University of Algarve, Faro, Portugal
| | - Hasan Tatari
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Namnabat
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mehdi Hadadzadeh
- Department of Cardiac Surgery, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Ali Navabi Shirazi
- Department of Pediatric Cardiac Surgery, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
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
|
Grueso Cerón AL, Arturo-Terranova D, Satizábal Soto JM. Characterization of genomic variants associated with congenital heart disease in patients from southwestern Colombian. Heliyon 2024; 10:e23678. [PMID: 38187265 PMCID: PMC10767385 DOI: 10.1016/j.heliyon.2023.e23678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/25/2023] [Accepted: 12/09/2023] [Indexed: 01/09/2024] Open
Abstract
Congenital heart diseases correspond to errors during embryogenesis, generating structural and functional malformations. Congenital heart diseases are the most prevalent congenital malformations and are responsible for the highest infant morbidity and mortality. Among these cases, 8 % can be attributed to variants in genes associated with cardiac development. To establish the population frequency of genomic variants that cause congenital heart disease, a review of the scope of prevalent genes was carried out, complete exome sequencing results of 320 patients without suspicion of congenital heart disease were used, the exome sequencing is a technique based on DNA extraction using a Qiagen kit, with massive sequencing of Nextera TM libraries using an Illumina platform with 100X coverage, alignment with reference genome GRCh38/hg19, and analysis with the CRAVAT program; clinical characterization, significance classification, and gene interaction networks were performed. The scope analysis allowed to determine that the genes NKX2-5, TBX20, GATA4, NOTCH1, PTPN11 are the most prevalent, the variants with the highest allelic frequency were c.63A > G (0.2844), c.39T > C (0.3406), c.1132A > G (0.0406), c.1669+9T > C (0.3531) and c.854-30T > C (0.0875) respectively; 4 variants were reclassified by in silico tools, in the NKX2-5 gene c.335-311_335-303del from benign to probably pathogenic, in the NOTCH1 gene c.2354-24C > T from benign to pathogenic, and in the PTPN11 gene c.2354-24C > T and c.854-30T > C from benign to pathogenic. 17 % of intronic variants and 4.8 % of missense variants were identified. This work contributes to knowledge about the frequency with which genomic variants associated with congenital heart disease are present in the population, generating a tool for early diagnosis, early treatment, thus reducing morbidity and mortality, with a view to future universal molecular neonatal screening of congenital heart disease.
Collapse
Affiliation(s)
- Angie Lizeth Grueso Cerón
- Universidad del Valle, Faculty of Natural and Exact Sciences, Department of Biology, Biology Program, Cali, Valle del Cauca, Colombia
| | - Daniela Arturo-Terranova
- Universidad del Valle, Faculty of Health, School of Basic Sciences, Postgraduate in Biomedical Sciences, Cali, Valle del Cauca, Colombia
| | - José María Satizábal Soto
- Universidad del Valle, Faculty of Health, School of Basic Sciences, Postgraduate in Biomedical Sciences, Cali, Valle del Cauca, Colombia
| |
Collapse
|
11
|
Lee Y, Kim J. Unraveling the mystery of oligogenic inheritance under way? Mol Cells 2024; 47:100003. [PMID: 38376484 PMCID: PMC10880077 DOI: 10.1016/j.mocell.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 02/21/2024] Open
Affiliation(s)
- Yerim Lee
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| | - Jaesang Kim
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
12
|
Kovacs S, Scansen BA, Stern JA. The Genetics of Canine Pulmonary Valve Stenosis. Vet Clin North Am Small Anim Pract 2023; 53:1379-1391. [PMID: 37423844 DOI: 10.1016/j.cvsm.2023.05.014] [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] [Indexed: 07/11/2023]
Abstract
There have been recent advancements in understanding the genetic contribution to pulmonary valve stenosis (PS) in brachycephalic breeds such as the French Bulldog and Bulldog. The associated genes are transcriptions factors involved in cardiac development, which is comparable to the genes that cause PS in humans. However, validation studies and functional follow up is necessary before this information can be used for screening purposes.
Collapse
Affiliation(s)
- Samantha Kovacs
- Anatomic Pathology Service, School of Veterinary Medicine, University of California Davis, UC Davis VMTH, 1 Garrod Drive, Davis, CA 95616, USA.
| | - Brian A Scansen
- College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Veterinary Teaching Hospital, 300 West Drake Road, 1678 Campus Delivery, Fort Collins, CO 80523-1678, USA
| | - Joshua A Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, UC Davis VMTH, 1 Garrod Drive, Davis, CA 95616, USA
| |
Collapse
|
13
|
Selewa A, Luo K, Wasney M, Smith L, Sun X, Tang C, Eckart H, Moskowitz IP, Basu A, He X, Pott S. Single-cell genomics improves the discovery of risk variants and genes of atrial fibrillation. Nat Commun 2023; 14:4999. [PMID: 37591828 PMCID: PMC10435551 DOI: 10.1038/s41467-023-40505-5] [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: 02/11/2022] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
Genome-wide association studies (GWAS) have linked hundreds of loci to cardiac diseases. However, in most loci the causal variants and their target genes remain unknown. We developed a combined experimental and analytical approach that integrates single cell epigenomics with GWAS to prioritize risk variants and genes. We profiled accessible chromatin in single cells obtained from human hearts and leveraged the data to study genetics of Atrial Fibrillation (AF), the most common cardiac arrhythmia. Enrichment analysis of AF risk variants using cell-type-resolved open chromatin regions (OCRs) implicated cardiomyocytes as the main mediator of AF risk. We then performed statistical fine-mapping, leveraging the information in OCRs, and identified putative causal variants in 122 AF-associated loci. Taking advantage of the fine-mapping results, our novel statistical procedure for gene discovery prioritized 46 high-confidence risk genes, highlighting transcription factors and signal transduction pathways important for heart development. In summary, our analysis provides a comprehensive map of AF risk variants and genes, and a general framework to integrate single-cell genomics with genetic studies of complex traits.
Collapse
Affiliation(s)
- Alan Selewa
- Biophysical Sciences Graduate Program, The University of Chicago, Chicago, IL, 60637, USA
| | - Kaixuan Luo
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA
| | - Michael Wasney
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Linsin Smith
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiaotong Sun
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA
| | - Chenwei Tang
- The College, The University of Chicago, Chicago, IL, 60637, USA
| | - Heather Eckart
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Ivan P Moskowitz
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of Chicago, Chicago, IL, 60637, USA
| | - Anindita Basu
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| | - Xin He
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA.
| | - Sebastian Pott
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
14
|
Patel KK, Venkatesan C, Abdelhalim H, Zeeshan S, Arima Y, Linna-Kuosmanen S, Ahmed Z. Genomic approaches to identify and investigate genes associated with atrial fibrillation and heart failure susceptibility. Hum Genomics 2023; 17:47. [PMID: 37270590 DOI: 10.1186/s40246-023-00498-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023] Open
Abstract
Atrial fibrillation (AF) and heart failure (HF) contribute to about 45% of all cardiovascular disease (CVD) deaths in the USA and around the globe. Due to the complex nature, progression, inherent genetic makeup, and heterogeneity of CVDs, personalized treatments are believed to be critical. To improve the deciphering of CVD mechanisms, we need to deeply investigate well-known and identify novel genes that are responsible for CVD development. With the advancements in sequencing technologies, genomic data have been generated at an unprecedented pace to foster translational research. Correct application of bioinformatics using genomic data holds the potential to reveal the genetic underpinnings of various health conditions. It can help in the identification of causal variants for AF, HF, and other CVDs by moving beyond the one-gene one-disease model through the integration of common and rare variant association, the expressed genome, and characterization of comorbidities and phenotypic traits derived from the clinical information. In this study, we examined and discussed variable genomic approaches investigating genes associated with AF, HF, and other CVDs. We collected, reviewed, and compared high-quality scientific literature published between 2009 and 2022 and accessible through PubMed/NCBI. While selecting relevant literature, we mainly focused on identifying genomic approaches involving the integration of genomic data; analysis of common and rare genetic variants; metadata and phenotypic details; and multi-ethnic studies including individuals from ethnic minorities, and European, Asian, and American ancestries. We found 190 genes associated with AF and 26 genes linked to HF. Seven genes had implications in both AF and HF, which are SYNPO2L, TTN, MTSS1, SCN5A, PITX2, KLHL3, and AGAP5. We listed our conclusion, which include detailed information about genes and SNPs associated with AF and HF.
Collapse
Affiliation(s)
- Kush Ketan Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Cynthia Venkatesan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Saman Zeeshan
- Rutgers Cancer Institute of New Jersey, Rutgers University, 195 Little Albany St, New Brunswick, NJ, USA
| | - Yuichiro Arima
- Developmental Cardiology Laboratory, International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Kumamoto City, Kumamoto, Japan
| | - Suvi Linna-Kuosmanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Zeeshan Ahmed
- Department of Genetics and Genome Sciences, UConn Health, 400 Farmington Ave, Farmington, CT, USA.
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
15
|
Peña-Martínez EG, Rivera-Madera A, Pomales-Matos DA, Sanabria-Alberto L, Rosario-Cañuelas BM, Rodríguez-Ríos JM, Carrasquillo-Dones EA, Rodríguez-Martínez JA. Disease-associated non-coding variants alter NKX2-5 DNA-binding affinity. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194906. [PMID: 36690178 PMCID: PMC10013089 DOI: 10.1016/j.bbagrm.2023.194906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/30/2022] [Accepted: 01/02/2023] [Indexed: 01/22/2023]
Abstract
Genome-wide association studies (GWAS) have mapped over 90 % of disease- or trait-associated variants within the non-coding genome, like cis-regulatory elements (CREs). Non-coding single nucleotide polymorphisms (SNPs) are genomic variants that can change how DNA-binding regulatory proteins, like transcription factors (TFs), interact with the genome and regulate gene expression. NKX2-5 is a TF essential for proper heart development, and mutations affecting its function have been associated with congenital heart diseases (CHDs). However, establishing a causal mechanism between non-coding genomic variants and human disease remains challenging. To address this challenge, we identified 8475 SNPs predicted to alter NKX2-5 DNA-binding using a position weight matrix (PWM)-based predictive model. Five variants were prioritized for in vitro validation; four of them are associated with traits and diseases that impact cardiovascular health. The impact of these variants on NKX2-5 binding was evaluated with electrophoretic mobility shift assay (EMSA) using purified recombinant NKX2-5 homeodomain. Binding curves were constructed to determine changes in binding between variant and reference alleles. Variants rs7350789, rs7719885, rs747334, and rs3892630 increased binding affinity, whereas rs61216514 decreased binding by NKX2-5 when compared to the reference genome. Our findings suggest that differential TF-DNA binding affinity can be key in establishing a causal mechanism of pathogenic variants.
Collapse
|
16
|
Rudaka I, Vilne B, Isakova J, Kalejs O, Gailite L, Rots D. Genetic Basis of Early Onset Atrial Fibrillation in Patients without Risk Factors. J Cardiovasc Dev Dis 2023; 10:104. [PMID: 36975868 PMCID: PMC10057774 DOI: 10.3390/jcdd10030104] [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: 02/07/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common arrhythmia and typically occurs in elderly patients with other cardiovascular and extracardiac diseases. However, up to 15% of AF develops without any related risk factors. Recently, the role of genetic factors has been highlighted in this particular form of AF. AIMS The aims of this study were to determine the prevalence of pathogenic variants in early-onset AF in patients without known disease-related risk factors and to identify any structural cardiac abnormalities in these patients. MATERIALS AND METHODS We conducted exome sequencing and interpretation in 54 risk factor-free early-onset AF patients and further validated our findings in a similar AF patient cohort from the UK Biobank. RESULTS Pathogenic/likely pathogenic variants were found in 13/54 (24%) patients. The variants were identified in cardiomyopathy-related and not arrhythmia-related genes. The majority of the identified variants were TTN gene truncating variants (TTNtvs) (9/13 (69%) patients). We also observed two TTNtvs founder variants in the analysed population-c.13696C>T p.(Gln4566Ter) and c.82240C>T p.(Arg27414Ter). Pathogenic/likely pathogenic variants were found in 9/107 (8%) individuals from an independent similar AF patient cohort from the UK Biobank. In correspondence with our Latvian patients, only variants in cardiomyopathy-associated genes were identified. In five (38%) of the thirteen Latvian patients with pathogenic/likely pathogenic variants, dilation of one or both ventricles was identified on a follow-up cardiac magnetic resonance scan. CONCLUSIONS We observed a high prevalence of pathogenic/likely pathogenic variants in cardiomyopathy-associated genes in patients with risk factor-free early-onset AF. Moreover, our follow-up imaging data indicate that these types of patients are at risk of developing ventricular dilation. Furthermore, we identified two TTNtvs founder variants in our Latvian study population.
Collapse
Affiliation(s)
- Irina Rudaka
- Scientific Laboratory of Molecular Genetics, Rīga Stradiņš University, LV-1007 Riga, Latvia
- Latvian Cardiology Centre, Pauls Stradiņš Clinical University Hospital, LV-1002 Riga, Latvia
| | - Baiba Vilne
- Bioinformatics Laboratory, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Jekaterina Isakova
- Scientific Laboratory of Molecular Genetics, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Oskars Kalejs
- Latvian Cardiology Centre, Pauls Stradiņš Clinical University Hospital, LV-1002 Riga, Latvia
| | - Linda Gailite
- Scientific Laboratory of Molecular Genetics, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Dmitrijs Rots
- Scientific Laboratory of Molecular Genetics, Rīga Stradiņš University, LV-1007 Riga, Latvia
| |
Collapse
|
17
|
El-Medany A, Aziz S, Duncan E. NKX2-5 genetic mutation in a young woman with an atrial septal defect presenting with complete heart block: ICD or bradycardia pacemaker? BMJ Case Rep 2023; 16:e252523. [PMID: 36609421 PMCID: PMC9827188 DOI: 10.1136/bcr-2022-252523] [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] [Indexed: 01/09/2023] Open
Abstract
A woman in her 40s was admitted following syncope. The 12-lead ECG showed atrial fibrillation with slow ventricular response and suspected complete atrioventricular (AV) block. Cardiac monitoring demonstrated non-sustained monomorphic ventricular tachycardia (VT). Her medical history included surgical repair of an atrial septal defect (ASD) aged 4 years. The patient's mother died suddenly in her early 50s and also had an ASD. Given the patient's syncope, background of familial sudden cardiac death (SCD), suspicion of complete AV block and non-sustained VT, she received an implantable cardiac defibrillator (ICD). She underwent genetic testing, revealing a heterozygous NKX2-5 genetic mutation. The signature phenotype in NKX2-5 mutations is ASD with AV conduction disturbance and an increased risk of SCD secondary to ventricular arrhythmias or severe bradycardia. SCD has been described in NKX2-5 mutation carriers despite functioning permanent pacemakers (PPMs). Therefore, we propose implantation of a preventive ICD, as opposed to a PPM.
Collapse
Affiliation(s)
- Ahmed El-Medany
- Cardiology, Bristol Heart Institute, Bristol, UK
- Cardiology, Southmead Hospital, Bristol, UK
| | | | | |
Collapse
|
18
|
Transcriptomic Changes Associated with ERBB2 Overexpression in Colorectal Cancer Implicate a Potential Role of the Wnt Signaling Pathway in Tumorigenesis. Cancers (Basel) 2022; 15:cancers15010130. [PMID: 36612126 PMCID: PMC9817785 DOI: 10.3390/cancers15010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Colorectal cancer (CRC) remains the third most common cause of cancer mortality worldwide. Precision medicine using OMICs guided by transcriptomic profiling has improved disease diagnosis and prognosis by identifying many CRC targets. One such target that has been actively pursued is an erbb2 receptor tyrosine kinase 2 (ERBB2) (Human Epidermal Growth Factor Receptor 2 (HER2)), which is overexpressed in around 3-5% of patients with CRC worldwide. Despite targeted therapies against HER2 showing significant improvement in disease outcomes in multiple clinical trials, to date, no HER2-based treatment has been clinically approved for CRC. In this study we performed whole transcriptome ribonucleic acid (RNA) sequencing on 11 HER2+ and 3 HER2- CRC patients with advanced stages II, III and IV of the disease. In addition, transcriptomic profiling was carried out on CRC cell lines (HCT116 and HT29) and normal colon cell lines (CCD841 and CCD33), ectopically overexpressing ERBB2. Our analysis revealed transcriptomic changes involving many genes in both CRC cell lines overexpressing ERBB2 and in HER2+ patients, compared to normal colon cell lines and HER2- patients, respectively. Gene Set Enrichment Analysis indicated a role for HER2 in regulating CRC pathogenesis, with Wnt/β-catenin signaling being mediated via a HER2-dependent regulatory pathway impacting expression of the homeobox gene NK2 homeobox 5 (NKX2-5). Results from this study thus identified putative targets that are co-expressed with HER2 in CRC warranting further investigation into their role in CRC pathogenesis.
Collapse
|
19
|
Althali NJ, Hentges KE. Genetic insights into non-syndromic Tetralogy of Fallot. Front Physiol 2022; 13:1012665. [PMID: 36277185 PMCID: PMC9582763 DOI: 10.3389/fphys.2022.1012665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/13/2022] [Indexed: 10/17/2023] Open
Abstract
Congenital heart defects (CHD) include structural abnormalities of the heart or/and great vessels that are present at birth. CHD affects around 1% of all newborns worldwide. Tetralogy of Fallot (TOF) is the most prevalent cyanotic congenital cardiac abnormality, affecting three out of every 10,000 live infants with a prevalence rate of 5-10% of all congenital cardiac defects. The four hallmark characteristics of TOF are: right ventricular hypertrophy, pulmonary stenosis, ventricular septal defect, and overriding aorta. Approximately 20% of cases of TOF are associated with a known disease or chromosomal abnormality, with the remaining 80% of TOF cases being non-syndromic, with no known aetiology. Relatively few TOF patients have been studied, and little is known about critical causative genes for non-syndromic TOF. However, rare genetic variants have been identified as significant risk factors for CHD, and are likely to cause some cases of TOF. Therefore, this review aims to provide an update on well-characterized genes and the most recent variants identified for non-syndromic TOF.
Collapse
Affiliation(s)
- Nouf J. Althali
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Biology Department, Science College, King Khalid University, Abha, Saudi Arabia
| | - Kathryn E. Hentges
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
20
|
Su S, Xia S, He Y, Li J, Ma L, Chen X, Li J. A Phenotype and Genotype Case Report of a Neonate With Congenital Bilateral Coronary Artery Fistulas and Multiple Collateral Arteries. Front Cardiovasc Med 2022; 9:939551. [PMID: 35872895 PMCID: PMC9299261 DOI: 10.3389/fcvm.2022.939551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
We report a unique case of an 18-day-old girl with three coronary artery fistulas to the right atrium and right ventricle, respectively: three collateral arteries arising from the descending aorta and one from the right subclavian artery draining through a sac to the top of the right atrium, patent ductus arteriosus, and atrial septal defect. She presented symptoms of acute congestive heart failure. Cardiac catheterization and surgical interventions were performed to repair the defects. The patient recovered uneventfully and grew up well at 3 years of follow-up. Whole-genome sequencing (WES) in the patient, compared to her parents, showed 17 variants within 11 genes. Among these, only compound heterozygous mutation, c.T470G (p.L157R) and c.A1622G (p.D541G), in the DRC1 gene have been reportedly related to congenital heart disease and are the most likely causative in our patient.
Collapse
Affiliation(s)
- Shixin Su
- Clinical Physiology Laboratory, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Shuliang Xia
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Cardiovascular Surgery, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Ye He
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Department of Pediatric Surgery, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Jianbin Li
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Cardiac Intensive Care Unit, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Li Ma
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Cardiovascular Surgery, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Xinxin Chen
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Cardiovascular Surgery, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- *Correspondence: Xinxin Chen
| | - Jia Li
- Clinical Physiology Laboratory, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
- Jia Li
| |
Collapse
|
21
|
Yamada Y, Yasuda K, Hata Y, Nishida N, Hirono K. A Novel NKX2-5 Variant in a Child with Left Ventricular Noncompaction, Atrial Septal Defect, Atrioventricular Conduction Disorder, and Syncope. J Clin Med 2022; 11:jcm11113171. [PMID: 35683556 PMCID: PMC9181799 DOI: 10.3390/jcm11113171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/21/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022] Open
Abstract
The NKX2-5 gene encodes a transcription factor and is actively involved in heart formation and development. A pediatric case with its variant and left ventricular noncompaction (LVNC) has not been reported. A 12-year-old girl with a history of a surgery for atrial septal detect was referred because of syncope during exercise. The electrocardiogram showed atrioventricular block, and the echocardiogram revealed prominent trabeculations in the left ventricular wall, suggesting LVNC. A novel heterozygous variant in the NKX2-5 gene (NM_004387.1: c.255_256delCT, p.Phe86fs) was identified. NKX2-5 variants should be considered in cases with LVNC, congenital heart disease, arrhythmia, and syncope to prevent sudden cardiac death.
Collapse
Affiliation(s)
- Yuya Yamada
- Department of Cardiology, Aichi Children’s Health and Medical Center, Obu 474-8710, Japan; (Y.Y.); (K.Y.)
| | - Kazushi Yasuda
- Department of Cardiology, Aichi Children’s Health and Medical Center, Obu 474-8710, Japan; (Y.Y.); (K.Y.)
| | - Yukiko Hata
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; (Y.H.); (N.N.)
| | - Naoki Nishida
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; (Y.H.); (N.N.)
| | - Keiichi Hirono
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
- Correspondence: ; Tel.: +81-76-434-7313
| |
Collapse
|
22
|
Ma L, Xu J, Tang Q, Cao Y, Kong R, Li K, Liu J, Jiang L. SLC2A3
variants in familial and sporadic congenital heart diseases in a Chinese Yunnan population. J Clin Lab Anal 2022; 36:e24456. [PMID: 35466476 DOI: 10.1002/jcla.24456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Lijing Ma
- Faculty of Life Science and Technology Kunming University of Science and Technology Kunming China
- Department of Endocrinology The First People’s Hospital of Yunnan Province Kunming China
| | - Jiaxin Xu
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
| | - Qisheng Tang
- Regenerative Medicine Research Center The First People's Hospital of Yunnan Province Kunming China
| | - Yu Cao
- Department of Cardiovascular Surgery The First Peoples’ Hospital of Yunnan Province Kunming China
- Department of Cardiovascular Surgery The First Affiliated Hospital of Kunming University of Science and Technology Kunming China
| | - Ruize Kong
- Department of Vascular Surgery The First Peoples’ Hospital of Yunnan Province Kunming China
- Department of Vascular Surgery The First Affiliated Hospital of Kunming University of Science and Technology Kunming China
| | - Kunlin Li
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
| | - Jie Liu
- Regenerative Medicine Research Center The First People's Hospital of Yunnan Province Kunming China
| | - Lihong Jiang
- Department of Cardiovascular Surgery The First Peoples’ Hospital of Yunnan Province Kunming China
- Department of Cardiovascular Surgery The First Affiliated Hospital of Kunming University of Science and Technology Kunming China
| |
Collapse
|
23
|
Stutt N, Song M, Wilson MD, Scott IC. Cardiac specification during gastrulation - The Yellow Brick Road leading to Tinman. Semin Cell Dev Biol 2021; 127:46-58. [PMID: 34865988 DOI: 10.1016/j.semcdb.2021.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.
Collapse
Affiliation(s)
- Nathan Stutt
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Mengyi Song
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Michael D Wilson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ian C Scott
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
24
|
Dixit R, Narasimhan C, Balekundri VI, Agrawal D, Kumar A, Mohapatra B. Functional analysis of novel genetic variants of NKX2-5 associated with nonsyndromic congenital heart disease. Am J Med Genet A 2021; 185:3644-3663. [PMID: 34214246 DOI: 10.1002/ajmg.a.62413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 01/26/2023]
Abstract
NKX2-5, a master cardiac regulatory transcription factor was the first known genetic cause of congenital heart diseases (CHDs). To further investigate its role in CHD pathogenesis, we performed mutational screening of 285 CHD probands and 200 healthy controls. Five coding sequence variants were identified in six CHD cases (2.1%), including three in the N-terminal region (p.A61G, p.R95L, and p.E131K) and one each in homeodomain (HD) (p.A148E) and tyrosine-rich domain (p.P247A). Variant-p.A148E showed tertiary structure changes and differential DNA binding affinity of mutant compared to wild type. Two N-terminal variants-p.A61G and p.E131K along with HD variant p.A148E demonstrated significantly reduced transcriptional activity of Nppa and Actc1 promoters in dual luciferase promoter assay supported by their reduced expression in qRT-PCR. Nonetheless, variant p.R95L affected the synergy of NKX2-5 with serum response factor and TBX5 leading to significantly decreased Actc1 promoter activity depicting a distinctive role of this region. The aberrant expression of other target genes-Irx4, Mef2c, Bmp10, Myh6, Myh7, and Myocd is also observed in response to NKX2-5 variants, possibly due to the defective gene regulatory network. Severely impaired downstream promoter activities and abnormal expression of target genes due to N-terminal variants supports the emerging role of this region during cardiac-developmental pathways.
Collapse
Affiliation(s)
- Ritu Dixit
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Chitra Narasimhan
- Department of Pediatric Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, Karnataka, India
| | - Vijayalakshmi I Balekundri
- Super Speciality Hospital, Pradhan Mantri Swasthya Suraksha Yojana (PMSSY), Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Damyanti Agrawal
- Department of Cardiothoracic and Vascular Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
25
|
Morlanes-Gracia P, Antoniutti G, Alvarez-Rubio J, Torres-Juan L, Heine-Suñer D, Ripoll-Vera T. Case Report: A Novel NKX2-5 Mutation in a Family With Congenital Heart Defects, Left Ventricular Non-compaction, Conduction Disease, and Sudden Cardiac Death. Front Cardiovasc Med 2021; 8:691203. [PMID: 34277740 PMCID: PMC8280289 DOI: 10.3389/fcvm.2021.691203] [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: 04/05/2021] [Accepted: 06/01/2021] [Indexed: 01/27/2023] Open
Abstract
The NKX2-5 gene encodes for a transcription factor crucial for cardiac cell differentiation and proliferation. It was the first gene associated with congenital heart disease (CHD) in humans and has been linked to conduction disorders or cardiomyopathies. However, an overlapping phenotype is not frequent in the literature. We describe a family with a novel missense mutation in the NKX2-5 gene (p.Gln181Pro) with numerous antecedents with atrial septal defect (ASD), left ventricular non-compaction (LVNC), conduction disease, and sudden cardiac death (SCD).
Collapse
Affiliation(s)
| | | | - Jorge Alvarez-Rubio
- Hospital Universitario Son LLàtzer, Palma de Mallorca, Spain.,Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Laura Torres-Juan
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain.,Departmento de Genetica Clínica y Molecular, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Damian Heine-Suñer
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain.,Departmento de Genetica Clínica y Molecular, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Tomás Ripoll-Vera
- Hospital Universitario Son LLàtzer, Palma de Mallorca, Spain.,Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain.,Centro de Investigación Biomédica CIBEROBN, Madrid, Spain
| |
Collapse
|
26
|
Romano V, Gallinoro CM, Mottola R, Serio A, Di Meglio F, Castaldo C, Sirico F, Nurzynska D. Patent Foramen Ovale-A Not So Innocuous Septal Atrial Defect in Adults. J Cardiovasc Dev Dis 2021; 8:jcdd8060060. [PMID: 34070460 PMCID: PMC8228640 DOI: 10.3390/jcdd8060060] [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: 04/16/2021] [Revised: 05/14/2021] [Accepted: 05/22/2021] [Indexed: 12/11/2022] Open
Abstract
Patent foramen ovale (PFO) is a common congenital atrial septal defect with an incidence of 15–35% in the adult population. The development of the interatrial septum is a process that begins in the fourth gestational week and is completed only after birth. During intrauterine life, the foramen ovale allows the passage of highly oxygenated blood from the right to the left atrium and into the systemic arteries, thus bypassing the pulmonary circulation. In 75% of the general population, the foramen ovale closes after birth, and only an oval depression, called fossa ovalis, remains on the right side of the interatrial septum. Patent foramen ovale can be associated with various clinically important conditions, including migraine and stroke, or decompression illness in divers. The aim of this review is to summarize the PFO developmental and anatomical features and to discuss the clinical risks associated with this atrial septal defect in adults.
Collapse
Affiliation(s)
- Veronica Romano
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy; (V.R.); (C.M.G.); (R.M.); (A.S.); (F.D.M.); (C.C.)
| | - Carlo Maria Gallinoro
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy; (V.R.); (C.M.G.); (R.M.); (A.S.); (F.D.M.); (C.C.)
| | - Rosita Mottola
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy; (V.R.); (C.M.G.); (R.M.); (A.S.); (F.D.M.); (C.C.)
| | - Alessandro Serio
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy; (V.R.); (C.M.G.); (R.M.); (A.S.); (F.D.M.); (C.C.)
| | - Franca Di Meglio
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy; (V.R.); (C.M.G.); (R.M.); (A.S.); (F.D.M.); (C.C.)
| | - Clotilde Castaldo
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy; (V.R.); (C.M.G.); (R.M.); (A.S.); (F.D.M.); (C.C.)
| | - Felice Sirico
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy; (V.R.); (C.M.G.); (R.M.); (A.S.); (F.D.M.); (C.C.)
- Correspondence: (F.S.); (D.N.)
| | - Daria Nurzynska
- Department of Medicine, Surgery and Dentistry “ScuolaMedicaSalernitana”, University of Salerno, 84081 Baronissi, Italy
- Correspondence: (F.S.); (D.N.)
| |
Collapse
|
27
|
De Lange L, Vera L, Decloedt A, Van Steenkiste G, Vernemmen I, van Loon G. Prevalence and characteristics of ventricular septal defects in a non-racehorse equine population (2008-2019). J Vet Intern Med 2021; 35:1573-1581. [PMID: 33742468 PMCID: PMC8163134 DOI: 10.1111/jvim.16106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Ventricular septal defects (VSDs) are the most common congenital cardiac defect in horses. OBJECTIVES To identify prevalence, age, breed, and sex distribution of VSD and to describe associated clinical and ultrasonographic findings. ANIMALS Hospital-based population of 21 136 horses presented to the equine internal medicine department. METHODS Medical records over a 12-year period were reviewed for VSD confirmed by ultrasonography. Age, breed, sex, sport discipline, murmur, clinical signs, outcome, VSD type, VSD size, shunt velocity, cardiac dimensions, concomitant cardiac anomalies, and valvular regurgitations were recorded. RESULTS From 1894 horses that underwent echocardiography, 54 had a VSD: 42 as an isolated lesion and 12 as part of complex congenital heart disease (CHD). Median age was 5 years (range, 0-26) and 1 year (range, 0-8), respectively. Warmbloods and males were overrepresented. In the isolated VSD group, only 15% had associated clinical signs and most horses had a perimembranous VSD (pmVSD; 36/42). Horses with a pmVSD and clinical signs showed a significantly lower maximal shunt velocity (3.77 vs 5.20 m/s; P < .001), higher VSD/Aortic root (Ao) diameter (0.52 vs 0.38; P = .05), higher left atrium/Ao diameter (1.94 vs 1.22; P < .001), and higher pulmonary artery/Ao diameter (1.15 vs 0.88; P = .005) compared to horses without clinical signs. All horses with complex CHD had clinical signs and abnormal cardiac dimensions. CONCLUSION AND CLINICAL IMPORTANCE Most isolated VSD were diagnosed only at a later age and were not associated with clinical signs. Horses with complex CHD were more likely to have or develop clinical signs at younger age.
Collapse
Affiliation(s)
- Lisa De Lange
- Equine Cardioteam, Department of Large Animal Internal Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | - Lisse Vera
- Equine Cardioteam, Department of Large Animal Internal Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | - Annelies Decloedt
- Equine Cardioteam, Department of Large Animal Internal Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | - Glenn Van Steenkiste
- Equine Cardioteam, Department of Large Animal Internal Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | - Ingrid Vernemmen
- Equine Cardioteam, Department of Large Animal Internal Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | - Gunther van Loon
- Equine Cardioteam, Department of Large Animal Internal Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| |
Collapse
|
28
|
Wu Y, Jin X, Zhang Y, Zheng J, Yang R. Genetic and epigenetic mechanisms in the development of congenital heart diseases. WORLD JOURNAL OF PEDIATRIC SURGERY 2021; 4:e000196. [DOI: 10.1136/wjps-2020-000196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Congenital heart disease (CHD) is the most common of congenital cardiovascular malformations associated with birth defects, and it results in significant morbidity and mortality worldwide. The classification of CHD is still elusive owing to the complex pathogenesis of CHD. Advances in molecular medicine have revealed the genetic basis of some heart anomalies. Genes associated with CHD might be modulated by various epigenetic factors. Thus, the genetic and epigenetic factors are gradually accepted as important triggers in the pathogenesis of CHD. However, few literatures have comprehensively elaborated the genetic and epigenetic mechanisms of CHD. This review focuses on the etiology of CHD from genetics and epigenetics to discuss the role of these factors in the development of CHD. The interactions between genetic and epigenetic in the pathogenesis of CHD are also elaborated. Chromosome abnormalities and gene mutations in genetics, and DNA methylations, histone modifications and on-coding RNAs in epigenetics are summarized in detail. We hope the summative knowledge of these etiologies may be useful for improved diagnosis and further elucidation of CHD so that morbidity and mortality of children with CHD can be reduced in the near future.
Collapse
|
29
|
Transcriptional Regulation of Postnatal Cardiomyocyte Maturation and Regeneration. Int J Mol Sci 2021; 22:ijms22063288. [PMID: 33807107 PMCID: PMC8004589 DOI: 10.3390/ijms22063288] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
During the postnatal period, mammalian cardiomyocytes undergo numerous maturational changes associated with increased cardiac function and output, including hypertrophic growth, cell cycle exit, sarcomeric protein isoform switching, and mitochondrial maturation. These changes come at the expense of loss of regenerative capacity of the heart, contributing to heart failure after cardiac injury in adults. While most studies focus on the transcriptional regulation of embryonic or adult cardiomyocytes, the transcriptional changes that occur during the postnatal period are relatively unknown. In this review, we focus on the transcriptional regulators responsible for these aspects of cardiomyocyte maturation during the postnatal period in mammals. By specifically highlighting this transitional period, we draw attention to critical processes in cardiomyocyte maturation with potential therapeutic implications in cardiovascular disease.
Collapse
|
30
|
Miyamoto M, Gangrade H, Tampakakis E. Understanding Heart Field Progenitor Cells for Modeling Congenital Heart Diseases. Curr Cardiol Rep 2021; 23:38. [PMID: 33694131 DOI: 10.1007/s11886-021-01468-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Heart development is a meticulously coordinated process that involves the specification of two distinct populations of cardiac progenitor cells, namely the first and the second heart field. Disruption of heart field progenitors can result in congenital heart defects. In this review, we aim to describe the signaling pathways and transcription factors that link heart field development and congenital heart disease. RECENT FINDINGS Single-cell transcriptomics, lineage-tracing mouse models, and stem cell-based in vitro modeling of cardiogenesis have significantly improved the spatiotemporal characterization of cardiac progenitors. Additionally, novel functional genomic studies have now linked more genetic variants with congenital heart disease. Dysregulation of cardiac progenitor cells causes malformations that can be lethal. Ongoing research will continue to shed light on cardiac morphogenesis and help us better understand and treat patients with congenital heart disease.
Collapse
Affiliation(s)
- Matthew Miyamoto
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 835, Baltimore, MD, 21205, USA
| | - Harshi Gangrade
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 835, Baltimore, MD, 21205, USA
| | - Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Ross 835, Baltimore, MD, 21205, USA.
| |
Collapse
|
31
|
Yuan X, Scott IC, Wilson MD. Heart Enhancers: Development and Disease Control at a Distance. Front Genet 2021; 12:642975. [PMID: 33777110 PMCID: PMC7987942 DOI: 10.3389/fgene.2021.642975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
Bound by lineage-determining transcription factors and signaling effectors, enhancers play essential roles in controlling spatiotemporal gene expression profiles during development, homeostasis and disease. Recent synergistic advances in functional genomic technologies, combined with the developmental biology toolbox, have resulted in unprecedented genome-wide annotation of heart enhancers and their target genes. Starting with early studies of vertebrate heart enhancers and ending with state-of-the-art genome-wide enhancer discovery and testing, we will review how studying heart enhancers in metazoan species has helped inform our understanding of cardiac development and disease.
Collapse
Affiliation(s)
- Xuefei Yuan
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ian C. Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Michael D. Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
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: 9] [Impact Index Per Article: 1.8] [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
|
33
|
Lai G, Wang L, Li Z, Zhao Y. Homocysteine downregulates cardiac homeobox transcription factor NKX2.5 via IGFBP5. Am J Physiol Heart Circ Physiol 2020; 319:H1380-H1386. [PMID: 33035436 DOI: 10.1152/ajpheart.00347.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Homocysteine (Hcy) is an independent risk factor of congenital heart disease (CHD), but its exact underlying mechanism is unclear. In this study, we collected amniotic fluid (AF) supernatant samples from pregnant women carrying CHD-affected (n = 16) or normal (n = 16) fetuses. We found that Hcy concentrations were higher in the AF of the CHD group when compared with normal pregnancies. Also, Western blot showed that NK2 homeobox 5 (NKX2.5) was decreased and insulin-like growth factor binding protein 5 (IGFBP5) was increased in the AF of the CHD group. In the H9C2 cell culture experiment, 500 μmol/L Hcy downregulated NKX2.5 and upregulated IGFBP5. Real-time PCR and Western blot showed that NKX2.5 expression was reduced in H9C2 cells treated with IGFBP5. Luciferase reporter gene demonstrated that IGFBP5 decreased the transcription of the NKX2.5 promoter. Chromatin immunoprecipitation and electrophoretic mobility shift assay suggested that IGFBP5 binds to the NKX2.5 promoter region. Thus, the data indicated that one of the possible mechanisms by which Hcy is involved in CHD may be that Hcy inhibits NKX2.5 expression partly through IGFBP5.NEW & NOTEWORTHY We found that Hcy and IGFBP5 were increased, whereas NKX2.5 was decreased, in AF of CHD. Meanwhile, Hcy could upregulate IGFBP5 but downregulate NKX2.5, and IGFBP5 inhibited NKX2.5 expression in vitro. Moreover, IGFBP5 can bind to the NKX2.5 promoter region and reduce NKX2.5 transcriptional activity.
Collapse
Affiliation(s)
- Guangrui Lai
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Leitong Wang
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.,Department of Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, Liaoning, People's Republic of China
| | - Zhen Li
- Department of Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yanyan Zhao
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
34
|
Felemban SG, Vyas FS, Durose L, Hargreaves AJ, Dickenson JM. Phenyl Saligenin Phosphate Disrupts Cell Morphology and the Actin Cytoskeleton in Differentiating H9c2 Cardiomyoblasts and Human-Induced Pluripotent Stem-Cell-Derived Cardiomyocyte Progenitor Cells. Chem Res Toxicol 2020; 33:2310-2323. [PMID: 32786544 DOI: 10.1021/acs.chemrestox.0c00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have previously shown that phenyl saligenin phosphate (PSP), an organophosphorus compound which is classed as a weak inhibitor of acetylcholinesterase, triggered cytotoxicity in mitotic and differentiated H9c2 cardiomyoblasts. The aim of this study was to assess whether sublethal concentrations of PSP could disrupt the morphology of differentiating rat H9c2 cardiomyoblasts and human-induced pluripotent stem-cell-derived cardiomyocyte progenitor cells (hiPSC-CMs) and to assess the underlying cytoskeletal changes. PSP-induced changes in protein expression were monitored via Western blotting, immunocytochemistry, and proteomic analysis. PSP-mediated cytotoxicity was determined by measuring MTT reduction, LDH release, and caspase-3 activity. Sublethal exposure to PSP (3 μM) induced morphological changes in differentiating H9c2 cells (7, 9, and 13 days), reflected by reduced numbers of spindle-shaped cells. Moreover, this treatment (7 days) attenuated the expression of the cytoskeletal proteins cardiac troponin I, tropomyosin-1, and α-actin. Further proteomic analysis identified nine proteins (e.g., heat shock protein 90-β and calumenin) which were down-regulated by PSP exposure in H9c2 cells. To assess the cytotoxic effects of organophosphorus compounds in a human cell model, we determined their effects on human-induced pluripotent stem-cell-derived cardiomyocyte progenitor cells. Chlorpyrifos and diazinon-induced cytotoxicity (48 h) was evident only at concentrations >100 μM. By contrast, PSP exhibited cytotoxicity in hiPSC-CMs at a concentration of 25 μM following 48 h exposure. Finally, sublethal exposure to PSP (3 μM; 7 days) induced morphological changes and decreased the expression of cardiac troponin I, tropomyosin-1, and α-actin in hiPSC-CMs. In summary, our data suggest cardiomyocyte morphology is disrupted in both cell models by sublethal concentrations of PSP via modulation of cytoskeletal protein expression.
Collapse
Affiliation(s)
- Shatha G Felemban
- School of Science and Technology Nottingham Trent University Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Falguni S Vyas
- School of Science and Technology Nottingham Trent University Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Lyndsey Durose
- School of Science and Technology Nottingham Trent University Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Alan J Hargreaves
- School of Science and Technology Nottingham Trent University Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - John M Dickenson
- School of Science and Technology Nottingham Trent University Clifton Lane, Nottingham NG11 8NS, United Kingdom
| |
Collapse
|
35
|
Crystal Structures of Ternary Complexes of MEF2 and NKX2-5 Bound to DNA Reveal a Disease Related Protein-Protein Interaction Interface. J Mol Biol 2020; 432:5499-5508. [PMID: 32681840 DOI: 10.1016/j.jmb.2020.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 11/22/2022]
Abstract
MEF2 and NKX2-5 transcription factors interact with each other in cardiogenesis and are necessary for normal heart formation. Despite evidence suggesting that these two transcription factors function synergistically and possibly through direct physical interactions, molecular mechanisms by which they interact are not clear. Here we determined the crystal structures of ternary complexes of MEF2 and NKX2-5 bound to myocardin enhancer DNA in two crystal forms. These crystal structures are the first example of human MADS-box/homeobox ternary complex structures involved in cardiogenesis. Our structures reveal two possible modes of interactions between MEF2 and NKX2-5: MEF2 and NKX bind to adjacent DNA sites to recognize DNA in cis; and MEF2 and NKX bind to different DNA strands to interact with each other in trans via a conserved protein-protein interface observed in both crystal forms. Disease-related mutations are mapped to the observed protein-protein interface. Our structural studies provide a starting point to understand and further study the molecular mechanisms of the interactions between MEF2 and NKX2.5 and their roles in cardiogenesis.
Collapse
|
36
|
Wang H, Liu Y, Han S, Zi Y, Zhang Y, Kong R, Liu Z, Cai Z, Zhong C, Liu W, Li L, Jiang L. Nkx2-5 Regulates the Proliferation and Migration of H9c2 Cells. Med Sci Monit 2020; 26:e925388. [PMID: 32780729 PMCID: PMC7441744 DOI: 10.12659/msm.925388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background The protein NKX2–5 affects mammalian heart development. In mice, the disruption of Nkx2–5 has been associated with arrhythmias, abnormal myocardial contraction, abnormal cardiac morphogenesis, and death. However, the details of the mechanisms are unclear. This study was designed to investigate them. Material/Methods Rat cardiomyocytes from the H9c2 cell line were used in our study. First, we knocked down Nkx2–5 in the H9c2 cells and then validated consequent changes in cell proliferation and migration. We then used RNA sequencing to determine the changes in transcripts. Finally, we validated these results by quantitative reverse transcription-polymerase chain reaction. Results We confirmed that Nkx2–5 regulates the proliferation and migration of H9c2 cells. In our experiments, Nkx2–5 regulated the expression of genes related to proliferation, migration, heart development, and disease. Based on bioinformatics analysis, knockdown of Nkx2–5 caused differential expression of genes involved in cardiac development, calcium ion-related biological activity, the transforming growth factor (TGF)-β signaling pathway, pathways related to heart diseases, the MAPK signaling pathway, and other biological processes and signaling pathways. Conclusions Nkx2–5 may regulate proliferation and migration of the H9c2 cells through the genes Tgfb-2, Bmp10, Id2, Wt1, Hey1, and Cacna1g; rno-miR-1-3p; the TGF-β signaling pathway; the MAPK signaling pathway; as well as other genes and pathways.
Collapse
Affiliation(s)
- Hongshu Wang
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yong Liu
- Fuwai Yunnan Cardiovascular Hospital, Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Shen Han
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yunfeng Zi
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yayong Zhang
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Ruize Kong
- The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Zu Liu
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Zhibin Cai
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Chongbin Zhong
- Department of Thoracic Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, China (mainland)
| | - Wei Liu
- Department II of Hepatobillary Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, China (mainland)
| | - Lifeng Li
- Department of Thoracic Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, Yunnan, China (mainland)
| | - Lihong Jiang
- The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
37
|
Amodeo S, Vitrano G, Guardino M, Paci G, Corselli F, Antona V, Barrano G, Magliozzi M, Novelli A, Venezia R, Corsello G. What is the impact of a novel MED12 variant on syndromic conotruncal heart defects? Analysis of case report on two male sibs. Ital J Pediatr 2020; 46:98. [PMID: 32682435 PMCID: PMC7368728 DOI: 10.1186/s13052-020-00865-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/10/2020] [Indexed: 12/29/2022] Open
Abstract
Background Syndromic congenital heart disease accounts for 30% of cases and can be determined by genetic, environmental or multifactorial causes. In many cases the etiology remains uncertain. Many known genes are responsible for specific morphopathogenetic mechanisms during the development of the heart whose alteration can determine specific phenotypes of cardiac malformations. Case presentation We report on two cases of association of conotruncal heart defect with facial dysmorphisms in sibs. In both cases the malformations’ identification occurred by ultrasound in the prenatal period. It was followed by prenatal invasive diagnosis. The genetic analysis revealed no rearrangements in Array-CGH test, while gene panel sequencing identified a new hemizygous variant of uncertain significance (c.887G > A; p.Arg296Gln) in the MED12 gene, located on the X chromosome and inherited from the healthy mother. Conclusion No other reports about the involvement of MED12 gene in syndromic conotruncal heart defects are actually available from the literature and the international genomic databases. This novel variant is a likely pathogenic variant of uncertain significance and it could broaden the spectrum of genes involved in the development of congenital heart diseases and the phenotypic range of MED12-related disorders.
Collapse
Affiliation(s)
- Silvia Amodeo
- Department of Obstetrics and Gynecology, University Hospital Policlinico P. Giaccone, Via Alfonso Giordano 3, Palermo, Italy
| | - Giuseppe Vitrano
- Department of Obstetrics and Gynecology, University Hospital Policlinico P. Giaccone, Via Alfonso Giordano 3, Palermo, Italy
| | - Melania Guardino
- Department of Neonatology and NICU, University Hospital Policlinico P. Giaccone, Palermo, Italy.
| | - Giuseppe Paci
- Department of Obstetrics and Gynecology, University Hospital Policlinico P. Giaccone, Via Alfonso Giordano 3, Palermo, Italy
| | - Fulvio Corselli
- Department of Obstetrics and Gynecology, University Hospital Policlinico P. Giaccone, Via Alfonso Giordano 3, Palermo, Italy
| | - Vincenzo Antona
- Department of Neonatology and NICU, University Hospital Policlinico P. Giaccone, Palermo, Italy
| | - Giuseppe Barrano
- San Pietro Fatebenefratelli Hospital, UOSD Medical Genetics, Rome, Italy
| | - Monia Magliozzi
- Bambino Gesù Children's Hospital, Laboratory of Medical Genetics, Rome, Italy
| | - Antonio Novelli
- Bambino Gesù Children's Hospital, Laboratory of Medical Genetics, Rome, Italy
| | - Renato Venezia
- Department of Obstetrics and Gynecology, University Hospital Policlinico P. Giaccone, Via Alfonso Giordano 3, Palermo, Italy
| | - Giovanni Corsello
- Department of Neonatology and NICU, University Hospital Policlinico P. Giaccone, Palermo, Italy
| |
Collapse
|
38
|
Laforest B, Dai W, Tyan L, Lazarevic S, Shen KM, Gadek M, Broman MT, Weber CR, Moskowitz IP. Atrial fibrillation risk loci interact to modulate Ca2+-dependent atrial rhythm homeostasis. J Clin Invest 2020; 129:4937-4950. [PMID: 31609246 DOI: 10.1172/jci124231] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/16/2019] [Indexed: 12/30/2022] Open
Abstract
Atrial fibrillation (AF), defined by disorganized atrial cardiac rhythm, is the most prevalent cardiac arrhythmia worldwide. Recent genetic studies have highlighted a major heritable component and identified numerous loci associated with AF risk, including the cardiogenic transcription factor genes TBX5, GATA4, and NKX2-5. We report that Tbx5 and Gata4 interact with opposite signs for atrial rhythm controls compared with cardiac development. Using mouse genetics, we found that AF pathophysiology caused by Tbx5 haploinsufficiency, including atrial arrhythmia susceptibility, prolonged action potential duration, and ectopic cardiomyocyte depolarizations, were all rescued by Gata4 haploinsufficiency. In contrast, Nkx2-5 haploinsufficiency showed no combinatorial effect. The molecular basis of the TBX5/GATA4 interaction included normalization of intra-cardiomyocyte calcium flux and expression of calcium channel genes Atp2a2 and Ryr2. Furthermore, GATA4 and TBX5 showed antagonistic interactions on an Ryr2 enhancer. Atrial rhythm instability caused by Tbx5 haploinsufficiency was rescued by a decreased dose of phospholamban, a sarco/endoplasmic reticulum Ca2+-ATPase inhibitor, consistent with a role for decreased sarcoplasmic reticulum calcium flux in Tbx5-dependent AF susceptibility. This work defines a link between Tbx5 dose, sarcoplasmic reticulum calcium flux, and AF propensity. The unexpected interactions between Tbx5 and Gata4 in atrial rhythm control suggest that evaluating specific interactions between genetic risk loci will be necessary for ascertaining personalized risk from genetic association data.
Collapse
Affiliation(s)
| | | | - Leonid Tyan
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | | | | | - Michael T Broman
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | - Ivan P Moskowitz
- Department of Pediatrics, Pathology, and Human Genetics.,Department of Pathology, and
| |
Collapse
|
39
|
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
|
40
|
Abstract
Various strategies have been applied to replace the loss of cardiomyocytes in order to restore reduced cardiac function and prevent the progression of heart disease. Intensive research efforts in the field of cellular reprogramming and cell transplantation may eventually lead to efficient in vivo applications for the treatment of cardiac injuries, representing a novel treatment strategy for regenerative medicine. Modulation of cardiac transcription factor (TF) networks by chemical entities represents another viable option for therapeutic interventions. Comprehensive screening projects have revealed a number of molecular entities acting on molecular pathways highly critical for cellular lineage commitment and differentiation, including compounds targeting Wnt- and transforming growth factor beta (TGFβ)-signaling. Furthermore, previous studies have demonstrated that GATA4 and NKX2-5 are essential TFs in gene regulation of cardiac development and hypertrophy. For example, both of these TFs are required to fully activate mechanical stretch-responsive genes such as atrial natriuretic peptide and brain natriuretic peptide (BNP). We have previously reported that the compound 3i-1000 efficiently inhibited the synergy of the GATA4-NKX2-5 interaction. Cellular effects of 3i-1000 have been further characterized in a number of confirmatory in vitro bioassays, including rat cardiac myocytes and animal models of ischemic injury and angiotensin II-induced pressure overload, suggesting the potential for small molecule-induced cardioprotection.
Collapse
Affiliation(s)
- Mika J. Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| |
Collapse
|
41
|
Kalayinia S, Maleki M, Mahdavi M, Mahdieh N. A novel de novo dominant mutation of NOTCH1 gene in an Iranian family with non-syndromic congenital heart disease. J Clin Lab Anal 2019; 34:e23147. [PMID: 31867804 PMCID: PMC7171333 DOI: 10.1002/jcla.23147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/10/2019] [Accepted: 11/16/2019] [Indexed: 12/16/2022] Open
Abstract
Background Congenital heart disease (CHD) is the most common birth defect which can arises from different genetic defects. The genetic heterogeneity of this disease leads to restricted success in candidate genes screening method. Emerging approaches such as next‐generation sequencing (NGS)‐based genetic analysis might provide a better understating of CHD etiology in the patients who are left undiagnosed. To this aim, in this study, we survived the causes of CHD in an Iranian family who was consanguineous and had two affected children. Methods Affected individuals of this family were checked previously by PCR‐direct sequencing for six candidate genes (NKX2‐5, ZIC3, NODAL, FOXH1, GJA1, GATA4) and had not revealed any reported CHD causative mutations. Whole‐exome sequencing (WES) was performed on this family probond to determine the underlying cause of CHD, and the identified variants were confirmed and segregated by Sanger sequencing. Results We identified one heterozygous missense mutation, c.T6797C (p.Phe2266Ser), in the NOTCH1 gene, which seems to be the most probably disease causing of this family patients. This mutation was found to be novel and not reported on 1000 Genomes Project, dbSNP, and ExAC. Conclusion Worldwide, mutations in NOTCH1 gene are considered as one of the most known causes of CHD. The found NOTCH1 variant in this family affected individuals was the first report from Iran. Yet again, this result indicates the importance of NOTCH1 screening in CHD patients.
Collapse
Affiliation(s)
- Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nejat Mahdieh
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Kalayinia S, Ghasemi S, Mahdieh N. A comprehensive in silico analysis, distribution and frequency of human Nkx2-5 mutations; A critical gene in congenital heart disease. J Cardiovasc Thorac Res 2019; 11:287-299. [PMID: 31824610 PMCID: PMC6891041 DOI: 10.15171/jcvtr.2019.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
Introduction: Congenital heart disease (CHD) affects 1% to 2 % of live births. The Nkx2-5 gene, is known as the significant heart marker during embryonic evolution and it is also necessary for the survival of cardiomyocytes and homeostasis in adulthood. In this study, Nkx2-5 mutations are investigated to identify the frequency, distribution, functional consequences of mutations by using computational tools.
Methods: A complete literature search was conducted to find Nkx2-5 mutations using the following key words: Nkx2-5 and/or CHD and mutations. The mutations were in silico analyzed using tools which predict the pathogenicity of the variants. A picture of Nkx2-5 protein and functional or structural effects of its variants were also figured using I-TASSER and STRING.
Results: A total number of 105 mutations from 18 countries were introduced. The most (24.1%) and the least (1.49%) frequency of Nkx2-5 mutations were observed in Europe and Africa, respectively. The c.73C>T and c.533C>T mutations are distributed worldwide. c.325G>T (62.5%) and c.896A>G (52.9%) had the most frequency. The most numbers of Nkx2-5 mutations were reported from Germany. The c.541C>T had the highest CADD score (Phred score = 38) and the least was for c.380C>A (Phred score=0.002). 41.9% of mutations were predicted as potentially pathogenic by all prediction tools.
Conclusion: This is the first report of the Nkx2-5 mutations evaluation in the worldwide. Given that the high frequency of mutation in Germany, and also some mutations were seen only in this country, therefore, presumably the main origin of Nkx2-5 mutations arise from Germany.
Collapse
Affiliation(s)
- Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Serwa Ghasemi
- Department of Biology, School of Basic Sciences, Islamic Azad University Research Tehran Branch, Tehran, Iran
| | - Nejat Mahdieh
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Safari-Arababadi A, Behjati-Ardakani M, Kalantar SM, Jaafarinia M. The Contribution of Gene Mutations to the Pathogenesisof Tetralogy of Fallot. INTERNATIONAL JOURNAL OF BASIC SCIENCE IN MEDICINE 2019. [DOI: 10.15171/ijbsm.2019.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Congenital heart disease (CHD) is considered as an important and developing area in the medical community. Since these patients can reach maturity and have children, the role of genetic determinants in increasing risk of CHD is extremely evident among children of these patients. Because genetic studies related to CHD are increasing, and each day the role of new genetic markers is more and more clarified, this review re-examined the effects of gene mutations in the pathogenesis of tetralogy of Fallot (TOF) as an important pathological model among other CHDs. Due to the complexity of heart development, it is not astonishing that numerous signaling pathways and transcription factors, and many genes are involved in pathogenesis of TOF. This review focuses on the jag1, nkx2.5, gata4, zfpm2/fog2 and cited2 genes previously reported to be involved in TOF.
Collapse
Affiliation(s)
- Amin Safari-Arababadi
- Department of Molecular Genetics, Fars Science and Research Branch, Islamic Azad University, Shiraz, Iran
- Department of Molecular Genetics, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | | | - Seyed Mehdi Kalantar
- Genetic and Reproductive Unit, Recurrent Abortion Research Centre, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mojtaba Jaafarinia
- Department of Molecular Genetics, Fars Science and Research Branch, Islamic Azad University, Shiraz, Iran
- Department of Molecular Genetics, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|
44
|
Kalayinia S, Maleki M, Rokni-Zadeh H, Changi-Ashtiani M, Ahangar H, Biglari A, Shahani T, Mahdieh N. GATA4 screening in Iranian patients of various ethnicities affected with congenital heart disease: Co-occurrence of a novel de novo translocation (5;7) and a likely pathogenic heterozygous GATA4 mutation in a family with autosomal dominant congenital heart disease. J Clin Lab Anal 2019; 33:e22923. [PMID: 31115957 PMCID: PMC6757118 DOI: 10.1002/jcla.22923] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/20/2019] [Accepted: 05/07/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is the most common birth defect and a major health problem around the world. However, its exact etiology has remained unclear. Among various genetic contributing factors, GATA4 transcription factor plays a significant role in the CHD pathogenesis. In this study, GATA4 coding sequence was screened in Iranian patients of various ethnicities. METHODS Sixty six individuals with familial CHD referred to our center were recruited in this study. After receiving written informed consent from each individual or their parents, chromosomal analyses and GATA4 variant screening were performed. Pathogenicity of the suspected variants was evaluated using available online software tools: CADD, Mutation Taster, SIFT, and PolyPhen-2. RESULTS A total of twelve GATA4 variants were detected including five intronic, 2 exonic and 3 polymorphisms as well as 2 missense mutations, the c.1220C>A and c.1309G>A. Unlike the c.1220C>A, the likely pathogenic heterozygous c.1309G>A has not been previously associated with any phenotype. Here, we not only report, for the first time, a c.1309G>A-related CHD, but also report a novel de novo balanced translocation, 46,XY,t(5;7)(qter13;qter11), in the same patient which may have influenced the disease severity. CONCLUSION From screening GATA4 sequence in 66 Iranian patients of various ethnicities, we conclude that cytogenetic analysis and PCR-direct sequencing of different candidate genes may not be the best approach for genetic diagnosis in CHD. Applying novel approaches such as next-generation sequencing (NGS) may provide a better understating of genetic contributing factors in CHD patients for whom conventional methods could not reveal any genetic causative factor.
Collapse
Affiliation(s)
- Samira Kalayinia
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran.,Cardiogenetics Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetics Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Rokni-Zadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Majid Changi-Ashtiani
- School of Mathematics, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Hassan Ahangar
- Department of Cardiology, Mousavi Hospital, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran.,Department of Cardiology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Alireza Biglari
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Tina Shahani
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Nejat Mahdieh
- Cardiogenetics Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Wang H, Liu Y, Li Y, Wang W, Li L, Meng M, Xie Y, Zhang Y, Yunfeng Z, Han S, Zeng J, Hou Z, Jiang L. Analysis of NKX2-5 in 439 Chinese Patients with Sporadic Atrial Septal Defect. Med Sci Monit 2019; 25:2756-2763. [PMID: 30982828 PMCID: PMC6481236 DOI: 10.12659/msm.916052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background The NKX2 gene family is made up of core transcription factors that are involved in the morphogenesis of the vertebrate heart. NKx2-5 plays a pivotal role in mouse cardiogenesis, and mutations in NKx2-5 result in an abnormal structure and function of the heart, including atrial septal defect and cardiac electrophysiological abnormalities. Material/Methods To investigate the genetic variation of NKX2-5 in Chinese patients with sporadic atrial septal defect, we sequenced the full length of the NKX2-5 gene in the participants of the study. Four hundred thirty-nine patients and 567 healthy unrelated individuals were recruited. Genomic DNA was extracted from the peripheral blood leukocytes of the participants. DNA samples from the participants were amplified by multiplex PCR and sequenced on an Illumina HiSeq platform. Variations were detected by comparison with a standard reference genome and annotation with a variant effect predictor. Results Thirty variations were detected in Chinese patients with sporadic atrial septal defect, and 6 single nucleotide polymorphisms (SNPs) had a frequency greater than 1%. Among the 30 variations, the SNPs rs2277923 and rs3729753 were extremely prominent, with a high frequency and odds ratio in patients. Conclusions Single nucleotide variations are the prominent genetic variations of NKX2-5 in Chinese patients with sporadic atrial septal defect. The SNPs rs2277923 and rs3729753 are prominent single nucleotide variations (SNVs) in Chinese patients with sporadic atrial septal defect.
Collapse
Affiliation(s)
- Hongshu Wang
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland).,Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Yong Liu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Yaxiong Li
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Wenju Wang
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland).,Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Lin Li
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland).,Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Mingyao Meng
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yanhua Xie
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yayong Zhang
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Zi Yunfeng
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Shen Han
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland).,Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Jianying Zeng
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland).,Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - ZongLiu Hou
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland).,Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (mainland).,Key Laboratory of Cardiovascular Disease of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Lihong Jiang
- The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
46
|
Double de novo mutations in dilated cardiomyopathy with cardiac arrest. J Electrocardiol 2018; 53:40-43. [PMID: 30611920 DOI: 10.1016/j.jelectrocard.2018.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/16/2018] [Accepted: 12/21/2018] [Indexed: 01/27/2023]
Abstract
Here we report the identification of two novel mutations in a previously asymptomatic young man who suffered an out-of-hospital sudden cardiac arrest. During following evaluation, diagnosis of early stage dilated cardiomyopathy was established, while electrocardiogram monitoring showed frequent complex ventricular arrhythmias, incomplete right bundle branch block and prolonged QT duration. No reversible causes explaining the clinical presentation were established and an automatic implantable cardioverter defibrillator was therefore implanted. Heterozygous mutations in human protein coding genes NKX2-5 and RBM20 are associated with a wide array of pathological phenotypes some of which are sudden cardiac death, unexplained syncope and either combined or isolated congenital heart diseases such as dilated cardiomyopathy.
Collapse
|
47
|
张 慧, 陈 名, 方 涛, 张 甜, 倪 文. [Establishment and verification of a mouse model of Gata4 gene H435Y mutation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1245-1249. [PMID: 30377126 PMCID: PMC6744054 DOI: 10.3969/j.issn.1673-4254.2018.10.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To establish a mouse model of H435Y mutation of Gata4 gene using CRISPR/Cas9- mediated gene targeting. METHODS The single-stranded guide RNA (sgRNA) specific to the H435Y loci of Gata4 gene was designed based on the sequence of Gata4 gene. After activity assessment, the active sgRNA and Cas9 were in vitro transcribed into RNA and microinjected along with the donor DNA fragment with point mutations into fertilized mouse eggs. The microinjected eggs were transferred into pseudopregnant mice to obtain the F0 generation mice with the target Gata4 gene mutation confirmed by PCR and gene sequencing. Gata4 gene mutations in the offsprings of the F0 generation mice were analyzed. RESULTS Gene sequencing confirmed the successful establishment of mouse models carrying H435Y mutation of Gata4 gene in 4 of the F0 generation mice. The positive F0 generation mice were crossed with wild-type C57BL/6J mice to obtain the F1 generation mice, and PCR confirmed the presence of H435Y mutations of Gata4 gene in 6 of the F1 mice. Then F2 generation mice were obtained by F1 generation matting with each other. PCR showed that H435Y mutation of Gata4 gene in F2 mice was found, indicating the mousemodel of Gata4 gene mutation in H435Y was established and propagated successfully. CONCLUSIONS We successfully established Gata4 gene H435Y mutant mouse models using CRISPR/Cas9 technique.
Collapse
Affiliation(s)
- 慧 张
- />安徽医科大学附属省立医院儿科,安徽 合肥 230001Department of Pediatrics, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 23001, China
| | - 名武 陈
- />安徽医科大学附属省立医院儿科,安徽 合肥 230001Department of Pediatrics, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 23001, China
| | - 涛 方
- />安徽医科大学附属省立医院儿科,安徽 合肥 230001Department of Pediatrics, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 23001, China
| | - 甜 张
- />安徽医科大学附属省立医院儿科,安徽 合肥 230001Department of Pediatrics, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 23001, China
| | - 文泉 倪
- />安徽医科大学附属省立医院儿科,安徽 合肥 230001Department of Pediatrics, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 23001, China
| |
Collapse
|
48
|
Williams MR, Perry JC. Arrhythmias and conduction disorders associated with atrial septal defects. J Thorac Dis 2018; 10:S2940-S2944. [PMID: 30305954 DOI: 10.21037/jtd.2018.08.27] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Atrial septal defects (ASDs) are the most common form of congenital heart disease. There are 4 embryologic types of ASDs, and rhythm considerations vary based on type. ASDs have left-to-right shunt and primarily right-sided volume overload. This leads to electrical remodeling that may predispose patients to atrial tachyarrhythmias and conduction disorders. Risk for arrhythmias is increased with late age of ASD repair, shunt size, other factors such as pulmonary hypertension and comorbid conditions. Arrhythmia incidence is decreased after ASD closure, but remains elevated compared to general population. Medical and procedural therapy for arrhythmias should consider type and timing of ASD repair. Conduction disorders are rare. Sinus node dysfunction may be seen with late age of repair and large shunt size. Sinus venosus ASD exhibits a higher rate of sinus node dysfunction, especially with older surgical techniques. Ostium primum ASD has higher risk of spontaneous or post-operative AV block, though this is rare with current surgical techniques. Risk of AV block with surgical repair or device closure of secundum ASD is rare. Familial ASDs and other forms of congenital heart disease may be seen with mutations in associated myocardial transcription factors NKX2.5, GATA4, TBX6, along with conduction disorders such as AV block.
Collapse
Affiliation(s)
- Matthew R Williams
- Division of Cardiology, Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego, San Diego, CA 92123, USA
| | - James C Perry
- Division of Cardiology, Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego, San Diego, CA 92123, USA
| |
Collapse
|
49
|
Jhaveri S, Aziz PF, Saarel E. Expanding the electrical phenotype of NKX2-5 mutations: Ventricular tachycardia, atrial fibrillation, and complete heart block within one family. HeartRhythm Case Rep 2018; 4:530-533. [PMID: 30479953 PMCID: PMC6241164 DOI: 10.1016/j.hrcr.2018.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Simone Jhaveri
- Department of Pediatric Cardiology, Cleveland Clinic Children's, Cleveland, Ohio
| | - Peter F Aziz
- Department of Pediatric Cardiology, Cleveland Clinic Children's, Cleveland, Ohio
| | - Elizabeth Saarel
- Department of Pediatric Cardiology, Cleveland Clinic Children's, Cleveland, Ohio
| |
Collapse
|
50
|
Khatami M, Mazidi M, Taher S, Heidari MM, Hadadzadeh M. Novel Point Mutations in the NKX2.5 Gene in Pediatric Patients with Non-Familial Congenital Heart Disease. ACTA ACUST UNITED AC 2018; 54:medicina54030046. [PMID: 30344277 PMCID: PMC6122093 DOI: 10.3390/medicina54030046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 01/25/2023]
Abstract
Background and objective: Congenital heart disease (CHD) is the most common birth abnormality in the structure or function of the heart that affects approximately 1% of all newborns. Despite its prevalence and clinical importance, the etiology of CHD remains mainly unknown. Somatic and germline mutations in cardiac specific transcription factor genes have been identified as the factors responsible for various forms of CHD, particularly ventricular septal defects (VSDs), tetralogy of Fallot (TOF), and atrial septal defects (ASDs). p. NKX2.5 is a homeodomain protein that controls many of the physiological processes in cardiac development including specification and proliferation of cardiac precursors. The aim of our study was to evaluate the NKX2.5 gene mutations in sporadic pediatric patients with clinical diagnosis of congenital heart malformations. Materials and methods: In this study, we investigated mutations of the NKX2.5 gene’s coding region in 105 Iranian pediatric patients with non-familial CHD by polymerase chain reaction-single stranded conformation polymorphism (PCR-SSCP) and direct sequencing. Results: We observed a total of four mutations, of which, two were novel DNA sequence variants in the coding region of exon 1 (c. 95 A > T and c. 93 A > T) and two others were previously reported as single-nucleotide polymorphisms (SNPs), namely rs72554028 (c. 2357 G > A) and rs3729753 (c. 606 G > C) in exon 2. Further, observed mutations are completely absent in normal healthy individuals (n = 92). Conclusion: These results suggest that NKX2.5 mutations are highly rare in CHD patients. However, in silico analysis proves that c.95 A > T missense mutation in NKX2.5 gene is probably pathogenic and may be contributing to the risk of sporadic CHD in the Iranian population.
Collapse
Affiliation(s)
- Mehri Khatami
- Department of Biology, Faculty of Science, Yazd University, Yazd 8915818411, Iran.
| | - Mansoureh Mazidi
- Department of Biology, Faculty of Science, Yazd University, Yazd 8915818411, Iran.
| | - Shabnam Taher
- Department of Biology, Faculty of Science, Yazd University, Yazd 8915818411, Iran.
| | | | - Mehdi Hadadzadeh
- Department of Cardiac Surgery, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd 8915818411, Iran.
| |
Collapse
|