1
|
Shi H, Zhang Z, Shen A, Ding T, Zhao R, Shi Y, Zhao J, Cai K, Wang F. Maternal di(2-ethylhexyl) phthalate exposure increases the risk of congenital heart disease in mice offspring. Pediatr Res 2025:10.1038/s41390-025-03997-z. [PMID: 40089607 DOI: 10.1038/s41390-025-03997-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/04/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
ABSTRCT BACKGROUD: Epidemiological data suggest that maternal occupational exposure to mixed phthalates comprising di(2-ethylhexyl) phthalate (DEHP) increases the risk of congenital heart disease (CHD). In this study, we used mice as an animal model to validate impact of first-trimester DEHP exposure on the risk of CHD in offspring, to elucidate the possible mechanisms and to provide a potential feasible intervention. METHODS AND RESULTS Eight-week-old C57BL/6J pregnant mice were randomly divided into standard and DEHP diet groups. The incidence of CHD in DEHP diet group offspring was up to 14.41% observed via Hematoxylin-eosin (HE) staining. Quantitative PCR analysis revealed that expression of key genes involved in cardiogenesis were suppressed at the transcriptional level, which may be due to decreased nuclear translocation of p65. The inhibition of DEHP on key genes was rescued to some extent by choline through driving p65 into nuclear. In the mice, supplementation of choline during DEHP exposure reduced the incidence of CHD in offspring from 14.41% to 4.63%. CONCLUSIONS Our study demonstrates that mice first-trimester DEHP exposure significantly increases the risk of CHD in the offspring via inhibiting mRNA levels of key genes in cardiogenesis, and choline could protect against the pathogenesis. IMPACT Our study provides key mechanistic insights into the risk of CHD by DEHP exposure during early pregnancy, and provides choline as a potentially effective intervention. DEHP suppressed the expression of key genes involved in embryonic cardiac septum development at the transcriptional level via inhibiting nuclear translocation of p65. Choline can play a role in rescuing the inhibition of DEHP on cardiogenesis genes via driving p65 translocate into the nuclear.
Collapse
Affiliation(s)
- Haiqun Shi
- Pediatric Heart Center, NHC Key Laboratory of Neonatal Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Zehua Zhang
- Pediatric Heart Center, NHC Key Laboratory of Neonatal Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Anna Shen
- Pediatric Heart Center, NHC Key Laboratory of Neonatal Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Tong Ding
- Pediatric Heart Center, NHC Key Laboratory of Neonatal Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Rui Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yan Shi
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ke Cai
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Feng Wang
- Pediatric Heart Center, NHC Key Laboratory of Neonatal Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
| |
Collapse
|
2
|
Meng X, Du W, Sun Z. Fine particulate matter‑induced cardiac developmental toxicity (Review). Exp Ther Med 2025; 29:6. [PMID: 39534282 PMCID: PMC11552469 DOI: 10.3892/etm.2024.12756] [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: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Fine particulate matter (PM2.5) has become an important risk factor threatening human health. Epidemiological and toxicological investigations have revealed that PM2.5 not only leads to cardiovascular dysfunction, but it also gives rise to various adverse health effects on the human body, such as cardiovascular and cerebrovascular diseases, cancers, neurodevelopmental disorders, depression and autism. PM2.5 is able to penetrate both respiratory and placental barriers, thereby resulting in negative effects on fetal development. A large body of epidemiological evidences has suggested that gestational exposure to PM2.5 increases the incidence of congenital diseases in offspring, including congenital heart defects. In addition, animal model studies have revealed that gestational exposure to PM2.5 can disrupt normal heart development in offspring, although the potential molecular mechanisms have yet to be fully elucidated. The aim of the present review was to provide a brief overview of what is currently known regarding the molecular mechanisms underlying cardiac developmental toxicity in offspring induced by gestational exposure to PM2.5.
Collapse
Affiliation(s)
- Xiangjiang Meng
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Weiyuan Du
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Zongli Sun
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| |
Collapse
|
3
|
Zhang L, Zhou J. Zebrafish: A smart tool for heart disease research. JOURNAL OF FISH BIOLOGY 2024; 105:1487-1500. [PMID: 37824489 DOI: 10.1111/jfb.15585] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/07/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
The increasing prevalence of heart disease poses a significant threat to human survival and safety. However, the current treatments available for heart disease are quite limited. Therefore, it is important to utilize suitable animal models that can accurately simulate the physiological characteristics of heart disease. This would help improve our understanding of this disease and aid in the development of new treatment methods and drugs. Zebrafish heart not only exhibits similarities to mammalian hearts, but they also share ~70% of homologous genes with humans. Utilizing zebrafish as an alternative to expensive and time-consuming mammalian models offers numerous advantages. Zebrafish models can be easily established and maintained, and compound screening and genetic methods allow for the development of various economical and easily controlled zebrafish and zebrafish embryonic heart disease models in a short period of time. Consequently, zebrafish have become a powerful tool for exploring the pathological mechanisms of heart disease and identifying new effective genes. In this review, we summarize recent studies on different zebrafish models of heart disease. We also describe the techniques and protocols used to develop zebrafish models of myocardial infarction, heart failure, and congenital heart disease, including surgical procedures, forward and reverse genetics, and drug and combination screening. This review aims to promote the utilization of zebrafish models in investigating diverse pathological mechanisms of heart disease, enhancing our knowledge and comprehension of heart disease, and offering novel insights and objectives for exploring the prevention and treatment of heart disease.
Collapse
Affiliation(s)
- Lantian Zhang
- Education Branch, Chongqing Publishing Group, Chongqing, China
| | - Jinrun Zhou
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| |
Collapse
|
4
|
Rosca I, Popescu DE, Jura AMC, Turenschi A, Filip C, Dinulescu A, Constantin AT. The Critical Role of Pulse Oximetry Screening for Congenital Heart Disease and Patent Ductus Arteriosus in Newborns. Cureus 2024; 16:e75388. [PMID: 39781152 PMCID: PMC11709140 DOI: 10.7759/cureus.75388] [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] [Accepted: 12/08/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Congenital heart disease (CHD) is diagnosed with high prevalence. Pulse oximetry and clinical examination are screening tools to aid in obtaining a CHD diagnosis. MATERIALS AND METHODS We conducted a retrospective longitudinal study over three years, screening 1188 newborns admitted to the neonatal intensive care unit (NICU) during the first 72 hours of life. RESULTS Two hundred one newborns were diagnosed with CHD. Almost all (n=198) had a positive screening test during the 72 hours, while three had a negative screening test but were later diagnosed using echocardiography. The prevalence rate of CHD was 16.67%. A confusion matrix was constructed with 198 true positives (TP), 3 false negatives (FN), no false positives (FP), and 987 true negatives (TN). The sensitivity of the screening test was 98.51%, while the specificity was 100%, with no false positives. In our study, a total of nine deaths occurred due to CHD or complications of CHD. Using the Kaplan-Meier method, the survival probability appeared to be slightly above 0.9 (95.5%), indicating that almost all patients survived past 72 hours after being diagnosed with CHD. CONCLUSION Pulse oximetry is an effective, low-cost, and easy-to-use method that is useful for assessing and diagnosing CHD.
Collapse
Affiliation(s)
- Ioana Rosca
- Neonatology, University of Medicine and Pharmacy Carol Davila, Bucharest, ROU
- Neonatology, Clinic Hospital of Obstetrics and Gynecology Prof. Dr. Panait Sirbu, Bucharest, ROU
| | - Daniela-Eugenia Popescu
- Obstetrics-Gynecology and Neonatology, Victor Babeş University of Medicine and Pharmacy, Timisoara, ROU
| | - Ana Maria Cristina Jura
- Obstetrics-Gynecology and Neonatology, Victor Babeş University of Medicine and Pharmacy, Timisoara, ROU
| | | | - Cristina Filip
- Pediatric Cardiology, Marie Curie Children's Hospital, Bucharest, ROU
| | - Alexandru Dinulescu
- Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Andreea T Constantin
- Pediatrics, Alessandrescu-Rusescu National Institute of Mother and Child Health, Bucharest, ROU
| |
Collapse
|
5
|
Nawaz K, Alifah N, Hussain T, Hameed H, Ali H, Hamayun S, Mir A, Wahab A, Naeem M, Zakria M, Pakki E, Hasan N. From genes to therapy: A comprehensive exploration of congenital heart disease through the lens of genetics and emerging technologies. Curr Probl Cardiol 2024; 49:102726. [PMID: 38944223 DOI: 10.1016/j.cpcardiol.2024.102726] [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: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Congenital heart disease (CHD) affects approximately 1 % of live births worldwide, making it the most common congenital anomaly in newborns. Recent advancements in genetics and genomics have significantly deepened our understanding of the genetics of CHDs. While the majority of CHD etiology remains unclear, evidence consistently indicates that genetics play a significant role in its development. CHD etiology holds promise for enhancing diagnosis and developing novel therapies to improve patient outcomes. In this review, we explore the contributions of both monogenic and polygenic factors of CHDs and highlight the transformative impact of emerging technologies on these fields. We also summarized the state-of-the-art techniques, including targeted next-generation sequencing (NGS), whole genome and whole exome sequencing (WGS, WES), single-cell RNA sequencing (scRNA-seq), human induced pluripotent stem cells (hiPSCs) and others, that have revolutionized our understanding of cardiovascular disease genetics both from diagnosis perspective and from disease mechanism perspective in children and young adults. These molecular diagnostic techniques have identified new genes and chromosomal regions involved in syndromic and non-syndromic CHD, enabling a more defined explanation of the underlying pathogenetic mechanisms. As our knowledge and technologies continue to evolve, they promise to enhance clinical outcomes and reduce the CHD burden worldwide.
Collapse
Affiliation(s)
- Khalid Nawaz
- Department of Medical Laboratory Technology, Khyber Medical University, Peshawar, 25100, Khyber Pakhtunkhwa, Pakistan
| | - Nur Alifah
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar, 90245, Republic of Indonesia
| | - Talib Hussain
- Women Dental College, Khyber Medical University, Abbottabad, 22080, Khyber Pakhtunkhwa, Pakistan
| | - Hamza Hameed
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485, Punjab, Pakistan
| | - Haider Ali
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Shah Hamayun
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485, Punjab, Pakistan
| | - Awal Mir
- Department of Medical Laboratory Technology, Khyber Medical University, Peshawar, 25100, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Naeem
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Mohammad Zakria
- Advanced Center for Genomic Technologies, Khyber Medical University, Peshawar, 25100, Khyber Pakhtunkhwa, Pakistan
| | - Ermina Pakki
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar, 90245, Republic of Indonesia
| | - Nurhasni Hasan
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar, 90245, Republic of Indonesia.
| |
Collapse
|
6
|
Liang J, He X, Wang Y. Cardiomyocyte proliferation and regeneration in congenital heart disease. PEDIATRIC DISCOVERY 2024; 2:e2501. [PMID: 39308981 PMCID: PMC11412308 DOI: 10.1002/pdi3.2501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/25/2024] [Indexed: 09/25/2024]
Abstract
Despite advances in prenatal screening and a notable decrease in mortality rates, congenital heart disease (CHD) remains the most prevalent congenital disorder in newborns globally. Current therapeutic surgical approaches face challenges due to the significant rise in complications and disabilities. Emerging cardiac regenerative therapies offer promising adjuncts for CHD treatment. One novel avenue involves investigating methods to stimulate cardiomyocyte proliferation. However, the mechanism of altered cardiomyocyte proliferation in CHD is not fully understood, and there are few feasible approaches to stimulate cardiomyocyte cell cycling for optimal healing in CHD patients. In this review, we explore recent progress in understanding genetic and epigenetic mechanisms underlying defective cardiomyocyte proliferation in CHD from development through birth. Targeting cell cycle pathways shows promise for enhancing cardiomyocyte cytokinesis, division, and regeneration to repair heart defects. Advancements in human disease modeling techniques, CRISPR-based genome and epigenome editing, and next-generation sequencing technologies will expedite the exploration of abnormal machinery governing cardiomyocyte differentiation, proliferation, and maturation across diverse genetic backgrounds of CHD. Ongoing studies on screening drugs that regulate cell cycling are poised to translate this nascent technology of enhancing cardiomyocyte proliferation into a new therapeutic paradigm for CHD surgical interventions.
Collapse
Affiliation(s)
- Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Xingyu He
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
7
|
Khan MH, Ahsan A, Mehta F, Kanawala A, Mondal R, Dilshad A, Akbar A. Precision Medicine in Congenital Heart Disease, Rheumatic Heart Disease, and Kawasaki Disease of Children: An Overview of Literature. Cardiol Rev 2024:00045415-990000000-00257. [PMID: 39819650 DOI: 10.1097/crd.0000000000000709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Congenital heart disease and common acquired heart diseases like Kawasaki disease and rheumatic heart disease are prevalent cardiovascular conditions in children worldwide. Despite the availability of treatment options, they continue to be significant contributors to morbidity and mortality. Advancements in early diagnosis, improvements in treatment approaches, and overcoming resistance to available treatments are crucial to reduce morbidity. Researchers have turned to precision medicine to tackle these challenges. We aimed to analyze the existing literature concerning the utilization of precision medicine in congenital heart disease, rheumatic heart disease, and Kawasaki disease. The emphasis is placed on comprehending the key themes explored in these studies and evaluating the present state of their clinical integration. The central theme of most studies revolves around the examination of genetic factors. Despite promising research outcomes, limitations in these studies indicate that the clinical implementation of precision medicine in these conditions remains a distant prospect, necessitating additional exploration and attention to confounding factors.
Collapse
Affiliation(s)
- Muhammad Hamza Khan
- From the Department of Internal Medicine, Karachi Medical and Dental College, Karachi, Pakistan
| | - Areeba Ahsan
- Department of Internal Medicine, Foundation University School of Health Sciences, Islamabad, Pakistan
| | - Fena Mehta
- Department of Internal Medicine, Smt. NHL Municipal Medical College, Ahmedabad, India
| | - Arundati Kanawala
- Department of Internal Medicine, Smt. Kashibai Navale Medical College and General Hospital, Pune, India
| | - Riddhi Mondal
- Department of Internal Medicine, Jagannath Gupta Institute of Medical Sciences and Hospital, Kolkata, India
| | - Aamna Dilshad
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Anum Akbar
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
8
|
Zhang DW, Zhu YB, Zhou SJ, Chen XH, Li HB, Liu WJ, Wu ZQ, Chen Q, Cao H. Maternal cardiovascular health in early pregnancy and the risk of congenital heart defects in offspring. BMC Pregnancy Childbirth 2024; 24:325. [PMID: 38671408 PMCID: PMC11047036 DOI: 10.1186/s12884-024-06529-5] [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: 10/25/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is the predominant birth defect. This study aimed to explore the association between maternal cardiovascular health (CVH) and the CHD risk in offspring. METHODS We used the prospective data from the Fujian Birth Cohort Study, collected from March 2019 to December 2022 on pregnant women within 14 weeks of gestation. Overall maternal CVH was assessed by seven CVH metrics (including physical activity, smoking, sleep duration, body mass index, blood pressure, total cholesterol, and fasting plasma glucose), with each metric classified as ideal, intermediate or poor with specific points. Participants were further allocated into high, moderate and low CVH categories based on the cumulative CVH score. The association with offspring CHD was determined with log-binominal regression models. RESULTS A total of 19810 participants aged 29.7 (SD: 3.9) years were included, with 7846 (39.6%) classified as having high CVH, 10949 (55.3%) as having moderate CVH, and 1015 (5.1%) as having low CVH. The average offspring CHD rate was 2.52%, with rates of 2.35%, 2.52% and 3.84% across the high, moderate and low CVH categories, respectively (P = 0.02). Adjusted relative risks (RRs) of having offspring CHD were 0.64 (95% CI: 0.45-0.90, P = 0.001) for high CVH and 0.67 (95% CI: 0.48-0.93, P = 0.02) for moderate CVH compared to low CVH. For individual metrics, only ideal total cholesterol was significantly associated with lower offspring CHD (RR: 0.73, 95% CI: 0.59-0.83, P = 0.002). CONCLUSIONS Pregnant women of high or moderate CVH categories in early pregnancy had reduced risks of CHD in offspring, compared to those of low CVH. It is important to monitor and improve CVH during pre-pregnancy counseling and early prenatal care.
Collapse
Affiliation(s)
- Dan-Wei Zhang
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.966 Hengyu Road, Jinan District, Fuzhou, 350014, People's Republic of China
| | - Yi-Bing Zhu
- Division of Birth Cohort Study, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, People's Republic of China
| | - Si-Jia Zhou
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.966 Hengyu Road, Jinan District, Fuzhou, 350014, People's Republic of China
| | - Xiu-Hua Chen
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.966 Hengyu Road, Jinan District, Fuzhou, 350014, People's Republic of China
| | - Hai-Bo Li
- Division of Birth Cohort Study, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, People's Republic of China
| | - Wen-Juan Liu
- Division of Birth Cohort Study, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, People's Republic of China
- Division of Birth Cohort Study, Fujian Children's Hospital, Fuzhou, People's Republic of China
| | - Zheng-Qin Wu
- Division of Birth Cohort Study, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, People's Republic of China
- Division of Birth Cohort Study, Fujian Obstetrics and Gynecology Hospital, Fuzhou, People's Republic of China
| | - Qiang Chen
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.966 Hengyu Road, Jinan District, Fuzhou, 350014, People's Republic of China.
| | - Hua Cao
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.966 Hengyu Road, Jinan District, Fuzhou, 350014, People's Republic of China.
| |
Collapse
|
9
|
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
|
10
|
Favara G, Maugeri A, Magnano San Lio R, Barchitta M, Agodi A. Exploring Gene-Diet Interactions for Mother-Child Health: A Systematic Review of Epidemiological Studies. Nutrients 2024; 16:994. [PMID: 38613027 PMCID: PMC11013682 DOI: 10.3390/nu16070994] [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: 02/27/2024] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Maternal-child health suggests the critical impact of maternal nutrition during the pre-conception and gestational periods, with some genetic variants also playing a significant role. Our systematic review provides an overview of epidemiological studies exploring the interactions between genetic variants, maternal dietary habits, and neonatal and/or maternal pregnancy outcomes. METHODS From its inception until June 2023, we conducted a comprehensive literature search on PubMed, Embase, and Web of Science databases. RESULTS On a total of 29 epidemiological studies, 11 studies were conducted to explore the interplay between genetic variants and dietary factors, focusing on the risks associated with gestational diabetes mellitus, hypertensive disorders of pregnancy, recurrent spontaneous abortion, recurrent pregnancy loss, iron deficiency anemia, and gestational weight gain. Concerning neonatal outcomes, six studies investigated the interplay between genetic variants, dietary factors, and anthropometric measures, while eight studies delved into abnormal embryonic development, two studies focused on preterm birth, and two studies explored other neonatal outcomes. CONCLUSIONS Deeply understanding gene-diet interactions could be useful in developing highly personalized approaches to maternal and child nutrition, as well as in exploring the potential implications in disease prevention and the promotion of the long-term well-being of both mothers and their offspring.
Collapse
Affiliation(s)
| | | | | | | | - Antonella Agodi
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, 95123 Catania, Italy; (G.F.); (A.M.); (R.M.S.L.); (M.B.)
| |
Collapse
|
11
|
Nakano H, Nakano A. The role of metabolism in cardiac development. Curr Top Dev Biol 2024; 156:201-243. [PMID: 38556424 DOI: 10.1016/bs.ctdb.2024.01.005] [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: 04/02/2024]
Abstract
Metabolism is the fundamental process that sustains life. The heart, in particular, is an organ of high energy demand, and its energy substrates have been studied for more than a century. In recent years, there has been a growing interest in understanding the role of metabolism in the early differentiation of pluripotent stem cells and in cancer research. Studies have revealed that metabolic intermediates from glycolysis and the tricarboxylic acid cycle act as co-factors for intracellular signal transduction, playing crucial roles in regulating cell behaviors. Mitochondria, as the central hub of metabolism, are also under intensive investigation regarding the regulation of their dynamics. The metabolic environment of the fetus is intricately linked to the maternal metabolic status, and the impact of the mother's nutrition and metabolic health on fetal development is significant. For instance, it is well known that maternal diabetes increases the risk of cardiac and nervous system malformations in the fetus. Another notable example is the decrease in the risk of neural tube defects when pregnant women are supplemented with folic acid. These examples highlight the profound influence of the maternal metabolic environment on the fetal organ development program. Therefore, gaining insights into the metabolic environment within developing fetal organs is critical for deepening our understanding of normal organ development. This review aims to summarize recent findings that build upon the historical recognition of the environmental and metabolic factors involved in the developing embryo.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States; Cardiology Division, Department of Medicine, UCLA, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States; Molecular Biology Institute, UCLA, Los Angeles, CA, United States; Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
12
|
Masuda Y, Nagayasu Y, Murakami H, Nishie R, Morita N, Hashida S, Daimon A, Nunode M, Maruoka H, Yoo M, Sano T, Odanaka Y, Fujiwara S, Fujita D, Okamoto N, Ohmichi M. Triple repeated fetal congenital heart disease linked to PLD1 mutation: a case report. J Med Case Rep 2023; 17:411. [PMID: 37770978 PMCID: PMC10540367 DOI: 10.1186/s13256-023-04149-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Congenital heart disease occurs in approximately 1 in 100 cases. Although sibling occurrence is high (3-9%), the causative genes for this disease are still being elucidated. PLD1 (Phospholipase D1) is a recently discovered gene; however, few case reports have been published on it. In this report, we describe a case of triplicate fetal congenital heart disease that was diagnosed as a PDL1 mutation. Our objective is to explore the clinical manifestations of PLD1 mutations in this particular case. CASE PRESENTATION A 32-year-old Japanese woman (gravida, para 0) was introduced since fetus four chamber view was not clear and was diagnosed with ductus arteriosus-dependent left ventricular single ventricle and pulmonary atresia at 21 weeks and 1 day of gestation during her first pregnancy. Artificial abortion using Gemeprost was performed at 21 weeks and 5 days of gestation. The second pregnancy was diagnosed as pulmonary atresia with intact ventricular septum with cardiomegaly, a cardiothoracic area ratio of more than 35%, and a circulatory shunt at 13 weeks and 3 days of gestation. Subsequently, intrauterine fetal death was confirmed at 14 weeks and 3 days of gestation. Regarding the third pregnancy, fetal ultrasonography at 11 weeks and 5 days of gestation showed mild fetal hydrops and moderate tricuspid valve regurgitation. At 16 weeks and 5 days of gestation, the fetus was suspected to have a left ventricular-type single ventricle, trace right ventricle, pulmonary atresia with intact ventricular septum, or cardiomyopathy. Cardiac function gradually declined at 26 weeks of gestation, and intrauterine fetal death was confirmed at 27 weeks and 5 days of gestation. The fourth pregnancy resulted in a normal heart with good progression and no abnormal baby. We submitted the first and second fetuses' umbilical cord, third fetus' placenta, and the fourth fetus' blood to genetic testing using whole exome analysis with next generation sequencing. Genetic analysis identified hemizygous PLD1 mutations in the first, second, and third fetuses. The fourth fetus was heterozygous. In addition, the parents were heterozygous for PLD1. This case is based on three consecutive cases of homozygosity for the PLD1 gene in the sibling cases and the fetuses with recurrent right ventricular valve dysplasia. This will elucidate the cause of recurrent congenital heart disease and intrauterine fetal death and may serve as an indicator for screening the next fetus. To date, homozygous mutations in PLD1 that repeat three times in a row are not reported, only up to two times. The novelty of this report is that it was repeated three times, followed by a heterozygous live birth. CONCLUSIONS This report is consistent with previous reports that mutations in PLD1 cause right ventricular valve dysplasia. However, there have been few case reports of PLD1 mutations, and we hope that this report will contribute to elucidate the causes of congenital heart disease, especially right ventricular valve dysplasia, and that the accumulation of such information will provide more detailed information on PLD1 mutations in heart disease.
Collapse
Affiliation(s)
- Yuki Masuda
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
- Department of Obstetrics and Gynecology, Saiseikai Suita Hospital, Suita, Japan
| | - Yoko Nagayasu
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan.
| | - Hikaru Murakami
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Ruri Nishie
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Natsuko Morita
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Sosuke Hashida
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Atsushi Daimon
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Misa Nunode
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Hiroshi Maruoka
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Masae Yoo
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Takumi Sano
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Yutaka Odanaka
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Satoe Fujiwara
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Daisuke Fujita
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| |
Collapse
|
13
|
Pei G, Chen L, Wang Y, He C, Fu C, Wei Q. Role of miR-182 in cardiovascular and cerebrovascular diseases. Front Cell Dev Biol 2023; 11:1181515. [PMID: 37228653 PMCID: PMC10203221 DOI: 10.3389/fcell.2023.1181515] [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: 03/07/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
The treatment of cardiovascular and cerebrovascular diseases have undergone major advances in recent decades, allowing for a more effective prevention of cardiovascular and cerebrovascular events. However, cardiac and cerebral atherothrombotic complications still account for substantial morbidity and mortality worldwide. Novel therapeutic strategies are critical to improve patient outcomes following cardiovascular diseases. miRNAs are small non-coding RNAs, that regulate gene expression. Here, we discuss the role of miR-182 in regulating myocardial proliferation, migration, hypoxia, ischemia, apoptosis and hypertrophy in atherosclerosis, CAD, MI, I/R injury, organ transplant, cardiac hypertrophy, hypertension, heart failure, congenital heart disease and cardiotoxicity. Besides, we also summarize the current progress of miR-182 therapeutics in clinical development and discuss challenges that will need to be overcome to enter the clinic for patients with cardiac disease.
Collapse
Affiliation(s)
- Gaiqin Pei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Li Chen
- Department of Rehabilitation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Yang Wang
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Chengqi He
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Quan Wei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Zhang Y, Chen Y, Xu K, Xia S, Aihaiti A, Zhu M, Wang C. Exposure of embryos to phenanthrene impacts the cardiac development in F1 zebrafish larvae and potential reasons. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:52369-52379. [PMID: 36840880 DOI: 10.1007/s11356-023-26165-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/23/2023] [Indexed: 06/18/2023]
Abstract
To explore the impact of embryonic exposure to phenanthrene (Phe), a typical tricyclic polycyclic aromatic hydrocarbon, on cardiac development in next generation, fertilized zebrafish embryos were exposed to 0.05, 0.5, 5 and 50 nM Phe for 96 h, and then transferred to clear water and raised to adulthood. The cardiac development in F1 larvae generated by adult females or males mated with unexposed zebrafish was assessed. Malformation and dysfunction of the heart, such as increased heart rate, arrhythmia, enlarged heart and abnormal contraction, were shown in both paternal and maternal F1 larvae. A greater impact on the distance between the sinus venosus and bulbus arteriosus was exhibited in maternal F1 larvae, while paternal F1 larvae displayed a more severe impact on heart rate and arrhythmia. The transcription of genes related to cardiac development was disturbed in F1 larvae. DNA methylation levels in the promoter of some genes were associated with their transcription. The expression of acetylated histone H3K9Ac and H3K14Ac in maternal F1 larvae was no significantly changed, but was significantly downregulated in paternal F1 larvae, which might be associated with the downregulated transcription of tbx5. These results indicate that exposure to Phe during embryogenesis adversely affects cardiac development in F1 generation, and the effects and toxic mechanisms showed sex-linked hereditary differences, highlighting the risk of Phe exposure in early life to heart health in next generation.
Collapse
Affiliation(s)
- Ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Ke Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Siyu Xia
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Ailifeire Aihaiti
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Mingxia Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China.
| |
Collapse
|
15
|
Lu E, Wu L, Chen B, Xu S, Fu Z, Wu Y, Wu Y, Gu H. Maternal Serum tRNA-Derived Fragments (tRFs) as Potential Candidates for Diagnosis of Fetal Congenital Heart Disease. J Cardiovasc Dev Dis 2023; 10:jcdd10020078. [PMID: 36826574 PMCID: PMC9968204 DOI: 10.3390/jcdd10020078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is one of the most predominant birth defects that causes infant death worldwide. The timely and successful surgical treatment of CHD on newborns after delivery requires accurate detection and reliable diagnosis during pregnancy. However, there are no biomarkers that can serve as an early diagnostic factor for CHD patients. tRNA-derived fragments (tRFs) have been reported to play an important role in the occurrence and progression of numerous diseases, but their roles in CHD remains unknown. METHODS High-throughput sequencing was performed on the peripheral blood of pregnant women with an abnormal fetal heart and a normal fetal heart, and 728 differentially expressed tRFs/tiRNAs were identified, among which the top 18 tRFs/tiRNAs were selected as predictive biomarkers of CHD. Then, a quantitative reverse transcriptase polymerase chain reaction verified the expression of tRFs/tiRNAs in more clinical samples, and the correlation between tRFs/tiRNAs abnormalities and CHD was analyzed. RESULTS tRF-58:74-Gly-GCC-1 and tiRNA-1:35-Leu-CAG-1-M2 may be promising biomarkers. Through further bioinformatics analysis, we predicted that TRF-58:744-GLy-GCC-1 could induce CHD by influencing biological metabolic processes. CONCLUSIONS Our results provide a theoretical basis for the abnormally expressed tRF-58:74-Gly-GCC-1 in maternal peripheral blood as a new potential biomarker for the accurate diagnosis of CHD during pregnancy.
Collapse
Affiliation(s)
- Enkang Lu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Central Laboratory of Jiangsu Provincial Maternal and Child Health Care Hospital, Maternal and Child Branch of the First Affiliated Hospital of Nanjing Medical University, Nanjing 210036, China
| | - Lijun Wu
- Department of Ultrasound, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Bin Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shipeng Xu
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Ziyi Fu
- Central Laboratory of Jiangsu Provincial Maternal and Child Health Care Hospital, Maternal and Child Branch of the First Affiliated Hospital of Nanjing Medical University, Nanjing 210036, China
| | - Yun Wu
- Department of Ultrasound, Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Nanjing 210004, China
- Correspondence: (Y.W.); (Y.W.); (H.G.); Tel.: +86-189-0518-0170 (Y.W.); +86-139-5194-5999 (Y.W.); +86-139-0159-2427 (H.G.)
| | - Yanhu Wu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Correspondence: (Y.W.); (Y.W.); (H.G.); Tel.: +86-189-0518-0170 (Y.W.); +86-139-5194-5999 (Y.W.); +86-139-0159-2427 (H.G.)
| | - Haitao Gu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Correspondence: (Y.W.); (Y.W.); (H.G.); Tel.: +86-189-0518-0170 (Y.W.); +86-139-5194-5999 (Y.W.); +86-139-0159-2427 (H.G.)
| |
Collapse
|
16
|
do Amaral Lopes SAV, Guimarães ICB, de Oliva Costa SF, Acosta AX, Abe-Sandes K, Mendes CMC. Risk factors for critical and complex congenital heart diseases: Case-control study. PROGRESS IN PEDIATRIC CARDIOLOGY 2023. [DOI: 10.1016/j.ppedcard.2022.101612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
17
|
Suo MJ, Chen WC, Xu ZQ, Tian GX, Li T, Li P, Sheng W, Huang GY, Ma XJ. X-linked BCOR variants identified in Chinese Han patients with congenital heart disease. J Gene Med 2023; 25:e3461. [PMID: 36314054 DOI: 10.1002/jgm.3461] [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: 08/15/2022] [Revised: 10/08/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Congenital heart disease (CHD) frequently manifests as a complex phenotype and approximately one-third of cases may be caused by genetic factors. BCOR, an X-linked gene encoding the corepressor of BCL6, has been demonstrated to be closely involved in human heart development. However, whether BCOR variants represent the genetic etiology underlying CHD needs further investigation. METHODS We performed whole exome sequencing on CHD nuclear families and identified a candidate gene, BCOR, by robust bioinformatic analysis and medical literature searches. Targeted DNA sequencing of the candidate gene was conducted and then the association between variants and the risk of developing CHD was analyzed. The effects of BCOR mutations on gene expression, localization, protein interaction, and signaling pathways were evaluated in vitro. RESULTS We identified a BCOR hemizygous missense variant (c.1448C>T, p.Pro483Leu) in a male proband presented with CHD/heterotaxy. Sanger sequencing confirmed that this variant was inherited from his asymptomatic mother. Interestingly, through literature searches, we observed another novel BCOR hemizygous missense variant (c.1619G>A, p.Arg540Gln) in a CHD patient with heterotaxy, supporting the pathogenic evidence of BCOR variants. Functional experiments conducted in vitro revealed that the variant p.Pro483Leu altered the subcellular localization of BCOR protein, disrupted its interaction with BCL6, and significantly promoted cell proliferation, whereas the variant p.Arg540Gln displayed no obvious effects. Nevertheless, transcriptional analysis revealed that down-regulation of BCOR substantially enhanced the activities of mitogen-activated protein and phosphoinositide 3-kinase-AKT signaling pathways, which are closely attributed to heart development. Targeted sequencing of 932 sporadic CHD patients enriched nine variants of BCOR predicted as likely rare and damaging and a septal defect was present in 81.8% (9/11) of them, including the two probands, which was consistent with the possible phenotype caused by BCOR defects. CONCLUSIONS The findings of the present study indicate that variants in BCOR may predispose individuals to CHD in the Chinese Han population.
Collapse
Affiliation(s)
- Mei-Jiao Suo
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei-Cheng Chen
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Zi-Qing Xu
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Gui-Xiang Tian
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Ting Li
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Ping Li
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei Sheng
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Guo-Ying Huang
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Xiao-Jing Ma
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
18
|
Wang YJ, Zhang X, Lam CK, Guo H, Wang C, Zhang S, Wu JC, Snyder M, Li J. Systems analysis of de novo mutations in congenital heart diseases identified a protein network in the hypoplastic left heart syndrome. Cell Syst 2022; 13:895-910.e4. [PMID: 36167075 PMCID: PMC9671831 DOI: 10.1016/j.cels.2022.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/14/2022] [Accepted: 09/02/2022] [Indexed: 01/26/2023]
Abstract
Despite a strong genetic component, only a few genes have been identified in congenital heart diseases (CHDs). We introduced systems analyses to uncover the hidden organization on biological networks of mutations in CHDs and leveraged network analysis to integrate the protein interactome, patient exomes, and single-cell transcriptomes of the developing heart. We identified a CHD network regulating heart development and observed that a sub-network also regulates fetal brain development, thereby providing mechanistic insights into the clinical comorbidities between CHDs and neurodevelopmental conditions. At a small scale, we experimentally verified uncharacterized cardiac functions of several proteins. At a global scale, our study revealed developmental dynamics of the network and observed its association with the hypoplastic left heart syndrome (HLHS), which was further supported by the dysregulation of the network in HLHS endothelial cells. Overall, our work identified previously uncharacterized CHD factors and provided a generalizable framework applicable to studying many other complex diseases. A record of this paper's Transparent Peer Review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yuejun Jessie Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, the Bakar Computational Health Sciences Institute, the Parker Institute for Cancer Immunotherapy, and the Department of Neurology, School of Medicine, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
| | - Xicheng Zhang
- Department of Genetics and the Center for Genomics and Personalized Medicine, School of Medicine, Stanford University, 291 Campus Dr., Stanford, CA 94305, USA
| | - Chi Keung Lam
- Stanford Cardiovascular Institute, School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA; Department of Medicine, Division of Cardiology, School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Hongchao Guo
- Stanford Cardiovascular Institute, School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA; Department of Medicine, Division of Cardiology, School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA
| | - Cheng Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, the Bakar Computational Health Sciences Institute, the Parker Institute for Cancer Immunotherapy, and the Department of Neurology, School of Medicine, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
| | - Sai Zhang
- Department of Genetics and the Center for Genomics and Personalized Medicine, School of Medicine, Stanford University, 291 Campus Dr., Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA; Department of Medicine, Division of Cardiology, School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA; Department of Radiology, Stanford University School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA
| | - Michael Snyder
- Department of Genetics and the Center for Genomics and Personalized Medicine, School of Medicine, Stanford University, 291 Campus Dr., Stanford, CA 94305, USA; Stanford Cardiovascular Institute, School of Medicine, Stanford University, 265 Campus Dr., Stanford, CA 94305, USA.
| | - Jingjing Li
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, the Bakar Computational Health Sciences Institute, the Parker Institute for Cancer Immunotherapy, and the Department of Neurology, School of Medicine, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA.
| |
Collapse
|
19
|
Wu X, Chen Y, Luz A, Hu G, Tokar EJ. Cardiac Development in the Presence of Cadmium: An in Vitro Study Using Human Embryonic Stem Cells and Cardiac Organoids. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:117002. [PMID: 36321828 PMCID: PMC9628677 DOI: 10.1289/ehp11208] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 05/24/2023]
Abstract
BACKGROUND Exposure to cadmium (Cd) is associated with cardiovascular diseases. Maternal Cd exposure is a significant risk factor for congenital heart disease. However, mechanisms of Cd on developmental cardiotoxicity are not well defined. OBJECTIVES We evaluated the effects of Cd on the different stages (mesoderm, cardiac induction, cardiac function) of cardiac development using an early embryo development in vitro model and two- or three-dimensional (2- or 3D) cardiomyocyte and cardiac organoid formation models mimicking early cardiac development. METHODS Embryonic stem cells (ESCs) form 3D aggregates, called embryoid bodies, that recapitulate events involved with early embryogenesis (e.g., germ layer formation). This model was used for early germ layer formation and signaling pathway identification. The 2D cardiomyocyte differentiation from the NKX2-5eGFP/w human ESCs model was used to explore the effects of Cd exposure on cardiomyocyte formation and to model mesoderm differentiation and cardiac induction, allowing us to explore different developmental windows of Cd toxicity. The 3D cardiac organoid model was used in evaluating the effects of Cd exposure on contractility and cardiac development. RESULTS Cd (0.6μM; 110 ppb) lowered the differentiation of embryoid bodies to mesoderm via suppression of Wnt/β-catenin-signaling pathways. During early mesoderm induction, the mesoderm-associated transcription factors MESP1 and EOMES showed a transient up-regulation, which decreased later in the cardiac induction stage. Cd (0.15μM) lowered mesoderm formation and cardiac induction through suppression of the transcription factors and mesoderm marker genes HAND1, SNAI2, HOPX, and the cardiac-specific genes NKX2-5, GATA4, troponin T, and alpha-actinin. In addition, Cd-induced histone modifications for both gene activation (H3K4me3) and repression (H3K27me3), which play vital roles in regulating mesoderm commitment markers. The effects of Cd inhibition on cardiomyocyte differentiation were confirmed in 3D cardiac organoids. DISCUSSION In conclusion, using a human ESC-derived 2D/3D in vitro differentiation model system and cardiac organoids, we demonstrated that low-dose Cd suppressed mesoderm formation through mesoderm gene histone modification, thus inhibiting cardiomyocyte differentiation and cardiac induction. The studies provide valuable insights into cellular events and molecular mechanisms associated with Cd-induced congenital heart disease. https://doi.org/10.1289/EHP11208.
Collapse
Affiliation(s)
- Xian Wu
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute for Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Yichang Chen
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute for Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Anthony Luz
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute for Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Erik J. Tokar
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute for Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| |
Collapse
|
20
|
Schroeder AM, Nielsen T, Lynott M, Vogler G, Colas AR, Bodmer R. Nascent polypeptide-Associated Complex and Signal Recognition Particle have cardiac-specific roles in heart development and remodeling. PLoS Genet 2022; 18:e1010448. [PMID: 36240221 PMCID: PMC9604979 DOI: 10.1371/journal.pgen.1010448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 10/26/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022] Open
Abstract
Establishing a catalog of Congenital Heart Disease (CHD) genes and identifying functional networks would improve our understanding of its oligogenic underpinnings. Our studies identified protein biogenesis cofactors Nascent polypeptide-Associated Complex (NAC) and Signal-Recognition-Particle (SRP) as disease candidates and novel regulators of cardiac differentiation and morphogenesis. Knockdown (KD) of the alpha- (Nacα) or beta-subunit (bicaudal, bic) of NAC in the developing Drosophila heart disrupted cardiac developmental remodeling resulting in a fly with no heart. Heart loss was rescued by combined KD of Nacα with the posterior patterning Hox gene Abd-B. Consistent with a central role for this interaction in cardiogenesis, KD of Nacα in cardiac progenitors derived from human iPSCs impaired cardiac differentiation while co-KD with human HOXC12 and HOXD12 rescued this phenotype. Our data suggest that Nacα KD preprograms cardioblasts in the embryo for abortive remodeling later during metamorphosis, as Nacα KD during translation-intensive larval growth or pupal remodeling only causes moderate heart defects. KD of SRP subunits in the developing fly heart produced phenotypes that targeted specific segments and cell types, again suggesting cardiac-specific and spatially regulated activities. Together, we demonstrated directed function for NAC and SRP in heart development, and that regulation of NAC function depends on Hox genes.
Collapse
Affiliation(s)
- Analyne M. Schroeder
- Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- * E-mail: (AMS); (RB)
| | - Tanja Nielsen
- Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Michaela Lynott
- Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Georg Vogler
- Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Alexandre R. Colas
- Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- * E-mail: (AMS); (RB)
| |
Collapse
|
21
|
Miao Q, Dunn S, Wen SW, Lougheed J, Sharif F, Walker M. Associations of congenital heart disease with deprivation index by rural-urban maternal residence: a population-based retrospective cohort study in Ontario, Canada. BMC Pediatr 2022; 22:476. [PMID: 35931992 PMCID: PMC9356510 DOI: 10.1186/s12887-022-03498-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The risk of congenital heart disease (CHD) has been found to vary by maternal socioeconomic status (SES) and rural-urban residence. In this study, we examined associations of CHD with two maternal SES indicators and stratified the analysis by maternal rural-urban residence. METHODS This was a population-based retrospective cohort study. We included all singleton stillbirths and live hospital births from April 1, 2012 to March 31, 2018 in Ontario, Canada. We linked the BORN Information System and Canadian Institute for Health Information databases. Multivariable logistic regression models were used to examine associations of CHD with material deprivation index (MDI), social deprivation index (SDI), and maternal residence while adjusting for maternal age at birth, assisted reproductive technology, obesity, pre-pregnancy maternal health conditions, mental health illness before and during pregnancy, substance use during pregnancy, and infant's sex. MDI and SDI were estimated at a dissemination area level in Ontario and were categorized into quintiles (Q1-Q5). RESULTS This cohort study included 798,173 singletons. In maternal urban residence, the p trend (Cochran-Armitage test) was less than 0.0001 for both MDI and SDI; while for rural residence, it was 0.002 and 0.98, respectively. Infants living in the most materially deprived neighbourhoods (MDI Q5) had higher odds of CHD (aOR: 1.21, 95% CI: 1.12-1.29) compared to Q1. Similarly, infants living in the most socially deprived neighbourhoods (SDI Q5) had an 18% increase in the odds of CHD (aOR: 1.18, 95% CI: 1.1-1.26) compared to Q1. Rural infants had a 13% increase in the odds of CHD compared to their urban counterparts. After stratifying by maternal rural-urban residence, we still detected higher odds of CHD with two indices in urban residence but only MDI in rural residence. CONCLUSION Higher material and social deprivation and rural residence were associated with higher odds of CHD. Health interventions and policies should reinforce the need for optimal care for all families, particularly underprivileged families in both rural and urban regions. Future studies should further investigate the effect of social deprivation on the risk of CHD development.
Collapse
Affiliation(s)
- Qun Miao
- BORN Ontario, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada. .,Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada. .,School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Canada.
| | - Sandra Dunn
- BORN Ontario, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada.,Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada.,School of Nursing, University of Ottawa, Ottawa, Ontario, Canada
| | - Shi Wu Wen
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Canada.,OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Obstetrics & Gynecology, University of Ottawa, Faculty of Medicine, Ottawa, Canada
| | - Jane Lougheed
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada.,Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Fayza Sharif
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Canada
| | - Mark Walker
- BORN Ontario, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada.,Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Canada.,OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Obstetrics & Gynecology, University of Ottawa, Faculty of Medicine, Ottawa, Canada
| |
Collapse
|
22
|
Tan M, Wang X, Liu H, Peng X, Yang Y, Yu H, Xu L, Li J, Cao H. Genetic Diagnostic Yield and Novel Causal Genes of Congenital Heart Disease. Front Genet 2022; 13:941364. [PMID: 35910219 PMCID: PMC9326225 DOI: 10.3389/fgene.2022.941364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Congenital heart disease (CHD) is the most common congenital malformation in fetuses and neonates, which also represents a leading cause of mortality. Although significant progress has been made by emerging advanced technologies in genetic etiology diagnosis, the causative genetic mechanisms behind CHD remain poorly understood and more than half of CHD patients lack a genetic diagnosis. Unlike carefully designed large case-control cohorts by multicenter trials, we designed a reliable strategy to analyze case-only cohorts to utilize clinical samples sufficiently. Combined low-coverage whole-genome sequencing (WGS) and whole-exome sequencing (WES) were simultaneously conducted in a patient-only cohort for identifying genetic etiologies and exploring candidate, or potential causative CHD-related genes. A total of 121 sporadic CHD patients were recruited and 34.71% (95% CI, 26.80 to 43.56) was diagnosed with genetic etiologies by low-coverage WGS and WES. Chromosomal abnormalities and damaging variants of CHD-related genes could explain 24.79% (95% CI, 17.92 to 33.22) and 18.18% (95% CI, 12.26 to 26.06) of CHD patients, separately, and 8.26% (95% CI, 4.39 to 14.70) of them have simultaneously detected two types of variants. Deletion of chromosome 22q11.2 and pathogenic variants of the COL3A1 gene were the most common recurrent variants of chromosomal abnormalities and gene variants, respectively. By in-depth manual interpretation, we identified eight candidate CHD-causing genes. Based on rare disease-causing variants prediction and interaction analysis with definitive CHD association genes, we proposed 86 genes as potential CHD-related genes. Gene Ontology (GO) enrichment analysis of the 86 genes revealed regulation-related processes were significantly enriched and processes response to regulation of muscle adaptation might be one of the underlying molecular mechanisms of CHD. Our findings and results provide new insights into research strategies and underlying mechanisms of CHD.
Collapse
Affiliation(s)
- Meihua Tan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Genomics Co., Ltd, Shenzhen, China
| | - Xinrui Wang
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Hongjie Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Peng
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - You Yang
- BGI Genomics Co., Ltd, Shenzhen, China
| | - Haifei Yu
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital, Fuzhou, China
- Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, Affiliated Hospital of Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, China
- *Correspondence: Liangpu Xu, ; Jia Li, ; Hua Cao,
| | - Jia Li
- BGI Genomics Co., Ltd, Shenzhen, China
- Hebei Industrial Technology Research Institute of Genomics in Maternal and Child Health, Shijiazhuang BGI Genomics Co., Ltd, Shijiazhuang, China
- *Correspondence: Liangpu Xu, ; Jia Li, ; Hua Cao,
| | - Hua Cao
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Liangpu Xu, ; Jia Li, ; Hua Cao,
| |
Collapse
|
23
|
Liu Y, Zhang Q, Yang L, Tian W, Yang Y, Xie Y, Li J, Yang L, Gao Y, Xu Y, Liu J, Wang Y, Yan J, Li G, Shen Y, Qi Z. Metformin Attenuates Cardiac Hypertrophy Via the HIF-1α/PPAR-γ Signaling Pathway in High-Fat Diet Rats. Front Pharmacol 2022; 13:919202. [PMID: 35833024 PMCID: PMC9271627 DOI: 10.3389/fphar.2022.919202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 12/19/2022] Open
Abstract
Coronary artery disease (CAD) and cardiac hypertrophy (CH) are two main causes of ischemic heart disease. Acute CAD may lead to left ventricular hypertrophy (LVH). Long-term and sustained CH is harmful and can gradually develop into cardiac insufficiency and heart failure. It is known that metformin (Met) can alleviate CH; however, the molecular mechanism is not fully understood. Herein, we used high-fat diet (HFD) rats and H9c2 cells to induce CH and clarify the potential mechanism of Met on CH. We found that Met treatment significantly decreased the cardiomyocyte size, reduced lactate dehydrogenase (LDH) release, and downregulated the expressions of hypertrophy markers ANP, VEGF-A, and GLUT1 either in vivo or in vitro. Meanwhile, the protein levels of HIF-1α and PPAR-γ were both decreased after Met treatment, and administrations of their agonists, deferoxamine (DFO) or rosiglitazone (Ros), markedly abolished the protective effect of Met on CH. In addition, DFO treatment upregulated the expression of PPAR-γ, whereas Ros treatment did not affect the expression of HIF-1α. In conclusion, Met attenuates CH via the HIF-1α/PPAR-γ signaling pathway.
Collapse
Affiliation(s)
- Yuansheng Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Qian Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Lei Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Tianjin, China
| | - Wencong Tian
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yinan Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yuhang Xie
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yachen Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Guoxun Li
- Xinjiang Production and Construction Corps Hospital, Urumqi, China
- Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| | - Yanna Shen
- Department of Microbiology, School of Laboratory Medicine, Tianjin Medical University, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Xinjiang Production and Construction Corps Hospital, Urumqi, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Guoxun Li, ; Yanna Shen, ; Zhi Qi,
| |
Collapse
|
24
|
Sonawane AR, Aikawa E, Aikawa M. Connections for Matters of the Heart: Network Medicine in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:873582. [PMID: 35665246 PMCID: PMC9160390 DOI: 10.3389/fcvm.2022.873582] [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: 02/11/2022] [Accepted: 04/19/2022] [Indexed: 01/18/2023] Open
Abstract
Cardiovascular diseases (CVD) are diverse disorders affecting the heart and vasculature in millions of people worldwide. Like other fields, CVD research has benefitted from the deluge of multiomics biomedical data. Current CVD research focuses on disease etiologies and mechanisms, identifying disease biomarkers, developing appropriate therapies and drugs, and stratifying patients into correct disease endotypes. Systems biology offers an alternative to traditional reductionist approaches and provides impetus for a comprehensive outlook toward diseases. As a focus area, network medicine specifically aids the translational aspect of in silico research. This review discusses the approach of network medicine and its application to CVD research.
Collapse
Affiliation(s)
- Abhijeet Rajendra Sonawane
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Nana L, Lu L, Zhen L, Ying D, Meixian W, Zhao J, Zeng S, Hong K, Yanping W, Jun Z, Jianxin Z, Ping Y. The effect of maternal polycyclic aromatic hydrocarbons exposure and methylation levels of CHDs-candidate genes on the risk of congenital heart diseases. Prenat Diagn 2022; 42:1142-1154. [PMID: 35556253 DOI: 10.1002/pd.6167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/27/2022] [Accepted: 05/08/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To evaluate the impact of maternal exposure to polycyclic aromatic hydrocarbons (PAHs) and methylation levels of CHDs-candidate genes on the risk of congenital heart diseases (CHDs), and the effect of PAHs exposure on DNA methylation states. METHODS A case-control study involving 60 mother -fetus pairs was performed by measuring 1-OHPG concentration in maternal urine and methylation levels of 20 CHDs-candidate genes in cord bloods. Logistic regression models were applied to determine the effect of maternal PAHs exposure and fetal methylation levels on the risk of CHDs. Spearman correlation was performed to correlate PAHs exposure and methylation levels. RESULTS Maternal higher PAHs exposure was associated with the risk of CHDs (aOR = 3.245, 95% CI: 1.060, 9.937) or some subtypes. The methylation levels of 23 amplicons within 11 genes exhibited significant differences between CHDs and controls. Higher methylation of NKX2-5_M1 was associated with decreased risk of CHDs (aOR=0.182, 95% CI:0.034, 0.983). No significant correlations were found between 1-OHPG concentration and methylation levels of NKX2-5_M1. CONCLUSIONS Maternal PAHs exposure was linked with CHDs. Higher methylation of the upstream sequence of NKX2-5 promoter decreased the risk of CHDs. There was no correlation between maternal PAHs exposure and the methylation level of NKX2-5. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Li Nana
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Li Lu
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Liu Zhen
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Deng Ying
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Wang Meixian
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Jinju Zhao
- Department of Gynecology and Obstetrics, Xichang people's Hospital, Xichang, China
| | - Shengli Zeng
- Department of Gynecology and Obstetrics, Rongchang Maternal and Child care Hospital, Chongqing, China
| | - Kang Hong
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Wang Yanping
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Zhu Jun
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Zhao Jianxin
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Yu Ping
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Sec.3 No.17, South RenMin Road, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Yang P, Yang Y, He X, Sun P, Zhang Y, Song X, Tian Y, Zong T, Ma J, Chen X, Lv Q, Yu T, Jiang Z. miR-153-3p Targets βII Spectrin to Regulate Formaldehyde-Induced Cardiomyocyte Apoptosis. Front Cardiovasc Med 2022; 8:764831. [PMID: 34977182 PMCID: PMC8714842 DOI: 10.3389/fcvm.2021.764831] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Formaldehyde (FA) is ubiquitous in the environment and can be transferred to the fetus through placental circulation, causing miscarriage and congenital heart disease (CHD). Studies have shown that βII spectrin is necessary for cardiomyocyte survival and differentiation, and its loss leads to heart development defects and cardiomyocyte apoptosis. Additionally, previous studies have demonstrated that miRNA is essential in heart development and remodeling. However, whether miRNA regulates FA-induced CHD and cardiomyocyte apoptosis remains unclear. Methods: Using commercially available rat embryonic cardiomyocytes and a rat model of fetal cardiomyocyte apoptosis. Real-time quantitative PCR (RT-qPCR) and Western blot were performed to examine the level of miR-153-3p, βII spectrin, caspase 7, cleaved caspase7, Bax, Bcl-2 expression in embryonic cardiomyocytes and a rat model of fetal cardiomyocyte apoptosis. Apoptotic cell populations were evaluated by flow cytometry and Tunel. Luciferase activity assay and RNA pull-down assay were used to detect the interaction between miR-153-3p and βII spectrin. Masson's trichrome staining detects the degree of tissue fibrosis. Fluorescence in situ hybridization (FISH) and Immunohistochemistry were used to detect the expression of miR-153-3p and βII spectrin in tissues. Results: Using commercially available rat embryonic cardiomyocytes and a rat model of fetal cardiomyocyte apoptosis, our studies indicate that miR-153-3p plays a regulatory role by directly targeting βII spectrin to promote cardiomyocyte apoptosis. miR-153-3p mainly regulates cardiomyocyte apoptosis by regulating the expression of caspase7, further elucidating the importance of apoptosis in heart development. Finally, the results with our animal model revealed that targeting the miR-153-3p/βII spectrin pathway effectively regulated FA-induced damage during heart development. Recovery experiments with miR-153-3p antagomir resulted in the reversal of FA-induced cardiomyocyte apoptosis and fetal cardiac fibrosis. Conclusion: This study investigated the molecular mechanism underpinning the role of βII spectrin in FA-induced CHD and the associated upstream miRNA pathway. The study findings suggest that miR-153-3p may provide a potential target for the clinical diagnosis and treatment of CHD.
Collapse
Affiliation(s)
- Panyu Yang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao, China
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pin Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Zhang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoxia Song
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yu Tian
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tingyu Zong
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianmin Ma
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaofei Chen
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qifeng Lv
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhirong Jiang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Akiel M. The genetic architecture behind congenital heart disease: A review of genetic and epigenetic factors. JOURNAL OF NATURE AND SCIENCE OF MEDICINE 2022. [DOI: 10.4103/jnsm.jnsm_126_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
28
|
Rosenthal SB, Willsey HR, Xu Y, Mei Y, Dea J, Wang S, Curtis C, Sempou E, Khokha MK, Chi NC, Willsey AJ, Fisch KM, Ideker T. A convergent molecular network underlying autism and congenital heart disease. Cell Syst 2021; 12:1094-1107.e6. [PMID: 34411509 PMCID: PMC8602730 DOI: 10.1016/j.cels.2021.07.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/10/2021] [Accepted: 07/28/2021] [Indexed: 12/29/2022]
Abstract
Patients with neurodevelopmental disorders, including autism, have an elevated incidence of congenital heart disease, but the extent to which these conditions share molecular mechanisms remains unknown. Here, we use network genetics to identify a convergent molecular network underlying autism and congenital heart disease. This network is impacted by damaging genetic variants from both disorders in multiple independent cohorts of patients, pinpointing 101 genes with shared genetic risk. Network analysis also implicates risk genes for each disorder separately, including 27 previously unidentified genes for autism and 46 for congenital heart disease. For 7 genes with shared risk, we create engineered disruptions in Xenopus tropicalis, confirming both heart and brain developmental abnormalities. The network includes a family of ion channels, such as the sodium transporter SCN2A, linking these functions to early heart and brain development. This study provides a road map for identifying risk genes and pathways involved in co-morbid conditions.
Collapse
Affiliation(s)
- Sara Brin Rosenthal
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuxiao Xu
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuan Mei
- Division of Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jeanselle Dea
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sheng Wang
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Charlotte Curtis
- Division of Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emily Sempou
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Neil C Chi
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Arthur Jeremy Willsey
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Kathleen M Fisch
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
29
|
Escarda-Castro E, Herráez MP, Lombó M. Effects of bisphenol A exposure during cardiac cell differentiation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 286:117567. [PMID: 34126515 DOI: 10.1016/j.envpol.2021.117567] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/21/2021] [Accepted: 06/06/2021] [Indexed: 06/12/2023]
Abstract
Heart development requires a precise temporal regulation of gene expression in cardiomyoblasts. Therefore, the transcriptional changes in differentiating cells can lead to congenital heart diseases. Although the genetic mutations underlie most of these alterations, exposure to environmental contaminants, such as bisphenol A (BPA), has been recently considered as a risk factor as well. In this study we investigated the genotoxic and epigenotoxic effects of BPA throughout cardiomyocyte differentiation. H9c2 cells (rat myoblasts) were exposed to 10 and 30 μM BPA before and during the last two days of cardiac-driven differentiation. Then, we have analysed the phenotypic and molecular modifications (at transcriptional, genetic and epigenetic level). The results showed that treated myoblasts developed a skeletal muscle cell-like phenotype. The transcriptional changes induced by BPA in genes codifying proteins involved in heart differentiation and function depend on the window of exposure to BPA. The exposure before differentiation repressed the expression of heart transcription factors (Hand2 and Gata4), whereas exposure during differentiation reduced the expression of cardiac-specific genes (Tnnt2, Myom2, Sln, and Atp2a1). Additionally, significant effects were observed regarding DNA damage and histone acetylation levels after the two periods of BPA exposure: in cells exposed to the toxicant the percentage of DNA repair foci (formed by the co-localization of γH2AX and 53BP1) increased in a dose-dependent manner, whereas the treatment with the toxicant triggered a decrease in the epigenetic marks H3K9ac and H3K27ac. Our in vitro results reveal that BPA seriously interferes with the process of cardiomyocyte differentiation, which could be related to the reported in vivo effects of this toxicant on cardiogenesis.
Collapse
Affiliation(s)
- Enrique Escarda-Castro
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - María Paz Herráez
- Department of Molecular Biology, Faculty of Biology, University of León, Campus Vegazana s/n, León, 24071, Spain
| | - Marta Lombó
- Department of Animal Reproduction, INIA, Av. Puerta de Hierro, 18, Madrid, Spain.
| |
Collapse
|
30
|
Yasuhara J, Garg V. Genetics of congenital heart disease: a narrative review of recent advances and clinical implications. Transl Pediatr 2021; 10:2366-2386. [PMID: 34733677 PMCID: PMC8506053 DOI: 10.21037/tp-21-297] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022] Open
Abstract
Congenital heart disease (CHD) is the most common human birth defect and remains a leading cause of mortality in childhood. Although advances in clinical management have improved the survival of children with CHD, adult survivors commonly experience cardiac and non-cardiac comorbidities, which affect quality of life and prognosis. Therefore, the elucidation of genetic etiologies of CHD not only has important clinical implications for genetic counseling of patients and families but may also impact clinical outcomes by identifying at-risk patients. Recent advancements in genetic technologies, including massively parallel sequencing, have allowed for the discovery of new genetic etiologies for CHD. Although variant prioritization and interpretation of pathogenicity remain challenges in the field of CHD genomics, advances in single-cell genomics and functional genomics using cellular and animal models of CHD have the potential to provide novel insights into the underlying mechanisms of CHD and its associated morbidities. In this review, we provide an updated summary of the established genetic contributors to CHD and discuss recent advances in our understanding of the genetic architecture of CHD along with current challenges with the interpretation of genetic variation. Furthermore, we highlight the clinical implications of genetic findings to predict and potentially improve clinical outcomes in patients with CHD.
Collapse
Affiliation(s)
- Jun Yasuhara
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Vidu Garg
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA.,Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
31
|
Nakano H, Fajardo VM, Nakano A. The role of glucose in physiological and pathological heart formation. Dev Biol 2021; 475:222-233. [PMID: 33577830 PMCID: PMC8107118 DOI: 10.1016/j.ydbio.2021.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/30/2020] [Accepted: 01/29/2021] [Indexed: 02/08/2023]
Abstract
Cells display distinct metabolic characteristics depending on its differentiation stage. The fuel type of the cells serves not only as a source of energy but also as a driver of differentiation. Glucose, the primary nutrient to the cells, is a critical regulator of rapidly growing embryos. This metabolic change is a consequence as well as a cause of changes in genetic program. Disturbance of fetal glucose metabolism such as in diabetic pregnancy is associated with congenital heart disease. In utero hyperglycemia impacts the left-right axis establishment, migration of cardiac neural crest cells, conotruncal formation and mesenchymal formation of the cardiac cushion during early embryogenesis and causes cardiac hypertrophy in late fetal stages. In this review, we focus on the role of glucose in cardiogenesis and the molecular mechanisms underlying heart diseases associated with hyperglycemia.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Viviana M Fajardo
- Department of Pediatrics, Division of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
Davis K, Azarcon P, Hickenlooper S, Bia R, Horiuchi E, Szulik MW, Franklin S. The role of demethylases in cardiac development and disease. J Mol Cell Cardiol 2021; 158:89-100. [PMID: 34081951 DOI: 10.1016/j.yjmcc.2021.05.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 12/16/2022]
Abstract
Heart failure is a worldwide health condition that currently has limited noninvasive treatments. Heart disease includes both structural and molecular remodeling of the heart which is driven by alterations in gene expression in the cardiomyocyte. Therefore, understanding the regulatory mechanisms which instigate these changes in gene expression and constitute the foundation for pathological remodeling may be beneficial for developing new treatments for heart disease. These gene expression changes are largely preceded by epigenetic alterations to chromatin, including the post-translational modification of histones such as methylation, which alters chromatin to be more or less accessible for transcription factors or regulatory proteins to bind and modify gene expression. Methylation was once thought to be a permanent mark placed on histone or non-histone targets by methyltransferases, but is now understood to be a reversible process after the discovery of the first demethylase, KDM1A/LSD1. Since this time, it has been shown that demethylases play key roles in embryonic development, in maintaining cellular homeostasis and disease progression. However, the role of demethylases in the fetal and adult heart remains largely unknown. In this review, we have compiled data on the 33 mammalian demethylases that have been identified to date and evaluate their expression in the embryonic and adult heart as well as changes in expression in the failing myocardium using publicly available RNA-sequencing and proteomic datasets. Our analysis detected expression of 14 demethylases in the normal fetal heart, and 5 demethylases in the normal adult heart. Moreover, 8 demethylases displayed differential expression in the diseased human heart compared to healthy hearts. We then examined the literature regarding these demethylases and provide phenotypic information of 13 demethylases that have been functionally interrogated in some way in the heart. Lastly, we describe the 6 arginine and lysine residues on histones which have been shown to be methylated but have no corresponding demethylase identified which removes these methyl marks. Overall, this review highlights our current knowledge on the role of demethylases, their importance in cardiac development and pathophysiology and provides evidence for the use of pharmacological inhibitors to combat disease.
Collapse
Affiliation(s)
- Kathryn Davis
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America.
| | - Presley Azarcon
- School of Medicine, University of Utah, Salt Lake City, UT, United States of America
| | - Samuel Hickenlooper
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Ryan Bia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Emilee Horiuchi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Marta W Szulik
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Sarah Franklin
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America.
| |
Collapse
|
33
|
Developmental and lifelong dioxin exposure induces measurable changes in cardiac structure and function in adulthood. Sci Rep 2021; 11:10378. [PMID: 34001975 PMCID: PMC8129097 DOI: 10.1038/s41598-021-89825-w] [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: 02/08/2021] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
Congenital heart disease (CHD) is the most common congenital abnormality. A precise etiology for CHD remains elusive, but likely results from interactions between genetic and environmental factors during development, when the heart adapts to physiological and pathophysiological conditions. Further, it has become clearer that early exposure to toxins that do not result in overt CHD may be associated with adverse cardiac outcomes that are not manifested until later life. Previously, interference with endogenous developmental functions of the aryl hydrocarbon receptor (AHR), either by gene ablation or by in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a potent AHR ligand, was shown to cause structural, molecular and functional cardiac abnormalities and altered heart physiology in mouse embryos. Here, we show that continuous exposure to TCDD from fertilization throughout adulthood caused male mice to underperform at exercise tolerance tests compared to their control and female counterparts, confirming previous observations of a sexually dimorphic phenotype. Renin-angiotensin stimulation by angiotensin II (Ang II) caused measurable increases in blood pressure and left ventricle mass, along with decreased end diastolic volume and preserved ejection fraction. Interestingly, TCDD exposure caused measurable reductions in the myocardial hypertrophic effects of Ang II, suggesting that endogenous AHR signaling present in adulthood may play a role in the pathogenesis of hypertrophy. Overall, the findings reported in this pilot study highlight the complex systems underlying TCDD exposure in the development of cardiac dysfunction in later life.
Collapse
|
34
|
Ozcan T, Kikano S, Plummer S, Strainic J, Ravishankar S. The Association of Fetal Congenital Cardiac Defects and Placental Vascular Malperfusion. Pediatr Dev Pathol 2021; 24:187-192. [PMID: 33491545 DOI: 10.1177/1093526620986497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Abnormal early angiogenesis appears to impact both placental disorders and fetal congenital heart defects (CHD). We sought to assess the association of placental perfusion defects (PPD) and fetal (CHD). METHODS Singleton pregnancies with isolated severe fetal CHD were compared to controls without congenital anomalies or maternal malperfusion (MVM) risk factors. CHD was categorized into group 1: single left ventricle morphology and transposition of the great vessels (TGA) and group 2: single right ventricle and two ventricle morphology. Malperfusion was defined as fetal vascular malperfusion (FVM), MVM, and both FVM and MVM. RESULTS PPD was increased for all CHD (n = 47), CHD with or without risk factors, and CHD groups compared to controls (n = 92). Overall CHD cases and CHD with risk factors had an increased risk of FVM (30% and 80% vs 14%), and MVM (43% and 50% vs 21%), respectively. MVM rates were similar in CHD with and without maternal risk factors. FVM (38% vs 14%) and MVM (44% vs 21%) were increased in Group 1. MVM (42% vs 21%) and both FVM and MVM (16% vs 3%) were increased in Group 2. CONCLUSIONS PPD risk is increased in severe isolated fetal CHD. The highest risk is seen in fetal CHD with maternal risk factors.
Collapse
Affiliation(s)
- Tulin Ozcan
- Division of Maternal Fetal Medicine, Department of OB/GYN, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Sandra Kikano
- Department of Pediatrics, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Sarah Plummer
- Divison of Pediatric Cardiology, Department of Pediatrics, University Hospitals Case Medical Center, Cleveland, Ohio
| | - James Strainic
- Divison of Pediatric Cardiology, Department of Pediatrics, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Sanjita Ravishankar
- Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio
| |
Collapse
|
35
|
Quantitative proteome comparison of human hearts with those of model organisms. PLoS Biol 2021; 19:e3001144. [PMID: 33872299 PMCID: PMC8084454 DOI: 10.1371/journal.pbio.3001144] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 04/29/2021] [Accepted: 02/12/2021] [Indexed: 01/23/2023] Open
Abstract
Delineating human cardiac pathologies and their basic molecular mechanisms relies on research conducted in model organisms. Yet translating findings from preclinical models to humans present a significant challenge, in part due to differences in cardiac protein expression between humans and model organisms. Proteins immediately determine cellular function, yet their large-scale investigation in hearts has lagged behind those of genes and transcripts. Here, we set out to bridge this knowledge gap: By analyzing protein profiles in humans and commonly used model organisms across cardiac chambers, we determine their commonalities and regional differences. We analyzed cardiac tissue from each chamber of human, pig, horse, rat, mouse, and zebrafish in biological replicates. Using mass spectrometry–based proteomics workflows, we measured and evaluated the abundance of approximately 7,000 proteins in each species. The resulting knowledgebase of cardiac protein signatures is accessible through an online database: atlas.cardiacproteomics.com. Our combined analysis allows for quantitative evaluation of protein abundances across cardiac chambers, as well as comparisons of cardiac protein profiles across model organisms. Up to a quarter of proteins with differential abundances between atria and ventricles showed opposite chamber-specific enrichment between species; these included numerous proteins implicated in cardiac disease. The generated proteomics resource facilitates translational prospects of cardiac studies from model organisms to humans by comparisons of disease-linked protein networks across species. This study provides protein abundance profiles for thousands of proteins across cardiac chambers for humans and five commonly used model organisms. This quantitative proteomics dataset represents the most comprehensive such resource to date, and can be queried via a web browser to identify the most appropriate model organism for future studies.
Collapse
|
36
|
Ahmadi AR, Sabri MR, Navabi ZS, Ghaderian M, Dehghan B, Mahdavi C, Khodarahmi S. Early Results of the Persian Registry of Cardiovascular Disease/Congenital Heart Disease (PROVE/CHD) in Isfahan. J Tehran Heart Cent 2021; 15:158-164. [PMID: 34178084 PMCID: PMC8217188 DOI: 10.18502/jthc.v15i4.5941] [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] [Indexed: 11/24/2022] Open
Abstract
Background: In 2016, a prospective registry for pediatric patients with congenital heart disease (CHD) was established in Isfahan, Iran. Data on pediatric CHD in Iran are scant; accordingly, we aimed to report the early results of the Persian Registry Of cardioVascular diseasE (PROVE/CHD) Registry in Isfahan. Methods: All patients with CHD and associated defects diagnosed by pediatric cardiologists were assessed via echocardiography for inclusion in the present study between late 2016 and August 2019. The participants' sociodemographic characteristics, maternal history, birth history, medical history, current clinical presentations in the clinic or hospital, paraclinical data, cardiac diagnoses based on the International Classification of Diseases, 10th Revision (ICD-10), disease management plans, and medications were entered into a questionnaire by the subjects' parents/legal custodians and physicians and then transferred to the PROVE/CHD Registry. Results: The PROVE/CHD registry encompasses 1252 patients with CHD (49.9% male) at a mean age of 6.50±6.36 years. The most frequent cardiac diagnoses were ventricular septal defect (39.3%), atrial septal defect (29.7%), patent ductus arteriosus (25.4%), pulmonary stenosis (11.0%), tetralogy of Fallot (6.1%), coarctation of the aorta (5.4%), and aortic stenosis (5.1%), respectively. The most frequent interventions were patent ductus arteriosus closure (4.3%), atrial septal defect closure (3.6%), pulmonary valvuloplasty (2.2%), coarctation of the aorta angioplasty (1.9%), and ventricular septal defect closure (1.1%), correspondingly. The approximate corresponding rates of corrective and palliative surgeries were 32.0% and 13.1%. The corrective surgeries were mainly comprised of ventricular septal defect closure (7.8%), patent ductus arteriosus closure (7.3%), atrial septal defect closure (5.1%), and tetralogy of Fallot repair (3.8%), respectively. The palliative surgeries mainly consisted of the Glenn shunt (9.0%) and pulmonary artery banding (3.6%). Conclusion: The PROVE/CHD Registry collects data on pediatric patients with CHD. The results of this registry can provide epidemiological data and a set of homogeneously defined cases for further studies.
Collapse
Affiliation(s)
- Ali Reza Ahmadi
- Pediatric Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Reza Sabri
- Pediatric Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zohreh Sadat Navabi
- Pediatric Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Ghaderian
- Pediatric Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahar Dehghan
- Pediatric Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Chehreh Mahdavi
- Pediatric Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Somaieh Khodarahmi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
37
|
Lou XY, Hou TT, Liu SY, Xu HM, Lin F, Tang X, MacLeod SL, Cleves MA, Hobbs CA. Innovative approach to identify multigenomic and environmental interactions associated with birth defects in family-based hybrid designs. Genet Epidemiol 2021; 45:171-189. [PMID: 32996630 PMCID: PMC8495752 DOI: 10.1002/gepi.22363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 11/09/2022]
Abstract
Genes, including those with transgenerational effects, work in concert with behavioral, environmental, and social factors via complex biological networks to determine human health. Understanding complex relationships between causal factors underlying human health is an essential step towards deciphering biological mechanisms. We propose a new analytical framework to investigate the interactions between maternal and offspring genetic variants or their surrogate single nucleotide polymorphisms (SNPs) and environmental factors using family-based hybrid study design. The proposed approach can analyze diverse genetic and environmental factors and accommodate samples from a variety of family units, including case/control-parental triads, and case/control-parental dyads, while minimizing potential bias introduced by population admixture. Comprehensive simulations demonstrated that our innovative approach outperformed the log-linear approach, the best available method for case-control family data. The proposed approach had greater statistical power and was capable to unbiasedly estimate the maternal and child genetic effects and the effects of environmental factors, while controlling the Type I error rate against population stratification. Using our newly developed approach, we analyzed the associations between maternal and fetal SNPs and obstructive and conotruncal heart defects, with adjustment for demographic and lifestyle factors and dietary supplements. Fourteen and 11 fetal SNPs were associated with obstructive and conotruncal heart defects, respectively. Twenty-seven and 17 maternal SNPs were associated with obstructive and conotruncal heart defects, respectively. In addition, maternal body mass index was a significant risk factor for obstructive defects. The proposed approach is a powerful tool for interrogating the etiological mechanism underlying complex traits.
Collapse
Affiliation(s)
- Xiang-Yang Lou
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ting-Ting Hou
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Shou-Ye Liu
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Hai-Ming Xu
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Feng Lin
- Institute of Bioinformatics and Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Xinyu Tang
- The US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Mario A. Cleves
- Department of Pediatrics, Morsani College of Medicine, Health Informatics Institute, University of South Florida, Tampa, Florida, USA
| | - Charlotte A. Hobbs
- Rady Children’s Institute for Genomic Medicine, San Diego, California, USA
| |
Collapse
|
38
|
Mullen M, Zhang A, Lui GK, Romfh AW, Rhee JW, Wu JC. Race and Genetics in Congenital Heart Disease: Application of iPSCs, Omics, and Machine Learning Technologies. Front Cardiovasc Med 2021; 8:635280. [PMID: 33681306 PMCID: PMC7925393 DOI: 10.3389/fcvm.2021.635280] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Congenital heart disease (CHD) is a multifaceted cardiovascular anomaly that occurs when there are structural abnormalities in the heart before birth. Although various risk factors are known to influence the development of this disease, a full comprehension of the etiology and treatment for different patient populations remains elusive. For instance, racial minorities are disproportionally affected by this disease and typically have worse prognosis, possibly due to environmental and genetic disparities. Although research into CHD has highlighted a wide range of causal factors, the reasons for these differences seen in different patient populations are not fully known. Cardiovascular disease modeling using induced pluripotent stem cells (iPSCs) is a novel approach for investigating possible genetic variants in CHD that may be race specific, making it a valuable tool to help solve the mystery of higher incidence and mortality rates among minorities. Herein, we first review the prevalence, risk factors, and genetics of CHD and then discuss the use of iPSCs, omics, and machine learning technologies to investigate the etiology of CHD and its connection to racial disparities. We also explore the translational potential of iPSC-based disease modeling combined with genome editing and high throughput drug screening platforms.
Collapse
Affiliation(s)
- McKay Mullen
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Angela Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
| | - George K. Lui
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - Anitra W. Romfh
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - June-Wha Rhee
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Department of Radiology, Stanford University, Stanford, CA, United States
| |
Collapse
|
39
|
Pintacuda G, Martín JM, Eggan KC. Mind the translational gap: using iPS cell models to bridge from genetic discoveries to perturbed pathways and therapeutic targets. Mol Autism 2021; 12:10. [PMID: 33557935 PMCID: PMC7869517 DOI: 10.1186/s13229-021-00417-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) comprises a group of neurodevelopmental disorders characterized by impaired social interactions as well as the presentation of restrictive and repetitive behaviors. ASD is highly heritable but genetically heterogenous with both common and rare genetic variants collaborating to predispose individuals to the disorder. In this review, we synthesize recent efforts to develop human induced pluripotent stem cell (iPSC)-derived models of ASD-related phenotypes. We firstly address concerns regarding the relevance and validity of available neuronal iPSC-derived models. We then critically evaluate the robustness of various differentiation and cell culture protocols used for producing cell types of relevance to ASD. By exploring iPSC models of ASD reported thus far, we examine to what extent cellular and neuronal phenotypes with potential relevance to ASD can be linked to genetic variants found to underlie it. Lastly, we outline promising strategies by which iPSC technology can both enhance the power of genetic studies to identify ASD risk factors and nominate pathways that are disrupted across groups of ASD patients that might serve as common points for therapeutic intervention.
Collapse
Affiliation(s)
- Greta Pintacuda
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Jacqueline M Martín
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Kevin C Eggan
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
40
|
Song Y, Chen W, Huang Z, Tian G, Li M, Zhao Z, Feng Z, Wu F, Qian M, Ma X, Sheng W, Huang G. A Non-coding HES1 Variant Predisposes Children to Congenital Heart Disease in Chinese Population. Front Cell Dev Biol 2021; 9:631942. [PMID: 33585489 PMCID: PMC7876461 DOI: 10.3389/fcell.2021.631942] [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: 11/21/2020] [Accepted: 01/07/2021] [Indexed: 11/13/2022] Open
Abstract
Background: As a key component in the NOTCH signaling pathway, HES1 plays an important role in vertebrate heart development. Variants in the HES1 coding sequence are known to be associated with congenital heart disease (CHD). However, little is known about HES1 non-coding sequence variants and their association with the risk of developing CHD. Method and Results: We initially analyzed the non-coding sequence of the HES1 gene in 12 unrelated CHD families by direct sequencing and identified a previously unreported promoter region variant (NM_005524.4: c.-1279-1278 insAC, rs148941464) in the HES1 gene in four CHD families. The homozygous variant in patients was inherited from carrier parents with normal phenotypes, indicating a likely recessive genetic model. Given that the HES1 gene is predicted to be likely to exhibit haploinsufficiency (%HI: 11.44), we hypothesized that the HES1 homozygous variant is a genetic risk factor underlying CHD. We then carried out sequencing of this HES1 variant in 629 sporadic non-syndromic CHD cases and 696 healthy controls and performed association analysis. Interestingly, we observed a significant association of the homozygous HES1 promoter variant with CHD (18.92% of cases vs. 9.91% of controls; OR: 2.291, 95% CI: 1.637-3.207, p = 9.72 × 10-7). No significant association with CHD was observed for the HES1 promoter heterozygous variant (p > 0.05). However, association analysis tests of the HES1 homozygous variant with each subtype of CHD revealed that this homozygous variant was strongly associated with transposition of the great arteries (TGA) (OR: 3.726, 95% CI: 1.745-7.956, p = 0.0003). Moreover, the prevalence of HES1 homozygous variants in CHD patients with TGA (27.66%) was significantly higher than that in patients with other CHD subtypes or controls. Similar results were observed in a replication group of TGA (n = 64). Functional studies demonstrated that the homozygous variant in the HES1 promoter can disrupt its ability to bind RXRA, an inhibitory transcription factor, which results in abnormally high expression of the HES1 gene, indicating that this variant harbors gain-of-function effects. Conclusions: Our findings reveal that the non-coding homozygous variant in the HES1 promoter has a gain-of-function effect and is associated with an increased risk of CHD development, especially the severe TGA subtype.
Collapse
Affiliation(s)
- Yangliu Song
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China
| | - Weicheng Chen
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China
| | - Zitong Huang
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Guixiang Tian
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Mengru Li
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Zhengshan Zhao
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Zhiyu Feng
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China
| | - Feizhen Wu
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China
| | - Maoxiang Qian
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China
| | - Xiaojing Ma
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China
| | - Wei Sheng
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Guoying Huang
- Institutes of Biomedical Sciences, and Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| |
Collapse
|
41
|
de Gannes M, Ko CI, Zhang X, Biesiada J, Niu L, Koch SE, Medvedovic M, Rubinstein J, Puga A. Dioxin Disrupts Dynamic DNA Methylation Patterns in Genes That Govern Cardiomyocyte Maturation. Toxicol Sci 2020; 178:325-337. [PMID: 33017471 DOI: 10.1093/toxsci/kfaa153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Congenital heart disease (CHD), the leading birth defect worldwide, has a largely unknown etiology, likely to result from complex interactions between genetic and environmental factors during heart development, at a time when the heart adapts to diverse physiological and pathophysiological conditions. Crucial among these is the regulation of cardiomyocyte development and postnatal maturation, governed by dynamic changes in DNA methylation. Previous work from our laboratory has shown that exposure to the environmental toxicant tetrachlorodibenzo-p-dioxin (TCDD) disrupts several molecular networks responsible for heart development and function. To test the hypothesis that the disruption caused by TCDD in the heart results from changes in DNA methylation and gene expression patterns of cardiomyocytes, we established a stable mouse embryonic stem cell line expressing a puromycin resistance selectable marker under control of the cardiomyocyte-specific Nkx2-5 promoter. Differentiation of these cells in the presence of puromycin induces the expression of a large suite of cardiomyocyte-specific markers. To assess the consequences of TCDD treatment on gene expression and DNA methylation in these cardiomyocytes, we subjected them to transcriptome and methylome analyses in the presence of TCDD. Unlike control cardiomyocytes maintained in vehicle, the TCDD-treated cardiomyocytes showed extensive gene expression changes, with a significant correlation between differential RNA expression and DNA methylation in 111 genes, many of which are key elements of pathways that regulate cardiovascular development and function. Our findings provide an important clue toward the elucidation of the complex interactions between genetic and epigenetic mechanisms after developmental TCDD exposure that may contribute to CHD.
Collapse
Affiliation(s)
- Matthew de Gannes
- Department of Environmental Health and Center for Environmental Genetics
| | - Chia-I Ko
- Department of Environmental Health and Center for Environmental Genetics
| | - Xiang Zhang
- Department of Environmental Health and Center for Environmental Genetics
| | - Jacek Biesiada
- Department of Environmental Health and Center for Environmental Genetics
| | - Liang Niu
- Department of Environmental Health and Center for Environmental Genetics
| | - Sheryl E Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Mario Medvedovic
- Department of Environmental Health and Center for Environmental Genetics
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics
| |
Collapse
|
42
|
Izarzugaza JMG, Ellesøe SG, Doganli C, Ehlers NS, Dalgaard MD, Audain E, Dombrowsky G, Banasik K, Sifrim A, Wilsdon A, Thienpont B, Breckpot J, Gewillig M, Brook JD, Hitz MP, Larsen LA, Brunak S. Systems genetics analysis identifies calcium-signaling defects as novel cause of congenital heart disease. Genome Med 2020; 12:76. [PMID: 32859249 PMCID: PMC7453558 DOI: 10.1186/s13073-020-00772-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 08/07/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Congenital heart disease (CHD) occurs in almost 1% of newborn children and is considered a multifactorial disorder. CHD may segregate in families due to significant contribution of genetic factors in the disease etiology. The aim of the study was to identify pathophysiological mechanisms in families segregating CHD. METHODS We used whole exome sequencing to identify rare genetic variants in ninety consenting participants from 32 Danish families with recurrent CHD. We applied a systems biology approach to identify developmental mechanisms influenced by accumulation of rare variants. We used an independent cohort of 714 CHD cases and 4922 controls for replication and performed functional investigations using zebrafish as in vivo model. RESULTS We identified 1785 genes, in which rare alleles were shared between affected individuals within a family. These genes were enriched for known cardiac developmental genes, and 218 of these genes were mutated in more than one family. Our analysis revealed a functional cluster, enriched for proteins with a known participation in calcium signaling. Replication in an independent cohort confirmed increased mutation burden of calcium-signaling genes in CHD patients. Functional investigation of zebrafish orthologues of ITPR1, PLCB2, and ADCY2 verified a role in cardiac development and suggests a combinatorial effect of inactivation of these genes. CONCLUSIONS The study identifies abnormal calcium signaling as a novel pathophysiological mechanism in human CHD and confirms the complex genetic architecture underlying CHD.
Collapse
Affiliation(s)
- Jose M G Izarzugaza
- Department of Health Technology, Technical University of Denmark, Kemitorvet, DK-2800, Kgs. Lyngby, Denmark
| | - Sabrina G Ellesøe
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark
| | - Canan Doganli
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark
| | - Natasja Spring Ehlers
- Department of Health Technology, Technical University of Denmark, Kemitorvet, DK-2800, Kgs. Lyngby, Denmark
| | - Marlene D Dalgaard
- Department of Health Technology, Technical University of Denmark, Kemitorvet, DK-2800, Kgs. Lyngby, Denmark
- DTU Multi Assay Core (DMAC), Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark
| | - Enrique Audain
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
| | - Gregor Dombrowsky
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark
| | - Alejandro Sifrim
- Wellcome Trust Sanger Institute, Cambridge, UK
- Centre for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Anna Wilsdon
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Bernard Thienpont
- Centre for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jeroen Breckpot
- Centre for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Marc Gewillig
- Pediatric Cardiology Unit, University Hospitals Leuven, Leuven, Belgium
| | - J David Brook
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Marc-Phillip Hitz
- Department of Congenital Heart Disease and Pediatric Cardiology, Universitätsklinikum Schleswig-Holstein Kiel, Kiel, Germany
- Wellcome Trust Sanger Institute, Cambridge, UK
- Institute of Human Genetics, Christian-Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Lars A Larsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark.
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
43
|
Hu CY, Huang K, Fang Y, Yang XJ, Ding K, Jiang W, Hua XG, Huang DY, Jiang ZX, Zhang XJ. Maternal air pollution exposure and congenital heart defects in offspring: A systematic review and meta-analysis. CHEMOSPHERE 2020; 253:126668. [PMID: 32278917 DOI: 10.1016/j.chemosphere.2020.126668] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 05/21/2023]
Abstract
BACKGROUND Congenital heart defects (CHDs) has a multifactorial causation with a strong genetic component and many environmental triggers. Emerging body of empirical studies suggest that air pollution is an important contributor to the development of CHDs, however, there still remains some controversy over the current evidence, and to the authors' knowledge, no studies have reviewed the most recent evidence. OBJECTIVES We performed a systematic review and meta-analysis of epidemiological literature to investigate the relationship between maternal air pollution exposure and CHDs risk in offspring. The presence of heterogeneity and publication bias across available studies were also examined. METHODS An extensive literature search of epidemiological studies pertaining to air pollution and CHDs, published in English language, until August 1, 2019 was conducted. Summary risk estimates of pollution-outcome combinations were calculated for i) risk per specific increment of concentration and ii) risk at high versus low exposure level in each study using fixed-effect model or random-effects model. RESULTS A total of 26 studies were finally included. In the meta-analyses, high versus low carbon monoxide (CO) exposure was associated with an increased risk of tetralogy of Fallot [odds ratio (OR) = 1.21, 95% confidence interval (CI): 1.04-1.41], yet particulate matter ≤ 5 μm (PM2.5) exposure was marginally associated with it. Increased risk of atrial septal defects (ASDs) was found for each 10 μg/m3 and 10 ppb increment in particulate matter ≤ 10 μm (PM10) and ozone (O3) exposure, respectively (OR = 1.04, 95% CI: 1.00-1.09; OR = 1.09, 95% CI: 1.02-1.17). Categorical nitrogen dioxide (NO2) exposure was associated with an increased risk of coarctation of the aorta (OR for high versus low = 1.14, 95% CI: 1.02-1.26). Analyses for other combinations yielded none statistically significant associations. Sensitive analyses showed similar findings. CONCLUSIONS The summary effect estimates from this study suggest statistically significant associations between increased risk of specific CHDs subtypes and PM2.5, PM10, NO2, CO, and O3 exposures. Further studies, especially conducted in developing countries, with improvements in exposure assessing, outcome harmonizing, and mechanistic understanding are needed to elaborate the suggestive associations.
Collapse
Affiliation(s)
- Cheng-Yang Hu
- Department of Humanistic Medicine, School of Humanistic Medicine, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China
| | - Kai Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China
| | - Yuan Fang
- Department of Public health, Erasmus MC University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, the Netherlands
| | - Xiao-Jing Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China
| | - Kun Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China
| | - Wen Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China
| | - Xiao-Guo Hua
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China
| | - Da-Yan Huang
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, 15# Yimin Road, Hefei, 230001, China
| | - Zheng-Xuan Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, 678# Furong Road, Hefei, 230601, China.
| | - Xiu-Jun Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81# Meishan Road, Hefei, 230032, China.
| |
Collapse
|
44
|
Yadav A, Vidal M, Luck K. Precision medicine - networks to the rescue. Curr Opin Biotechnol 2020; 63:177-189. [PMID: 32199228 PMCID: PMC7308189 DOI: 10.1016/j.copbio.2020.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
Genetic variants are often not predictive of the phenotypic outcome. Individuals carrying the same pathogenic variant, associated with Mendelian or complex disease, can manifest to different extents, from severe-to-mild to no disease. Improving the accuracy of predicted clinical manifestations of genetic variants has emerged as one of the biggest challenges in precision medicine, which can only be addressed by understanding the mechanisms underlying genotype-phenotype relationships. Efforts to understand the molecular basis of these relationships have identified complex systems of interacting biomolecules that underlie cellular function. Here, we review recent advances in how modeling cellular systems as networks of interacting proteins has fueled identification of disease-associated processes, delineation of underlying molecular mechanisms, and prediction of the pathogenicity of variants. This review is intended to be inspiring for clinicians, geneticists, and network biologists alike who aim to jointly advance our understanding of human disease and accelerate progress toward precision medicine.
Collapse
Affiliation(s)
- Anupama Yadav
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Katja Luck
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Current address: Institute of Molecular Biology, Mainz, Germany.
| |
Collapse
|
45
|
Phillips JM, Longoria JN. Addressing the neurodevelopmental needs of children and adolescents with congenital heart disease: A review of the existing intervention literature. Child Neuropsychol 2020; 26:433-459. [PMID: 31672097 DOI: 10.1080/09297049.2019.1682131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 10/14/2019] [Indexed: 10/25/2022]
Abstract
Congenital heart disease (CHD) is among the most prevalent birth defects in the United States. Given that children with CHD are at risk for differences with development, learning, and psychosocial functioning, effective intervention becomes a central tenant of recommendations following neuropsychological consultation and evaluation. The primary focus of this review is to summarize available interventions for children and adolescents with CHD. The existing CHD literature has concentrated on early developmental services, psychopharmacological treatment, and need for academic supports. The literature is limited with regard to intervention studies that target cognitive deficits and psychosocial difficulties. To address this discrepancy, efficacious interventions that have been used to mitigate these concerns within other medical groups are also discussed in an effort to provide options for alternative recommendations and services. The current paper is intended to facilitate comprehensive care for cardiac patients by providing clinicians with a review of the available intervention literature, as well as potential interventions that may serve as supplemental strategies in the interim.
Collapse
Affiliation(s)
- Jannel M Phillips
- Department of Behavioral Health, Henry Ford Health System, Detroit, MI, USA
| | | |
Collapse
|
46
|
P M, M AJ, L D. Assessment of Patient Dose with Special Look at Pediatrics during Cardiovascular Imaging. J Biomed Phys Eng 2020; 10:51-58. [PMID: 32158711 PMCID: PMC7036411 DOI: 10.31661/jbpe.v0i0.902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/10/2018] [Indexed: 11/16/2022]
Abstract
Background During interventional cardiology processes, patients especially women and children receive high radiation doses due to their sensitivity. Objective In this study, we evaluated a pediatric patient dose separately in those undergone intervention cardiac procedure. Material and Methods In this cross sectional study, a public hospital with 252 patients, Entrance Skin Dose (ESD) and Dose-Area Product (DAP) were recorded. Prior to the beginning of fluoroscopic procedure, the chest thickness and Body Mass Index (BMI) of patients were measured. Furthermore, kV, mAs, angle of tube and time of angiography and angioplasty were recorded. Results Children ratio to all patients underwent the cardiovascular imaging was 1.8. The means of patients' ESD, DAP and fluoroscopy time were 178.3±17 mGy, 1123.6±11 μGycm2 and 281.4±181.2 s, respectively for coronary angiography. The females were 96.8 under 30 years and their dose mean was 276±37 mGy, 368±24 μGycm2 for ESD and DAP received, respectively with 376s fluoroscopy time. Mean mAs was 359±34 and kV was 71.23±2.7. Above all, a direct and significant correlation was found between the patients' chest thickness with kV (p=0.037, r = 0.11) and mAs (p<0.001, r = 0.28) variations. Conclusion The results demonstrated that the number of children referred to the cardiology department and also the dose rate received by them during this test was higher than the data provided for children in developing countries. Paying attention to the children's perception of high-fluorescence time is necessary in comparison with total angiography time in order to reduce the number of radiation injuries among pediatrics.
Collapse
Affiliation(s)
- Mehnati P
- PhD, Cardiovascular Research Center, Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghari Jafarabadi M
- PhD, Department of Statistics and Epidemiology, Faculty of Health, Road Traffic Injury Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Danaee L
- MSc, Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
47
|
Burggren WW. Phenotypic Switching Resulting From Developmental Plasticity: Fixed or Reversible? Front Physiol 2020; 10:1634. [PMID: 32038303 PMCID: PMC6987144 DOI: 10.3389/fphys.2019.01634] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/27/2019] [Indexed: 12/19/2022] Open
Abstract
The prevalent view of developmental phenotypic switching holds that phenotype modifications occurring during critical windows of development are "irreversible" - that is, once produced by environmental perturbation, the consequent juvenile and/or adult phenotypes are indelibly modified. Certainly, many such changes appear to be non-reversible later in life. Yet, whether animals with switched phenotypes during early development are unable to return to a normal range of adult phenotypes, or whether they do not experience the specific environmental conditions necessary for them to switch back to the normal range of adult phenotypes, remains an open question. Moreover, developmental critical windows are typically brief, early periods punctuating a much longer period of overall development. This leaves open additional developmental time for reversal (correction) of a switched phenotype resulting from an adverse environment early in development. Such reversal could occur from right after the critical window "closes," all the way into adulthood. In fact, examples abound of the capacity to return to normal adult phenotypes following phenotypic changes enabled by earlier developmental plasticity. Such examples include cold tolerance in the fruit fly, developmental switching of mouth formation in a nematode, organization of the spinal cord of larval zebrafish, camouflage pigmentation formation in larval newts, respiratory chemosensitivity in frogs, temperature-metabolism relations in turtles, development of vascular smooth muscle and kidney tissue in mammals, hatching/birth weight in numerous vertebrates,. More extreme cases of actual reversal (not just correction) occur in invertebrates (e.g., hydrozoans, barnacles) that actually 'backtrack' along normal developmental trajectories from adults back to earlier developmental stages. While developmental phenotypic switching is often viewed as a permanent deviation from the normal range of developmental plans, the concept of developmental phenotypic switching should be expanded to include sufficient plasticity allowing subsequent correction resulting in the normal adult phenotype.
Collapse
Affiliation(s)
- Warren W. Burggren
- Developmental Integrative Biology, Department of Biological Sciences, University of North Texas, Denton, TX, United States
| |
Collapse
|
48
|
Dasouki MJ, Wakil SM, Al-Harazi O, Alkorashy M, Muiya NP, Andres E, Hagos S, Aldusery H, Dzimiri N, Colak D. New Insights into the Impact of Genome-Wide Copy Number Variations on Complex Congenital Heart Disease in Saudi Arabia. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 24:16-28. [PMID: 31855513 DOI: 10.1089/omi.2019.0165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Congenital heart diseases (CHDs) are complex traits that manifest in diverse clinical phenotypes such as the Tetralogy of Fallot (TOF), valvular and ventricular/atrial septal defects. Genetic mechanisms of CHDs have remained largely unclear to date. Copy number variations (CNVs) have been implicated in many complex diseases but their impact has not been examined extensively in various forms of CHD lesions. We report in this study, to the best of our knowledge, the largest cohort of Saudi Arab CHD patients to date who were evaluated using genome-wide CNV analysis. In a sample of 134 Saudi Arab patients with CHD, 66 exhibited pathogenic or likely pathogenic CNVs. Notably, 21 copy number gains and 11 copy number losses were detected that encompassed 141 genes and 146 genes, respectively. The most frequent gains were on 17q21.31, 8p11.21, and 22q11.23, whereas the losses were primarily localized to 16p11.2. Interestingly, all lesions have had gains at 17q21.31. Septal defects had also gains at 8p11.21 and 22q11.23, valvular lesions at 8p11.21, 22q11.23, and 2q13, and TOF at 16p11.2. Functional and network analyses demonstrated that cardiovascular and nervous system development and function as well as cell death/survival were most significantly associated with CNVs, thus highlighting the potentially important genes likely to be involved in CHD, including NPHP1, PLCB1, KANSL1, and NR3C1. In conclusion, this genome-wide analysis identifies a high frequency of CNVs mostly in patients with septal defects, primarily influencing cardiovascular developmental and functional pathways, thereby offering a deeper insight into the complex networks involved in CHD pathogenesis.
Collapse
Affiliation(s)
- Majed J Dasouki
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Salma M Wakil
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Olfat Al-Harazi
- Departments Biostatistics, Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Maarab Alkorashy
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nzioka P Muiya
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Editha Andres
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Samya Hagos
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Haya Aldusery
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nduna Dzimiri
- Departments Genetics and Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Dilek Colak
- Departments Biostatistics, Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
49
|
Affiliation(s)
- Palle Duun Rohde
- Department of Molecular Biology & Genetics, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
50
|
Genetics of Congenital Heart Disease. Biomolecules 2019; 9:biom9120879. [PMID: 31888141 PMCID: PMC6995556 DOI: 10.3390/biom9120879] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Congenital heart disease (CHD) is one of the most common birth defects. Studies in animal models and humans have indicated a genetic etiology for CHD. About 400 genes have been implicated in CHD, encompassing transcription factors, cell signaling molecules, and structural proteins that are important for heart development. Recent studies have shown genes encoding chromatin modifiers, cilia related proteins, and cilia-transduced cell signaling pathways play important roles in CHD pathogenesis. Elucidating the genetic etiology of CHD will help improve diagnosis and the development of new therapies to improve patient outcomes.
Collapse
|