1
|
Weeks O, Gao X, Basu S, Galdieri J, Chen K, Burns CG, Burns CE. Embryonic alcohol exposure in zebrafish predisposes adults to cardiomyopathy and diastolic dysfunction. Cardiovasc Res 2024; 120:1607-1621. [PMID: 38900908 PMCID: PMC11535724 DOI: 10.1093/cvr/cvae139] [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: 12/21/2023] [Revised: 05/01/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
AIMS Fetal alcohol spectrum disorders (FASDs) impact up to 0.8% of the global population. However, cardiovascular health outcomes in adult patients, along with predictive biomarkers for cardiac risk stratification, remain unknown. Our aim was to utilize a longitudinal cohort study in an animal model to evaluate the impact of embryonic alcohol exposure (EAE) on cardiac structure, function, and transcriptional profile across the lifespan. METHODS AND RESULTS Using zebrafish, we characterized the aftereffects of EAE in adults binned by congenital heart defect (CHD) severity. Chamber sizes were quantified on dissected adult hearts to identify structural changes indicative of cardiomyopathy. Using echocardiography, we quantified systolic function based on ejection fraction and longitudinal strain, and diastolic function based on ventricular filling dynamics, ventricular wall movement, and estimated atrial pressures. Finally, we performed RNA-sequencing on EAE ventricles and assessed how differentially expressed genes (DEGs) correlated with cardiac function. Here, we demonstrate that EAE causes cardiomyopathy and diastolic dysfunction through persistent alterations to ventricular wall structure and gene expression. Following abnormal ventricular morphogenesis, >30% of all EAE adults developed increased atrial-to-ventricular size ratios, abnormal ventricular filling dynamics, and reduced myocardial wall relaxation during early diastole despite preserved systolic function. RNA-sequencing of the EAE ventricle revealed novel and heart failure-associated genes (slc25a33, ankrd9, dusp2, dusp4, spry4, eya4, and edn1) whose expression levels were altered across the animal's lifespan or correlated with the degree of diastolic dysfunction detected in adulthood. CONCLUSION Our study identifies EAE as a risk factor for adult-onset cardiomyopathy and diastolic dysfunction, regardless of CHD status, and suggests novel molecular indicators of adult EAE-induced heart disease.
Collapse
Affiliation(s)
- Olivia Weeks
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Xinlei Gao
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Sandeep Basu
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Jennifer Galdieri
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Kaifu Chen
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| |
Collapse
|
2
|
Dong J, Hao T. Association of maternal and paternal risk factors with risk of congenital heart disease in infants: a case-control study. Ir J Med Sci 2024; 193:95-99. [PMID: 37249792 DOI: 10.1007/s11845-023-03409-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023]
Abstract
OBJECTIVE We aimed to explore maternal and paternal risk factors with risk of congenital heart disease in infants. METHODS A total of 125 congenital heart disease (CHD) infants and 125 controls were included in Heping Hospital Affiliated to Changzhi Medical College, Shanxi, China. Subjects were diagnosed between Jan 1, 2016 and Dec 31, 2021 in the present study. All the characteristics were collected with questionnaire by face-to-face interview, including maternal and paternal risk factors. Conditional logistic regression was conducted to explore the risk factors with risk of congenital heart disease in infants. RESULTS For maternal risk factors, we found that age, number of pregnancies, systolic blood pressure (SBP), diastolic blood pressure (DBP), and body mass index (BMI) were risk factors for CHD infants, and the ORs (95%CIs) were 1.15 (1.06-1.23) for age, 1.13 (1.02-1.29) for SBP, 1.06 (1.02-1.18) for DBP, 1.22 (1.16-1.31) for BMI. Compared with one pregnancy, the ORs (95%CIs) were 1.17 (1.05-1.29) for two pregnancies and 1.25 (1.16-1.47) for more pregnancies. For paternal risk factors, we found that age (OR = 1.07, 95%CI = 1.01-1.19), smoking (OR = 1.11, 95%CI = 1.03-1.27), drinking (OR = 1.04, 95%CI = 1.02-1.19), and BMI (OR = 1.15, 95%CI = 1.03-1.28) were risk factors for CHD infants. CONCLUSION We found that age, number of pregnancies, SBP, DBP, and BMI are maternal risk factors for CHD infants. And age, smoking, drinking, and BMI are paternal risk factors for CHD infants.
Collapse
Affiliation(s)
- Jianxia Dong
- Department of Preventive Health Care, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Tianyou Hao
- Department of Preventive Health Care, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| |
Collapse
|
3
|
Ling S, Blackburn BJ, Jenkins MW, Watanabe M, Ford SM, Lapierre-Landry M, Rollins AM. Segmentation of beating embryonic heart structures from 4-D OCT images using deep learning. BIOMEDICAL OPTICS EXPRESS 2023; 14:1945-1958. [PMID: 37206115 PMCID: PMC10191668 DOI: 10.1364/boe.481657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/29/2023] [Accepted: 02/20/2023] [Indexed: 05/21/2023]
Abstract
Optical coherence tomography (OCT) has been used to investigate heart development because of its capability to image both structure and function of beating embryonic hearts. Cardiac structure segmentation is a prerequisite for the quantification of embryonic heart motion and function using OCT. Since manual segmentation is time-consuming and labor-intensive, an automatic method is needed to facilitate high-throughput studies. The purpose of this study is to develop an image-processing pipeline to facilitate the segmentation of beating embryonic heart structures from a 4-D OCT dataset. Sequential OCT images were obtained at multiple planes of a beating quail embryonic heart and reassembled to a 4-D dataset using image-based retrospective gating. Multiple image volumes at different time points were selected as key-volumes, and their cardiac structures including myocardium, cardiac jelly, and lumen, were manually labeled. Registration-based data augmentation was used to synthesize additional labeled image volumes by learning transformations between key-volumes and other unlabeled volumes. The synthesized labeled images were then used to train a fully convolutional network (U-Net) for heart structure segmentation. The proposed deep learning-based pipeline achieved high segmentation accuracy with only two labeled image volumes and reduced the time cost of segmenting one 4-D OCT dataset from a week to two hours. Using this method, one could carry out cohort studies that quantify complex cardiac motion and function in developing hearts.
Collapse
Affiliation(s)
- Shan Ling
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brecken J. Blackburn
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael W. Jenkins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michiko Watanabe
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Cleveland, Ohio, USA
| | - Stephanie M. Ford
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Cleveland, Ohio, USA
- Division of Neonatology, Rainbow Babies and Children’s Hospital, Cleveland, Ohio, USA
| | - Maryse Lapierre-Landry
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Andrew M. Rollins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
4
|
Wu L, Li N, Liu Y. Association Between Maternal Factors and Risk of Congenital Heart Disease in Offspring: A Systematic Review and Meta-Analysis. Matern Child Health J 2023; 27:29-48. [PMID: 36344649 PMCID: PMC9867685 DOI: 10.1007/s10995-022-03538-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION This study aimed to summarize the evidence describing the relationship between maternal factors during gestation and risk of congenital heart disease (CHD) in offspring. METHODS PubMed, EMBASE, and the Cochrane Library were searched for potentially relevant reports from inception to May 2021. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) calculated by the random-effects model were used to evaluate the association between maternal factors and CHD risk. RESULTS There was a significant association between CHD risk and obesity in pregnancy (OR 1.29, 95% CI 1.22-1.37; P < 0.001), smoking in pregnancy (OR 1.16, 95% CI 1.07-1.25; P < 0.001), maternal diabetes (OR 2.65, 95% CI 2.20-3.19; P < 0.001), and exposure of pregnant women to organic solvents (OR 1.82, 95% CI 1.23-2.70; P = 0.003). No correlations were revealed between CHD susceptibility and advanced maternal age (OR 1.04, 95% CI 0.96-1.12; P = 0.328), underweight (OR 1.02, 95% CI 0.96-1.08; P = 0.519), alcohol intake in pregnancy (OR 1.08, 95% CI 0.95-1.22; P = 0.251), coffee intake (OR 1.18, 95% CI 0.97-1.44; P = 0.105), and exposure to irradiation (OR 1.80, 95% CI 0.85-3.80; P = 0.125). DISCUSSION Maternal factors including maternal obesity, smoking in pregnancy, maternal diabetes and exposure to organic solvents might predispose the offspring to CHD risk.
Collapse
Affiliation(s)
- Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Na Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yong Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
5
|
Ling S, Chen J, Lapierre-Landry M, Suh J, Liu Y, Jenkins MW, Watanabe M, Ford SM, Rollins AM. Automated endocardial cushion segmentation and cellularization quantification in developing hearts using optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2022; 13:5599-5615. [PMID: 36733755 PMCID: PMC9872882 DOI: 10.1364/boe.467629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 06/18/2023]
Abstract
Of all congenital heart defects (CHDs), anomalies in heart valves and septa are among the most common and contribute about fifty percent to the total burden of CHDs. Progenitors to heart valves and septa are endocardial cushions formed in looping hearts through a multi-step process that includes localized expansion of cardiac jelly, endothelial-to-mesenchymal transition, cell migration and proliferation. To characterize the development of endocardial cushions, previous studies manually measured cushion size or cushion cell density from images obtained using histology, immunohistochemistry, or optical coherence tomography (OCT). Manual methods are time-consuming and labor-intensive, impeding their applications in cohort studies that require large sample sizes. This study presents an automated strategy to rapidly characterize the anatomy of endocardial cushions from OCT images. A two-step deep learning technique was used to detect the location of the heart and segment endocardial cushions. The acellular and cellular cushion regions were then segregated by K-means clustering. The proposed method can quantify cushion development by measuring the cushion volume and cellularized fraction, and also map 3D spatial organization of the acellular and cellular cushion regions. The application of this method to study the developing looping hearts allowed us to discover a spatial asymmetry of the acellular cardiac jelly in endocardial cushions during these critical stages, which has not been reported before.
Collapse
Affiliation(s)
- Shan Ling
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jiawei Chen
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Maryse Lapierre-Landry
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Junwoo Suh
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yehe Liu
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael W. Jenkins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michiko Watanabe
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Cleveland, Ohio, USA
- Division of Neonatology, Rainbow Babies and Children’s Hospital, Cleveland, Ohio, USA
| | - Stephanie M. Ford
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Cleveland, Ohio, USA
- Division of Neonatology, Rainbow Babies and Children’s Hospital, Cleveland, Ohio, USA
| | - Andrew M. Rollins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Sankar S, Jayabalan M, Venkatesh S, Ibrahim M. Effect of hyperglycemia on tbx5a and nppa gene expression and its correlation to structural and functional changes in developing zebrafish heart. Cell Biol Int 2022; 46:2173-2184. [PMID: 36069519 DOI: 10.1002/cbin.11901] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022]
Abstract
The objective of the current study is to analyze the effects of gestational diabetes on structural and functional changes in correlation with these two essential regulators of developing hearts in vivo using zebrafish embryos. We employed fertilized zebrafish embryos exposed to a hyperglycemic condition of 25 mM glucose for 96 h postfertilization. The embryos were subjected to various structural and functional analyses in a time-course manner. The data showed that exposure to high glucose significantly affected the embryo's size, heart length, heart rate, and looping of the heart compared to the control. Further, we observed an increased incidence of ventricular standstill and valvular regurgitation with a marked reduction of peripheral blood flow in the high glucose-exposed group compared to the control. In addition, the histological data showed that the high-glucose exposure markedly reduced the thickness of the wall and the number of cardiomyocytes in both atrium and ventricles. We also observed striking alterations in the pericardium like edema, increase in diameter with thinning of the wall compared to the control group. Interestingly, the expression of tbx5a and nppa was increased in the early development and found to be repressed in the later stage of development in the hyperglycemic group compared to the control. In conclusion, the developing heart is more susceptible to hyperglycemia in the womb, thereby showing various developmental defects possibly by altering the expression of crucial gene regulators such as tbx5a and nppa.
Collapse
Affiliation(s)
- Suruthi Sankar
- Department of Anatomy, Dr. ALM Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India
| | - Monisha Jayabalan
- Department of Anatomy, Dr. ALM Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India
| | - Sundararajan Venkatesh
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, WV, United States
| | - Muhammed Ibrahim
- Department of Anatomy, Dr. ALM Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India
| |
Collapse
|
7
|
Abraham S, Lindo C, Peoples J, Cox A, Lytle E, Nguyen V, Mehta M, Alvarez JD, Yooseph S, Pacher P, Ebert SN. Maternal binge alcohol consumption leads to distinctive acute perturbations in embryonic cardiac gene expression profiles. Alcohol Clin Exp Res 2022; 46:1433-1448. [PMID: 35692084 DOI: 10.1111/acer.14880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/09/2022] [Accepted: 06/01/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Excessive alcohol consumption during pregnancy is associated with high risk of congenital heart defects, but it is unclear how alcohol specifically affects heart development during the acute aftermath of a maternal binge drinking episode. We hypothesize that administration of a single maternal binge dose of alcohol to pregnant mice at embryonic day 9.5 (E9.5) causes perturbations in the expression patterns of specific genes in the developing heart in the acute period (1-3 days) following the binge episode. To test this hypothesis and identify strong candidate ethanol-sensitive target genes of interest, we adapted a mouse binge alcohol model that is associated with a high incidence of congenital heart defects as described below. METHODS/RESULTS Pregnant mice were administered a single dose of alcohol (2.5 g/kg in saline) or control (saline alone) via oral gavage. To evaluate the impact of maternal binge alcohol on cardiac gene expression profiles, we isolated embryonic hearts from both groups (n = 5/group) at 24, 48, and 72 h post-gavage for transcriptomic analyses. RNA was extracted and evaluated using quantitative RNA-sequencing (RNA-Seq) methods. To identify a cohort of binge-altered cardiac genes, we set the threshold for change at >2.0-fold difference with adjusted p < 0.05 versus control. RNA-Seq analysis of cardiac gene expression revealed that of the 17 genes that were altered within the first 48 h post-binge, with the largest category consisting of transcription factors (Alx1, Alx4, HoxB7, HoxD8, and Runx2), followed by signaling molecules (Adamts18, Dkk2, Rtl1, and Wnt7a). Furthermore, multiple comparative and pathway analyses suggested that several of the candidate genes identified through differential RNA-Seq analysis may interact through certain common pathways. To investigate this further, we performed gene-specific qPCR analyses for three representative candidate targets: Runx2, Wnt7a, and Mlxipl. Notably, only Wnt7a showed significantly (p < 0.05) decreased expression in response to maternal binge alcohol in the qPCR assays. CONCLUSIONS These findings identify Wnt7a and a short list of potential other candidate genes and pathways for further study, which could provide mechanistic insights into how maternal binge alcohol consumption produces congenital cardiac malformations.
Collapse
Affiliation(s)
- Shani Abraham
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Chad Lindo
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Jessica Peoples
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Amanda Cox
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Erika Lytle
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Vu Nguyen
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Meeti Mehta
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Jose D Alvarez
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Shibu Yooseph
- Department of Computer Science, Genomics and Bioinformatics Cluster, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute of Alcohol and Alcohol Abuse (NIAAA), The National Institutes of Health (NIH), Rockville, Maryland, USA
| | - Steven N Ebert
- Division of Metabolic and Cardiovascular Science, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
8
|
Pai VP, Levin M. HCN2 Channel-induced Rescue of Brain, Eye, Heart, and Gut Teratogenesis Caused by Nicotine, Ethanol, and Aberrant Notch Signaling. Wound Repair Regen 2022; 30:681-706. [PMID: 35662339 DOI: 10.1111/wrr.13032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/28/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022]
Abstract
Organogenesis is a complex process that can be disrupted by embryonic exposure to teratogens or mutation-induced alterations in signaling pathways, both of which result in organ mispatterning. Building on prior work in Xenopus laevis that showed that increased HCN2 ion channel activity rescues nicotine-induced brain & eye morphogenesis, we demonstrate much broader HCN2-based rescue of organ patterning defects. Induced HCN2 expression in both local or distant tissues can rescue CNS (brain & eye) as well as non-CNS (heart, & gut) organ defects induced by three different teratogenic conditions: nicotine exposure, ethanol exposure, or aberrant Notch protein. Rescue can also be induced by small-molecule HCN2 channel activators, even with delayed treatment initiation. Our results suggest that HCN2 (likely mediated by bioelectric signals) can be an effective regulator of organogenesis from all three germ layers (ectoderm, mesoderm, and endoderm) and reveal non-cell-autonomous influences on organ formation that work at considerable distance during embryonic development. These results suggest molecular bioelectric strategies for repair that could be explored in the future for regenerative medicine. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Vaibhav P Pai
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
9
|
Saad H, Sinclair M, Bunting B. Maternal sociodemographic characteristics, early pregnancy behaviours, and livebirth outcomes as congenital heart defects risk factors - Northern Ireland 2010-2014. BMC Pregnancy Childbirth 2021; 21:759. [PMID: 34758755 PMCID: PMC8579547 DOI: 10.1186/s12884-021-04223-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/19/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Congenital Heart Defects (CHD) is the most commonly occurring congenital anomaly in Europe and a major paediatric health care concern. Investigations are needed to enable identification of CHD risk factors as studies have given conflicting results. This study aim was to identify maternal sociodemographic characteristics, behaviours, and birth outcomes as risk factors for CHD. This was a population based, data linkage cohort study using anonymised data from Northern Ireland (NI) covering the period 2010-2014. The study cohort composed of 94,067 live births with an outcome of 1162 cases of CHD using the International Statistical Classification of Diseases and Related Health Problems (ICD)-10 codes and based on the European Surveillance of Congenital Anomalies (EUROCAT) grouping system for CHD. CHD cases were obtained from the HeartSuite database (HSD) at the Royal Belfast Hospital for Sick Children (RBHSC), maternal data were extracted from the Northern Ireland Maternity System (NIMATS), and medication data were extracted from the Enhanced Prescribing Database (EPD). STATA version 14 was used for the statistical analysis in this study, Odds Ratio (OR), 95% Confident intervals (CI), P value, and logistic regression were used in the analysis. Ethical approval was granted from the National Health Service (NHS) Research Ethics Committee. RESULT In this study, a number of potential risk factors were assessed for statistically significant association with CHD, however only certain risk factors demonstrated a statistically significant association with CHD which included: gestational age at first booking (AOR = 1.21; 95% CI = 1.04-1.41; P < 0.05), family history of CHD or congenital abnormalities and syndromes (AOR = 4.14; 95% CI = 2.47-6.96; P < 0.05), woman's smoking in pregnancy (AOR = 1.22; 95% CI = 1.04-1.43; P < 0.05), preterm birth (AOR = 3.01; 95% CI = 2.44-3.01; P < 0.05), multiple births (AOR = 1.89; 95% CI = 1.58-2.60; P < 0.05), history of abortion (AOR = 1.12; 95% CI = 1.03-1.28; P < 0.05), small for gestational age (SGA) (AOR = 1.44; 95% CI = 1.22-1.78; P < 0.05), and low birth weight (LBW) (AOR = 3.10; 95% CI = 2.22-3.55; P < 0.05). Prescriptions and redemptions of antidiabetic (AOR = 2.68; 95% CI = 1.85-3.98; P < 0.05), antiepileptic (AOR = 1.77; 95% CI = 1.10-2.81; P < 0.05), and dihydrofolate reductase inhibitors (DHFRI) (AOR = 2.13; 95% CI = 1.17-5.85; P < 0.05) in early pregnancy also showed evidence of statistically significant association with CHD. CONCLUSION The results of this study suggested that there are certain maternal sociodemographic characteristics, behaviours and birth outcomes that are statistically significantly associated with higher risk of CHD. Appropriate prevention policy to target groups with higher risk for CHD may help to reduce CHD prevalence. These results are important for policy makers, obstetricians, cardiologists, paediatricians, midwives and the public.
Collapse
Affiliation(s)
- Hafi Saad
- Maternal Fetal and Infant Research Centre, Ulster University, Jordanstown, UK.
| | - Marlene Sinclair
- Maternal Fetal and Infant Research Centre, Ulster University, Jordanstown, UK
| | | |
Collapse
|
10
|
Montaña-Jimenez LP, Lasalvia P, Diaz Puentes M, Olaya-C M. Congenital heart defects and umbilical cord abnormalities, an unknown association? J Neonatal Perinatal Med 2021; 15:81-88. [PMID: 34542034 DOI: 10.3233/npm-210799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Few studies exist that research the association between umbilical cord characteristics with cardiac malformations. In this study, we describe a population of newborns with congenital heart defects (CHD) and the frequency of presentation of umbilical cord (UC) alterations, based upon the hypothesis that the continuity of the cardio-placental circuit can be affected by similar noxas during early development. METHODS We carried out a descriptive study at a hospital in Bogota based on clinical records from newborns with congenital heart disease with placental and UC pathology results. Group analyses were done according to the major categories of the ICD-10. RESULTS We analyzed 122 cases and found that the most frequent alterations where hypercoiling (27.9%) and abnormal UC insertion (16.4%). Additionally, in almost every group of CHD, more than 65%of patients had some type of cord alteration. CONCLUSION We discovered a high frequency of UC alterations in patients with CHD. This outcome suggests that a possible association exists between the two phenomena, further research is needed.
Collapse
Affiliation(s)
- L P Montaña-Jimenez
- Hospital Universitario San Ignacio, Bogotá D.C, Colombia.,Pontificia Universidad Javeriana, Bogotá D.C, Colombia
| | | | | | - M Olaya-C
- Hospital Universitario San Ignacio, Bogotá D.C, Colombia.,Pontificia Universidad Javeriana, Bogotá D.C, Colombia
| |
Collapse
|
11
|
Ling S, Jenkins MW, Watanabe M, Ford SM, Rollins AM. Prenatal ethanol exposure impairs the conduction delay at the atrioventricular junction in the looping heart. Am J Physiol Heart Circ Physiol 2021; 321:H294-H305. [PMID: 34142884 PMCID: PMC8526336 DOI: 10.1152/ajpheart.00107.2021] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/27/2022]
Abstract
The etiology of ethanol-related congenital heart defects has been the focus of much study, but most research has concentrated on cellular and molecular mechanisms. We have shown with optical coherence tomography (OCT) that ethanol exposure led to increased retrograde flow and smaller atrioventricular (AV) cushions compared with controls. Since AV cushions play a role in patterning the conduction delay at the atrioventricular junction (AVJ), this study aims to investigate whether ethanol exposure alters the AVJ conduction in early looping hearts and whether this alteration is related to the decreased cushion size. Quail embryos were exposed to a single dose of ethanol at gastrulation, and Hamburger-Hamilton stage 19-20 hearts were dissected for imaging. Cardiac conduction was measured using an optical mapping microscope and we imaged the endocardial cushions using OCT. Our results showed that, compared with controls, ethanol-exposed embryos exhibited abnormally fast AVJ conduction and reduced cushion size. However, this increased conduction velocity (CV) did not strictly correlate with decreased cushion volume and thickness. By matching the CV map to the cushion-size map along the inflow heart tube, we found that the slowest conduction location was consistently at the atrial side of the AVJ, which had the thinner cushions, not at the thickest cushion location at the ventricular side as expected. Our findings reveal regional differences in the AVJ myocardium even at this early stage in heart development. These findings reveal the early steps leading to the heterogeneity and complexity of conduction at the mature AVJ, a site where arrhythmias can be initiated.NEW & NOTEWORTHY To the best of our knowledge, this is the first study investigating the impact of ethanol exposure on the early cardiac conduction system. Our results showed that ethanol-exposed embryos exhibited abnormally fast atrioventricular conduction. In addition, our findings, in CV measurements and endocardial cushion thickness, reveal regional differences in the AVJ myocardium even at this early stage in heart development, suggesting that the differentiation and maturation at this site are complex and warrant further studies.
Collapse
Affiliation(s)
- Shan Ling
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Michael W Jenkins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Michiko Watanabe
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Stephanie M Ford
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children's Hospital, Cleveland, Ohio
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Andrew M Rollins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
12
|
Dyer LA, Rugonyi S. Fetal Blood Flow and Genetic Mutations in Conotruncal Congenital Heart Disease. J Cardiovasc Dev Dis 2021; 8:90. [PMID: 34436232 PMCID: PMC8397097 DOI: 10.3390/jcdd8080090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022] Open
Abstract
In congenital heart disease, the presence of structural defects affects blood flow in the heart and circulation. However, because the fetal circulation bypasses the lungs, fetuses with cyanotic heart defects can survive in utero but need prompt intervention to survive after birth. Tetralogy of Fallot and persistent truncus arteriosus are two of the most significant conotruncal heart defects. In both defects, blood access to the lungs is restricted or non-existent, and babies with these critical conditions need intervention right after birth. While there are known genetic mutations that lead to these critical heart defects, early perturbations in blood flow can independently lead to critical heart defects. In this paper, we start by comparing the fetal circulation with the neonatal and adult circulation, and reviewing how altered fetal blood flow can be used as a diagnostic tool to plan interventions. We then look at known factors that lead to tetralogy of Fallot and persistent truncus arteriosus: namely early perturbations in blood flow and mutations within VEGF-related pathways. The interplay between physical and genetic factors means that any one alteration can cause significant disruptions during development and underscore our need to better understand the effects of both blood flow and flow-responsive genes.
Collapse
Affiliation(s)
- Laura A. Dyer
- Department of Biology, University of Portland, Portland, OR 97203, USA;
| | - Sandra Rugonyi
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
13
|
Ford SM, Pedersen CJ, Ford MR, Kim JW, Karunamuni GH, McPheeters MT, Jawaid S, Jenkins MW, Rollins AM, Watanabe M. Folic acid prevents functional and structural heart defects induced by prenatal ethanol exposure. Am J Physiol Heart Circ Physiol 2021. [DOI: 10.1152/ajpheart.00817.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
State-of-the-art biophotonic tools captured blood flow and endocardial cushion volumes in tiny beating quail embryo hearts, an accessible model for studying four-chambered heart development. Both hemodynamic flow and endocardial cushion volumes were altered with ethanol exposure but normalized when folic acid was introduced with ethanol. Folic acid supplementation preserved hemodynamic function that is intimately involved in sculpting the heart from the earliest stages of heart development.
Collapse
Affiliation(s)
- Stephanie M. Ford
- Division of Neonatology, Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Cameron J. Pedersen
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew R. Ford
- Department of Ophthalmology, Cole Eye Institute, Cleveland Clinic, Cleveland Ohio
| | - Jun W. Kim
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ganga H. Karunamuni
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew T. McPheeters
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Safdar Jawaid
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michael W. Jenkins
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew M. Rollins
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michiko Watanabe
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
14
|
Jawaid S, Strainic JP, Kim J, Ford MR, Thrane L, Karunamuni GH, Sheehan MM, Chowdhury A, Gillespie CA, Rollins AM, Jenkins MW, Watanabe M, Ford SM. Glutathione Protects the Developing Heart from Defects and Global DNA Hypomethylation Induced by Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2021; 45:69-78. [PMID: 33206417 PMCID: PMC8865806 DOI: 10.1111/acer.14511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is caused by prenatal alcohol exposure (PAE), the intake of ethanol (C2 H5 OH) during pregnancy. Features of FASD cover a range of structural and functional defects including congenital heart defects (CHDs). Folic acid and choline, contributors of methyl groups to one-carbon metabolism (OCM), prevent CHDs in humans. Using our avian model of FASD, we have previously reported that betaine, another methyl donor downstream of choline, prevents CHDs. The CHD preventions are substantial but incomplete. Ethanol causes oxidative stress as well as depleting methyl groups for OCM to support DNA methylation and other epigenetic alterations. To identify more compounds that can safely and effectively prevent CHDs and other effects of PAE, we tested glutathione (GSH), a compound that regulates OCM and is known as a "master antioxidant." METHODS/RESULTS Quail embryos injected with a single dose of ethanol at gastrulation exhibited congenital defects including CHDs similar to those identified in FASD individuals. GSH injected simultaneously with ethanol not only prevented CHDs, but also improved survival and prevented other PAE-induced defects. Assays of hearts at 8 days (HH stage 34) of quail development, when the heart normally has developed 4-chambers, showed that this single dose of PAE reduced global DNA methylation. GSH supplementation concurrent with PAE normalized global DNA methylation levels. The same assays performed on quail hearts at 3 days (HH stage 19-20) of development, showed no difference in global DNA methylation between controls, ethanol-treated, GSH alone, and GSH plus ethanol-treated cohorts. CONCLUSIONS GSH supplementation shows promise to inhibit effects of PAE by improving survival, reducing the incidence of morphological defects including CHDs, and preventing global hypomethylation of DNA in heart tissues.
Collapse
Affiliation(s)
- Safdar Jawaid
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - James P. Strainic
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Jun Kim
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Matthew R. Ford
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Lars Thrane
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Ganga H. Karunamuni
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Megan M. Sheehan
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Amrin Chowdhury
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Brecksville-Broadview Heights High School, Broadview Heights OH 44147
| | - Caitlyn A. Gillespie
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Fisk University, Nashville TN 37208
| | - Andrew M. Rollins
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Michael W. Jenkins
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Michiko Watanabe
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Stephanie M Ford
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| |
Collapse
|
15
|
Sun SN, Jiang Q, Lu D, Gui YH. [Effect of dhfr gene overexpression on ethanol-induced abnormal cardiovascular development in zebrafish embryos]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:916-922. [PMID: 32800042 PMCID: PMC7441502 DOI: 10.7499/j.issn.1008-8830.2006079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/15/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To study the effect of dhfr gene overexpression on ethanol-induced abnormal cardiac and vascular development in zebrafish embryos and underlying mechanisms. METHODS dhfr mRNA was transcribed in vitro and microinjected into zebrafish fertilized eggs to induce the overexpression of dhfr gene, and the efficiency of overexpression was verified. Wild-type zebrafish were divided into a control group, an ethanol group, and an ethanol+dhfr overexpression group (microinjection of 6 nL dhfr mRNA). The embryonic development was observed for each group. The transgenic zebrafish Tg (cmlc2:mcherry) with heart-specific red fluorescence was used to observe atrial and ventricular development. Fluorescence microscopy was performed to observe the development of cardiac outflow tract and blood vessels. Heart rate and ventricular shortening fraction were used to assess cardiac function. Gene probes were constructed, and embryo in situ hybridization and real-time PCR were used to measure the expression of nkx2.5, tbx1, and flk-1 in the embryo. RESULTS Compared with the ethanol group, the ethanol+dhfr overexpression group had a significant reduction in the percentage of abnormal embryonic development and a significant increase in the percentage of embryonic survival (P<0.05), with significant improvements in the abnormalities of the atrium, ventricle, outflow tract, and blood vessels and cardiac function. Compared with the control group, the ethanol group had significant reductions in the expression of nkx2.5, tbx1, and flk-1 (P<0.05), and compared with the ethanol group, the ethanol+dhfr overexpression group had significant increases in the expression of nkx2.5, tbx1, and flk-1 (P<0.05), which were still lower than their expression in the control group. CONCLUSIONS The overexpression of the dhfr gene can partially improve the abnormal development of embryonic heart and blood vessels induced by ethanol, possibly by upregulating the decreased expression of nkx2.5, tbx1, and flk-1 caused by ethanol.
Collapse
Affiliation(s)
- Shu-Na Sun
- Department of Cardiology, Children's Hospital, Fudan University, Shanghai 201102, China.
| | | | | | | |
Collapse
|
16
|
Sun SN, Jiang Q, Lu D, Gui YH. [Effect of dhfr gene overexpression on ethanol-induced abnormal cardiovascular development in zebrafish embryos]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:916-922. [PMID: 32800042 PMCID: PMC7441502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/15/2020] [Indexed: 11/13/2023]
Abstract
OBJECTIVE To study the effect of dhfr gene overexpression on ethanol-induced abnormal cardiac and vascular development in zebrafish embryos and underlying mechanisms. METHODS dhfr mRNA was transcribed in vitro and microinjected into zebrafish fertilized eggs to induce the overexpression of dhfr gene, and the efficiency of overexpression was verified. Wild-type zebrafish were divided into a control group, an ethanol group, and an ethanol+dhfr overexpression group (microinjection of 6 nL dhfr mRNA). The embryonic development was observed for each group. The transgenic zebrafish Tg (cmlc2:mcherry) with heart-specific red fluorescence was used to observe atrial and ventricular development. Fluorescence microscopy was performed to observe the development of cardiac outflow tract and blood vessels. Heart rate and ventricular shortening fraction were used to assess cardiac function. Gene probes were constructed, and embryo in situ hybridization and real-time PCR were used to measure the expression of nkx2.5, tbx1, and flk-1 in the embryo. RESULTS Compared with the ethanol group, the ethanol+dhfr overexpression group had a significant reduction in the percentage of abnormal embryonic development and a significant increase in the percentage of embryonic survival (P<0.05), with significant improvements in the abnormalities of the atrium, ventricle, outflow tract, and blood vessels and cardiac function. Compared with the control group, the ethanol group had significant reductions in the expression of nkx2.5, tbx1, and flk-1 (P<0.05), and compared with the ethanol group, the ethanol+dhfr overexpression group had significant increases in the expression of nkx2.5, tbx1, and flk-1 (P<0.05), which were still lower than their expression in the control group. CONCLUSIONS The overexpression of the dhfr gene can partially improve the abnormal development of embryonic heart and blood vessels induced by ethanol, possibly by upregulating the decreased expression of nkx2.5, tbx1, and flk-1 caused by ethanol.
Collapse
Affiliation(s)
- Shu-Na Sun
- Department of Cardiology, Children's Hospital, Fudan University, Shanghai 201102, China.
| | | | | | | |
Collapse
|
17
|
Chen Z, Li S, Guo L, Peng X, Liu Y. Prenatal alcohol exposure induced congenital heart diseases: From bench to bedside. Birth Defects Res 2020; 113:521-534. [PMID: 32578335 DOI: 10.1002/bdr2.1743] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/27/2022]
Abstract
Alcohol consumption is increasing worldwide. Many child-bearing-aged women consume alcohol during pregnancy, intentionally or unintentionally, thereby increasing the potential risk for severe congenital diseases. Congenital heart disease (CHD) is the most common birth defect worldwide and can result from both hereditary and acquired factors. Prenatal alcohol exposure (PAE) is considered a key factor that leads to teratogenesis in CHD and its specific phenotypes, especially defects of the cardiac septa, cardiac valves, cardiac canals, and great arteries, adjacent to the chambers, both in animal experiments and clinical retrospective studies. The mechanisms underlying CHD and its phenotypes caused by PAE are associated with changes in retinoic acid biosynthesis and its signaling pathway, apoptosis and defective function of cardiac neural crest cells, disturbance of the Wntβ-catenin signaling pathway, suppression of bone morphogenetic protein (BMP) signaling, and other epigenetic mechanisms. Drug supplements and early diagnosis can help prevent PAE from inducing CHDs.
Collapse
Affiliation(s)
- Zhiyan Chen
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Sheng Li
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Linghong Guo
- Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China
| | - Xu Peng
- Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China
| | - Yin Liu
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China.,Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China.,Department of Anesthesiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Follow Me! A Tale of Avian Heart Development with Comparisons to Mammal Heart Development. J Cardiovasc Dev Dis 2020; 7:jcdd7010008. [PMID: 32156044 PMCID: PMC7151090 DOI: 10.3390/jcdd7010008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/16/2020] [Accepted: 02/21/2020] [Indexed: 12/19/2022] Open
Abstract
Avian embryos have been used for centuries to study development due to the ease of access. Because the embryos are sheltered inside the eggshell, a small window in the shell is ideal for visualizing the embryos and performing different interventions. The window can then be covered, and the embryo returned to the incubator for the desired amount of time, and observed during further development. Up to about 4 days of chicken development (out of 21 days of incubation), when the egg is opened the embryo is on top of the yolk, and its heart is on top of its body. This allows easy imaging of heart formation and heart development using non-invasive techniques, including regular optical microscopy. After day 4, the embryo starts sinking into the yolk, but still imaging technologies, such as ultrasound, can tomographically image the embryo and its heart in vivo. Importantly, because like the human heart the avian heart develops into a four-chambered heart with valves, heart malformations and pathologies that human babies suffer can be replicated in avian embryos, allowing a unique developmental window into human congenital heart disease. Here, we review avian heart formation and provide comparisons to the mammalian heart.
Collapse
|
19
|
Courchaine K, Rugonyi S. Optical coherence tomography for in vivo imaging of endocardial to mesenchymal transition during avian heart development. BIOMEDICAL OPTICS EXPRESS 2019; 10:5989-5995. [PMID: 31799059 PMCID: PMC6865111 DOI: 10.1364/boe.10.005989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/20/2019] [Accepted: 10/28/2019] [Indexed: 05/08/2023]
Abstract
The endocardial to mesenchymal transition (EndMT) that occurs in endocardial cushions during heart development is critical for proper heart septation and formation of the heart's valves. In EndMT, cells delaminate from the endocardium and migrate into the previously acellular endocardial cushions. Optical coherence tomography (OCT) imaging uses the optical properties of tissues for contrast, and during early development OCT can differentiate cellular versus acellular tissues. Here we show that OCT can be used to non-invasively track EndMT progression in vivo in the outflow tract cushions of chicken embryos. This enables in vivo studies to elucidate factors leading to cardiac malformations.
Collapse
Affiliation(s)
- Katherine Courchaine
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave. Mail/Code:CH13B, Portland, OR 97239, USA
| | - Sandra Rugonyi
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave. Mail/Code:CH13B, Portland, OR 97239, USA
| |
Collapse
|
20
|
Deniz E, Jonas S, Khokha MK, Choma MA. Quantitative Phenotyping of Xenopus Embryonic Heart Pathophysiology Using Hemoglobin Contrast Subtraction Angiography to Screen Human Cardiomyopathies. Front Physiol 2019; 10:1197. [PMID: 31620018 PMCID: PMC6763566 DOI: 10.3389/fphys.2019.01197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 09/03/2019] [Indexed: 01/09/2023] Open
Abstract
Congenital heart disease (CHD) is a significant cause of mortality in infants and adults. Currently human genomic analysis has identified a number of candidate genes in these patients. These genes span diverse categories of gene function suggesting that despite the similarity in cardiac lesion, the underlying pathophysiology may be different. In fact, patients with similar CHDs can have drastically different outcomes, including a dramatic decrease in myocardial function. To test these human candidate genes for their impact on myocardial function, we need efficient animals models of disease. For this purpose, we paired Xenopus tropicalis with our microangiography technique, hemoglobin contrast subtraction angiography (HCSA). To demonstrate the gene-teratogen-physiology relationship, we modeled human cardiomyopathy in tadpoles. First we depleted the sarcomeric protein myosin heavy chain 6 (myh6) expression using morpholino oligos. Next, we exposed developing embryos to the teratogen ethanol and in both conditions showed varying degrees of cardiac dysfunction. Our results demonstrate that HCSA can distinguish biomechanical phenotypes in the context of gene dysfunction or teratogen. This approach can be used to screen numerous candidate CHD genes or suspected teratogens for their effect on cardiac function.
Collapse
Affiliation(s)
- Engin Deniz
- Department of Pediatrics, Yale University, New Haven, CT, United States
| | - Stephan Jonas
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Mustafa K Khokha
- Department of Pediatrics, Yale University, New Haven, CT, United States.,Department of Genetics, Yale University, New Haven, CT, United States
| | - Michael A Choma
- Department of Pediatrics, Yale University, New Haven, CT, United States.,Department of Diagnostic Radiology, Yale University, New Haven, CT, United States.,Department of Biomedical Engineering, Yale University, New Haven, CT, United States.,Department of Applied Physics, Yale University, New Haven, CT, United States
| |
Collapse
|
21
|
Ninh VK, El Hajj EC, Mouton AJ, Gardner JD. Prenatal Alcohol Exposure Causes Adverse Cardiac Extracellular Matrix Changes and Dysfunction in Neonatal Mice. Cardiovasc Toxicol 2019; 19:389-400. [PMID: 30684169 PMCID: PMC7261018 DOI: 10.1007/s12012-018-09503-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Fetal alcohol syndrome (FAS) is the most severe condition of fetal alcohol spectrum disorders (FASD) and is associated with congenital heart defects. However, more subtle defects such as ventricular wall thinning and cardiac compliance may be overlooked in FASD. Our studies focus on the role of cardiac fibroblasts in the neonatal heart, and how they are affected by prenatal alcohol exposure (PAE). We hypothesize that PAE affects fibroblast function contributing to dysregulated collagen synthesis, which leads to cardiac dysfunction. To investigate these effects, pregnant C57/BL6 mice were intraperitoneally injected with 2.9 g EtOH/kg dose to achieve a blood alcohol content of approximately 0.35 on gestation days 6.75 and 7.25. Pups were sacrificed on neonatal day 5 following echocardiography measurements of left ventricular (LV) chamber dimension and function. Hearts were used for primary cardiac fibroblast isolation or protein expression analysis. PAE animals had thinner ventricular walls than saline exposed animals, which was associated with increased LV wall stress and decreased ejection fraction. In isolated fibroblasts, PAE decreased collagen I/III ratio and increased gene expression of profibrotic markers, including α-smooth muscle actin and lysyl oxidase. Notch1 signaling was assessed as a possible mechanism for fibroblast activation, and indicated that gene expression of Notch1 receptor and downstream Hey1 transcription factor were increased. Cardiac tissue analysis revealed decreased collagen I/III ratio and increased protein expression of α-smooth muscle actin and lysyl oxidase. However, Notch1 signaling components decreased in whole heart tissue. Our study demonstrates that PAE caused adverse changes in the cardiac collagen profile and a decline in cardiac function in the neonatal heart.
Collapse
Affiliation(s)
- Van K Ninh
- Department of Physiology, LSU Health Sciences Center, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Elia C El Hajj
- Department of Physiology, LSU Health Sciences Center, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Alan J Mouton
- Department of Physiology, LSU Health Sciences Center, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Jason D Gardner
- Department of Physiology, LSU Health Sciences Center, 1901 Perdido Street, New Orleans, LA, 70112, USA.
| |
Collapse
|
22
|
Cook JC, Lynch ME, Coles CD. Association Analysis: Fetal Alcohol Spectrum Disorder and Hypertension Status in Children and Adolescents. Alcohol Clin Exp Res 2019; 43:1727-1733. [DOI: 10.1111/acer.14121] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 05/27/2019] [Indexed: 11/26/2022]
|
23
|
Dong Y, Zhang Y, Tong S, Jiang Z, Xu Z, Li X, Wang W. Analysis of the Seasonal Trend of Congenital Heart Defects. J Pediatr 2019; 207:29-33.e1. [PMID: 30922502 DOI: 10.1016/j.jpeds.2018.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To determine the seasonal trend of congenital heart defects (CHDs) in China using hospital-based clinical data. STUDY DESIGN We included 40 501 patients with CHD hospitalized at the Shanghai Children Medical Center between 2006 and 2017. The birth rate of CHD in each month was adjusted by sex, year of birth, and monthly birth rate of the general population. Negative binomial regression models were used to assess the seasonal trend of CHD. RESULTS The included patients consisted of 22 600 boys (55.8%), resulting in a male-to-female ratio of 1.26:1. Among subtypes of CHDs, ventricular septal defects and atrial septal defects were the most common, accounting for 39.7% and 12.6%, respectively. A statistically significant seasonal trend in the monthly birth rate of patients with CHDs was found; the highest relative rate of CHD was found in October and the lowest in April. After adjusting for the potential confounders, the highest relative rate of CHD was found in October and the lowest in November. CONCLUSIONS There seems to be a significant monthly birth rate variation of CHDs in China. The highest relative rate of CHDs occurred in October, suggesting possible maternal exposure to environmental hazards from January to March. These hazards may include air pollution, virus infection, and unhealthy lifestyle behaviors during the Spring Festival.
Collapse
Affiliation(s)
- Yuanyuan Dong
- Child Health Advocacy Institute, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, PR China
| | - Yunting Zhang
- Child Health Advocacy Institute, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, PR China
| | - Shilu Tong
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia; Department of Clinical Epidemiology and Biostatistics, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, PR China
| | - Zhongyi Jiang
- Child Health Advocacy Institute, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, PR China
| | - Zhiwei Xu
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia
| | - Xinyue Li
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT
| | - Wei Wang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, PR China.
| |
Collapse
|
24
|
Liu Y, Broberg MCG, Watanabe M, Rollins AM, Jenkins MW. SLIME: robust, high-speed 3D microvascular mapping. Sci Rep 2019; 9:893. [PMID: 30696870 PMCID: PMC6351571 DOI: 10.1038/s41598-018-37313-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/30/2018] [Indexed: 01/08/2023] Open
Abstract
Three dimensional (3D) microvascular imaging of cubic millimeter to centimeter size volumes often requires much time and expensive instruments. By combining optical clearing with a novel scatter-based optical coherence tomography (OCT) contrast agent, we have greatly extended OCT imaging depth in excised tissues while maintaining a simple and low cost approach that does not require in-depth OCT knowledge. The new method enables fast 3D microvascular mapping in large tissue volumes, providing a promising tool for investigating organ level microvascular abnormalities in large cohorts.
Collapse
Affiliation(s)
- Yehe Liu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Meredith C G Broberg
- Department of Pediatrics, Case Western Reserve University, Cleveland, USA.,Division of Pediatric Critical Care, UH Rainbow Babies & Children's Hospital, Cleveland, USA
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University, Cleveland, USA
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Michael W Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA. .,Department of Pediatrics, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
25
|
Courchaine K, Rykiel G, Rugonyi S. Influence of blood flow on cardiac development. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 137:95-110. [PMID: 29772208 PMCID: PMC6109420 DOI: 10.1016/j.pbiomolbio.2018.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/06/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
Abstract
The role of hemodynamics in cardiovascular development is not well understood. Indeed, it would be remarkable if it were, given the dauntingly complex array of intricately synchronized genetic, molecular, mechanical, and environmental factors at play. However, with congenital heart defects affecting around 1 in 100 human births, and numerous studies pointing to hemodynamics as a factor in cardiovascular morphogenesis, this is not an area in which we can afford to remain in the dark. This review seeks to present the case for the importance of research into the biomechanics of the developing cardiovascular system. This is accomplished by i) illustrating the basics of some of the highly complex processes involved in heart development, and discussing the known influence of hemodynamics on those processes; ii) demonstrating how altered hemodynamic environments have the potential to bring about morphological anomalies, citing studies in multiple animal models with a variety of perturbation methods; iii) providing examples of widely used technological innovations which allow for accurate measurement of hemodynamic parameters in embryos; iv) detailing the results of studies in avian embryos which point to exciting correlations between various hemodynamic manipulations in early development and phenotypic defect incidence in mature hearts; and finally, v) stressing the relevance of uncovering specific biomechanical pathways involved in cardiovascular formation and remodeling under adverse conditions, to the potential treatment of human patients. The time is ripe to unravel the contributions of hemodynamics to cardiac development, and to recognize their frequently neglected role in the occurrence of heart malformation phenotypes.
Collapse
Affiliation(s)
- Katherine Courchaine
- Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland OR, USA
| | - Graham Rykiel
- Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland OR, USA
| | - Sandra Rugonyi
- Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland OR, USA.
| |
Collapse
|
26
|
Raghunathan R, Wu C, Singh M, Liu CH, Miranda RC, Larin KV. Evaluating the effects of maternal alcohol consumption on murine fetal brain vasculature using optical coherence tomography. JOURNAL OF BIOPHOTONICS 2018; 11:e201700238. [PMID: 29292845 PMCID: PMC6292438 DOI: 10.1002/jbio.201700238] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/28/2017] [Indexed: 05/09/2023]
Abstract
Prenatal alcohol exposure (PAE) can result in a range of anomalies including brain and behavioral dysfunctions, collectively termed fetal alcohol spectrum disorder. PAE during the 1st and 2nd trimester is common, and research in animal models has documented significant neural developmental deficits associated with PAE during this period. However, little is known about the immediate effects of PAE on fetal brain vasculature. In this study, we used in utero speckle variance optical coherence tomography, a high spatial- and temporal-resolution imaging modality, to evaluate dynamic changes in microvasculature of the 2nd trimester equivalent murine fetal brain, minutes after binge-like maternal alcohol exposure. Acute binge-like PAE resulted in a rapid (<1 hour) and significant decrease (P < .001) in vessel diameter as compared to the sham group. The data show that a single binge-like maternal alcohol exposure resulted in swift vasoconstriction in fetal brain vessels during the critical period of neurogenesis.
Collapse
Affiliation(s)
- Raksha Raghunathan
- Department of Biomedical Engineering, University of Houston, Houston, Texas
| | - Chen Wu
- Department of Biomedical Engineering, University of Houston, Houston, Texas
| | - Manmohan Singh
- Department of Biomedical Engineering, University of Houston, Houston, Texas
| | - Chih-Hao Liu
- Department of Biomedical Engineering, University of Houston, Houston, Texas
| | - Rajesh C. Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, College Station, Texas
| | - Kirill V. Larin
- Department of Biomedical Engineering, University of Houston, Houston, Texas
- Interdisciplinary Laboratory of Biophotonics, Tomsk State University, Tomsk, Russia
- Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
- Correspondence: Kirill V. Larin, Department of Biomedical Engineering, University of Houston, Houston, TX.
| |
Collapse
|
27
|
Feng Y, Cai J, Tong X, Chen R, Zhu Y, Xu B, Mo X. Non-inheritable risk factors during pregnancy for congenital heart defects in offspring: A matched case-control study. Int J Cardiol 2018; 264:45-52. [PMID: 29685690 DOI: 10.1016/j.ijcard.2018.04.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/04/2018] [Accepted: 04/02/2018] [Indexed: 11/26/2022]
Affiliation(s)
- Yu Feng
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China; Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Jun Cai
- Ministry of Education Key Laboratory for Earth System Modelling, Department of Earth System Science, Tsinghua University, Beijing 100084, China; Joint Center for Global Change Studies, Beijing 100875, China
| | - Xing Tong
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Runsen Chen
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Yu Zhu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Bing Xu
- Ministry of Education Key Laboratory for Earth System Modelling, Department of Earth System Science, Tsinghua University, Beijing 100084, China; Joint Center for Global Change Studies, Beijing 100875, China.
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
28
|
Elahi S, Gu S, Thrane L, Rollins AM, Jenkins MW. Complex regression Doppler optical coherence tomography. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-8. [PMID: 29704328 PMCID: PMC5920204 DOI: 10.1117/1.jbo.23.4.046009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/09/2018] [Indexed: 05/06/2023]
Abstract
We introduce a new method to measure Doppler shifts more accurately and extend the dynamic range of Doppler optical coherence tomography (OCT). The two-point estimate of the conventional Doppler method is replaced with a regression that is applied to high-density B-scans in polar coordinates. We built a high-speed OCT system using a 1.68-MHz Fourier domain mode locked laser to acquire high-density B-scans (16,000 A-lines) at high enough frame rates (∼100 fps) to accurately capture the dynamics of the beating embryonic heart. Flow phantom experiments confirm that the complex regression lowers the minimum detectable velocity from 12.25 mm / s to 374 μm / s, whereas the maximum velocity of 400 mm / s is measured without phase wrapping. Complex regression Doppler OCT also demonstrates higher accuracy and precision compared with the conventional method, particularly when signal-to-noise ratio is low. The extended dynamic range allows monitoring of blood flow over several stages of development in embryos without adjusting the imaging parameters. In addition, applying complex averaging recovers hidden features in structural images.
Collapse
Affiliation(s)
- Sahar Elahi
- Case Western Reserve University, Department of Pediatrics, Cleveland, Ohio, United States
| | - Shi Gu
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, Ohio, United States
| | - Lars Thrane
- Case Western Reserve University, Department of Pediatrics, Cleveland, Ohio, United States
| | - Andrew M. Rollins
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, Ohio, United States
| | - Michael W. Jenkins
- Case Western Reserve University, Department of Pediatrics, Cleveland, Ohio, United States
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, Ohio, United States
- Address all correspondence to: Michael W. Jenkins, E-mail:
| |
Collapse
|
29
|
Flentke GR, Smith SM. The avian embryo as a model for fetal alcohol spectrum disorder. Biochem Cell Biol 2017; 96:98-106. [PMID: 29024604 DOI: 10.1139/bcb-2017-0205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Prenatal alcohol exposure (PAE) remains a leading preventable cause of structural birth defects and permanent neurodevelopmental disability. The chicken (Gallus gallus domesticus) is a powerful embryological research model, and was possibly the first in which the teratogenicity of alcohol was demonstrated. Pharmacologically relevant exposure to alcohol in the range of 20-70 mmol/L (20-80 mg/egg) disrupt the growth of chicken embryos, morphogenesis, and behavior, and the resulting phenotypes strongly parallel those of mammalian models. The avian embryo's direct accessibility has enabled novel insights into the teratogenic mechanisms of alcohol. These include the contribution of IGF1 signaling to growth suppression, the altered flow dynamics that reshape valvuloseptal morphogenesis and mediate its cardiac teratogenicity, and the suppression of Wnt and Shh signals thereby disrupting the migration, expansion, and survival of the neural crest, and underlie its characteristic craniofacial deficits. The genetic diversity within commercial avian strains has enabled the identification of unique loci, such as ribosome biogenesis, that modify vulnerability to alcohol. This venerable research model is equally relevant for the future, as the application of technological advances including CRISPR, optogenetics, and biophotonics to the embryo's ready accessibility creates a unique model in which investigators can manipulate and monitor the embryo in real-time to investigate the effect of alcohol on cell fate.
Collapse
Affiliation(s)
- George R Flentke
- UNC-Nutrition Research Institute and Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA.,UNC-Nutrition Research Institute and Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Susan M Smith
- UNC-Nutrition Research Institute and Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA.,UNC-Nutrition Research Institute and Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
30
|
Karunamuni G, Sheehan MM, Doughman YQ, Gu S, Sun J, Li Y, Strainic JP, Rollins AM, Jenkins MW, Watanabe M. Supplementation with the Methyl Donor Betaine Prevents Congenital Defects Induced by Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2017; 41:1917-1927. [PMID: 28888041 DOI: 10.1111/acer.13495] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 08/29/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Despite decades of public education about dire consequences of prenatal alcohol exposure (PAE), drinking alcohol during pregnancy remains prevalent. As high as 40% of live-born infants exposed to alcohol during gestation and diagnosed with fetal alcohol syndrome have congenital heart defects that can be life-threatening. In animal models, the methyl donor betaine, found in foods such as wheat bran, quinoa, beets, and spinach, ameliorated neurobehavioral deficits associated with PAE, but effects on heart development are unknown. METHODS Previously, we modeled a binge drinking episode during the first trimester in avian embryos. Here, we investigated whether betaine could prevent adverse effects of alcohol on heart development. Embryos exposed to ethanol (EtOH) with and without an optimal dose of betaine (5 μM) were analyzed at late developmental stages. Cardiac morphology parameters were rapidly analyzed and quantified using optical coherence tomography. DNA methylation at early stages was detected by immunofluorescent staining for 5-methylcytosine in sections of embryos treated with EtOH or cotreated with betaine. RESULTS Compared to EtOH-exposed embryos, betaine-supplemented embryos had higher late-stage survival rates and fewer gross head and body defects than seen after alcohol exposure alone. Betaine also reduced the incidence of late-stage cardiac defects such as absent vessels, abnormal atrioventricular (AV) valves, and hypertrophic ventricles. Furthermore, betaine cotreatment brought measurements of great vessel diameters, interventricular septum thickness, and AV leaflet volumes in betaine-supplemented embryos close to control values. Early-stage 5-methycytosine staining revealed that DNA methylation levels were reduced by EtOH exposure and normalized by co-administration with betaine. CONCLUSIONS This is the first study demonstrating efficacy of the methyl donor betaine in alleviating cardiac defects associated with PAE. These findings highlight the therapeutic potential of low-concentration betaine doses in mitigating PAE-induced birth defects and have implications for prenatal nutrition policies, especially for women who may not be responsive to folate supplementation.
Collapse
Affiliation(s)
- Ganga Karunamuni
- Department of Pediatrics, Congenital Heart Collaborative, UH Rainbow Babies and Children's Hospital, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Megan M Sheehan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Yong Qiu Doughman
- Department of Pediatrics, Congenital Heart Collaborative, UH Rainbow Babies and Children's Hospital, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Jiayang Sun
- Department of Population and Quantitative Health Sciences, Center for Statistical Research, Computing and Collaboration, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Youjun Li
- Department of Population and Quantitative Health Sciences, Center for Statistical Research, Computing and Collaboration, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - James P Strainic
- Department of Pediatrics, Congenital Heart Collaborative, UH Rainbow Babies and Children's Hospital, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Michael W Jenkins
- Department of Pediatrics, Congenital Heart Collaborative, UH Rainbow Babies and Children's Hospital, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Michiko Watanabe
- Department of Pediatrics, Congenital Heart Collaborative, UH Rainbow Babies and Children's Hospital, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
31
|
Ford SM, McPheeters MT, Wang YT, Ma P, Gu S, Strainic J, Snyder C, Rollins AM, Watanabe M, Jenkins MW. Increased regurgitant flow causes endocardial cushion defects in an avian embryonic model of congenital heart disease. CONGENIT HEART DIS 2017; 12:322-331. [PMID: 28211263 PMCID: PMC5467887 DOI: 10.1111/chd.12443] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/06/2016] [Accepted: 12/08/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The relationship between changes in endocardial cushion and resultant congenital heart diseases (CHD) has yet to be established. It has been shown that increased regurgitant flow early in embryonic heart development leads to endocardial cushion defects, but it remains unclear how abnormal endocardial cushions during the looping stages might affect the fully septated heart. The goal of this study was to reproducibly alter blood flow in vivo and then quantify the resultant effects on morphology of endocardial cushions in the looping heart and on CHDs in the septated heart. METHODS Optical pacing was applied to create regurgitant flow in embryonic hearts, and optical coherence tomography (OCT) was utilized to quantify regurgitation and morphology. Embryonic quail hearts were optically paced at 3 Hz (180 bpm, well above intrinsic rate 60-110 bpm) at stage 13 of development (3-4 weeks human) for 5 min. Pacing fatigued the heart and led to at least 1 h of increased regurgitant flow. Resultant morphological changes were quantified with OCT imaging at stage 19 (cardiac looping-4-5 weeks human) or stage 35 (4 chambered heart-8 weeks human). RESULTS All paced embryos imaged at stage 19 displayed structural changes in cardiac cushions. The amount of regurgitant flow immediately after pacing was inversely correlated with cardiac cushion size 24-h post pacing (P value < .01). The embryos with the most regurgitant flow and smallest cushions after pacing had a decreased survival rate at 8 days (P < .05), indicating that those most severe endocardial cushion defects were lethal. Of the embryos that survived to stage 35, 17/18 exhibited CHDs including valve defects, ventricular septal defects, hypoplastic ventricles, and common AV canal. CONCLUSION The data illustrate a strong inverse relationship in which regurgitant flow precedes abnormal and smaller cardiac cushions, resulting in the development of CHDs.
Collapse
Affiliation(s)
- Stephanie M Ford
- Rainbow Babies and Children's Hospital Division of Neonatology, University Hospitals, Cleveland, Ohio, USA
| | - Matthew T McPheeters
- Department of Pediatric Cardiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Yves T Wang
- Case Western Reserve University Department of Biomedical Engineering, Cleveland, Ohio, USA
| | - Pei Ma
- Case Western Reserve University Department of Biomedical Engineering, Cleveland, Ohio, USA
| | - Shi Gu
- Case Western Reserve University Department of Biomedical Engineering, Cleveland, Ohio, USA
| | - James Strainic
- Rainbow Babies and Children's Hospital Division of Pediatric Cardiology, University Hospitals, Cleveland, Ohio, USA
| | - Christopher Snyder
- Rainbow Babies and Children's Hospital Division of Pediatric Cardiology, University Hospitals, Cleveland, Ohio, USA
| | - Andrew M Rollins
- Case Western Reserve University Department of Biomedical Engineering, Cleveland, Ohio, USA
| | - Michiko Watanabe
- Department of Pediatric Cardiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Michael W Jenkins
- Department of Pediatric Cardiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
32
|
Peterson LM, Gu S, Karunamuni G, Jenkins MW, Watanabe M, Rollins AM. Embryonic aortic arch hemodynamics are a functional biomarker for ethanol-induced congenital heart defects [Invited]. BIOMEDICAL OPTICS EXPRESS 2017; 8:1823-1837. [PMID: 28663868 PMCID: PMC5480583 DOI: 10.1364/boe.8.001823] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/27/2017] [Accepted: 01/27/2017] [Indexed: 05/19/2023]
Abstract
The great arteries develop from symmetrical aortic arch arteries which are extensively remodeled. These events are vulnerable to perturbations. Hemodynamic forces have a significant role in this remodeling. In this study, optical coherence tomography (OCT) visualized live avian embryos for staging and measuring pharyngeal arch morphology. Measurements acquired with our orientation-independent, dual-angle Doppler OCT technique revealed that ethanol exposure leads to higher absolute blood flow, shear stress, and retrograde flow. Ethanol-exposed embryos had smaller cardiac neural crest (CNC) derived pharyngeal arch mesenchyme and fewer migrating CNC-derived cells. These differences in forces and CNC cell numbers could explain the abnormal aortic arch remodeling.
Collapse
Affiliation(s)
- Lindsy M. Peterson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Ganga Karunamuni
- Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Michael W. Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Michiko Watanabe
- Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Andrew M. Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| |
Collapse
|
33
|
Ma P, Gu S, Karunamuni GH, Jenkins MW, Watanabe M, Rollins AM. Cardiac neural crest ablation results in early endocardial cushion and hemodynamic flow abnormalities. Am J Physiol Heart Circ Physiol 2016; 311:H1150-H1159. [PMID: 27542407 PMCID: PMC5130492 DOI: 10.1152/ajpheart.00188.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/17/2016] [Indexed: 12/22/2022]
Abstract
Cardiac neural crest cell (CNCC) ablation creates congenital heart defects (CHDs) that resemble those observed in many syndromes with craniofacial and cardiac consequences. The loss of CNCCs causes a variety of great vessel defects, including persistent truncus arteriosus and double-outlet right ventricle. However, because of the lack of quantitative volumetric measurements, less severe defects, such as great vessel size changes and valve defects, have not been assessed. Also poorly understood is the role of abnormal cardiac function in the progression of CNCC-related CHDs. CNCC ablation was previously reported to cause abnormal cardiac function in early cardiogenesis, before the CNCCs arrive in the outflow region of the heart. However, the affected functional parameters and how they correlate with the structural abnormalities were not fully characterized. In this study, using a CNCC-ablated quail model, we contribute quantitative phenotyping of CNCC ablation-related CHDs and investigate abnormal early cardiac function, which potentially contributes to late-stage CHDs. Optical coherence tomography was used to assay early- and late-stage embryos and hearts. In CNCC-ablated embryos at four-chambered heart stages, great vessel diameter and left atrioventricular valve leaflet volumes are reduced. Earlier, at cardiac looping stages, CNCC-ablated embryos exhibit abnormally twisted bodies, abnormal blood flow waveforms, increased retrograde flow percentage, and abnormal cardiac cushions. The phenotypes observed in this CNCC-ablation model were also strikingly similar to those found in an established avian fetal alcohol syndrome model, supporting the contribution of CNCC dysfunction to the development of alcohol-induced CHDs.
Collapse
Affiliation(s)
- Pei Ma
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio; and
| | - Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio; and
| | - Ganga H Karunamuni
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Michael W Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio; and
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio; and
| |
Collapse
|
34
|
Bulk A, Bark D, Johnson B, Garrity D, Dasi LP. Mechanisms influencing retrograde flow in the atrioventricular canal during early embryonic cardiogenesis. J Biomech 2016; 49:3162-3167. [DOI: 10.1016/j.jbiomech.2016.07.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 07/23/2016] [Accepted: 07/26/2016] [Indexed: 12/21/2022]
|
35
|
Impaired ADAMTS9 secretion: A potential mechanism for eye defects in Peters Plus Syndrome. Sci Rep 2016; 6:33974. [PMID: 27687499 PMCID: PMC5043182 DOI: 10.1038/srep33974] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/06/2016] [Indexed: 01/15/2023] Open
Abstract
Peters Plus syndrome (PPS), a congenital disorder of glycosylation, results from recessive mutations affecting the glucosyltransferase B3GLCT, leading to congenital corneal opacity and diverse extra-ocular manifestations. Together with the fucosyltransferase POFUT2, B3GLCT adds Glucoseβ1-3Fucose disaccharide to a consensus sequence in thrombospondin type 1 repeats (TSRs) of several proteins. Which of these target proteins is functionally compromised in PPS is unknown. We report here that haploinsufficiency of murine Adamts9, encoding a secreted metalloproteinase with 15 TSRs, leads to congenital corneal opacity and Peters anomaly (persistent lens-cornea adhesion), which is a hallmark of PPS. Mass spectrometry of recombinant ADAMTS9 showed that 9 of 12 TSRs with the O-fucosylation consensus sequence carried the Glucoseβ1-3Fucose disaccharide and B3GLCT knockdown reduced ADAMTS9 secretion in HEK293F cells. Together, the genetic and biochemical findings imply a dosage-dependent role for ADAMTS9 in ocular morphogenesis. Reduced secretion of ADAMTS9 in the absence of B3GLCT is proposed as a mechanism of Peters anomaly in PPS. The functional link between ADAMTS9 and B3GLCT established here also provides credence to their recently reported association with age-related macular degeneration.
Collapse
|
36
|
Embryonic Ethanol Exposure Dysregulates BMP and Notch Signaling, Leading to Persistent Atrio-Ventricular Valve Defects in Zebrafish. PLoS One 2016; 11:e0161205. [PMID: 27556898 PMCID: PMC4996461 DOI: 10.1371/journal.pone.0161205] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/01/2016] [Indexed: 12/30/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD), birth defects associated with ethanol exposure in utero, includes a wide spectrum of congenital heart defects (CHDs), the most prevalent of which are septal and conotruncal defects. Zebrafish FASD model was used to dissect the mechanisms underlying FASD-associated CHDs. Embryonic ethanol exposure (3–24 hours post fertilization) led to defects in atrio-ventricular (AV) valvulogenesis beginning around 37 hpf, a morphogenetic event that arises long after ethanol withdrawal. Valve leaflets of the control embryos comprised two layers of cells confined at the compact atrio-ventricular canal (AVC). Ethanol treated embryos had extended AVC and valve forming cells were found either as rows of cells spanning the AVC or as unorganized clusters near the AV boundary. Ethanol exposure reduced valve precursors at the AVC, but some ventricular cells in ethanol treated embryos exhibited few characteristics of valve precursors. Late staged larvae and juvenile fish exposed to ethanol during embryonic development had faulty AV valves. Examination of AVC morphogenesis regulatory networks revealed that early ethanol exposure disrupted the Bmp signaling gradient in the heart during valve formation. Bmp signaling was prominent at the AVC in controls, but ethanol-exposed embryos displayed active Bmp signaling throughout the ventricle. Ethanol exposure also led to mislocalization of Notch signaling cells in endocardium during AV valve formation. Normally, highly active Notch signaling cells were organized at the AVC. In ethanol-exposed embryos, highly active Notch signaling cells were dispersed throughout the ventricle. At later stages, ethanol-exposed embryos exhibited reduced Wnt/β-catenin activity at the AVC. We conclude that early embryonic ethanol exposure alters Bmp, Notch and other signaling activities during AVC differentiation leading to faulty valve morphogenesis and valve defects persist in juvenile fish.
Collapse
|
37
|
Lu Q, Yao Y, Hu Z, Hu C, Song Q, Ye J, Xu C, Wang AZ, Chen Q, Wang QK. Angiogenic Factor AGGF1 Activates Autophagy with an Essential Role in Therapeutic Angiogenesis for Heart Disease. PLoS Biol 2016; 14:e1002529. [PMID: 27513923 PMCID: PMC4981375 DOI: 10.1371/journal.pbio.1002529] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/12/2016] [Indexed: 01/13/2023] Open
Abstract
AGGF1 is an angiogenic factor with therapeutic potential to treat coronary artery disease (CAD) and myocardial infarction (MI). However, the underlying mechanism for AGGF1-mediated therapeutic angiogenesis is unknown. Here, we show for the first time that AGGF1 activates autophagy, a housekeeping catabolic cellular process, in endothelial cells (ECs), HL1, H9C2, and vascular smooth muscle cells. Studies with Atg5 small interfering RNA (siRNA) and the autophagy inhibitors bafilomycin A1 (Baf) and chloroquine demonstrate that autophagy is required for AGGF1-mediated EC proliferation, migration, capillary tube formation, and aortic ring-based angiogenesis. Aggf1+/- knockout (KO) mice show reduced autophagy, which was associated with inhibition of angiogenesis, larger infarct areas, and contractile dysfunction after MI. Protein therapy with AGGF1 leads to robust recovery of myocardial function and contraction with increased survival, increased ejection fraction, reduction of infarct areas, and inhibition of cardiac apoptosis and fibrosis by promoting therapeutic angiogenesis in mice with MI. Inhibition of autophagy in mice by bafilomycin A1 or in Becn1+/- and Atg5 KO mice eliminates AGGF1-mediated angiogenesis and therapeutic actions, indicating that autophagy acts upstream of and is essential for angiogenesis. Mechanistically, AGGF1 initiates autophagy by activating JNK, which leads to activation of Vps34 lipid kinase and the assembly of Becn1-Vps34-Atg14 complex involved in the initiation of autophagy. Our data demonstrate that (1) autophagy is essential for effective therapeutic angiogenesis to treat CAD and MI; (2) AGGF1 is critical to induction of autophagy; and (3) AGGF1 is a novel agent for treatment of CAD and MI. Our data suggest that maintaining or increasing autophagy is a highly innovative strategy to robustly boost the efficacy of therapeutic angiogenesis. Treatment with the angiogenic factor AGGF1 dramatically improves survival and cardiac function in mouse models for coronary artery disease and myocardial infarction by activating autophagy and angiogenesis. Coronary artery disease is the number one killer disease worldwide. Recently, therapeutic angiogenesis has been proposed as an attractive new strategy for treating this and other ischemic diseases. This study establishes the angiogenic factor AGGF1 as a novel target and agent that can successfully treat coronary artery disease and acute myocardial infarction and dramatically improve survival and cardiac function in mouse models. We present the unexpected finding that AGGF1 has these effects via activating autophagy, and that autophagy is essential for therapeutic angiogenesis in animals. We find that AGGF1 is a novel master regulator of autophagy not only in endothelial cells but also in all other cell types examined in the study. Mechanistically, AGGF1 activates autophagy by activating JNK, which leads to activation of the Vps34 lipid kinase and assembly of the Becn1-Vps34-Atg14 complex involved in the initiation of autophagy. The study thus provides a link connecting the therapeutic angiogenesis and autophagy pathways in heart disease.
Collapse
MESH Headings
- Angiogenic Proteins/genetics
- Angiogenic Proteins/metabolism
- Angiogenic Proteins/pharmacology
- Animals
- Autophagy/drug effects
- Autophagy/genetics
- Autophagy/physiology
- Autophagy-Related Protein 5/genetics
- Autophagy-Related Protein 5/metabolism
- Beclin-1/genetics
- Beclin-1/metabolism
- Blotting, Western
- Cell Line
- Cells, Cultured
- Enzyme Inhibitors/pharmacology
- Heart Diseases/drug therapy
- Heart Diseases/genetics
- Heart Diseases/metabolism
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/physiology
- Humans
- Macrolides/pharmacology
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Physiologic/drug effects
- Recombinant Proteins/metabolism
- Recombinant Proteins/pharmacology
Collapse
Affiliation(s)
- Qiulun Lu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Zhenkun Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Changqing Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Qixue Song
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Jian Ye
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Annabel Z. Wang
- Duke University, Durham, North Carolina, United States of America
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Qing Kenneth Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail: ;
| |
Collapse
|
38
|
Linask KK, Han M. Acute alcohol exposure during mouse gastrulation alters lipid metabolism in placental and heart development: Folate prevention. ACTA ACUST UNITED AC 2016; 106:749-60. [PMID: 27296863 PMCID: PMC5094567 DOI: 10.1002/bdra.23526] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/29/2016] [Accepted: 04/28/2016] [Indexed: 12/20/2022]
Abstract
Background Embryonic acute exposure to ethanol (EtOH), lithium, and homocysteine (HCy) induces cardiac defects at the time of exposure; folic acid (FA) supplementation protects normal cardiogenesis (Han et al., 2009, 2012; Serrano et al., 2010). Our hypothesis is that EtOH exposure and FA protection relate to lipid and FA metabolism during mouse cardiogenesis and placentation. Methods On the morning of conception, pregnant C57BL/6J mice were placed on either of two FA‐containing diets: a 3.3 mg health maintenance diet or a high FA diet of 10.5 mg/kg. Mice were injected a binge level of EtOH, HCy, or saline on embryonic day (E) 6.75, targeting gastrulation. On E15.5, cardiac and umbilical blood flow were examined by ultrasound. Embryonic cardiac tissues were processed for gene expression of lipid and FA metabolism; the placenta and heart tissues for neutral lipid droplets, or for medium chain acyl‐dehydrogenase (MCAD) protein. Results EtOH exposure altered lipid‐related gene expression on E7.5 in comparison to control or FA‐supplemented groups and remained altered on E15.5 similarly to changes with HCy, signifying FA deficiency. In comparison to control tissues, the lipid‐related acyl CoA dehydrogenase medium length chain gene and its protein MCAD were altered with EtOH exposure, as were neutral lipid droplet localization in the heart and placenta. Conclusion EtOH altered gene expression associated with lipid and folate metabolism, as well as neutral lipids, in the E15.5 abnormally functioning heart and placenta. In comparison to controls, the high FA diet protected the embryo and placenta from these effects allowing normal development. Birth Defects Research (Part A) 106:749–760, 2016. © 2016 The Authors Birth Defects Research Part A: Clinical and Molecular Teratology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kersti K Linask
- Department of Pediatrics, USF Morsani College of Medicine, Tampa and St. Petersburg, Florida.
| | - Mingda Han
- Department of Pediatrics, USF Morsani College of Medicine, Tampa and St. Petersburg, Florida
| |
Collapse
|
39
|
Blech-Hermoni Y, Sullivan CB, Jenkins MW, Wessely O, Ladd AN. CUG-BP, Elav-like family member 1 (CELF1) is required for normal myofibrillogenesis, morphogenesis, and contractile function in the embryonic heart. Dev Dyn 2016; 245:854-73. [PMID: 27144987 DOI: 10.1002/dvdy.24413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/27/2016] [Accepted: 04/27/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND CUG-BP, Elav-like family member 1 (CELF1) is a multifunctional RNA binding protein found in a variety of adult and embryonic tissues. In the heart, CELF1 is found exclusively in the myocardium. However, the roles of CELF1 during cardiac development have not been completely elucidated. RESULTS Myofibrillar organization is disrupted and proliferation is reduced following knockdown of CELF1 in cultured chicken primary embryonic cardiomyocytes. In vivo knockdown of Celf1 in developing Xenopus laevis embryos resulted in myofibrillar disorganization and a trend toward reduced proliferation in heart muscle, indicating conserved roles for CELF1 orthologs in embryonic cardiomyocytes. Loss of Celf1 also resulted in morphogenetic abnormalities in the developing heart and gut. Using optical coherence tomography, we showed that cardiac contraction was impaired following depletion of Celf1, while heart rhythm remained unperturbed. In contrast to cardiac muscle, loss of Celf1 did not disrupt myofibril organization in skeletal muscle cells, although it did lead to fragmentation of skeletal muscle bundles. CONCLUSIONS CELF1 is required for normal myofibril organization, proliferation, morphogenesis, and contractile performance in the developing myocardium. Developmental Dynamics 245:854-873, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yotam Blech-Hermoni
- Program in Cell Biology, Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Connor B Sullivan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Michael W Jenkins
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Oliver Wessely
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Andrea N Ladd
- Program in Cell Biology, Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
40
|
Raghunathan R, Singh M, Dickinson ME, Larin KV. Optical coherence tomography for embryonic imaging: a review. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:50902. [PMID: 27228503 PMCID: PMC4881290 DOI: 10.1117/1.jbo.21.5.050902] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/25/2016] [Indexed: 05/18/2023]
Abstract
Embryogenesis is a highly complex and dynamic process, and its visualization is crucial for understanding basic physiological processes during development and for identifying and assessing possible defects, malformations, and diseases. While traditional imaging modalities, such as ultrasound biomicroscopy, micro-magnetic resonance imaging, and micro-computed tomography, have long been adapted for embryonic imaging, these techniques generally have limitations in their speed, spatial resolution, and contrast to capture processes such as cardiodynamics during embryogenesis. Optical coherence tomography (OCT) is a noninvasive imaging modality with micrometer-scale spatial resolution and imaging depth up to a few millimeters in tissue. OCT has bridged the gap between ultrahigh resolution imaging techniques with limited imaging depth like confocal microscopy and modalities, such as ultrasound sonography, which have deeper penetration but poorer spatial resolution. Moreover, the noninvasive nature of OCT has enabled live imaging of embryos without any external contrast agents. We review how OCT has been utilized to study developing embryos and also discuss advances in techniques used in conjunction with OCT to understand embryonic development.
Collapse
Affiliation(s)
- Raksha Raghunathan
- University of Houston, Department of Biomedical Engineering, 3517 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
| | - Manmohan Singh
- University of Houston, Department of Biomedical Engineering, 3517 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
| | - Mary E. Dickinson
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, One Baylor Plaza- BCM335, Houston, Texas 77030, United States
| | - Kirill V. Larin
- University of Houston, Department of Biomedical Engineering, 3517 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, One Baylor Plaza- BCM335, Houston, Texas 77030, United States
| |
Collapse
|
41
|
Wen Z, Yu D, Zhang W, Fan C, Hu L, Feng Y, Yang L, Wu Z, Chen R, Yin KJ, Mo X. Association between alcohol consumption during pregnancy and risks of congenital heart defects in offspring: meta-analysis of epidemiological observational studies. Ital J Pediatr 2016; 42:12. [PMID: 26843087 PMCID: PMC4739085 DOI: 10.1186/s13052-016-0222-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/27/2016] [Indexed: 01/19/2023] Open
Abstract
Background To explore the association between maternal alcohol consumption and/or binge drinking and congenital heart defects (CHDs), we conducted a meta-analysis for more sufficient evidence on this issue. Methods We searched Medline, EMBASE, and the Cochrane Library from their inceptions to December 2014 for case-control and cohort studies that assessed the association between maternal alcohol consumption and CHD risk. Study-specific relative risk estimates were calculated using random-effect or fixed-effect models. Results A total of 19 case-control studies and 4 cohort studies were included in the meta-analysis. We observed a null association between maternal alcohol consumption during pregnancy and the risk of CHDs. Even in the analysis of different trimesters of pregnancy, we found little association between the two. Conclusions This meta-analysis suggests that maternal alcohol consumption is modestly not associated with the risk of CHDs. However, further investigation is needed to confirm this conclusion.
Collapse
Affiliation(s)
- Zhongyuan Wen
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Di Yu
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Weiyan Zhang
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Changfeng Fan
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Liang Hu
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Yu Feng
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Lei Yang
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Zeyu Wu
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Runsen Chen
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| | - Ke-Jie Yin
- Department of Neurology, Center for the Study of Nervous System Injury, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing, 210008, China.
| |
Collapse
|
42
|
Coram RJ, Stillwagon SJ, Guggilam A, Jenkins MW, Swanson MS, Ladd AN. Muscleblind-like 1 is required for normal heart valve development in vivo. BMC DEVELOPMENTAL BIOLOGY 2015; 15:36. [PMID: 26472242 PMCID: PMC4608261 DOI: 10.1186/s12861-015-0087-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/09/2015] [Indexed: 12/26/2022]
Abstract
Background Development of the valves and septa of the heart depends on the formation and remodeling of the endocardial cushions in the atrioventricular canal and outflow tract. These cushions are populated by mesenchyme produced from the endocardium by epithelial-mesenchymal transition (EMT). The endocardial cushions are remodeled into the valves at post-EMT stages via differentiation of the mesenchyme and changes in the extracellular matrix (ECM). Transforming growth factor β (TGFβ) signaling has been implicated in both the induction of EMT in the endocardial cushions and the remodeling of the valves at post-EMT stages. We previously identified the RNA binding protein muscleblind-like 1 (MBNL1) as a negative regulator of TGFβ signaling and EMT in chicken endocardial cushions ex vivo. Here, we investigate the role of MBNL1 in endocardial cushion development and valvulogenesis in Mbnl1∆E3/∆E3 mice, which are null for MBNL1 protein. Methods Collagen gel invasion assays, histology, immunohistochemistry, real-time RT-PCR, optical coherence tomography, and echocardiography were used to evaluate EMT and TGFβ signaling in the endocardial cushions, and morphogenesis, ECM composition, and function of the heart valves. Results As in chicken, the loss of MBNL1 promotes precocious TGFβ signaling and EMT in the endocardial cushions. Surprisingly, this does not lead to the production of excess mesenchyme, but later valve morphogenesis is aberrant. Adult Mbnl1∆E3/∆E3 mice exhibit valve dysmorphia with elevated TGFβ signaling, changes in ECM composition, and increased pigmentation. This is accompanied by a high incidence of regurgitation across both inflow and outflow valves. Mbnl1∆E3/∆E3 mice also have a high incidence of ostium secundum septal defects accompanied by atrial communication, but do not develop overt cardiomyopathy. Conclusions Together, these data indicate that MBNL1 plays a conserved role in negatively regulating TGFβ signaling, and is required for normal valve morphogenesis and homeostasis in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0087-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ryan J Coram
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Present Address: Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA.
| | - Samantha J Stillwagon
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Present Address: Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Anuradha Guggilam
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Michael W Jenkins
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Maurice S Swanson
- Department of Molecular Genetics & Microbiology, College of Medicine, Center for NeuroGenetics and the Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Andrea N Ladd
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
43
|
Alcohol-induced histone H3K9 hyperacetylation and cardiac hypertrophy are reversed by a histone acetylases inhibitor anacardic acid in developing murine hearts. Biochimie 2015; 113:1-9. [DOI: 10.1016/j.biochi.2015.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/11/2015] [Indexed: 01/04/2023]
|
44
|
Sun J, Chen X, Chen H, Ma Z, Zhou J. Maternal Alcohol Consumption before and during Pregnancy and the Risks of Congenital Heart Defects in Offspring: A Systematic Review and Meta-analysis. CONGENIT HEART DIS 2015; 10:E216-24. [PMID: 26032942 DOI: 10.1111/chd.12271] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Epidemiologic studies have reported conflicting results regarding maternal alcohol consumption before and during pregnancy, and the risk of congenital heart defects (CHDs). However, a systematic review and meta-analysis of the association between maternal alcohol consumption and CHDs in offspring has not been conducted. DESIGN We searched MEDLINE and EMBASE for articles catalogued between their inception and February 16, 2015; we identified relevant published studies that assessed the association between maternal alcohol consumption and CHD risk. Two authors independently assessed the eligibility of the retrieved articles and extracted data from them. Study-specific relative risk estimates were pooled by random-effects or fixed-effects models. RESULTS From the 1527 references, a total of 19 case-control studies and four cohort studies were enrolled in this meta-analysis. The summary of 23 studies related to CHDs indicated an overall pooled relative risk of 1.13 (95% confidence interval: 0.96, 1.29) among mothers drinking before or during pregnancy. Statistically significant heterogeneity was detected (Q = 196.61, P < .001, I(2) = 88.8%) with no publication bias (Egger's test: P = .157). We conducted stratified and meta-regression analyses to identify the origin of the heterogeneity among studies. CONCLUSION In summary, this meta-analysis provided no positive association between maternal alcohol consumption and risk of CHDs.
Collapse
Affiliation(s)
- Jian Sun
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Anesthesiology, Huai'an Maternal and Child Health Hospital, Huai'an, Jiangsu, China
| | - Xiaoling Chen
- Department of Nephrology, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Huajun Chen
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengliang Ma
- Department of Anesthesiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jianwei Zhou
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
45
|
Gu S, Wang YT, Ma P, Werdich AA, Rollins AM, Jenkins MW. Mapping conduction velocity of early embryonic hearts with a robust fitting algorithm. BIOMEDICAL OPTICS EXPRESS 2015; 6:2138-57. [PMID: 26114034 PMCID: PMC4473749 DOI: 10.1364/boe.6.002138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/27/2015] [Accepted: 04/27/2015] [Indexed: 05/23/2023]
Abstract
Cardiac conduction maturation is an important and integral component of heart development. Optical mapping with voltage-sensitive dyes allows sensitive measurements of electrophysiological signals over the entire heart. However, accurate measurements of conduction velocity during early cardiac development is typically hindered by low signal-to-noise ratio (SNR) measurements of action potentials. Here, we present a novel image processing approach based on least squares optimizations, which enables high-resolution, low-noise conduction velocity mapping of smaller tubular hearts. First, the action potential trace measured at each pixel is fit to a curve consisting of two cumulative normal distribution functions. Then, the activation time at each pixel is determined based on the fit, and the spatial gradient of activation time is determined with a two-dimensional (2D) linear fit over a square-shaped window. The size of the window is adaptively enlarged until the gradients can be determined within a preset precision. Finally, the conduction velocity is calculated based on the activation time gradient, and further corrected for three-dimensional (3D) geometry that can be obtained by optical coherence tomography (OCT). We validated the approach using published activation potential traces based on computer simulations. We further validated the method by adding artificially generated noise to the signal to simulate various SNR conditions using a curved simulated image (digital phantom) that resembles a tubular heart. This method proved to be robust, even at very low SNR conditions (SNR = 2-5). We also established an empirical equation to estimate the maximum conduction velocity that can be accurately measured under different conditions (e.g. sampling rate, SNR, and pixel size). Finally, we demonstrated high-resolution conduction velocity maps of the quail embryonic heart at a looping stage of development.
Collapse
Affiliation(s)
- Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yves T Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, 44016, USA
| | - Pei Ma
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Andreas A Werdich
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Michael W Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, 44016, USA
| |
Collapse
|
46
|
Karunamuni G, Gu S, Doughman YQ, Noonan AI, Rollins AM, Jenkins MW, Watanabe M. Using optical coherence tomography to rapidly phenotype and quantify congenital heart defects associated with prenatal alcohol exposure. Dev Dyn 2015; 244:607-18. [PMID: 25546089 DOI: 10.1002/dvdy.24246] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The most commonly used method to analyze congenital heart defects involves serial sectioning and histology. However, this is often a time-consuming process where the quantification of cardiac defects can be difficult due to problems with accurate section registration. Here we demonstrate the advantages of using optical coherence tomography, a comparatively new and rising technology, to phenotype avian embryo hearts in a model of fetal alcohol syndrome where a binge-like quantity of alcohol/ethanol was introduced at gastrulation. RESULTS The rapid, consistent imaging protocols allowed for the immediate identification of cardiac anomalies, including ventricular septal defects and misaligned/missing vessels. Interventricular septum thicknesses and vessel diameters for three of the five outflow arteries were also significantly reduced. Outflow and atrioventricular valves were segmented using image processing software and had significantly reduced volumes compared to controls. This is the first study to our knowledge that has 3D reconstructed the late-stage cardiac valves in precise detail to examine their morphology and dimensions. CONCLUSIONS We believe, therefore, that optical coherence tomography, with its ability to rapidly image and quantify tiny embryonic structures in high resolution, will serve as an excellent and cost-effective preliminary screening tool for developmental biologists working with a variety of experimental/disease models.
Collapse
Affiliation(s)
- Ganga Karunamuni
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | | | | | | | | | | | | |
Collapse
|
47
|
Linask KK, Han M, Bravo-Valenzuela NJM. Changes in vitelline and utero-placental hemodynamics: implications for cardiovascular development. Front Physiol 2014; 5:390. [PMID: 25426076 PMCID: PMC4227466 DOI: 10.3389/fphys.2014.00390] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/21/2014] [Indexed: 12/31/2022] Open
Abstract
Analyses of cardiovascular development have shown an important interplay between heart function, blood flow, and morphogenesis of heart structure during the formation of a four-chambered heart. It is known that changes in vitelline and placental blood flow seemingly contribute substantially to early cardiac hemodynamics. This suggests that in order to understand mammalian cardiac structure-hemodynamic functional relationships, blood flow from the extra-embryonic circulation needs to be taken into account and its possible impact on cardiogenesis defined. Previously published Doppler ultrasound analyses and data of utero-placental blood flow from human studies and those using the mouse model are compared to changes observed with environmental exposures that lead to cardiovascular anomalies. Use of current concepts and models related to mechanotransduction of blood flow and fluid forces may help in the future to better define the characteristics of normal and abnormal utero-placental blood flow and the changes in the biophysical parameters that may contribute to congenital heart defects. Evidence from multiple studies is discussed to provide a framework for future modeling of the impact of experimental changes in blood flow on the mouse heart during normal and abnormal cardiogenesis.
Collapse
Affiliation(s)
- Kersti K Linask
- Department of Pediatrics, Morsani College of Medicine, Children's Research Institute, University of South Florida Health St. Petersburg, FL, USA
| | - Mingda Han
- Department of Pediatrics, Morsani College of Medicine, Children's Research Institute, University of South Florida Health St. Petersburg, FL, USA
| | | |
Collapse
|
48
|
Kowalski WJ, Pekkan K, Tinney JP, Keller BB. Investigating developmental cardiovascular biomechanics and the origins of congenital heart defects. Front Physiol 2014; 5:408. [PMID: 25374544 PMCID: PMC4204442 DOI: 10.3389/fphys.2014.00408] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 10/02/2014] [Indexed: 11/24/2022] Open
Abstract
Innovative research on the interactions between biomechanical load and cardiovascular (CV) morphogenesis by multiple investigators over the past 3 decades, including the application of bioengineering approaches, has shown that the embryonic heart adapts both structure and function in order to maintain cardiac output to the rapidly growing embryo. Acute adaptive hemodynamic mechanisms in the embryo include the redistribution of blood flow within the heart, dynamic adjustments in heart rate and developed pressure, and beat to beat variations in blood flow and vascular resistance. These biomechanically relevant events occur coincident with adaptive changes in gene expression and trigger adaptive mechanisms that include alterations in myocardial cell growth and death, regional and global changes in myocardial architecture, and alterations in central vascular morphogenesis and remodeling. These adaptive mechanisms allow the embryo to survive these biomechanical stresses (environmental, maternal) and to compensate for developmental errors (genetic). Recent work from numerous laboratories shows that a subset of these adaptive mechanisms is present in every developing multicellular organism with a “heart” equivalent structure. This chapter will provide the reader with an overview of some of the approaches used to quantify embryonic CV functional maturation and performance, provide several illustrations of experimental interventions that explore the role of biomechanics in the regulation of CV morphogenesis including the role of computational modeling, and identify several critical areas for future investigation as available experimental models and methods expand.
Collapse
Affiliation(s)
- William J Kowalski
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA
| | - Kerem Pekkan
- Department of Biomedical Engineering, Carnegie Mellon University Pittsburgh, PA, USA
| | - Joseph P Tinney
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA
| | - Bradley B Keller
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA ; Department of Biomedical Engineering, Carnegie Mellon University Pittsburgh, PA, USA
| |
Collapse
|
49
|
Karunamuni GH, Ma P, Gu S, Rollins AM, Jenkins MW, Watanabe M. Connecting teratogen-induced congenital heart defects to neural crest cells and their effect on cardiac function. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2014; 102:227-50. [PMID: 25220155 PMCID: PMC4238913 DOI: 10.1002/bdrc.21082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 08/26/2014] [Indexed: 12/26/2022]
Abstract
Neural crest cells play many key roles in embryonic development, as demonstrated by the abnormalities that result from their specific absence or dysfunction. Unfortunately, these key cells are particularly sensitive to abnormalities in various intrinsic and extrinsic factors, such as genetic deletions or ethanol-exposure that lead to morbidity and mortality for organisms. This review discusses the role identified for a segment of neural crest in regulating the morphogenesis of the heart and associated great vessels. The paradox is that their derivatives constitute a small proportion of cells to the cardiovascular system. Findings supporting that these cells impact early cardiac function raises the interesting possibility that they indirectly control cardiovascular development at least partially through regulating function. Making connections between insults to the neural crest, cardiac function, and morphogenesis is more approachable with technological advances. Expanding our understanding of early functional consequences could be useful in improving diagnosis and testing therapies.
Collapse
Affiliation(s)
- Ganga H. Karunamuni
- Department of Pediatrics, Case Western Reserve University School of Medicine, Case Medical Center Division of Pediatric Cardiology, Rainbow Babies and Children’s Hospital, Cleveland OH 44106
| | - Pei Ma
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Andrew M. Rollins
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Michael W. Jenkins
- Department of Pediatrics, Case Western Reserve University School of Medicine, Case Medical Center Division of Pediatric Cardiology, Rainbow Babies and Children’s Hospital, Cleveland OH 44106
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University School of Medicine, Case Medical Center Division of Pediatric Cardiology, Rainbow Babies and Children’s Hospital, Cleveland OH 44106
| |
Collapse
|
50
|
Joya X, Garcia-Algar O, Salat-Batlle J, Pujades C, Vall O. Advances in the development of novel antioxidant therapies as an approach for fetal alcohol syndrome prevention. ACTA ACUST UNITED AC 2014; 103:163-77. [PMID: 25131946 DOI: 10.1002/bdra.23290] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/08/2014] [Accepted: 07/08/2014] [Indexed: 01/14/2023]
Abstract
Ethanol is the most common human teratogen, and its consumption during pregnancy can produce a wide range of abnormalities in infants known as fetal alcohol spectrum disorder (FASD). The major characteristics of FASD can be divided into: (i) growth retardation, (ii) craniofacial abnormalities, and (iii) central nervous system (CNS) dysfunction. FASD is the most common cause of nongenetic mental retardation in Western countries. Although the underlying molecular mechanisms of ethanol neurotoxicity are not completely determined, the induction of oxidative stress is believed to be one central process linked to the development of the disease. Currently, there is no known effective strategy for prevention (other than alcohol avoidance) or treatment. In the present review we will provide the state of art in the evidence for the use of antioxidants as a potential therapeutic strategy for the treatment using whole-embryo and culture cells models of FASD. We conclude that the imbalance of the intracellular redox state contributes to the pathogenesis observed in FASD models, and we suggest that antioxidant therapy can be considered a new efficient strategy to mitigate the effects of prenatal ethanol exposure.
Collapse
Affiliation(s)
- Xavier Joya
- Unitat de Recerca Infància i Entorn (URIE), Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain; Red de Salud Materno-Infantil y del Desarrollo (SAMID), Programa RETICS, Instituto Carlos III, Madrid, Spain
| | | | | | | | | |
Collapse
|