51
|
Zeng X, Shi S, Sun Y, Feng Y, Tan L, Lin R, Li J, Duan H, Shu Q, Li H. A time-aware attention model for prediction of acute kidney injury after pediatric cardiac surgery. J Am Med Inform Assoc 2022; 30:94-102. [PMID: 36287639 PMCID: PMC9748588 DOI: 10.1093/jamia/ocac202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Acute kidney injury (AKI) is a common complication after pediatric cardiac surgery, and the early detection of AKI may allow for timely preventive or therapeutic measures. However, current AKI prediction researches pay less attention to time information among time-series clinical data and model building strategies that meet complex clinical application scenario. This study aims to develop and validate a model for predicting postoperative AKI that operates sequentially over individual time-series clinical data. MATERIALS AND METHODS A retrospective cohort of 3386 pediatric patients extracted from PIC database was used for training, calibrating, and testing purposes. A time-aware deep learning model was developed and evaluated from 3 clinical perspectives that use different data collection windows and prediction windows to answer different AKI prediction questions encountered in clinical practice. We compared our model with existing state-of-the-art models from 3 clinical perspectives using the area under the receiver operating characteristic curve (ROC AUC) and the area under the precision-recall curve (PR AUC). RESULTS Our proposed model significantly outperformed the existing state-of-the-art models with an improved average performance for any AKI prediction from the 3 evaluation perspectives. This model predicted 91% of all AKI episodes using data collected at 24 h after surgery, resulting in a ROC AUC of 0.908 and a PR AUC of 0.898. On average, our model predicted 83% of all AKI episodes that occurred within the different time windows in the 3 evaluation perspectives. The calibration performance of the proposed model was substantially higher than the existing state-of-the-art models. CONCLUSIONS This study showed that a deep learning model can accurately predict postoperative AKI using perioperative time-series data. It has the potential to be integrated into real-time clinical decision support systems to support postoperative care planning.
Collapse
Affiliation(s)
- Xian Zeng
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Shanshan Shi
- CICU, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuhan Sun
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yuqing Feng
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Linhua Tan
- CICU, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ru Lin
- CICU, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Li
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Huilong Duan
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Qiang Shu
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Haomin Li
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
52
|
Genome Editing and Myocardial Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1396:53-73. [PMID: 36454459 DOI: 10.1007/978-981-19-5642-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Congenital heart disease (CHD) has a strong genetic etiology, making it a likely candidate for therapeutic intervention using genetic editing. Complex genetics involving an orchestrated series of genetic events and over 400 genes are responsible for myocardial development. Cooperation is required from a vast series of genetic networks, and mutations in such can lead to CHD and cardiovascular abnormalities, affecting up to 1% of all live births. Genome editing technologies are becoming better studied and with time and improved logistics, CHD could be a prime therapeutic target. Syndromic, nonsyndromic, and cases of familial inheritance all involve identifiable causative mutations and thus have the potential for genome editing therapy. Mouse models are well-suited to study and predict clinical outcome. This review summarizes the anatomical and genetic timeline of myocardial development in both mice and humans, the potential of gene editing in typical CHD categories, as well as the use of mice thus far in reproducing models of human CHD and correcting the mutations that create them.
Collapse
|
53
|
Aryafar M, Mahdavi M, Shahzadi H, Nasrollahzadeh J. Effect of feeding with standard or higher-density formulas on anthropometric measures in children with congenital heart defects after corrective surgery: a randomized clinical trial. Eur J Clin Nutr 2022; 76:1713-1718. [PMID: 35906331 DOI: 10.1038/s41430-022-01186-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVES The aim of the present study was to assess the efficacy of feeding a higher-density formula (HDF) in infant with congenital heart disease (CHD). METHODS In a parallel randomized trial, infants (6 to 12 months) who underwent CHD corrective surgery received either a standard-density formula (SDF, 67 kcal /100 ml) or an HDF (90 kcal/100 ml) after discharge from the intensive care unit for 8 weeks. In addition to the formula, infants could receive breast milk or complementary food. Anthropometry, biochemistry, and formula intake were collected. RESULT Sixty-four infants completed the study (n = 32 in each group). All infants gained weight. The mean ± standard deviation (SD) of weight z score at baseline and week-8 were -2.38 ± 10.04 to -1.38 + 0.97 in the SDF group and -2.69 ± 1.19 to -0.89 ± 0.90 in the HDF group (between-group p = 0.0001). Both groups gained length, but showed a decline in length z-score which was significant in the SDF group but not significant in the HDF group. Mid-upper arm circumference and its z score improved in both groups, with more improvement in the HDF group. Serum albumin level was higher in the HDF than the SDF group at week-8, but no significant between-group differences were observed in hemoglobin, serum ferritin, or iron. Symptoms of gastrointestinal intolerance were not reported, but parents of 4 infants in the HDF group complained of their infants' constipation. CONCLUSION Feeding infants using a concentrated formula could increase infants' weight gain and growth, and improve the nutritional status after CHD surgery.
Collapse
Affiliation(s)
- Maryam Aryafar
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Shahzadi
- Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Nasrollahzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
54
|
Isaak A, Mesropyan N, Hart C, Zhang S, Kravchenko D, Endler C, Katemann C, Weber O, Pieper CC, Kuetting D, Attenberger U, Dabir D, Luetkens JA. Non-contrast free-breathing 3D cardiovascular magnetic resonance angiography using REACT (relaxation-enhanced angiography without contrast) compared to contrast-enhanced steady-state magnetic resonance angiography in complex pediatric congenital heart disease at 3T. J Cardiovasc Magn Reson 2022; 24:55. [PMID: 36384752 PMCID: PMC9670549 DOI: 10.1186/s12968-022-00895-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND To evaluate the great vessels in young children with complex congenital heart disease (CHD) using non-contrast cardiovascular magnetic resonance angiography (CMRA) based on three-dimensional relaxation-enhanced angiography without contrast (REACT) in comparison to contrast-enhanced steady-state CMRA. METHODS In this retrospective study from April to July 2021, respiratory- and electrocardiogram-gated native REACT CMRA was compared to contrast-enhanced single-phase steady-state CMRA in children with CHD who underwent CMRA at 3T under deep sedation. Vascular assessment included image quality (1 = non-diagnostic, 5 = excellent), vessel diameter, and diagnostic findings. For statistical analysis, paired t-test, Pearson correlation, Bland-Altman analysis, Wilcoxon test, and intraclass correlation coefficients (ICC) were applied. RESULTS Thirty-six young children with complex CHD (median 4 years, interquartile range, 2-5; 20 males) were included. Native REACT CMRA was obtained successfully in all patients (mean scan time: 4:22 ± 1:44 min). For all vessels assessed, diameters correlated strongly between both methods (Pearson r = 0.99; bias = 0.04 ± 0.61 mm) with high interobserver reproducibility (ICC: 0.99 for both CMRAs). Native REACT CMRA demonstrated comparable overall image quality to contrast-enhanced CMRA (3.9 ± 1.0 vs. 3.8 ± 0.9, P = 0.018). With REACT CMRA, better image quality was obtained at the ascending aorta (4.8 ± 0.5 vs. 4.3 ± 0.8, P < 0.001), coronary roots (e.g., left: 4.1 ± 1.0 vs. 3.3 ± 1.1, P = 0.001), and inferior vena cava (4.6 ± 0.5 vs. 3.2 ± 0.8, P < 0.001). In all patients, additional vascular findings were assessed equally with native REACT CMRA and the contrast-enhanced reference standard (n = 6). CONCLUSION In young children with complex CHD, REACT CMRA can provide gadolinium-free high image quality, accurate vascular measurements, and equivalent diagnostic quality compared to standard contrast-enhanced CMRA.
Collapse
Affiliation(s)
- Alexander Isaak
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany.
| | - Narine Mesropyan
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Christopher Hart
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Pediatric Cardiology, University Hospital Bonn, Bonn, Germany
| | - Shuo Zhang
- Philips GmbH Market DACH, Hamburg, Germany
| | - Dmitrij Kravchenko
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Christoph Endler
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | | | | | - Claus C Pieper
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Daniel Kuetting
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Darius Dabir
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Julian A Luetkens
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| |
Collapse
|
55
|
Wehrle FM, Bartal T, Adams M, Bassler D, Hagmann CF, Kretschmar O, Natalucci G, Latal B. Similarities and Differences in the Neurodevelopmental Outcome of Children with Congenital Heart Disease and Children Born Very Preterm at School Entry. J Pediatr 2022; 250:29-37.e1. [PMID: 35660491 DOI: 10.1016/j.jpeds.2022.05.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To describe the similarities and differences in the neurodevelopmental outcome of children with congenital heart disease (CHD) undergoing cardiopulmonary bypass surgery compared with children born very preterm (VPT) at school entry. STUDY DESIGN IQ, motor abilities, behavior, and therapy use were assessed in 155 children with CHD as part of a prospective, single-center, longitudinal study, and in 251 children born VPT as part of a national follow-up register at the same center. Group differences were tested using independent t-tests and χ2-tests. Equivalence testing was used to investigate similarities between the groups. RESULTS Mild (ie, 70 ≤ IQ < 85) and severe intellectual impairments (ie, IQ < 70) occurred in 17.4% and 4.5% of children with CHD compared with 22.1% and 5.5% in children VPT, respectively. Motor and behavioral functions were impaired in 57.0% and 15.3% of children with CHD compared with 37.8% and 11.5% of children born VPT, respectively. Children with CHD had poorer global motor abilities (d = -0.26) and poorer dynamic balance (d = -0.62) than children born VPT, and children born VPT had poorer fine motor abilities than children with CHD (d = 0.34; all P < .023). Peer problems were statistically similar between the groups (P = .020). Therapies were less frequent in children with CHD compared with children born VPT (23.4% vs 40.3%; P < .001). CONCLUSIONS Children with CHD undergoing cardiopulmonary bypass surgery and children born VPT share an overall risk for neurodevelopmental impairments that manifest in different domains. Despite this, children with CHD receive fewer therapies, indicating a lack of awareness of the neurodevelopmental burden these children face.
Collapse
Affiliation(s)
- Flavia M Wehrle
- Child Development Center, University Children's Hospital Zurich, Zurich, Switzerland; Department of Neonatology and Intensive Care, University Children's Hospital Zurich, Zurich, Switzerland; Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Timm Bartal
- Child Development Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Mark Adams
- Newborn Research, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland
| | - Dirk Bassler
- Newborn Research, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland; University of Zurich, Zurich, Switzerland
| | - Cornelia F Hagmann
- Department of Neonatology and Intensive Care, University Children's Hospital Zurich, Zurich, Switzerland; Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland; University of Zurich, Zurich, Switzerland
| | - Oliver Kretschmar
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland; University of Zurich, Zurich, Switzerland; Department of Cardiology, University Children's Hospital, Zurich, Switzerland
| | - Giancarlo Natalucci
- Newborn Research, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland; University of Zurich, Zurich, Switzerland; Family Larsson-Rosenquist Center for Neurodevelopment, Growth and Nutrition of the Newborn, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland
| | - Beatrice Latal
- Child Development Center, University Children's Hospital Zurich, Zurich, Switzerland; Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland; University of Zurich, Zurich, Switzerland
| |
Collapse
|
56
|
Konkel T, Kroll KH, Goertz MT, Lavoie J, Bagli SP, Kogutkiewicz K, Kostroski R, Scott L, Stoll P, Andres J, Saudek D, Handler SS, Brosig CL. Screening for traumatic stress in children and adolescents with congenital heart disease. PROGRESS IN PEDIATRIC CARDIOLOGY 2022. [DOI: 10.1016/j.ppedcard.2022.101579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
57
|
Depletion of VGLL4 Causes Perinatal Lethality without Affecting Myocardial Development. Cells 2022; 11:cells11182832. [PMID: 36139407 PMCID: PMC9496954 DOI: 10.3390/cells11182832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital heart disease is one of the leading causes of pediatric morbidity and mortality, thus highlighting the importance of deciphering the molecular mechanisms that control heart development. As the terminal transcriptional effectors of the Hippo-YAP pathway, YAP and TEAD1 form a transcriptional complex that regulates the target gene expression and depletes either of these two genes in cardiomyocytes, thus resulting in cardiac hypoplasia. Vestigial-like 4 (VGLL4) is a transcriptional co-factor that interacts with TEAD and suppresses the YAP/TEAD complex by competing against YAP for TEAD binding. To understand the VGLL4 function in the heart, we generated two VGLL4 loss-of-function mouse lines: a germline Vgll4 depletion allele and a cardiomyocyte-specific Vgll4 depletion allele. The whole-body deletion of Vgll4 caused defective embryo development and perinatal lethality. The analysis of the embryos at day 16.5 revealed that Vgll4 knockout embryos had reduced body size, malformed tricuspid valves, and normal myocardium. Few whole-body Vgll4 knockout pups could survive up to 10 days, and none of them showed body weight gain. In contrast to the whole-body Vgll4 knockout mutants, cardiomyocyte-specific Vgll4 knockout mice had no noticeable heart growth defects and had normal heart function. In summary, our data suggest that VGLL4 is required for embryo development but dispensable for myocardial growth.
Collapse
|
58
|
Lasso A, Herz C, Nam H, Cianciulli A, Pieper S, Drouin S, Pinter C, St-Onge S, Vigil C, Ching S, Sunderland K, Fichtinger G, Kikinis R, Jolley MA. SlicerHeart: An open-source computing platform for cardiac image analysis and modeling. Front Cardiovasc Med 2022; 9:886549. [PMID: 36148054 PMCID: PMC9485637 DOI: 10.3389/fcvm.2022.886549] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
Cardiovascular disease is a significant cause of morbidity and mortality in the developed world. 3D imaging of the heart's structure is critical to the understanding and treatment of cardiovascular disease. However, open-source tools for image analysis of cardiac images, particularly 3D echocardiographic (3DE) data, are limited. We describe the rationale, development, implementation, and application of SlicerHeart, a cardiac-focused toolkit for image analysis built upon 3D Slicer, an open-source image computing platform. We designed and implemented multiple Python scripted modules within 3D Slicer to import, register, and view 3DE data, including new code to volume render and crop 3DE. In addition, we developed dedicated workflows for the modeling and quantitative analysis of multi-modality image-derived heart models, including heart valves. Finally, we created and integrated new functionality to facilitate the planning of cardiac interventions and surgery. We demonstrate application of SlicerHeart to a diverse range of cardiovascular modeling and simulation including volume rendering of 3DE images, mitral valve modeling, transcatheter device modeling, and planning of complex surgical intervention such as cardiac baffle creation. SlicerHeart is an evolving open-source image processing platform based on 3D Slicer initiated to support the investigation and treatment of congenital heart disease. The technology in SlicerHeart provides a robust foundation for 3D image-based investigation in cardiovascular medicine.
Collapse
Affiliation(s)
- Andras Lasso
- Laboratory for Percutaneous Surgery, School of Computing, Queen's University, Kingston, ON, Canada
| | - Christian Herz
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hannah Nam
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Alana Cianciulli
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | | | - Simon Drouin
- Software and Information Technology Engineering, École de Technologie Supérieure, Montreal, QC, Canada
| | | | - Samuelle St-Onge
- Software and Information Technology Engineering, École de Technologie Supérieure, Montreal, QC, Canada
| | - Chad Vigil
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Stephen Ching
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Kyle Sunderland
- Laboratory for Percutaneous Surgery, School of Computing, Queen's University, Kingston, ON, Canada
| | - Gabor Fichtinger
- Laboratory for Percutaneous Surgery, School of Computing, Queen's University, Kingston, ON, Canada
| | - Ron Kikinis
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Matthew A. Jolley
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States,Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, United States,*Correspondence: Matthew A. Jolley
| |
Collapse
|
59
|
Shi HY, Xie MS, Yang CX, Huang RT, Xue S, Liu XY, Xu YJ, Yang YQ. Identification of SOX18 as a New Gene Predisposing to Congenital Heart Disease. Diagnostics (Basel) 2022; 12:diagnostics12081917. [PMID: 36010266 PMCID: PMC9406965 DOI: 10.3390/diagnostics12081917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital heart disease (CHD) is the most frequent kind of birth deformity in human beings and the leading cause of neonatal mortality worldwide. Although genetic etiologies encompassing aneuploidy, copy number variations, and mutations in over 100 genes have been uncovered to be involved in the pathogenesis of CHD, the genetic components predisposing to CHD in most cases remain unclear. We recruited a family with CHD from the Chinese Han population in the present investigation. Through whole-exome sequencing analysis of selected family members, a new SOX18 variation, namely NM_018419.3:c.349A>T; p.(Lys117*), was identified and confirmed to co-segregate with the CHD phenotype in the entire family by Sanger sequencing analysis. The heterozygous variant was absent from the 384 healthy volunteers enlisted as control individuals. Functional exploration via luciferase reporter analysis in cultivated HeLa cells revealed that Lys117*-mutant SOX18 lost transactivation on its target genes NR2F2 and GATA4, two genes responsible for CHD. Moreover, the genetic variation terminated the synergistic activation between SOX18 and NKX2.5, another gene accountable for CHD. The findings strongly indicate SOX18 as a novel gene contributing to CHD, which helps address challenges in the clinical genetic diagnosis and prenatal prophylaxis of CHD.
Collapse
Affiliation(s)
- Hong-Yu Shi
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, China
| | - Meng-Shi Xie
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| |
Collapse
|
60
|
Sheng SP, Feinberg JL, Bostrom JA, Tang Y, Sweeney G, Pierre A, Katz ES, Whiteson JH, Haas F, Dodson JA, Halpern DG. Adherence and Exercise Capacity Improvements of Patients With Adult Congenital Heart Disease Participating in Cardiac Rehabilitation. J Am Heart Assoc 2022; 11:e023896. [PMID: 35929458 PMCID: PMC9496295 DOI: 10.1161/jaha.121.023896] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background As the number of adults with congenital heart disease increases because of therapeutic advances, cardiac rehabilitation (CR) is increasingly being used in this population after cardiac procedures or for reduced exercise tolerance. We aim to describe the adherence and exercise capacity improvements of patients with adult congenital heart disease (ACHD) in CR. Methods and Results This retrospective study included patients with ACHD in CR at New York University Langone Rusk Rehabilitation from 2013 to 2020. We collected data on patient characteristics, number of sessions attended, and functional testing results. Pre‐CR and post‐CR metabolic equivalent task, exercise time, and maximal oxygen uptake were assessed. In total, 89 patients with ACHD (mean age, 39.0 years; 54.0% women) participated in CR. Referral indications were reduced exercise tolerance for 42.7% and post–cardiac procedure (transcatheter or surgical) for the remainder. Mean number of sessions attended was 24.2, and 42 participants (47.2%) completed all 36 CR sessions. Among participants who completed the program as well as pre‐CR and post‐CR functional testing, metabolic equivalent task increased by 1.3 (95% CI, 0.7–1.9; baseline mean, 8.1), exercise time increased by 66.4 seconds (95% CI, 21.4–111.4 seconds; baseline mean, 536.1 seconds), and maximal oxygen uptake increased by 2.5 mL/kg per minute (95% CI, 0.7–4.2 mL/kg per minute; baseline mean, 20.2 mL/kg per minute). Conclusions On average, patients with ACHD who completed CR experienced improvements in exercise capacity. Efforts to increase adherence would allow more patients with ACHD to benefit.
Collapse
Affiliation(s)
- S Peter Sheng
- Department of Medicine New York University Grossman School of Medicine New York NY 10016
| | - Jodi L Feinberg
- Leon H. Charney Division of Cardiology New York University Grossman School of Medicine New York NY 10016
| | - John A Bostrom
- Leon H. Charney Division of Cardiology New York University Grossman School of Medicine New York NY 10016
| | - Ying Tang
- Department of Physical Medicine and Rehabilitation New York University Grossman School of Medicine New York NY 10016
| | - Greg Sweeney
- Department of Physical Medicine and Rehabilitation New York University Grossman School of Medicine New York NY 10016
| | - Alicia Pierre
- Department of Physical Medicine and Rehabilitation New York University Grossman School of Medicine New York NY 10016
| | - Edward S Katz
- Leon H. Charney Division of Cardiology New York University Grossman School of Medicine New York NY 10016
| | - Jonathan H Whiteson
- Department of Physical Medicine and Rehabilitation New York University Grossman School of Medicine New York NY 10016
| | - François Haas
- Department of Physical Medicine and Rehabilitation New York University Grossman School of Medicine New York NY 10016
| | - John A Dodson
- Leon H. Charney Division of Cardiology New York University Grossman School of Medicine New York NY 10016
| | - Dan G Halpern
- Leon H. Charney Division of Cardiology New York University Grossman School of Medicine New York NY 10016
| |
Collapse
|
61
|
Rashkin SR, Cleves M, Shaw GM, Nembhard WN, Nestoridi E, Jenkins MM, Romitti PA, Lou XY, Browne ML, Mitchell LE, Olshan AF, Lomangino K, Bhattacharyya S, Witte JS, Hobbs CA. A genome-wide association study of obstructive heart defects among participants in the National Birth Defects Prevention Study. Am J Med Genet A 2022; 188:2303-2314. [PMID: 35451555 PMCID: PMC9283270 DOI: 10.1002/ajmg.a.62759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 01/19/2023]
Abstract
Obstructive heart defects (OHDs) share common structural lesions in arteries and cardiac valves, accounting for ~25% of all congenital heart defects. OHDs are highly heritable, resulting from interplay among maternal exposures, genetic susceptibilities, and epigenetic phenomena. A genome-wide association study was conducted in National Birth Defects Prevention Study participants (Ndiscovery = 3978; Nreplication = 2507), investigating the genetic architecture of OHDs using transmission/disequilibrium tests (TDT) in complete case-parental trios (Ndiscovery_TDT = 440; Nreplication_TDT = 275) and case-control analyses separately in infants (Ndiscovery_CCI = 1635; Nreplication_CCI = 990) and mothers (case status defined by infant; Ndiscovery_CCM = 1703; Nreplication_CCM = 1078). In the TDT analysis, the SLC44A2 single nucleotide polymorphism (SNP) rs2360743 was significantly associated with OHD (pdiscovery = 4.08 × 10-9 ; preplication = 2.44 × 10-4 ). A CAPN11 SNP (rs55877192) was suggestively associated with OHD (pdiscovery = 1.61 × 10-7 ; preplication = 0.0016). Two other SNPs were suggestively associated (p < 1 × 10-6 ) with OHD in only the discovery sample. In the case-control analyses, no SNPs were genome-wide significant, and, even with relaxed thresholds ( × discovery < 1 × 10-5 and preplication < 0.05), only one SNP (rs188255766) in the infant analysis was associated with OHDs (pdiscovery = 1.42 × 10-6 ; preplication = 0.04). Additional SNPs with pdiscovery < 1 × 10-5 were in loci supporting previous findings but did not replicate. Overall, there was modest evidence of an association between rs2360743 and rs55877192 and OHD and some evidence validating previously published findings.
Collapse
Affiliation(s)
- Sara R. Rashkin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, US
| | - Mario Cleves
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Health Informatics Institute, Tampa, FL 33612, US
| | - Gary M. Shaw
- Dept of Pediatrics, Stanford University, Stanford, CA 94305, US
| | - Wendy N. Nembhard
- University of Arkansas for Medical Sciences, Department of Epidemiology and Arkansas Center for Birth Defects and Prevention, University of Arkansas for Medical Sciences, Little Rock, AR 72205, US
| | - Eirini Nestoridi
- Massachusetts Center for Birth Defects Research and Prevention, Massachusetts Department of Public Health, Boston, MA 02108, US
| | - Mary M. Jenkins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30333, US
| | - Paul A. Romitti
- Department of Epidemiology, University of Iowa, Iowa City, IA 52242, US
| | - Xiang-Yang Lou
- Department of Biostatistics, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, FL 32603, US
| | - Marilyn L. Browne
- Birth Defects Research Section, New York State Department of Health, Albany, NY 12203, US; Department of Epidemiology and Biostatistics, School of Public Health, University at Albany, Rensselaer, NY 12114, US
| | - Laura E. Mitchell
- Department of Epidemiology, Human Genetics, and Environmental Sciences, UTHealth School of Public Health, Houston, TX 77030, US
| | - Andrew F. Olshan
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, US
| | | | - Sudeepa Bhattacharyya
- Bioinformatics and Data Science at University of Arkansas, Little Rock, AR 72204, US
| | - John S. Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, US
- These authors contributed equally to this work
| | - Charlotte A. Hobbs
- Rady Children’s Institute for Genomic Medicine, San Diego, CA 92123, US
- These authors contributed equally to this work
| | | |
Collapse
|
62
|
Manganese, Iron, Lead, and Zinc Levels and Haematological Profile among Welders in Bibiani Anhwiaso Bekwai District, Ghana. JOURNAL OF ENVIRONMENTAL AND PUBLIC HEALTH 2022; 2022:1508523. [PMID: 35859574 PMCID: PMC9293574 DOI: 10.1155/2022/1508523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 05/23/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022]
Abstract
Welders are exposed to metal ions or oxides through direct contact at occupational sites or indirectly through uptake from contaminated dust or air. This study was a case-control study designed to assess the levels of some heavy metals and the hematological profile of welders (cases) as compared to nonwelders (controls) from Bibiani Anhwiaso Bekwai District of Ghana, comparatively to determine whether their values are within acceptable international range. A quantitative-based survey using structured questionnaires was used to collect demographic data from purposively selected welders (n = 40) and nonwelders (n = 40) from the study area. Five (5 mL) blood samples were collected from the study participants and analyzed for blood cell count as well as levels of Mn, Fe, Pb, and Zn. There were no significant differences in the Mn, Zn, and Fe levels between the welders and nonwelders (
, 0.53 vs. 0.23 mg/L,
, 0.41 vs. 0.15,
, 1.82 vs. 1.11). The level of Pb was, however, significantly lower among welders compared to the nonwelders (
, 0.09 < 0.3 mg/L). The total white blood cell count did not differ significantly between welders and nonwelders (
, 5.16 vs. 4.85 × 109/L). However, the mixed cell fraction was significantly higher among welders compared to nonwelders (
, 0.34 × 109/L > 0.28 × 109/L). Red blood cell count and indices showed no significant differences between the welders and nonwelders. Hemoglobin levels in welders were, however, higher (14.47 g/dL) but this was not statistically significant compared to their nonwelder counterparts (13.85 g/dL). It was concluded from the study that welders in Bibiani Anhwiaso Bekwai District of the Western Region of Ghana had elevated levels of Pb in their bodies. This was associated with an increase in mixed white blood cell fraction platelets. However, the recorded levels were within the accepted physiological limits suggesting that the heavy metal exposure of welders had no clinically pathological significance.
Collapse
|
63
|
Schlapbach LJ, Gibbons KS, Horton SB, Johnson K, Long DA, Buckley DHF, Erickson S, Festa M, d’Udekem Y, Alphonso N, Winlaw DS, Delzoppo C, van Loon K, Jones M, Young PJ, Butt W, Schibler A. Effect of Nitric Oxide via Cardiopulmonary Bypass on Ventilator-Free Days in Young Children Undergoing Congenital Heart Disease Surgery: The NITRIC Randomized Clinical Trial. JAMA 2022; 328:38-47. [PMID: 35759691 PMCID: PMC9237803 DOI: 10.1001/jama.2022.9376] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/19/2022] [Indexed: 12/30/2022]
Abstract
Importance In children undergoing heart surgery, nitric oxide administered into the gas flow of the cardiopulmonary bypass oxygenator may reduce postoperative low cardiac output syndrome, leading to improved recovery and shorter duration of respiratory support. It remains uncertain whether nitric oxide administered into the cardiopulmonary bypass oxygenator improves ventilator-free days (days alive and free from mechanical ventilation). Objective To determine the effect of nitric oxide applied into the cardiopulmonary bypass oxygenator vs standard care on ventilator-free days in children undergoing surgery for congenital heart disease. Design, Setting, and Participants Double-blind, multicenter, randomized clinical trial in 6 pediatric cardiac surgical centers in Australia, New Zealand, and the Netherlands. A total of 1371 children younger than 2 years undergoing congenital heart surgery were randomized between July 2017 and April 2021, with 28-day follow-up of the last participant completed on May 24, 2021. Interventions Patients were assigned to receive nitric oxide at 20 ppm delivered into the cardiopulmonary bypass oxygenator (n = 679) or standard care cardiopulmonary bypass without nitric oxide (n = 685). Main Outcomes and Measures The primary end point was the number of ventilator-free days from commencement of bypass until day 28. There were 4 secondary end points including a composite of low cardiac output syndrome, extracorporeal life support, or death; length of stay in the intensive care unit; length of stay in the hospital; and postoperative troponin levels. Results Among 1371 patients who were randomized (mean [SD] age, 21.2 [23.5] weeks; 587 girls [42.8%]), 1364 (99.5%) completed the trial. The number of ventilator-free days did not differ significantly between the nitric oxide and standard care groups, with a median of 26.6 days (IQR, 24.4 to 27.4) vs 26.4 days (IQR, 24.0 to 27.2), respectively, for an absolute difference of -0.01 days (95% CI, -0.25 to 0.22; P = .92). A total of 22.5% of the nitric oxide group and 20.9% of the standard care group developed low cardiac output syndrome within 48 hours, needed extracorporeal support within 48 hours, or died by day 28, for an adjusted odds ratio of 1.12 (95% CI, 0.85 to 1.47). Other secondary outcomes were not significantly different between the groups. Conclusions and Relevance In children younger than 2 years undergoing cardiopulmonary bypass surgery for congenital heart disease, the use of nitric oxide via cardiopulmonary bypass did not significantly affect the number of ventilator-free days. These findings do not support the use of nitric oxide delivered into the cardiopulmonary bypass oxygenator during heart surgery. Trial Registration anzctr.org.au Identifier: ACTRN12617000821392.
Collapse
Affiliation(s)
- Luregn J. Schlapbach
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Paediatric Intensive Care Unit, Queensland Children’s Hospital, Children’s Health Queensland, Brisbane, Queensland, Australia
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kristen S. Gibbons
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Stephen B. Horton
- Cardiac Surgical Unit, Royal Children’s Hospital, Melbourne, Victoria, Australia
- Faculty of Medicine, Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Clinical Sciences Theme, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Kerry Johnson
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Paediatric Intensive Care Unit, Queensland Children’s Hospital, Children’s Health Queensland, Brisbane, Queensland, Australia
| | - Debbie A. Long
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- School of Nursing, Centre for Healthcare Transformation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - David H. F. Buckley
- Paediatric Intensive Care Unit, Starship Children’s Hospital, Auckland, New Zealand
| | - Simon Erickson
- Paediatric Critical Care, Perth Children’s Hospital, Western Australia and The University of Western Australia, Crawley, Western Australia, Australia
| | - Marino Festa
- Kids Critical Care Research, Paediatric Intensive Care Unit, Children’s Hospital at Westmead, Westmead, New South Wales, Australia
- Sydney Children’s Hospital Network, Sydney, New South Wales, Australia
| | - Yves d’Udekem
- Faculty of Medicine, Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Children’s National Hospital and The George Washington University School of Medicine and Health Sciences, Seattle, Washington
- Heart Research, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Nelson Alphonso
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Cardiac Surgery, Queensland Children's Hospital, Brisbane, Queensland, Australia
- School of Medicine, Children’s Health Clinical Unit, University of Queensland, Brisbane, Queensland, Australia
| | - David S. Winlaw
- Heart Centre for Children, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
- Sydney Children’s Hospital Network and Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Carmel Delzoppo
- Faculty of Medicine, Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Paediatric Intensive Care Unit, Royal Children’s Hospital Melbourne, Melbourne, Victoria, Australia
| | - Kim van Loon
- Department of Anaesthesiology, University Medical Center Utrecht, Wilhelmina Children’s Hospital, Utrecht, the Netherlands
| | - Mark Jones
- Institute of Evidence Based Healthcare, Bond University, Gold Coast, Australia
| | - Paul J. Young
- The Intensive Care Research Programme, Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Warwick Butt
- Faculty of Medicine, Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Clinical Sciences Theme, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Paediatric Intensive Care Unit, Royal Children’s Hospital Melbourne, Melbourne, Victoria, Australia
- Department of Critical Care, Melbourne Medical School University of Melbourne, Victoria, Australia
- Central Clinical School Faculty of Medicine Monash University, Melbourne, Victoria, Australia
| | - Andreas Schibler
- Critical Care Research Group, Wesley Medical Research, St Andrew’s War Memorial Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
64
|
Do children with congenital heart defects meet the vaccination recommendations? Immunisation in children with congenital heart defects. Cardiol Young 2022; 32:1143-1148. [PMID: 34569455 DOI: 10.1017/s1047951121003887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Congenital heart defects (CHDs) are the most common congenital malformations. Patients with CHD have a higher morbidity and mortality rate and are at greater risk for infectious diseases. The risk might even be higher if complex CHD occurs and if CHD is associated with additional co-morbidities. Therefore, immunisations in these children are essential. MATERIALS AND METHODS Individuals were recruited at the outpatient centre of the Department of Congenital Heart Defects and Pediatric Cardiology at the German Heart Center Munich in the time between February 2016 and February 2017. Included were children between 23 months and 17 years and a diagnosis of CHD. The vaccination certificate aimed to assess the immunization status. RESULTS In total, 657 children with CHD were included and analysed. Regarding primary immunisation, only 34 % (n = 221) of the children reached the complete vaccination status within the allowed catch-up time. Among these primary immunisation rates, vaccinations against Hepatitis B, Meningococci, Varicella and Pneumococci were found to have the lowest coverage with all being below 80%. The vaccination rate was partly influenced by the previously performed number of surgeries but not by the diagnosis of specific genetic diseases. At the age of school entry, the immunisation rate in children with CHD was also lower than in the comparable healthy population. CONCLUSION The vaccination coverage rate in children with CHD is lower than in comparable healthy children, although this is a vulnerable patient group. Further education of parents and treating physicians of children with CHD regarding vaccination is still needed.
Collapse
|
65
|
Meiman J, Zhang C, Holland B. Use of fetal tele-echo at small regional hospitals increases the rate of prenatal diagnosis of congenital heart disease. Prenat Diagn 2022; 42:1120-1132. [PMID: 35702985 DOI: 10.1002/pd.6196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/27/2022] [Accepted: 05/19/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND To improve the rate of prenatal diagnosis of Congenital heart disease (CHD) in Kentucky, four fetal tele-echocardiography sites were established at regional hospitals in Kentucky: Ashland in 2011, Paducah in 2014, Lexington in 2014, and Owensboro in 2016. METHODS A 13-year retrospective review of medical records at Norton Children's Hospital was performed to identify patients with CHD who had cardiac surgery or intervention prior to one year of age. The rate of prenatal diagnosis prior to establishing any fetal tele-echocardiography sites was compared to the rate of prenatal diagnosis after the sites were established. Independent t-tests were used to determine if there was a statistically significant increase in the rate of prenatal diagnosis. RESULTS 1287 patients had cardiac surgery or cath lab intervention prior to one year of age at our institution from June 2005 to December 2018 and were included in the analysis. Seventeen patients were excluded due to incomplete medical records. The rate of prenatal diagnosis prior to the implementation of the first fetal tele-echocardiography site was 13.8% and after the sites were established, the prenatal diagnosis rate was 39.7% (p < 0.01). CONCLUSION Increasing the number of fetal tele-echocardiography sites at small regional hospitals in Kentucky was associated with a statistically significant increase in the rate of prenatal diagnosis for patients less than 1 year of age who had cardiac surgery or cath lab intervention at Norton Children's Hospital in Louisville. Fetal tele-echocardiography is an effective method to improve the rate of prenatal diagnosis in regions served by small regional hospitals with limited access to fetal echocardiography.
Collapse
Affiliation(s)
- Jenna Meiman
- University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Charlie Zhang
- Department of Geography and Geosciences, University of Louisville, Louisville, Kentucky, USA
| | - Brian Holland
- Department of Pediatrics, Division of Pediatric Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
66
|
Pre-operative nutritional status and its association with short-term post-operative outcomes in Iranian children with CHD. Cardiol Young 2022; 33:579-589. [PMID: 35695155 DOI: 10.1017/s1047951122001305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Nutritional assessment appears to be an essential component of the evaluation of children with CHD undergoing surgery because nutritional status may impact corrective surgery-associated morbidity. METHODS A prospective single-centre cohort study with children between 6 and 24 months of age. Patients who had genetic syndromes or those who were premature or low birthweight at birth were excluded. Pre-operative nutritional parameters included anthropometric measurements and serum concentrations of total protein, vitamin D, iron, and ferritin. Outcome measures included ICU length of stay, mechanical ventilation, vasoactive-inotropic score, and duration of inotropes. Linear regression analysis was performed to determine whether pre-operative variables were associated with outcomes. RESULTS Analysis was performed on 120 patients (median age of 8 months), of whom 67 were male. Prior to surgery, 50.8% of patients had reduced (z ≤ -2.0) weight-for-age z score, 23.3% had reduced length-for-age z score, and 59.2% had reduced mid-upper arm circumference z score. Pre-operative serum total protein levels were 59.36 ± 9.16 g/L. Multiple regression analysis showed that low serum protein was associated with longer ICU length of stay and length of mechanical ventilation, while mid-upper arm circumference z score ≤ -2 was associated with longer ICU length of stay and mechanical ventilation and inotropes duration. CONCLUSIONS Pre-operative assessment of nutritional status by performing anthropometric and biochemical measurements including mid-upper arm circumference z score and serum protein concentrations in children undergoing CHD surgery appears to be predictors of some post-operative short-term outcomes and could be used as a guide to highlight patients needing appropriate perioperative nutritional interventions.
Collapse
|
67
|
Liu Y, Gao J, Xu M, Zhou Q, Zhang Z, Ye J, Li R. Circular RNA circ-RCCD promotes cardiomyocyte differentiation in mouse embryo development via recruiting YY1 to the promoter of MyD88. J Cell Mol Med 2022; 26:3616-3627. [PMID: 35692080 PMCID: PMC9258712 DOI: 10.1111/jcmm.17336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/01/2022] [Accepted: 04/06/2022] [Indexed: 11/27/2022] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect, affecting approximately 1% of live births. Genetic and environmental factors are leading factors to CHD, but the mechanism of CHD pathogenesis remains unclear. Circular RNAs (circRNAs) are kinds of endogenous non‐coding RNAs (ncRNAs) involved in a variety of physiological and pathological processes, especially in heart diseases. In this study, three significant differently expressed circRNA between maternal embryonic day (E) E13 and E17 was found by microarray assay. Among them, the content of circ‐RCCD increases with the development of heart and was enriched in primary cardiomyocytes of different species, which arouses our attention. Functional experiments revealed that inhibition of circ‐RCCD dramatically suppressed the formation of beating cell clusters, the fluorescence intensity of cardiac differentiation marker MF20, and the expression of the myocardial‐specific markers CTnT, Mef2c, and GATA4. Next, we found that circ‐RCCD was involved in cardiomyocyte differentiation through negative regulation of MyD88 expression. Further experiments proved that circ‐RCCD inhibited MyD88 levels by recruiting YY1 to the promoter of MyD88; circ‐RCCD inhibited nuclear translocation of YY1. These results reported that circ‐RCCD promoted cardiomyocyte differentiation by recruiting YY1 to the promoter of MyD88. And, this study provided a potential role and molecular mechanism of circ‐RCCD as a target for the treatment of CHD.
Collapse
Affiliation(s)
- Yiwen Liu
- Department of Pediatrics, Tongren Hospital, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfang Gao
- Department of Pediatrics, Tongren Hospital, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Nursing, Tongren Hospital, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianqian Zhou
- Department of Nursing, Tongren Hospital, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongxiao Zhang
- Department of Pediatrics, Tongren Hospital, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxin Ye
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Li
- Department of Nursing, Tongren Hospital, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
68
|
Cocomello L, Taylor K, Caputo M, Cornish RP, Lawlor DA. Health and Well-Being in Surviving Congenital Heart Disease Patients: An Umbrella Review With Synthesis of Best Evidence. Front Cardiovasc Med 2022; 9:870474. [PMID: 35757334 PMCID: PMC9226339 DOI: 10.3389/fcvm.2022.870474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background Advances in the management of congenital heart disease (CHD) patients have enabled improvement in long-term survival even for those with serious defects. Research priorities (for patients, families and clinicians) have shifted from a focus on how to improve survival to exploring long-term outcomes in patients with CHD. A comprehensive appraisal of available evidence could inform best practice to maximize health and well-being, and identify research gaps to direct further research toward patient and clinical need. We aimed to critically appraise all available published systematic reviews of health and well-being outcomes in adult patients with CHD. Methods We conducted an umbrella review, including any systematic reviews that assessed the association of having vs. not having CHD with any long-term health (physical or mental), social (e.g., education, occupation) or well-being [e.g., quality of life (QoL)] outcome in adulthood (≥18-years). Results Out of 1330 articles screened, we identified five systematic reviews of associations of CHD with adult outcomes. All but one (which studied QoL) explored health outcomes: one cardiovascular, two mental, and one mortality after transplant. CHD patients had a higher risk of stroke, coronary heart disease and heart failure, with the pooled relative risk (RR) for any outcome of 3.12 (95% CI: 3.01 to 3.24), with substantial heterogeneity (I2 = 99%) explained by the outcome being studied (stronger association for heart failure) and geography (stronger in Europe compared with other regions). CHD patients had a higher risk of anxiety (OR = 2.58 (1.45 to 4.59)], and higher mean scores for depression/anxiety symptoms (difference in means = -0.11 SD (-0.28 to 0.06), I2 = 94%)]. Compared with patients having a cardiac transplant for other (non-CHD) diseases, CHD patients had higher short-term mortality (RR at 30-days post-transplant = 2.18 [1.62 to 2.93)], with moderate heterogeneity (I2 = 41%) explained by previous surgery (higher mortality with prior Fontan/Glenn operation). All domains of QoL were lower in patients with Fontan's circulation than non-CHD adults. Conclusion Adults with CHD have poorer cardiovascular, mental health and QoL outcomes, and higher short-term mortality after transplant. The paucity of systematic reviews, in particular for outcomes such as education, occupation and lifestyles, highlights the need for this to be made a priority by funders and researchers. Systematic Review Registration [www.crd.york.ac.uk/prospero], identifier [CRD42020175034].
Collapse
Affiliation(s)
- Lucia Cocomello
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Bristol, United Kingdom
| | - Kurt Taylor
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Bristol, United Kingdom
| | | | - Rosie P. Cornish
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Bristol, United Kingdom
| | - Deborah A. Lawlor
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Bristol, United Kingdom
| |
Collapse
|
69
|
Nakae K, Ueno K, Shiokawa N, Takahashi Y, Kawamura J, Hazeki D, Imoto Y, Kawano Y. Pediatric Patients Undergoing a Fontan Operation or with a High RACHS-1 Score Require Monitoring for Chronic Kidney Disease in Early Childhood. Pediatr Cardiol 2022; 43:1020-1028. [PMID: 35028678 DOI: 10.1007/s00246-022-02817-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 01/06/2022] [Indexed: 11/25/2022]
Abstract
Although the number of pediatric patients with long-term survival following cardiac surgery is increasing, concerns regarding chronic kidney disease (CKD) after surgery are growing. We examined the frequency of and risk factors for pediatric CKD development in patients with congenital heart disease (CHD) at least 2 years after cardiac surgery. This was a cross-sectional study of 147 patients who underwent open-heart surgery for CHD at Kagoshima University Hospital from April 2010 to March 2017. Data on demographics, acute kidney injury after cardiac surgery, cyanotic heart disease, Fontan circulation, medications in the perioperative period, and Risk Adjustment for Congenital Heart Surgery-1 (RACHS-1) category were recorded. CKD was defined using the current classification system described in the National Kidney Foundation's Kidney Disease Outcomes Quality Initiative and assessed during early childhood within 2-3 years of cardiac surgery. Statistical analyses were performed using SPSS Statistics for Windows version 25.0. We consecutively enrolled 147 patients, of whom 22 (15.0%) had CKD, all with stage-2 severity. Among patients with CKD, a higher proportion underwent Fontan surgery (P < 0.001), a higher proportion had cyanotic heart disease (P = 0.009), and the RACHS-1 category was high (P = 0.003). Patients with CKD appeared more frequently than patients without CKD in RACHS-1 categories 3, 5, and 6. It is essential to evaluate renal function longitudinally and monitor for CKD, given that patients who underwent Fontan surgery or complicated surgery in infancy have a high rate of developing postoperative CKD in early childhood.
Collapse
Affiliation(s)
- Koji Nakae
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan.
| | - Kentaro Ueno
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Naohiro Shiokawa
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yoshihiro Takahashi
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Junpei Kawamura
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Daisuke Hazeki
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yutaka Imoto
- Department of Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoshifumi Kawano
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| |
Collapse
|
70
|
Azab B, Aburizeg D, Ji W, Jeffries L, Isbeih NJ, Al-Akily AS, Mohammad H, Osba YA, Shahin MA, Dardas Z, Hatmal MM, Al-Ammouri I, Lakhani S. TBX5 variant with the novel phenotype of mixed‑type total anomalous pulmonary venous return in Holt‑Oram Syndrome and variable intrafamilial heart defects. Mol Med Rep 2022; 25:210. [PMID: 35514310 PMCID: PMC9133962 DOI: 10.3892/mmr.2022.12726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/07/2022] [Indexed: 12/04/2022] Open
Abstract
Variants in T‑box transcription factor 5 (TBX5) can result in a wide phenotypic spectrum, specifically in the heart and the limbs. TBX5 has been implicated in causing non‑syndromic cardiac defects and Holt‑Oram syndrome (HOS). The present study investigated the underlying molecular etiology of a family with heterogeneous heart defects. The proband had mixed‑type total anomalous pulmonary venous return (mixed‑type TAPVR), whereas her mother had an atrial septal defect. Genetic testing through trio‑based whole‑exome sequencing was used to reveal the molecular etiology. A nonsense variant was identified in TBX5 (c.577G>T; p.Gly193*) initially showing co‑segregation with a presumably non‑syndromic presentation of congenital heart disease. Subsequent genetic investigations and more complete phenotyping led to the correct diagnosis of HOS, documenting the novel association of mixed‑type TAPVR with HOS. Finally, protein modeling of the mutant TBX5 protein that harbored this pathogenic nonsense variant (p.Gly193*) revealed a substantial drop in the quantity of non‑covalent bonds. The decrease in the number of non‑covalent bonds suggested that the resultant mutant dimer was less stable compared with the wild‑type protein, consequently affecting the protein's ability to bind DNA. The present findings extended the phenotypic cardiac defects associated with HOS; to the best of our knowledge, this is the first association of mixed‑type TAPVR with TBX5. Prior to the current analysis, the molecular association of TAPVR with HOS had never been documented; hence, this is the first genetic investigation to report the association between TAPVR and HOS. Furthermore, it was demonstrated that the null‑variants reported in the T‑box domain of TBX5 were associated with a wide range of cardiac and/or skeletal anomalies on both the inter‑and intrafamilial levels. In conclusion, genetic testing was highlighted as a potentially powerful approach in the prognostication of the proper diagnosis.
Collapse
Affiliation(s)
- Bilal Azab
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
- Correspondence to: Dr Bilal Azab, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 3959 Broadway, New York, NY 10032, USA, E-mail:
| | - Dunia Aburizeg
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Weizhen Ji
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06504, USA
| | - Lauren Jeffries
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06504, USA
| | - Nooredeen Jamal Isbeih
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Amal Saleh Al-Akily
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Hashim Mohammad
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Yousef Abu Osba
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Mohammad A. Shahin
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Zain Dardas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ma'mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Iyad Al-Ammouri
- Department of Pediatrics, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Saquib Lakhani
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06504, USA
- Dr Saquib Lakhani, Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, 1 Park Street, Wing West Pavilion, New Haven, CT 06504, USA, E-mail:
| |
Collapse
|
71
|
Goldshtrom N, Vasquez AM, Chaves DV, Bateman DA, Kalfa D, Levasseur S, Torres AJ, Bacha E, Krishnamurthy G. Outcomes after neonatal cardiac surgery: The impact of a dedicated neonatal cardiac program. J Thorac Cardiovasc Surg 2022; 165:2204-2211.e4. [PMID: 35927084 DOI: 10.1016/j.jtcvs.2022.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/26/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Prematurity is a risk factor for in-hospital mortality after cardiac surgery. The structure of intensive care unit models designed to deliver optimal care to neonates including those born preterm with critical congenital heart disease is unknown. The objective of this study was to evaluate in-hospital outcomes after cardiac surgery across gestational ages in an institution with a dedicated neonatal cardiac program. METHODS This study is a single-center, retrospective review of infants who underwent cardiac surgical interventions from our dedicated neonatal cardiac intensive care program between 2006 and 2017. We evaluated in-hospital mortality and morbidity rates across all gestational ages. RESULTS A total of 1238 subjects met inclusion criteria over a 11-year period. Overall in-hospital mortality after cardiac surgery was 6.1%. The mortality rate in very preterm infants (n = 68; <34 weeks' gestation at birth) was 17.6% (odds ratio, 3.52 [1.4-8.53]), versus 4.3% in full-term (n = 563; 39-40 weeks) referent/control infants. Very preterm infants with isolated congenital heart disease (without evidence of other affected organ systems) experienced a mortality rate of 10.5% after cardiac surgery. Neither the late preterm (34-36 6/7 weeks) nor the early term (37-38 6/7) groups had significantly increased odds of mortality compared with full-term infants. Seventy-eight percent of very preterm infants incurred a preoperative or postoperative complication (odds ratio, 4.78 [2.61-8.97]) compared with 35% of full-term infants. CONCLUSIONS In this study of a single center with a dedicated neonatal cardiac program, we report some of the lowest mortality and morbidity rates after cardiac surgery in preterm infants in the recent era. The potential survival advantage of this model is most striking for very preterm infants born with isolated congenital heart disease.
Collapse
|
72
|
Iacobazzi D, Alvino VV, Caputo M, Madeddu P. Accelerated Cardiac Aging in Patients With Congenital Heart Disease. Front Cardiovasc Med 2022; 9:892861. [PMID: 35694664 PMCID: PMC9177956 DOI: 10.3389/fcvm.2022.892861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/11/2022] [Indexed: 01/03/2023] Open
Abstract
An increasing number of patients with congenital heart disease (CHD) survive into adulthood but develop long-term complications including heart failure (HF). Cellular senescence, classically defined as stable cell cycle arrest, is implicated in biological processes such as embryogenesis, wound healing, and aging. Senescent cells have a complex senescence-associated secretory phenotype (SASP), involving a range of pro-inflammatory factors with important paracrine and autocrine effects on cell and tissue biology. While senescence has been mainly considered as a cause of diseases in the adulthood, it may be also implicated in some of the poor outcomes seen in patients with complex CHD. We propose that patients with CHD suffer from multiple repeated stress from an early stage of the life, which wear out homeostatic mechanisms and cause premature cardiac aging, with this term referring to the time-related irreversible deterioration of the organ physiological functions and integrity. In this review article, we gathered evidence from the literature indicating that growing up with CHD leads to abnormal inflammatory response, loss of proteostasis, and precocious age in cardiac cells. Novel research on this topic may inspire new therapies preventing HF in adult CHD patients.
Collapse
Affiliation(s)
| | | | | | - Paolo Madeddu
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
73
|
Peng R, Li B, Chen S, Shi Z, Yu L, Gao Y, Yang X, Lu L, Wang H. Deleterious Rare Mutations of GLI1 Dysregulate Sonic Hedgehog Signaling in Human Congenital Heart Disease. Front Cardiovasc Med 2022; 9:798033. [PMID: 35445092 PMCID: PMC9014293 DOI: 10.3389/fcvm.2022.798033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/03/2022] [Indexed: 11/22/2022] Open
Abstract
The Glioma-associated oncogene (Gli) family members of zinc finger DNA-binding proteins are core effectors of Sonic hedgehog (SHH) signaling pathway. Studies in model organisms have identified that the Gli genes play critical roles during organ development, including the heart, brain, kidneys, etc. Deleterious mutations in GLI genes have previously been revealed in several human developmental disorders, but few in congenital heart disease (CHD). In this study, the mutations in GLI1-3 genes were captured by next generation sequencing in human cohorts composed of 412 individuals with CHD and 213 ethnically matched normal controls. A total of 20 patient-specific nonsynonymous rare mutations in coding regions of human GLI1-3 genes were identified. Functional analyses showed that GLI1 c.820G> T (p.G274C) is a gain-of-function mutation, while GLI1 c.878G>A (p.R293H) and c.1442T>A (p.L481X) are loss-of-function mutations. Our findings suggested that deleterious rare mutations in GLI1 gene broke the balance of the SHH signaling pathway regulation and may constitute a great contribution to human CHD, which shed new light on understanding genetic mechanism of embryo cardiogenesis regulated by SHH signaling.
Collapse
Affiliation(s)
- Rui Peng
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Binbin Li
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, United States
| | - Shuxia Chen
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Zhiwen Shi
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Liwei Yu
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- SUNY Downstate Medical Center, Children's Hospital at Downstate, Brooklyn, NY, United States
| | - Yunqian Gao
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Yang
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Lei Lu
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hongyan Wang
- NHC Key Laboratory of Reproduction Regulation, State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Shanghai Institute of Planned Parenthood Research, Institute of Reproduction and Development, Children's Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| |
Collapse
|
74
|
Almonaem ERA, Soliman DR, El Sayed MAM, Ahmed IA, Abdelrahman EG. Association between SNP rs59382073 in TBX2 3′ UTR and susceptibility to congenital heart diseases. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
75
|
Zhang S, Qiu X, Wang T, Chen L, Li J, Diao J, Li Y, Qin J, Chen L, Jiang Y. Hypertensive Disorders in Pregnancy Are Associated With Congenital Heart Defects in Offspring: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2022; 9:842878. [PMID: 35419442 PMCID: PMC8995565 DOI: 10.3389/fcvm.2022.842878] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background Although research indicates an association between hypertensive disorders of pregnancy (HDP) and congenital heart defects (CHDs) in offspring, consistency is still lacking. Therefore, we aimed to synthesize the updated published epidemiologic evidence to estimate the association of maternal HDP with the risk of total CHDs and its phenotypes in offspring. Methods A systematic search of Web of Science Database, PubMed, and Embase were searched from inception through April 30, 2021 based on a preprepared protocol, and the reference lists were also manually searched. The combined risk estimates were calculated using either the fixed-effect models or random-effect models. Possible heterogeneity moderators were detected by subgroup, sensitivity analyses, and Galbraith plot. Results Twenty-four studies involving 477,839 CHDs cases among 40,394,699 participants were included in our meta-analysis. Mothers who had HDP exposure were significantly associated with an increased risk of total CHDs compared with non-exposure. When maternal HDP exposure was further subdivided into pre-eclampsia (OR = 1.79, 95% CI: 1.50–2.13), gestational hypertension (OR = 1.16, 95% CI: 1.02–1.31), and chronic hypertension (OR = 1.68, 95% CI: 1.49–1.89), a significantly increased risk of total CHDs were still presented. Furthermore, a statistically significant increased association was found between maternal HDP exposure and most CHD phenotypes. Besides, relevant heterogeneity moderators have been identified by subgroup and sensitivity analyses. Conclusion Our study suggested that maternal HDP exposure may be associated with an increase in the risk of CHDs in offspring. These findings highlight the need for greater surveillance of pregnant women with HDP exposure to allow early prevention that may be good for reducing the risk of CHDs in offspring. Clinical Trial Registration [www.ClinicalTrials.gov], identifier [CRD42021268093].
Collapse
Affiliation(s)
- Senmao Zhang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Xing Qiu
- Xiangya Nursing School of Central South University, Changsha, China
| | - Tingting Wang
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Letao Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Jinqi Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Jingyi Diao
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Yihuan Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Jiabi Qin
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| | - Lizhang Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
- *Correspondence: Lizhang Chen,
| | - Yurong Jiang
- Department of Obstetrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
- Yurong Jiang,
| |
Collapse
|
76
|
Wang Z, Qiao XH, Xu YJ, Liu XY, Huang RT, Xue S, Qiu HY, Yang YQ. SMAD1 Loss-of-Function Variant Responsible for Congenital Heart Disease. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9916325. [PMID: 35281600 PMCID: PMC8913148 DOI: 10.1155/2022/9916325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/16/2022] [Indexed: 12/22/2022]
Abstract
As the most common form of developmental malformation affecting the heart and endothoracic great vessels, congenital heart disease (CHD) confers substantial morbidity and mortality as well as socioeconomic burden on humans globally. Aggregating convincing evidence highlights the genetic origin of CHD, and damaging variations in over 100 genes have been implicated with CHD. Nevertheless, the genetic basis underpinning CHD remains largely elusive. In this study, via whole-exosome sequencing analysis of a four-generation family inflicted with autosomal-dominant CHD, a heterozygous SMAD1 variation, NM_005900.3: c.264C > A; p.(Tyr88∗), was detected and validated by Sanger sequencing analysis to be in cosegregation with CHD in the whole family. The truncating variation was not observed in 362 unrelated healthy volunteers employed as control persons. Dual-luciferase reporter gene assay in cultured COS7 cells demonstrated that Tyr88∗-mutant SMAD1 failed to transactivate the genes TBX20 and NKX2.5, two already well-established CHD-causative genes. Additionally, the variation nullified the synergistic transcriptional activation between SMAD1 and MYOCD, another recognized CHD-causative gene. These data indicate SMAD1 as a new gene responsible for CHD, which provides new insight into the genetic mechanism underlying CHD, suggesting certain significance for genetic risk assessment and precise antenatal prevention of the family members inflicted with CHD.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Xiao-Hui Qiao
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hai-Yan Qiu
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| |
Collapse
|
77
|
Zhang S, Liu X, Yang T, Wang T, Chen L, Qin J. Association of maternal dietary habits and ADIPOQ gene polymorphisms with the risk of congenital heart defects in offspring: a hospital-based case-control study. Eur J Clin Nutr 2022; 76:373-381. [PMID: 34230631 DOI: 10.1038/s41430-021-00969-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 05/27/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To estimate the association of maternal ADIPOQ gene, dietary habits in early pregnancy, and their interactions with the risk of congenital heart defects (CHDs) in offspring. METHODS A case-control study of 464 mothers of CHDs children and 504 mothers of healthy children was included. Maternal dietary habits and genetic polymorphisms of ADIPOQ were the main exposure of interest. Their independent effects and interactions in the development of CHDs were analyzed in our study. RESULTS The excessive consumption of pickled vegetables (aOR = 1.58, 95%CI: 1.17-2.12), smoked foods (aOR = 1.84, 95%CI:1.34-2.52), barbecued foods (aOR = 1.62, 95%CI: 1.09-2.39), fish and shrimp (aOR = 0.37, 95%CI: 0.27-50), and milk products (aOR = 0.64, 95%CI: 0.51-80) had a significant association with total CHDs risk. The polymorphisms of ADIPOQ gene at rs1501299 (T/T vs G/G: aOR = 0.27, 95%CI: 0.14-50; G/T vs G/G: aOR = 0.67, 95%CI: 0.46-98) and rs2241766 (G/G vs T/T: aOR = 4.35, 95%CI: 2.23-8.51; T/G vs T/T: aOR = 2.23, 95%CI: 1.51-3.28) showed a significant association with total CHDs risk. Likewise, our results found that maternal dietary habits and ADIPOQ genetic variants also were significantly related to the risk of specific CHDs phenotypes. In addition, gene-diet interaction revealed significant associations between the ADIPOQ gene and maternal dietary habits with total CHDs. CONCLUSIONS Maternal dietary habits, ADIPOQ gene, and their interactions show a significant association with the risk of CHDs. However, our study has some limitations, thus our findings need to be taken with caution, which highlights that more studies are required to further corroborate our findings.
Collapse
Affiliation(s)
- Senmao Zhang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, Hunan, China
| | - Xiaoying Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, Hunan, China
| | - Tubao Yang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, Hunan, China
| | - Tingting Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.,National Health Commission Key Laboratory for Birth Defect Research and Prevention, Changsha, Hunan, China
| | - Lizhang Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China. .,Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, Hunan, China.
| | - Jiabi Qin
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China. .,Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, Hunan, China. .,National Health Commission Key Laboratory for Birth Defect Research and Prevention, Changsha, Hunan, China. .,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
78
|
Mid-regional pro-adrenomedullin for diagnosing evolution after cardiac surgery in newborns: the PRONEW study. Eur J Pediatr 2022; 181:1017-1028. [PMID: 34686907 DOI: 10.1007/s00431-021-04278-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/13/2021] [Accepted: 09/25/2021] [Indexed: 10/20/2022]
Abstract
Newborns are the most vulnerable patients after cardiac surgery. Although mortality risk scores before surgery may help predict the risk of poor outcome, new tools are required, and biomarkers could add objective data to these tools. The aim of this study was to assess the ability of mid-regional pro-adrenomedullin (pro-ADM) and pro-atrial natriuretic peptide (pro-ANP) to predict poor outcome after cardiac surgery. This is a pilot diagnostic accuracy study that includes newborns and infants under 2 months admitted to an intensive care unit after cardiac surgery. Pro-ADM and pro-ANP were determined immediately upon admission. Poor outcome was defined as mortality, cardiac arrest, requiring extracorporeal support, requiring renal replacement therapy, or neurological injury. Forty-four patients were included. Twenty-six (59%) had a STAT category of ≥ 4. Ten patients (22.7%) presented a poor outcome, four of whom (9.1%) died. Pro-ADM was higher in patients with poor outcome (p = 0.024) and death (p = 0.012). Pro-ADM showed the best area under curve (AUC) for predicting poor outcome (0.735) and mortality alone (0.869). A pro-ADM of 2 nmol/L had a Sn of 75% and a Sp of 85% for predicting mortality. Pro-ADM > 2 nmol/L was independently associated with poor outcome (OR 5.8) and mortality (OR 14.1). Although higher pro-ANP values were associated with poor outcomes, no cut-off point were found. The combination of STAT ≥ 4 and the biomarkers did not enhance predictive power for poor outcome or mortality.Conclusion: Pro-ADM and pro-ANP determined immediately after surgery could be helpful for stratifying risk of poor outcome and mortality in newborns. What is Known: • Some congenital heart diseases must be corrected/palliated during the first days of life. A useful tool to predict the risk of severe complications has not been proposed. • Most unstable newborns would have higher values of biomarkers such as pro-ADM and pro-ANP related to shock and compensatory actions. What is New: • Pro-ADM and pro-ANP seem to be good biomarkers to predict poor outcome after cardiac surgery. A pro-ADM < 2 nmol/L would imply a low likelihood of a poor outcome. • Deepening the analysis of biomarkers can help in making decisions to prevent/treat complications.
Collapse
|
79
|
Wang H, Zhang Y, Ding W, Zhu Y, Lu H, Yue H, Dai X, Li D, Zhu X, Xu X. Trends and influencing factors of perinatal birth defects in Huai'an from 2008 to 2020. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:10-18. [PMID: 35462467 PMCID: PMC9109768 DOI: 10.3724/zdxbyxb-2021-0120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To analyze the incidence, trends and related factors of birth defects in Huai'an from 2008 to 2020. METHODS The surveillance data from maternal and child health system of Huai'an from 2008 to 2020 and Huai'an Statistical Yearbook were used for analysis. Taking the annual change percentage and average annual change percentage (AAPC) as the main outcome indicators, the JoinPoint regression analysis was performed to estimate the changing trend of birth defects from 2008 to 2020. Spearman correlation analysis was used to examine the association between birth defects and birth rate, marriage rate, proportion of women with advanced maternal age. RESULTS During 2008 to 2020, a total of 3414 cases of neonatal birth defects occurred in Huai'an, with an incidence of 4.6‰ (3414/736 608). The rate of perinatal birth defects in Huai'an showed an increasing trend (AAPC=8.8%, t=3.2, P<0.01), and the year of 2016 was a significant changing point. Among 24 types of birth defects, the incidence of congenital heart disease rose and became the most prevalent defect, while the incidence of neural tube malformations such as anencephaly, encephalocele and spina bifida was declined. The incidence of birth defect was negatively correlated with the birth rate ( r=-0.751, P<0.01), not correlated with marriage rate ( r=-0.516, P>0.05), and positively correlated with the proportion of women with advanced maternal age ( r=0.726, P<0.01). CONCLUSION The incidence of birth defects in Huai'an shows an increasing trend from 2008 to 2020 with congenital heart disease as the most common type of birth defect, and the increase of birth defects incidence is closely related with the increase of the proportion of women with advanced maternal age.
Collapse
Affiliation(s)
- Hui Wang
- 1. Department of Preventive Health Care, Huai'an Maternity and Child Health Care Hospital, Huai'an 223001, Jiangsu Province, China
| | - Yue Zhang
- 2. School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weijie Ding
- 1. Department of Preventive Health Care, Huai'an Maternity and Child Health Care Hospital, Huai'an 223001, Jiangsu Province, China
| | - Yi Zhu
- 2. School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- 3. School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongmei Lu
- 1. Department of Preventive Health Care, Huai'an Maternity and Child Health Care Hospital, Huai'an 223001, Jiangsu Province, China
| | - Hongni Yue
- 1. Department of Preventive Health Care, Huai'an Maternity and Child Health Care Hospital, Huai'an 223001, Jiangsu Province, China
| | - Xiaochen Dai
- 4. School of Public Health, University of Washington, Seattle 98195, USA
| | - Duanhui Li
- 2. School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoqin Zhu
- 1. Department of Preventive Health Care, Huai'an Maternity and Child Health Care Hospital, Huai'an 223001, Jiangsu Province, China
| | - Xiaolin Xu
- 2. School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- 5. Center for Clinical Big Data and Statistics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
80
|
Zanini M. Treatment Evolution and the Impact of Pre-Surgical Predictors on Outcomes of Patients with Congenital Heart Disease. Arq Bras Cardiol 2022; 118:420-421. [PMID: 35262575 PMCID: PMC8856684 DOI: 10.36660/abc.20220020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Maurice Zanini
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul,Porto Alegre, RS – Brasil
| |
Collapse
|
81
|
|
82
|
Blum KM, Mirhaidari G, Breuer CK. Tissue engineering: Relevance to neonatal congenital heart disease. Semin Fetal Neonatal Med 2022; 27:101225. [PMID: 33674254 PMCID: PMC8390581 DOI: 10.1016/j.siny.2021.101225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Congenital heart disease (CHD) represents a large clinical burden, representing the most common cause of birth defect-related death in the newborn. The mainstay of treatment for CHD remains palliative surgery using prosthetic vascular grafts and valves. These devices have limited effectiveness in pediatric patients due to thrombosis, infection, limited endothelialization, and a lack of growth potential. Tissue engineering has shown promise in providing new solutions for pediatric CHD patients through the development of tissue engineered vascular grafts, heart patches, and heart valves. In this review, we examine the current surgical treatments for congenital heart disease and the research being conducted to create tissue engineered products for these patients. While much research remains to be done before tissue engineering becomes a mainstay of clinical treatment for CHD patients, developments have been progressing rapidly towards translation of tissue engineering devices to the clinic.
Collapse
Affiliation(s)
- Kevin M Blum
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Childrens Hospital, Columbus, OH, USA; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| | - Gabriel Mirhaidari
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Childrens Hospital, Columbus OH, USA,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus OH, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Childrens Hospital, Columbus, OH, USA.
| |
Collapse
|
83
|
Diminished Endothelial Function but Normal Vascular Structure in Adults with Tetralogy of Fallot. J Clin Med 2022; 11:jcm11030493. [PMID: 35159945 PMCID: PMC8836741 DOI: 10.3390/jcm11030493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/01/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
The life expectancy of patients with Tetralogy of Fallot (ToF) has increased in recent years. As a result, other risk factors with later onset in life are in the focus of patient care. Endothelial function is an early indicator of cardiovascular risk and was investigated along further structural vessel properties. A total of 17 patients (41.7 ± 7.1 years, 8 women) with Tetralogy of Fallot were 1:2 matched for sex with 34 (38.9 ± 8.1 years, 16 women) healthy volunteers. Participants received an assessment of their endothelial function and a structural assessment of the aorta. Patients with ToF showed a reduced endothelial function determined by reactive hyperaemia index after adjusting for age, weight and height (ToF: 1.55 ± 0.31 vs. controls: 1.84 ± 0.47; p = 0.023). No differences in carotid intima-media thickness (cIMT) between the ToF and healthy controls (ToF: 0.542 ± 0.063 mm vs. controls: 0.521 ± 0.164 mm; p = 0.319) were found. Patients with ToF had reduced vascular function compared to healthy subjects. As the structural component is not affected, endothelial dysfunction seems not to have yet manifested itself as a morphological change. Nevertheless, long-term management of these patients should include vascular parameters.
Collapse
|
84
|
Blum KM, Zbinden JC, Ramachandra AB, Lindsey SE, Szafron JM, Reinhardt JW, Heitkemper M, Best CA, Mirhaidari GJM, Chang YC, Ulziibayar A, Kelly J, Shah KV, Drews JD, Zakko J, Miyamoto S, Matsuzaki Y, Iwaki R, Ahmad H, Daulton R, Musgrave D, Wiet MG, Heuer E, Lawson E, Schwarz E, McDermott MR, Krishnamurthy R, Krishnamurthy R, Hor K, Armstrong AK, Boe BA, Berman DP, Trask AJ, Humphrey JD, Marsden AL, Shinoka T, Breuer CK. Tissue engineered vascular grafts transform into autologous neovessels capable of native function and growth. COMMUNICATIONS MEDICINE 2022; 2:3. [PMID: 35603301 PMCID: PMC9053249 DOI: 10.1038/s43856-021-00063-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022] Open
Abstract
Background Tissue-engineered vascular grafts (TEVGs) have the potential to advance the surgical management of infants and children requiring congenital heart surgery by creating functional vascular conduits with growth capacity. Methods Herein, we used an integrative computational-experimental approach to elucidate the natural history of neovessel formation in a large animal preclinical model; combining an in vitro accelerated degradation study with mechanical testing, large animal implantation studies with in vivo imaging and histology, and data-informed computational growth and remodeling models. Results Our findings demonstrate that the structural integrity of the polymeric scaffold is lost over the first 26 weeks in vivo, while polymeric fragments persist for up to 52 weeks. Our models predict that early neotissue accumulation is driven primarily by inflammatory processes in response to the implanted polymeric scaffold, but that turnover becomes progressively mechano-mediated as the scaffold degrades. Using a lamb model, we confirm that early neotissue formation results primarily from the foreign body reaction induced by the scaffold, resulting in an early period of dynamic remodeling characterized by transient TEVG narrowing. As the scaffold degrades, mechano-mediated neotissue remodeling becomes dominant around 26 weeks. After the scaffold degrades completely, the resulting neovessel undergoes growth and remodeling that mimicks native vessel behavior, including biological growth capacity, further supported by fluid-structure interaction simulations providing detailed hemodynamic and wall stress information. Conclusions These findings provide insights into TEVG remodeling, and have important implications for clinical use and future development of TEVGs for children with congenital heart disease.
Collapse
Affiliation(s)
- Kevin M. Blum
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 USA
| | - Jacob C. Zbinden
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 USA
| | | | - Stephanie E. Lindsey
- Department of Pediatrics (Cardiology), Stanford University, Stanford, CA 94305 USA
- Institute for Computational and Mathematical Engineering (ICME), Stanford University, Stanford, CA 94305 USA
| | - Jason M. Szafron
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520 USA
| | - James W. Reinhardt
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Megan Heitkemper
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Cameron A. Best
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- The Ohio State University College of Medicine, Columbus, OH 43210 USA
| | - Gabriel J. M. Mirhaidari
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Yu-Chun Chang
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Anudari Ulziibayar
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - John Kelly
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Kejal V. Shah
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Joseph D. Drews
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Jason Zakko
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Shinka Miyamoto
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Cardiovascular Surgery at Tokyo Women’s Medical University, Tokyo, Japan
| | - Yuichi Matsuzaki
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Ryuma Iwaki
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Hira Ahmad
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Pediatric Colorectal and Pelvic Reconstructive Surgery, Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Robbie Daulton
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- University of Cincinnati College of Medicine 3230 Eden Ave, Cincinnati, OH 45267 USA
| | - Drew Musgrave
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Matthew G. Wiet
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- The Ohio State University College of Medicine, Columbus, OH 43210 USA
| | - Eric Heuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Emily Lawson
- The Ohio State University College of Medicine, Columbus, OH 43210 USA
| | - Erica Schwarz
- Department of Bioengineering, Stanford University, Stanford, CA 94304 USA
| | - Michael R. McDermott
- Center for Cardiovascular Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Rajesh Krishnamurthy
- Department of Radiology, Nationwide Children’s Hospital, Columbus, Ohio 43205 USA
| | | | - Kan Hor
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Aimee K. Armstrong
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Brian A. Boe
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Darren P. Berman
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Aaron J. Trask
- Center for Cardiovascular Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210 USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520 USA
| | - Alison L. Marsden
- Institute for Computational and Mathematical Engineering (ICME), Stanford University, Stanford, CA 94305 USA
- Department of Bioengineering, Stanford University, Stanford, CA 94304 USA
| | - Toshiharu Shinoka
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205 USA
- Department of Cardiothoracic Surgery, The Ohio State University College of Medicine, Columbus, OH 43205 USA
| | - Christopher K. Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| |
Collapse
|
85
|
Meyer M, Wang Y, Brudy L, Häcker AL, Schulz T, Weberruss H, Oberhoffer R, Ewert P, Müller J. Impaired grip strength in children with congenital heart disease. Arch Dis Child 2022; 107:47-51. [PMID: 34140308 DOI: 10.1136/archdischild-2020-319955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 05/30/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Grip strength is known to be reduced in adults with congenital heart disease (CHD). This study compared grip strength in paediatric patients with CHD with healthy controls and determined a possible association between grip strength and health-related physical fitness (HRPF). METHODS Grip strength and HRPF were assessed in 569 children (12.4 years, 95% CI 12.16 to 12.72; 238 girls) with various CHD and compared with 2551 healthy controls (11.4 years, 95% CI 11.3 to 11.5; 1424 girls). Grip strength was determined as the maximum value of three repetitions with each hand. HRPF was tested by five motor tasks (FITNESSGRAM) and converted into an SD score (z-score). RESULTS After adjusting for age, sex and weight, children with CHD showed significantly lower grip strength compared with healthy controls (CHD: 20.8 kg, 95% CI 20.4 to 21.2; controls: 24.5 kg, 95% CI 24.3 to 24.8). CHD subgroup analysis also revealed significantly lower grip strength than the controls, with the lowest values in patients with total cavopulmonary connection (19.1, 95% CI 18.0 to 20.2). Children with complex CHD showed the lowest values with 19.8 kg (95% CI 19.2 to 20.4), those with moderate 20.7 kg (95% CI 19.9 to 21.4) and those with simple 22.5 kg (95% CI 21.6 to 23.3), respectively. HRPF was also lower (z-score: -0.46, 95% CI -0.49 to -0.35) compared with healthy controls and poorly associated with grip strength (r=0.21). CONCLUSIONS Grip strength is already reduced in children with CHD and poorly associated with HRPF. This suggests that grip strength and HRPF are different domains and have to be assessed separately.
Collapse
Affiliation(s)
- Michael Meyer
- Institute of Preventive Pediatrics, Technical University of Munich, Munich, Germany .,Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Munich, Germany
| | - Yi Wang
- Institute of Preventive Pediatrics, Technical University of Munich, Munich, Germany
| | - Leon Brudy
- Institute of Preventive Pediatrics, Technical University of Munich, Munich, Germany.,Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Munich, Germany
| | - Anna-Luisa Häcker
- Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Munich, Germany
| | - Thorsten Schulz
- Institute of Preventive Pediatrics, Technical University of Munich, Munich, Germany
| | - Heidi Weberruss
- Institute of Preventive Pediatrics, Technical University of Munich, Munich, Germany
| | - Renate Oberhoffer
- Institute of Preventive Pediatrics, Technical University of Munich, Munich, Germany.,Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Munich, Germany
| | - Peter Ewert
- Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Munich, Germany
| | - Jan Müller
- Institute of Preventive Pediatrics, Technical University of Munich, Munich, Germany.,Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich, Munich, Germany
| |
Collapse
|
86
|
Chlif M, Ammar MM, Said NB, Sergey L, Ahmaidi S, Alassery F, Hamam H. Mechanism of Dyspnea during Exercise in Children with Corrected Congenital Heart Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:99. [PMID: 35010359 PMCID: PMC8751078 DOI: 10.3390/ijerph19010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
This study will evaluate cardiorespiratory and peripheral muscle function and their relationship with subjective dyspnea threshold after the surgical correction of congenital heart disease in children. Thirteen children with surgically repaired congenital heart disease were recruited. Each participant performed an incremental exercise test on a cycle ergometer until exhaustion. Gas exchanges were continuously sampled to measure the maximal aerobic parameters and ventilatory thresholds. The functional capacity of the subjects was assessed with a 6 min walk test. At the end of the exercise test, isokinetic Cybex Norm was used to evaluate the strength and endurance of the knee extensor muscle in the leg. Dyspnea was subjectively scored with a visual analog scale during the last 15 s of each exercise step. Oxygen consumption measured at the dyspnea score/VO2 relationship located at the dyspnea threshold, at which dyspnea suddenly increased. Results: The maximal and submaximal values of the parameters describing the exercise and the peripheral muscular performances were: VO2 Peak: 33.8 ± 8.9 mL·min-1·kg-1; HR: 174 ± 9 b·min-1; VEmax: 65.68 ± 15.9 L·min-1; P max: 117 ± 27 W; maximal voluntary isometric force MVIF: 120.8 ± 41.9 N/m; and time to exhaustion Tlim: 53 ± 21 s. Oxygen consumption measured at the dyspnea threshold was related to VO2 Peak (R2 = 0.74; p < 0.01), Tlim (R2 = 0.78; p < 0.01), and the distance achieved during the 6MWT (R2 = 0.57; p < 0.05). Compared to the theoretical maximal values for the power output, VO2, and HR, the surgical correction did not repair the exercise performance. After the surgical correction of congenital heart disease, exercise performance was impeded by alterations of the cardiorespiratory function and peripheral local factors. A subjective evaluation of the dyspnea threshold is a reliable criterion that allows the prediction of exercise capacity in subjects suffering from congenital heart disease.
Collapse
Affiliation(s)
- Mehdi Chlif
- EA 3300 “APS and Motor Patterns: Adaptations-Rehabilitation”, Picardie Jules Verne University, 80025 Amiens, France
- National Center of Medicine and Science in Sports (NCMSS), Tunisian Research Laboratory Sports Performance Optimization, Ave Med Ali Akid, El Menzah, Tunis 263, Tunisia;
| | - Mohamed Mustapha Ammar
- Exercise Physiology Department, College of Sport Sciences and Physical Activity, King Saud University, C.P. 22480, Riyadh 11495, Saudi Arabia;
| | - Noureddine Ben Said
- Department of Biomechanics and Motor Behavior, College of Sport Sciences and Physical Activity, King Saud University, C.P. 22480, Riyadh 11495, Saudi Arabia;
| | - Levushkin Sergey
- Federal State-Funded Scientific Institution “Institute of Developmental Physiology of the Russian Academy of Education”, Russian State University of Physical Culture, Sport, Youth and Tourism (SCOLIPE), 105122 Moscow, Russia;
| | - Said Ahmaidi
- National Center of Medicine and Science in Sports (NCMSS), Tunisian Research Laboratory Sports Performance Optimization, Ave Med Ali Akid, El Menzah, Tunis 263, Tunisia;
| | - Fawaz Alassery
- Department of Computer Engineering, College of Computers and Information Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Habib Hamam
- Faculty of Engineering, Moncton University, Moncton, NB E1A 3E9, Canada;
| |
Collapse
|
87
|
Infrastructure Availability for the Care of Congenital Heart Disease Patients and Its Influence on Case Volume, Complexity and Access Among Healthcare Institutions in 17 Middle-Income Countries. Glob Heart 2021; 16:75. [PMID: 34900566 PMCID: PMC8533658 DOI: 10.5334/gh.968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 08/31/2021] [Indexed: 11/20/2022] Open
Abstract
The care for patients with congenital heart disease (CHD) is multi-disciplinary and resource intensive. There is limited information about the infrastructure available among programs that care for CHD patients in low and middle-income countries (LMIC). A survey covering the entire care-pathway for CHD, from initial assessment to inpatient care and outpatient follow-up, was administered to institutions participating in the International Quality Improvement Collaborative for Congenital Heart Disease (IQIC). Surgical case complexity-mix was collected from the IQIC registry and estimated surgical capacity requirement was based on population data. The statistical association of selected infrastructure with case volume, case-complexity and percentage of estimated case-burden actually treated, was analyzed. Thirty-seven healthcare institutions in seventeen countries with median annual surgical volume of 361 (41-3503) operations completed the survey. There was a median of two (1-16) operating room/s (OR), nine (2-80) intensive care unit (ICU) beds, three (1-20) cardiac surgeons, five (3-30) OR nurses, four (2-35) anesthesiologists, four (1-25) perfusionists, 28 (5-194) ICU nurses, six (0-30) cardiologists and three (1-15) interventional cardiologists. Higher surgical volume was associated with higher OR availability (p = 0.007), number of surgeons (p = 0.002), OR nurses (0.008), anesthesiologists (p = 0.04), perfusionists (p = 0.001), ICU nurses (p < 0.001), years of experience of the most senior surgeon (p = 0.03) or cardiologist (p = 0.05), and ICU bed capacity (p = 0.001). Location in an upper-middle income country (P = 0.04), OR availability (p = 0.02), and number of cardiologists (p = 0.004) were associated with performing a higher percentage of complex cases. This study demonstrates an overall deficit in the infrastructure available for the care of CHD patients among the participating institutions. While there is considerable variation across institutions surveyed, deficits in infrastructure that requires long-term investment like operating rooms, intensive care capacity, and availability of trained staff, are associated with reduced surgical capacity and access to CHD care.
Collapse
|
88
|
Li S, Jin Y, Tang P, Liu X, Chai X, Dong J, Che X, Zhou Q, Ni M, Jin F. Maternal serum-derived exosomal lactoferrin as a marker in detecting and predicting ventricular septal defect in fetuses. Exp Biol Med (Maywood) 2021; 247:488-497. [PMID: 34871505 DOI: 10.1177/15353702211060517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Among different types of congenital heart diseases, ventricular septal defect is the most frequently diagnosed type and is frequently missed in early prenatal screening programs. Herein, we explored the role of maternal serum-derived exosomes in detecting and predicting ventricular septal defect in fetuses in the early stage of pregnancy. A total of 104 pregnant women consisting of 52 ventricular septal defect cases and 52 healthy controls were recruited. TMT/iTRAQ proteomic analysis uncovered 15 maternal serum exosomal proteins, which showed differential expression between ventricular septal defect and control groups. Among these, four down-regulated proteins, lactoferrin, SBSN, DCD, and MBD3, were validated by Western blot. The protein lactoferrin was additionally verified by ELISA which was able to distinguish ventricular septal defects from controls with area under the ROC curve (AUC) 0.804 (p < 0.001). Our findings reveal that lactoferrin in maternal serum-derived exosomes may be a potential biomarker for non-invasive prenatal diagnosis of fetal ventricular septal defects.
Collapse
Affiliation(s)
- Suping Li
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.,Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Yuxia Jin
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Ping Tang
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Xiaodan Liu
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Xiaojun Chai
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Jinhua Dong
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Xuan Che
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Qinqin Zhou
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Meidi Ni
- Department of Fetal Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing University Affiliated Women and Children Hospital, Jiaxing 314050, China
| | - Fan Jin
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
89
|
Steurer MA, Baer RJ, Chambers CD, Costello J, Franck LS, McKenzie-Sampson S, Pacheco-Werner TL, Rajagopal S, Rogers EE, Rand L, Jelliffe-Pawlowski LL, Peyvandi S. Mortality and Major Neonatal Morbidity in Preterm Infants with Serious Congenital Heart Disease. J Pediatr 2021; 239:110-116.e3. [PMID: 34454949 PMCID: PMC10866139 DOI: 10.1016/j.jpeds.2021.08.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/29/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the trends of 1-year mortality and neonatal morbidities in preterm infants with serious congenital heart disease (CHD). STUDY DESIGN This cohort study used a population-based administrative dataset of all liveborn infants of 26-36 weeks gestational age with serious CHD born in California between 2011 and 2017. We assessed 1-year mortality and major neonatal morbidities (ie, retinopathy of prematurity, bronchopulmonary dysplasia, necrotizing enterocolitis, intraventricular hemorrhage grade >2, and periventricular leukomalacia) across the study period and compared these outcomes with those in infants without CHD. RESULTS We identified 1921 preterm infants with serious CHD. The relative risk (RR) of death decreased by 10.6% for each year of the study period (RR, 0.89; 95% CI, 0.84-0.95), and the RR of major neonatal morbidity increased by 8.3% for each year (RR, 1.08; 95% CI, 1.02-1.15). Compared with preterm neonates without any CHD (n = 234 522), the adjusted risk difference (ARD) for mortality was highest at 32 weeks of gestational age (9.7%; 95% CI, 8.3%-11.2%), that for major neonatal morbidity was highest at 28 weeks (21.9%; 95% CI, 17.0%-26.9%), and that for the combined outcome was highest at 30 weeks (26.7%; 95% CI, 23.3%-30.1%). CONCLUSIONS Mortality in preterm neonates with serious CHD decreased over the last decade, whereas major neonatal morbidities increased. Preterm infants with a gestational age of 28-32 weeks have the highest mortality or morbidity compared with their peers without CHD. These results support the need for specialized and focused medical neonatal care in preterm neonates with serious CHD.
Collapse
Affiliation(s)
- Martina A Steurer
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA; California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA
| | - Rebecca J Baer
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA; Department of Pediatrics, University of California San Diego, La Jolla, CA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA
| | | | - Jean Costello
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA; California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA
| | - Linda S Franck
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA
| | - Safyer McKenzie-Sampson
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA; California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA
| | - Tania L Pacheco-Werner
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA; Central Valley Health Policy Institute, California State University Fresno, Fresno, CA
| | - Satish Rajagopal
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Elizabeth E Rogers
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA
| | - Larry Rand
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA
| | - Laura L Jelliffe-Pawlowski
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA; California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA
| | - Shabnam Peyvandi
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA
| |
Collapse
|
90
|
Bossers GPL, Günthel M, van der Feen DE, Hagdorn QAJ, Koop AMC, van Duijvenboden K, Barnett P, Borgdorff MAJ, Christoffels VM, Silljé HHW, Berger RMF, Bartelds B. Neuregulin-1 enhances cell-cycle activity, delays cardiac fibrosis, and improves cardiac performance in rat pups with right ventricular pressure load. J Thorac Cardiovasc Surg 2021; 164:e493-e510. [PMID: 34922752 DOI: 10.1016/j.jtcvs.2021.10.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Right ventricular (RV) failure is a leading cause of death in patients with congenital heart disease. RV failure is kept at bay during childhood. Limited proliferation of cardiomyocytes is present in the postnatal heart. We propose that cardiomyocyte proliferation improves RV adaptation to pressure load (PL). We studied adaptation in response to increased RV PL and the role of increased cardiomyocyte cell cycle activity (CCA) in rat pups growing into adulthood. METHODS We induced RV PL at day of weaning in rats (3 weeks; 30-40 g) by pulmonary artery banding and followed rats into adulthood (300 g). We performed histological analyses and RNA sequencing analysis. To study the effects of increased cardiomyocyte cell cycle activity, we administered neuregulin-1 (NRG1), a growth factor involved in cardiac development. RESULTS PL induced an increase in CCA, with subsequent decline of CCA (sham/PL at 4 weeks: 0.14%/0.83%; P = .04 and 8 weeks: 0.00%/0.00%; P = .484) and cardiac function (cardiac index: control/PL 4 weeks: 4.41/3.29; P = .468 and 8 weeks: 3.57/1.44; P = .024). RNA sequencing analysis revealed delayed maturation and increased CCA pathways. NRG1 stimulated CCA (PL vehicle/NRG1 at 2 weeks: 0.62%/2.28%; P = .003), improved cardiac function (cardiac index control vs vehicle/NRG1 at 2 weeks: 4.21 vs 3.07/4.17; P = .009/.705) and postponed fibrosis (control vs vehicle/NRG1 at 4 weeks: 1.66 vs 4.82%/2.97%; P = .009/.078) in RV PL rats during childhood. CONCLUSIONS RV PL during growth induces a transient CCA increase. Further CCA stimulation improves cardiac function and delays fibrosis. This proof-of-concept study shows that stimulation of CCA can improve RV adaptation to PL in the postnatal developing heart and might provide a new approach to preserve RV function in patients with congenital heart disease.
Collapse
Affiliation(s)
- Guido P L Bossers
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Marie Günthel
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Diederik E van der Feen
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Quint A J Hagdorn
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne-Marie C Koop
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karel van Duijvenboden
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Phil Barnett
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Marinus A J Borgdorff
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Beatrijs Bartelds
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
91
|
Derrick CJ, Pollitt EJG, Sanchez Sevilla Uruchurtu A, Hussein F, Grierson AJ, Noël ES. Lamb1a regulates atrial growth by limiting second heart field addition during zebrafish heart development. Development 2021; 148:272294. [PMID: 34568948 DOI: 10.1242/dev.199691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
During early vertebrate heart development, the heart transitions from a linear tube to a complex asymmetric structure, a morphogenetic process that occurs simultaneously with growth of the heart. Cardiac growth during early heart morphogenesis is driven by deployment of cells from the second heart field (SHF) into both poles of the heart. Laminin is a core component of the extracellular matrix and, although mutations in laminin subunits are linked with cardiac abnormalities, no role for laminin has been identified in early vertebrate heart morphogenesis. We identified tissue-specific expression of laminin genes in the developing zebrafish heart, supporting a role for laminins in heart morphogenesis. Analysis of heart development in lamb1a zebrafish mutant embryos reveals mild morphogenetic defects and progressive cardiomegaly, and that Lamb1a functions to limit heart size during cardiac development by restricting SHF addition. lamb1a mutants exhibit hallmarks of altered haemodynamics, and blocking cardiac contractility in lamb1a mutants rescues heart size and atrial SHF addition. Together, these results suggest that laminin mediates interactions between SHF deployment and cardiac biomechanics during heart morphogenesis and growth in the developing embryo.
Collapse
Affiliation(s)
| | - Eric J G Pollitt
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | | | - Farah Hussein
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
92
|
Khatami M, Ghorbani S, Adriani MR, Bahaloo S, Naeini MA, Heidari MM, Hadadzadeh M. Novel Point Mutations in 3'-Untranslated Region of GATA4 Gene Are Associated with Sporadic Non-syndromic Atrial and Ventricular Septal Defects. Curr Med Sci 2021; 42:129-143. [PMID: 34652630 DOI: 10.1007/s11596-021-2428-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/14/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Transcription factor GATA4 has significant roles in embryonic heart development. Mutations of GATA4 appear to be responsible for a wide variety of congenital heart defects (CHD). Despite the high prevalence of GATA4 mutations in CHD phenotypes, extensive studies have not been performed. The 3'-untranslated region (3'-UTR) of the GATA4 gene comprises regulatory motifs and microRNA binding sites that are critical for the appropriate gene expression, nuclear transportation, and regulation of translation, and stability of mRNA. This study aimed to evaluate the association between mutations in the 3'-UTR of the GATA4 gene and CHD risk among Iranian patients. METHODS We analyzed the coding region of exon 6 and the whole 3'-UTR of GATA4 in DNA isolated from 175 blood samples of CHD patients and 115 unrelated healthy individuals. The functional importance of the observed GATA4 mutations was evaluated using a variety of bioinformatics algorithms for assessment of nonsynonymous mutations and those observed in miRNA binding sites of 3'-UTR. RESULTS Twenty-one point mutations including one missense mutation (c.511A>G: p.Ser377Gly) in exon 6 and 20 nucleotide variations in 3'-UTR of GATA4 gene were identified in 65 of the 175 CHD patients. In our patients, we identified 12 novel sequence alterations and 8 single nucleotide polymorphisms in the 3'-UTR of GATA4. Most of them had statistically significant differences between CHD patients and controls. CONCLUSION Our results suggest that 3'-UTR variations of the GATA4 gene probably change microRNA binding sites and present an additional molecular risk factor for the susceptibility of CHD.
Collapse
Affiliation(s)
- Mehri Khatami
- Department of Biology, Faculty of Science, Yazd University, Yazd, 8915818411, Iran.
| | - Sajedeh Ghorbani
- Department of Biology, Faculty of Science, Yazd University, Yazd, 8915818411, Iran
| | | | - Sahar Bahaloo
- Department of Biology, Faculty of Science, Yazd University, Yazd, 8915818411, Iran
| | - Mehri Azami Naeini
- Department of Biology, Faculty of Science, Yazd University, Yazd, 8915818411, Iran
| | | | - Mehdi Hadadzadeh
- Department of Cardiac Surgery, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, 8915887856, Iran
| |
Collapse
|
93
|
Chang YC, Li J, Mirhaidari G, Zbinden J, Barker J, Blum K, Reinhardt J, Best C, Kelly J, Shoji T, Yi T, Breuer C. Zoledronate alters natural progression of tissue-engineered vascular grafts. FASEB J 2021; 35:e21849. [PMID: 34473380 DOI: 10.1096/fj.202001606rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/11/2021] [Accepted: 07/27/2021] [Indexed: 12/19/2022]
Abstract
Macrophages are a critical driver of neovessel formation in tissue-engineered vascular grafts (TEVGs), but also contribute to graft stenosis, a leading clinical trial complication. Macrophage depletion via liposomal delivery of clodronate, a first-generation bisphosphonate, mitigates stenosis, but simultaneously leads to a complete lack of tissue development in TEVGs. This result and the associated difficulty of utilizing liposomal delivery means that clodronate may not be an ideal means of preventing graft stenosis. Newer generation bisphosphonates, such as zoledronate, may have differential effects on graft development with more facile drug delivery. We sought to examine the effect of zoledronate on TEVG neotissue formation and its potential application for mitigating TEVG stenosis. Thus, mice implanted with TEVGs received zoledronate or no treatment and were monitored by serial ultrasound for graft dilation and stenosis. After two weeks, TEVGs were explanted for histological examination. The overall graft area and remaining graft material (polyglycolic-acid) were higher in the zoledronate treatment group. These effects were associated with a corresponding decrease in macrophage infiltration. In addition, zoledronate affected the deposition of collagen in TEVGs, specifically, total and mature collagen. These differences may be, in part, explained by a depletion of leukocytes within the bone marrow that subsequently led to a decrease in the number of tissue-infiltrating macrophages. TEVGs from zoledronate-treated mice demonstrated a significantly greater degree of smooth muscle cell presence. There was no statistical difference in graft patency between treatment and control groups. While zoledronate led to a decrease in the number of macrophages in the TEVGs, the severity of stenosis appears to have increased significantly. Zoledronate treatment demonstrates that the process of smooth muscle cell-mediated neointimal hyperplasia may occur separately from a macrophage-mediated mechanism.
Collapse
Affiliation(s)
- Yu-Chun Chang
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Junlang Li
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Gabriel Mirhaidari
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jacob Zbinden
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, Ohio, USA
| | - Jenny Barker
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Plastic and Reconstructive Surgery, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Kevin Blum
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, Ohio, USA
| | - James Reinhardt
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Cameron Best
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - John Kelly
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Toshihiro Shoji
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Tai Yi
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Christopher Breuer
- Center for Regenerative Medicine at the Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
94
|
Yan F, Liu H, Zhang H, Yi L, Wu Y, Deng C, Qiu Y, Ma X, Li Q, Yang F, Xu W, Tao J, Buonocore JJ, Zhan Y, Dai L. Association between maternal exposure to gaseous pollutants and atrial septal defect in China: A nationwide population-based study. ENVIRONMENTAL RESEARCH 2021; 200:111472. [PMID: 34097890 DOI: 10.1016/j.envres.2021.111472] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND The association between maternal exposure to gaseous air pollutants and congenital heart defects (CHD) remains unclear. The concentration-response relationship and the time windows of susceptibility to gaseous pollutants may vary by pollutant species and CHD subtypes. OBJECTIVE We aimed to examine the relationship between maternal exposures to four species of gaseous pollutants (NO2, O3, SO2, and CO) and atrial septal defect (ASD), which is a common subtype of CHD, and to determine the critical time windows of susceptibility for each gaseous pollutant. METHODS Among 1,253,633 infants born between October 1, 2013 and December 31, 2016 in China, 1937 newborns were diagnosed with isolated ASD, a prevalence of 1.55‰. Maternal exposures to the gaseous pollutants were estimated by matching the geocoded maternal addresses with the gridded ambient concentrations. The adjusted odds ratios (aOR) between exposures and ASD were quantified by using mixed-effects logistic regression models. RESULTS We found significantly positive associations between ASD and maternal exposures to NO2, O3, SO2, and CO during entire pregnancy, first-, second-, and third-trimester. However, no statistically significant association was found between maternal exposure to PM2.5, PM2.5-10 and ASD risk (P > 0.05). In the fully adjusted model with respect to average exposure over entire pregnancy, the adjusted odds ratios (aOR) for each 10 μg/m3 increment of NO2, O3, SO2 were 1.33 (95% CI: 1.22-1.45), 1.13 (95% CI: 1.10-1.16), 1.28 (95% CI: 1.20-1.35), respectively; the aOR for each 100 μg/m3 increment of CO was 1.10 (95% CI: 1.06-1.15). The observed concentration-response relationships varied by exposure periods and pollutants, with the strongest association for NO2 during the 1st-8th embryology weeks, for O3 during the third trimester, for SO2 during the second trimester, and for CO without obvious variation. CONCLUSIONS The findings suggest an increased risk of ASD in association with maternal exposures to four common gaseous pollutants. From the perspective of birth defects prevention and ASD risk mitigation, it is critical to reduce maternal exposure to gaseous pollutants especially during the most susceptible time windows.
Collapse
Affiliation(s)
- Fangyuan Yan
- Department of Environmental Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China; The Joint Laboratory for Pulmonary Development and Related Diseases, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hanmin Liu
- Department of Environmental Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China; The Joint Laboratory for Pulmonary Development and Related Diseases, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hanyue Zhang
- Department of Environmental Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Ling Yi
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yangyang Wu
- Department of Environmental Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Changfei Deng
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yang Qiu
- Department of Environmental Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xia Ma
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qi Li
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fumo Yang
- Department of Environmental Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China; National Engineering Research Center for Flue Gas Desulfurization, Chengdu, Sichuan, 610065, China
| | - Wenli Xu
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Tao
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jonathan J Buonocore
- Center for Climate, Health, and the Global Environment at Harvard T.H. Chan School of Public Health, Boston, MA, 02115, United States
| | - Yu Zhan
- Department of Environmental Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China; National Engineering Research Center for Flue Gas Desulfurization, Chengdu, Sichuan, 610065, China; Med-X Center for Informatics, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Li Dai
- The Joint Laboratory for Pulmonary Development and Related Diseases, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, 610041, China; National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Med-X Center for Informatics, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
95
|
Zeng X, Hu Y, Shu L, Li J, Duan H, Shu Q, Li H. Explainable machine-learning predictions for complications after pediatric congenital heart surgery. Sci Rep 2021; 11:17244. [PMID: 34446783 PMCID: PMC8390484 DOI: 10.1038/s41598-021-96721-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 08/12/2021] [Indexed: 11/23/2022] Open
Abstract
The quality of treatment and prognosis after pediatric congenital heart surgery remains unsatisfactory. A reliable prediction model for postoperative complications of congenital heart surgery patients is essential to enable prompt initiation of therapy and improve the quality of prognosis. Here, we develop an interpretable machine-learning-based model that integrates patient demographics, surgery-specific features and intraoperative blood pressure data for accurately predicting complications after pediatric congenital heart surgery. We used blood pressure variability and the k-means algorithm combined with a smoothed formulation of dynamic time wrapping to extract features from time-series data. In addition, SHAP framework was used to provide explanations of the prediction. Our model achieved the best performance both in binary and multi-label classification compared with other consensus-based risk models. In addition, this explainable model explains why a prediction was made to help improve the clinical understanding of complication risk and generate actionable knowledge in practice. The combination of model performance and interpretability is easy for clinicians to trust and provide insight into how they should respond before the condition worsens after pediatric congenital heart surgery.
Collapse
Affiliation(s)
- Xian Zeng
- The Children's Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China.,The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yaoqin Hu
- The Children's Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Liqi Shu
- Department of Neurology, Rhode Island Hospital, Brown University, Providence, USA
| | - Jianhua Li
- The Children's Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Huilong Duan
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Qiang Shu
- The Children's Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China.
| | - Haomin Li
- The Children's Hospital of Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
96
|
Chen X, Cai XM, Zhang MJ, Xu JH, Li H, Xu ZM. Pharmacokinetics of treprostinil in children with functional single-ventricle pulmonary arterial hypertension: a randomized controlled trial. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1163. [PMID: 34430604 PMCID: PMC8350654 DOI: 10.21037/atm-21-3188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/14/2021] [Indexed: 12/22/2022]
Abstract
Background Application of Treprostinil (TRE) in the patients with single ventricle (SV) physiology is very limited, and the optimal dose for children has not been determined. In this study, we aimed to analyze plasma samples to assess the attainment of clinically therapeutic concentrations of TRE and its efficacy and safety in the treatment of pediatric functional SV pulmonary arterial hypertension (FSV-PAH).. Methods Pediatric patients with FSV-PAH were recruited in this study. IV TRE at an initial rate of 5 ng/kg/min was administered through the femoral vein with an increase in rate to 10 ng/kg/min every 30 minuntil the aiming dose of 80 ng/kg/min had been reached. The drug was gradually discontinued after 12 h of treatment at a stable dose. The mean postoperative pulmonary artery pressure (mPAP), pulmonary-to-systemic arterial pressure ratio (Pp/Ps), and the ratio between arterial oxygen partial pressure and inhaled oxygen concentration (PaO2/FiO2) were used to evaluate the efficacy of TRE treatment. A multiple linear regression model was used to explore the relevant factors associated with TRE blood concentration. Results A total of eight patients were enrolled in the investigation, with an age range of 2.5-9.9 years. The median stable dose of TRE was 70 ng/kg/min with a range of 55-75 ng/kg/min. The median subliminal dose was 55 ng/kg/min with a range of 25-75 ng/kg/min. A linear relationship was established between the TRE dose and the plasma concentration. TRE blood concentrations were associated with dose and patient height. After TRE treatment, mPAP, Pp/Ps, and PaO2/FiO2 were significantly improved (P<0.05). Conclusions A linear relationship was found between the blood concentration of TRE and its dose. IV TRE was an effective therapy without serious side effects in pediatric patients with FSV-PAH. Trial Registration ClinicalTrials.gov Identifier: NCT02865733.
Collapse
Affiliation(s)
- Xi Chen
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Man Cai
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Jie Zhang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Han Xu
- Department of Pharmacy, Clinical Research Center, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Li
- Department of Pharmacy, Clinical Research Center, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuo-Ming Xu
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
97
|
Isaak A, Luetkens JA, Faron A, Endler C, Mesropyan N, Katemann C, Zhang S, Kupczyk P, Kuetting D, Attenberger U, Dabir D. Free-breathing non-contrast flow-independent cardiovascular magnetic resonance angiography using cardiac gated, magnetization-prepared 3D Dixon method: assessment of thoracic vasculature in congenital heart disease. J Cardiovasc Magn Reson 2021; 23:91. [PMID: 34275486 PMCID: PMC8287681 DOI: 10.1186/s12968-021-00788-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To evaluate a non-contrast respiratory- and electrocardiogram-gated 3D cardiovascular magnetic resonance angiography (CMRA) based on magnetization-prepared Dixon method (relaxation-enhanced angiography without contrast and triggering, REACT) for the assessment of the thoracic vasculature in congenital heart disease (CHD) patients. METHODS 70 patients with CHD (mean 28 years, range: 10-65 years) were retrospectively identified in this single-center study. REACT-CMRA was applied with respiratory- and cardiac-gating. Image quality (IQ) of REACT-CMRA was compared to standard non-gated multi-phase first-pass-CMRA and respiratory- and electrocardiogram-gated steady-state-CMRA. IQ of different vessels of interest (ascending aorta, left pulmonary artery, left superior pulmonary vein, right coronary ostium, coronary sinus) was independently assessed by two readers on a five-point Likert scale. Measurements of vessel diameters were performed in predefined anatomic landmarks (ascending aorta, left pulmonary artery, left superior pulmonary vein). Both readers assessed artifacts and vascular abnormalities. Friedman test, chi-squared test, and Bland-Altman method were used for statistical analysis. RESULTS Overall IQ score of REACT-CMRA was higher compared to first-pass-CMRA (3.5 ± 0.4 vs. 2.7 ± 0.4, P < 0.001) and did not differ from steady-state-CMRA (3.5 ± 0.4 vs. 3.5 ± 0.6, P = 0.99). Non-diagnostic IQ of the defined vessels of interest was observed less frequently on REACT-CMRA (1.7 %) compared to steady-state- (4.3 %, P = 0.046) or first-pass-CMRA (20.9 %, P < 0.001). Close agreements in vessel diameter measurements were observed between REACT-CMRA and steady-state-CMRA (e.g. ascending aorta, bias: 0.38 ± 1.0 mm, 95 % limits of agreement (LOA): - 1.62-2.38 mm). REACT-CMRA showed high intra- (bias: 0.04 ± 1.0 mm, 95 % LOA: - 1.9-2.0 mm) and interobserver (bias: 0.20 ± 1.1 mm, 95 % LOA: - 2.0-2.4 mm) agreements regarding vessel diameter measurements. Fat-water separation artifacts were observed in 11/70 (16 %) patients on REACT-CMRA but did not limit diagnostic utility. Six vascular abnormalities were detected on REACT-CMRA that were not seen on standard contrast-enhanced CMRA. CONCLUSIONS Non-contrast-enhanced cardiac-gated REACT-CMRA offers a high diagnostic quality for assessment of the thoracic vasculature in CHD patients.
Collapse
Affiliation(s)
- Alexander Isaak
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany.
| | - Julian A Luetkens
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Anton Faron
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Christoph Endler
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Narine Mesropyan
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | | | | | - Patrick Kupczyk
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Daniel Kuetting
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Darius Dabir
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Quantitative Imaging Lab Bonn (QILaB), Bonn, Germany
| |
Collapse
|
98
|
Diab NS, Barish S, Dong W, Zhao S, Allington G, Yu X, Kahle KT, Brueckner M, Jin SC. Molecular Genetics and Complex Inheritance of Congenital Heart Disease. Genes (Basel) 2021; 12:1020. [PMID: 34209044 PMCID: PMC8307500 DOI: 10.3390/genes12071020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/09/2023] Open
Abstract
Congenital heart disease (CHD) is the most common congenital malformation and the leading cause of mortality therein. Genetic etiologies contribute to an estimated 90% of CHD cases, but so far, a molecular diagnosis remains unsolved in up to 55% of patients. Copy number variations and aneuploidy account for ~23% of cases overall, and high-throughput genomic technologies have revealed additional types of genetic variation in CHD. The first CHD risk genotypes identified through high-throughput sequencing were de novo mutations, many of which occur in chromatin modifying genes. Murine models of cardiogenesis further support the damaging nature of chromatin modifying CHD mutations. Transmitted mutations have also been identified through sequencing of population scale CHD cohorts, and many transmitted mutations are enriched in cilia genes and Notch or VEGF pathway genes. While we have come a long way in identifying the causes of CHD, more work is required to end the diagnostic odyssey for all CHD families. Complex genetic explanations of CHD are emerging but will require increasingly sophisticated analysis strategies applied to very large CHD cohorts before they can come to fruition in providing molecular diagnoses to genetically unsolved patients. In this review, we discuss the genetic architecture of CHD and biological pathways involved in its pathogenesis.
Collapse
Affiliation(s)
- Nicholas S. Diab
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
| | - Syndi Barish
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
| | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065, USA
| | - Shujuan Zhao
- Department of Genetics, School of Medicine, Washington University, St. Louis, MO 63110, USA; (S.Z.); (X.Y.)
| | - Garrett Allington
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Xiaobing Yu
- Department of Genetics, School of Medicine, Washington University, St. Louis, MO 63110, USA; (S.Z.); (X.Y.)
- Department of Computer Science & Engineering, Washington University, St. Louis, MO 63130, USA
| | - Kristopher T. Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA;
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Martina Brueckner
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sheng Chih Jin
- Department of Genetics, School of Medicine, Washington University, St. Louis, MO 63110, USA; (S.Z.); (X.Y.)
- Department of Pediatrics, School of Medicine, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
99
|
Perioperative GABA Blood Concentrations in Infants with Cyanotic and Non-Cyanotic Congenital Heart Diseases. Diagnostics (Basel) 2021; 11:diagnostics11071149. [PMID: 34202425 PMCID: PMC8304774 DOI: 10.3390/diagnostics11071149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022] Open
Abstract
Perioperative stress detection in children with congenital heart disease (CHD), particularly in the brain, is still limited. Among biomarkers, γ-amino-aminobutyric acid (GABA) assessment in biological fluids appears to be promising for its regulatory action on the cardiovascular and cerebral systems. We aimed to investigate cyanotic (C) or non-cyanotic (N) CHD children for GABA blood level changes in the perioperative period. We conducted an observational study in 68 CHD infants (C: n = 33; N: n = 35) who underwent perioperative clinical, standard laboratory and monitoring parameter recordings and GABA assessment. Blood samples were drawn at five predetermined time-points before, during and after surgery. No significant perioperative differences were observed between groups in clinical and laboratory parameters. In C, perioperative GABA levels were significantly lower than N. Arterial oxygen saturation and blood concentration significantly differed between C and N children and correlated at cardiopulmonary by-pass (CPB) time-point with GABA levels. The present data showing higher hypoxia/hyperoxia-mediated GABA concentrations in C children suggest that they are more prone to perioperative cardiovascular and brain stress/damage. The findings suggest the usefulness of further investigations to detect the “optimal” oxygen concentration target in order to avoid the side effects associated with re-oxygenation during CPB.
Collapse
|
100
|
Tesson S, Butow PN, Marshall K, Fonagy P, Kasparian NA. Parent-child bonding and attachment during pregnancy and early childhood following congenital heart disease diagnosis. Health Psychol Rev 2021; 16:378-411. [PMID: 33955329 DOI: 10.1080/17437199.2021.1927136] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Diagnosis and treatment of congenital heart disease (CHD) can present challenges to the developing parent-child relationship due to periods of infant hospitalization and intensive medical care, parent-infant separations, child neurodevelopmental delay and feeding problems, and significant parent and child distress and trauma. Yet, the ways in which CHD may affect the parent-child relationship are not well-understood. We systematically reviewed the evidence on parental bonding, parent-child interaction, and child attachment following CHD diagnosis, according to a pre-registered protocol (CRD42019135687). Six electronic databases were searched for English-language studies comparing a cardiac sample (i.e., expectant parents or parents and their child aged 0-5 years with CHD) with a healthy comparison group on relational outcomes. Of 22 unique studies, most used parent-report measures (73%) and yielded mixed results for parental bonding and parent-child interaction quality. Observational results also varied, although most studies (4 of 6) found difficulties in parent-child interaction on one or more affective or behavioural domains (e.g., lower maternal sensitivity, lower infant responsiveness). Research on parental-fetal bonding, father-child relationships, and child attachment behaviour was lacking. Stronger evidence is needed to determine the nature, prevalence, and predictors of relational disruptions following CHD diagnosis, and to inform targeted screening, prevention, and early intervention programs for at-risk dyads.
Collapse
Affiliation(s)
- Stephanie Tesson
- School of Psychology, The University of Sydney, Sydney, Australia.,Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, Australia
| | - Phyllis N Butow
- School of Psychology, The University of Sydney, Sydney, Australia.,Psycho-Oncology Co-operative Research Group (PoCoG), The University of Sydney, Sydney, Australia
| | - Kate Marshall
- Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, Australia.,Discipline of Paediatrics, School of Women's and Children's Health, UNSW Medicine, The University of New South Wales, Sydney, Australia
| | - Peter Fonagy
- Research Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Nadine A Kasparian
- Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, Australia.,Cincinnati Children's Center for Heart Disease and Mental Health, Heart Institute and the Division of Behavioral Medicine & Clinical Psychology, Cincinnati Children's Hospital, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|