1
|
Hitz MP, Dombrowsky G, Melnik N, Vey C. Current and future diagnostics of congenital heart disease (CHD). MED GENET-BERLIN 2025; 37:95-102. [PMID: 40207043 PMCID: PMC11976401 DOI: 10.1515/medgen-2025-2008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Congenital heart defects (CHD) are one of the most common anomalies found among live births and represent a complex multifactorial condition. Given that more than 90 % of cases survive due to improved early treatment options (e.g., catheter intervention, surgical procedure, and improved intensive care), genotype-informed patient follow-up should consider lifelong treatment considering different types of comorbidities. Unfortunately, a thorough genetic workup is only offered to a minority of CHD patients. However, a comprehensive understanding of the genetic underpinnings combined with in-depth phenotyping would strengthen our knowledge regarding the impact of environmental (e.g., pre-gestational diabetes) and genetic causes ranging from aneuploidies to single variants and more complex inheritance patterns on early heart development. Therefore, comprehensive genetic analysis in these patients is an essential way of predicting the prognosis and recurrence risk in families and ultimately improving patients' quality of life due to better therapeutic options. In this review, we examine the different types of variants and genes of different molecular genetics techniques to assess the diagnostic yield in different CHD sub-phenotypes. Given the complex inheritance pattern observed in CHD, we also consider possible future methods and frameworks to improve diagnostics and allow for better genotype-phenotype correlation in this patient group. Predicting recurrence risk and prognosis in CHD patients will ultimately allow for better treatment and lifelong therapeutic outcomes for CHD patients.
Collapse
Affiliation(s)
- Marc-Phillip Hitz
- Carl von Ossietzky UniversityInstitute of Medical GeneticsRahel-Straus-Str. 1026133OldenburgGermany
| | - Gregor Dombrowsky
- Carl von Ossietzky UniversityInstitute of Medical GeneticsRahel-Straus-Str. 1026133OldenburgGermany
| | - Nico Melnik
- Carl von Ossietzky UniversityInstitute of Medical GeneticsRahel-Straus-Str. 1026133OldenburgGermany
| | - Chiara Vey
- Carl von Ossietzky UniversityInstitute of Medical GeneticsRahel-Straus-Str. 1026133OldenburgGermany
| |
Collapse
|
2
|
Morrish AM, O'Malley BR, Hilton DCK, Webb AE, Bennetts B, Sholler GF, Smith J, Blue GM. Outcomes and experiences of genetic testing in children with congenital heart disease. J Med Genet 2025; 62:335-344. [PMID: 40011010 DOI: 10.1136/jmg-2024-110553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Following genomic advances, genetic testing options for paediatric patients with congenital heart disease (CHD) have evolved significantly. A single-site audit was conducted to assess testing outcomes and a survey created to explore family experiences and preferences. METHOD All genetic tests ordered in postcardiac surgery patients with CHD at The Children's Hospital at Westmead between January 2017 and December 2021 were reviewed. Diagnostic yield, clinical and demographic factors, and testing trends over time were evaluated. Surveys were sent to parents of children who had met a clinical geneticist (n=112). RESULTS Genetic testing was completed in 607 individuals (74 molecular testing; 533 cytogenetic testing only). The diagnostic rate was 36% and 9%, respectively. Use of molecular testing significantly increased over time (p=0.033), but yield did not (p=0.288). Molecular testing yield was high in neonates (64%), and patients with extracardiac anomalies (40%) or relevant family history (40%). Brain (p=0.022), haematological/cancer (p≤0.001), immune (p≤0.001), endocrine (p≤0.001) anomalies and intellectual disability (p=0.027) were associated with a diagnosis following cytogenetic testing. Short stature was significantly associated with diagnostic yield following molecular testing (p=0.012). Survey respondents (n=28) reported a positive experience (p=0.013) with minimal decisional regret (p=0.322). CONCLUSION Cytogenetic testing remains an important first-tier test in CHD. Furthermore, molecular testing guided by a clinical geneticist generates a high rate of genetic diagnoses. Parents of children with CHD value genetic testing with little regret.
Collapse
Affiliation(s)
- Ansley M Morrish
- Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Department of Clinical Genetics, The Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Bridget R O'Malley
- Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Desiree C K Hilton
- Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Annabel E Webb
- Cerebral Palsy Alliance Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Bruce Bennetts
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Molecular Genetics Lab, The Sydney Children's Hospital Network, Sydney, New South Wales, Australia
| | - Gary F Sholler
- Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Janine Smith
- Department of Clinical Genetics, The Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Gillian M Blue
- Heart Centre for Children, The Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
De Robertis V, Stampalija T, Abuhamad AZ, Bosco M, Chaoui R, Formigoni C, Moon-Grady AJ, Paladini D, Pilu G, Ramezzana IG, Rychik J, Volpe P. Indications for fetal echocardiography: consensus and controversies among evidence-based national and international guidelines. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2025. [PMID: 40208627 DOI: 10.1002/uog.29224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/27/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025]
Abstract
OBJECTIVE Fetal echocardiography (FE) is an indication-driven examination for pregnant women with a fetus at high risk for congenital heart disease (CHD). Several familial, maternal and fetal factors are reported to increase the risk of CHD. The aim of this study was to highlight the existing differences in recommended indications for FE among recently published guidelines and consensuses of experts. METHODS Guidelines and expert consensuses published from January 2008 to October 2023 were identified through a systematic literature search. FE guidelines and consensus statements were excluded if not written in the English language and if indications for FE were not reported. All familial, maternal and fetal risk factors for CHD reported in the consensuses and guidelines were listed and comparisons were made between documents. The agreement or disagreement for each risk factor between guidelines and consensuses was classified as: complete agreement (all analyzed documents reported the same indication); partial agreement (all documents considered a risk factor as an indication, but with inconsistency in its definition); or complete disagreement (inconsistency between documents for the considered risk factor as an indication). RESULTS Six guidelines and expert consensuses that met the inclusion criteria were identified. Overall, a total of 17 risk factors were identified as an indication for FE. Complete agreement was reached for 3/17 (17.6%) risk factors, all of which are fetal risk factors (suspected CHD at the anomaly scan, presence of major fetal extracardiac abnormality and non-immune hydrops fetalis). Partial agreement was recorded for 8/17 (47.1%) risk factors (family history of CHD, increased nuchal translucency, multiple gestation, maternal diabetes mellitus, maternal phenylketonuria, maternal infection, maternal autoimmune disease and autoantibody positivity, and teratogen exposure). Complete disagreement was recorded for 6/17 (35.3%) risk factors (inherited genetic disease associated with CHD, fetal genetic anomaly, suspected abnormality of heart rate or rhythm, first-trimester sonographic markers of CHD, abnormality of umbilical cord and venous system, and use of assisted reproductive technology). CONCLUSIONS Areas of controversy regarding which CHD risk factors warrant FE were greater in quantity than were the areas of consensus. An internationally standardized agreement would be valuable for physicians and guideline developers. For many risk factors, further evidence is needed to justify their use as an indication for FE. © 2025 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- V De Robertis
- Fetal Medicine Unit, Di Venere Hospital, Bari, Italy
| | - T Stampalija
- Unit of Fetal Medicine and Prenatal Diagnosis, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - A Z Abuhamad
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - M Bosco
- Unit of Obstetrics and Gynecology, Department of Surgery, Dentistry, Pediatrics, and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - R Chaoui
- Center of Prenatal Diagnosis and Human Genetics, Berlin, Germany
| | | | - A J Moon-Grady
- Division of Cardiology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - D Paladini
- Fetal Medicine and Surgery Unit, IRCCS Istituto G. Gaslini, Genoa, Italy
| | - G Pilu
- Obstetric Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - I G Ramezzana
- Prenatal Diagnosis and Fetal Surgery Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - J Rychik
- Fetal Heart Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - P Volpe
- Fetal Medicine Unit, Di Venere Hospital, Bari, Italy
| |
Collapse
|
4
|
Helm BM, Wetherill L, Landis BJ, Ware SM. Dysmorphology-Based Prediction Model for Genetic Disorders in Infants With Congenital Heart Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004895. [PMID: 40151936 PMCID: PMC11999770 DOI: 10.1161/circgen.124.004895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/06/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Genetic disorders are prevalent in patients with congenital heart disease (CHD), but genetic evaluations are underutilized and nonstandardized. We sought to quantify a dysmorphology score and develop phenotype-based prediction models for genetic diagnoses in CHD. METHODS We used a test-negative case-control study of inpatient infants (<1 year) with CHD undergoing standardized genetic evaluations. We quantified a novel dysmorphology score and combined it with other clinical variables used in multivariable logistic regression models to predict genetic diagnoses identified by genetic testing. RESULTS Of 1008 patients, 24.1% (243/1008) had genetic diagnoses identified. About half of the cohort were either nondysmorphic or mildly dysmorphic with dysmorphology scores ≤2. There were higher dysmorphology scores according to CHD class (P=0.0007), extracardiac anomaly-positive status (P<0.0001), female sex (P=0.05), and genetic diagnosis identified (P<0.0001). Multivariable logistic regression models quantified this effect further: each +1 increase in the dysmorphology score was associated with a 17% to 20% increased risk of genetic diagnoses (odds ratios, 1.17-1.20, P<0.0001). Extracardiac anomaly-positive status remained a stronger predictor of genetic diagnoses (odds ratios, 2.81-3.39). Nonetheless, about 10% of the cohort were minimally dysmorphic (dysmorphology scores ≤2), had isolated CHD, and were found to have genetic diagnoses, indicating that dysmorphology-based screening can be used to risk-stratify but not exclude genetic diagnoses. CONCLUSIONS The dysmorphology score is a novel screen for patients with CHD at high risk of having genetic diagnoses identified by genetic testing, including disorders not easily recognized by clinicians. We used these results to develop predicted probability plots for genetic diagnoses in patients with CHD.
Collapse
Affiliation(s)
- Benjamin M. Helm
- Dept of Medical & Molecular Genetics, Indiana University School of Medicine
- Dept. of Epidemiology, Indiana University Fairbanks School of Public Health, Indianapolis, IN
| | - Leah Wetherill
- Dept of Medical & Molecular Genetics, Indiana University School of Medicine
| | | | - Stephanie M. Ware
- Dept of Medical & Molecular Genetics, Indiana University School of Medicine
- Dept of Pediatrics, Indiana University School of Medicine
| |
Collapse
|
5
|
Park Y, Heo J, Kang D, Gwak GY. Impact of Maternal Hepatitis B Virus Infection on Congenital Heart Disease Risk in Offspring: A National Cohort Study. J Viral Hepat 2025; 32:e14036. [PMID: 39588801 DOI: 10.1111/jvh.14036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/24/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024]
Abstract
Maternal hepatitis B virus (HBV) infection influence both maternal and fetal health. Recent studies reported increased congenital anomalies in offspring of HBV-infected mothers. This study investigated whether maternal HBV infection was associated with higher risk of congenital heart disease (CHD) in children. With the Korean National Health Insurance Service (K-NHIS) database, this retrospective cohort study included live births from 2005 to 2019, born to women under 40. Propensity score matching with a 1:3 ratio was conducted to compare HBV-infected mother's children with HBV-uninfected mother's children while adjusting for various maternal and pregnancy-related factors. Logistic regression models were used to estimate the risk. Of 2,673,059 eligible participants, 263,904 children were born to HBV-infected mothers. Risk estimation in this group showed a modestly increased risk of CHD (OR = 1.05, 95% CI = 1.02, 1.09). Notably, when pregnant mothers were treated with antiviral medication, there was an indication of reduced CHD risk, although this result was not statistically significant. The highest risk of CHD was observed in children who were themselves infected with HBV. The study indicates an association between maternal HBV infection and an increased CHD risk in offspring. The findings suggest the need to re-evaluate the timing of antiviral treatment during pregnancy to align more closely with early stages of fetal cardiac development. Further research is needed to understand the biological mechanisms of this association and to redefine clinical guidelines for managing HBV infection in pregnancy.
Collapse
Affiliation(s)
- Yewan Park
- Department of Internal Medicine, Kyung Hee University Hospital, Seoul, South Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Jihye Heo
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Danbee Kang
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Geum-Youn Gwak
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
6
|
Silva Delgado LE, Vasquez Cueva ET, Silva Delgado K, Maguiña Quispe JL, Silva E. Long-term outcomes after pediatric cardiac surgery: A 20-year single-institution study. Perfusion 2025:2676591251328926. [PMID: 40131175 DOI: 10.1177/02676591251328926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
PurposeThis study evaluated the long-term outcomes and factors associated with survival in patients undergoing cardiac surgery with cardiopulmonary bypass at a children's cardiac center.MethodsA retrospective cohort study was conducted between 2001 and 2020 Outcomes included early or late survival and associated risk factors for survival.ResultsSurvival had a mean of 16.78 years (95% CI 16.50-17.06). The survival rates at 1, 5, 10, and 19 years were 91, 88, and 88%, respectively. Cox regression analysis indicated that age HR = 0.39, 95% CI [0.26-0.59], the RACHS-1 scale HR = 9.92, 95% CI [3.67-26.79], open chest HR = 1, 90, 95% CI [1.20-3.00], cardiac arrest HR = 3.91, 95% CI [2.40-6.36] and peritoneal dialysis HR = 6.71, 95% CI [4, 33-10.38], were independently associated with worse survival.Conclusionslong-term survival of children who undergo cardiac surgery at our single center was satisfactory. Age, higher RACHS score, delayed sternal closure, cardiac arrest, and peritoneal dialysis were strong predictors of adverse outcomes affecting early and long-term survival of pediatric cardiac surgery.
Collapse
Affiliation(s)
| | | | | | | | - Eduardo Silva
- Pediatric Cardiovascular Intensive Care Unit, Instituto Nacional de Salud del Niño (INSN), Lima, Perú
| |
Collapse
|
7
|
Penaloza JS, Moreland B, Gaither JB, Landis BJ, Ware SM, McBride KL, White P. Identification of Long Noncoding RNA Candidate Disease Genes Associated With Clinically Reported Copy Number Variants in Congenital Heart Disease. J Am Heart Assoc 2025; 14:e039177. [PMID: 40079339 DOI: 10.1161/jaha.124.039177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/04/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Copy number variants (CNVs) contribute to 3% to 10% of isolated congenital heart disease (CHD) cases, yet their pathogenic roles remain unclear. Diagnostic efforts have focused on protein-coding genes, largely overlooking long noncoding RNAs (lncRNAs), which play key roles in development and disease. METHODS AND RESULTS We systematically analyzed lncRNAs overlapping clinically validated CNVs in 743 patients with CHD from the Cytogenomics of Cardiovascular Malformations Consortium. We identified heart-expressed lncRNAs, constructed a gene regulatory network using weighted gene coexpression network analysis, and identified gene modules associated with heart development. Functional enrichment and network analyses were used to identify lncRNAs that may be involved in heart development and potentially contribute to CHD. The code is stably archived at https://doi.org/10.5281/zenodo.13799779. We identified 18 lncRNA candidate genes within modules significantly correlated with heart tissue, highlighting their potential involvement in CHD pathogenesis. Notably, lncRNAs such as lnc-STK32C-3, lnc-TBX20-1, and CRMA demonstrated strong associations with known CHD genes. Strikingly, although only 7.6% of known CHD genes were affected by a CNV, 68.8% of the CNVs contained a lncRNA expressed in the heart. CONCLUSIONS Using weighted gene coexpression network analysis, we identified CNV-associated lncRNAs with potential relevance to CHD, underscoring the complexities of noncoding regions in disease pathogenesis. These findings suggest that lncRNAs may play a greater role in CHD than previously recognized, highlighting the need for broader genomic analyses that extend beyond protein-coding genes. This study provides a foundation for further exploration of lncRNAs in CHD, with potential implications for improved genetic characterization and diagnosis.
Collapse
Affiliation(s)
- Jacqueline S Penaloza
- The Office of Data Sciences The Abigail Wexner Research Institute Nationwide Children's Hospital Columbus OH USA
- The Steve and Cindy Rasmussen Institute for Genomic Medicine The Abigail Wexner Research Institute Nationwide Children's Hospital Columbus OH USA
| | - Blythe Moreland
- The Steve and Cindy Rasmussen Institute for Genomic Medicine The Abigail Wexner Research Institute Nationwide Children's Hospital Columbus OH USA
| | - Jeffrey B Gaither
- The Office of Data Sciences The Abigail Wexner Research Institute Nationwide Children's Hospital Columbus OH USA
| | - Benjamin J Landis
- Department of Pediatrics Indiana University School of Medicine Indianapolis IN USA
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN USA
| | - Stephanie M Ware
- Department of Pediatrics Indiana University School of Medicine Indianapolis IN USA
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN USA
| | - Kim L McBride
- Department of Medical Genetics Cumming School of Medicine University of Calgary Calgary Canada
| | - Peter White
- The Office of Data Sciences The Abigail Wexner Research Institute Nationwide Children's Hospital Columbus OH USA
- The Steve and Cindy Rasmussen Institute for Genomic Medicine The Abigail Wexner Research Institute Nationwide Children's Hospital Columbus OH USA
- Department of Pediatrics The Ohio State University College of Medicine Columbus OH USA
| |
Collapse
|
8
|
Dong W, Jin SC, Sierant MC, Lu Z, Li B, Lu Q, Morton SU, Zhang J, López-Giráldez F, Nelson-Williams C, Knight JR, Zhao H, Cao J, Mane S, Gruber PJ, Lek M, Goldmuntz E, Deanfield J, Giardini A, Mital S, Russell M, Gaynor JW, Cnota JF, Wagner M, Srivastava D, Bernstein D, Porter GA, Newburger J, Roberts AE, Yandell M, Yost HJ, Tristani-Firouzi M, Kim R, Seidman J, Chung WK, Gelb BD, Seidman CE, Lifton RP, Brueckner M. Recessive genetic contribution to congenital heart disease in 5,424 probands. Proc Natl Acad Sci U S A 2025; 122:e2419992122. [PMID: 40030011 PMCID: PMC11912448 DOI: 10.1073/pnas.2419992122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/14/2025] [Indexed: 03/15/2025] Open
Abstract
Variants with large effect contribute to congenital heart disease (CHD). To date, recessive genotypes (RGs) have commonly been implicated through anecdotal ascertainment of consanguineous families and candidate gene-based analysis; the recessive contribution to the broad range of CHD phenotypes has been limited. We analyzed whole exome sequences of 5,424 CHD probands. Rare damaging RGs were estimated to contribute to at least 2.2% of CHD, with greater enrichment among laterality phenotypes (5.4%) versus other subsets (1.4%). Among 108 curated human recessive CHD genes, there were 66 RGs, with 54 in 11 genes with >1 RG, 12 genes with 1 RG, and 85 genes with zero. RGs were more prevalent among offspring of consanguineous union (4.7%, 32/675) than among nonconsanguineous probands (0.7%, 34/4749). Founder variants in GDF1 and PLD1 accounted for 74% of the contribution of RGs among 410 Ashkenazi Jewish probands. We identified genome-wide significant enrichment of RGs in C1orf127, encoding a likely secreted protein expressed in embryonic mouse notochord and associated with laterality defects. Single-cell transcriptomes from gastrulation-stage mouse embryos revealed enrichment of RGs in genes highly expressed in the cardiomyocyte lineage, including contractility-related genes MYH6, UNC45B, MYO18B, and MYBPC3 in probands with left-sided CHD, consistent with abnormal contractile function contributing to these malformations. Genes with significant RG burden account for 1.3% of probands, more than half the inferred total. These results reveal the recessive contribution to CHD, and indicate that many genes remain to be discovered, with each likely accounting for a very small fraction of the total.
Collapse
Grants
- U01 HL128711 NHLBI NIH HHS
- RM1HG011014 HHS | NIH | National Human Genome Research Institute (NHGRI)
- Howard Hughes Medical Institute Howard Hughes Medical Institute (HHMI)
- U01 HL098162 NHLBI NIH HHS
- UL1 TR000003 NCATS NIH HHS
- 1UG1HL135680-01 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- CDI-FR-2021-926 Children's Discovery Institute (CDI)
- UO1-HL128711 HHS | NIH | NHLBI | Division of Intramural Research (DIR)
- UG1 HL135680 NHLBI NIH HHS
- UO1-HL098147 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R03 HD100883 NICHD NIH HHS
- RM1 HG011014 NHGRI NIH HHS
- U01 HL098153 NHLBI NIH HHS
- U01 HL131003 NHLBI NIH HHS
- 5U54HG006504 HHS | NIH | National Human Genome Research Institute (NHGRI)
- UO1-HL098162 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U01 HL153009 NHLBI NIH HHS
- R00 HL143036 NHLBI NIH HHS
- HL157653 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL157653 HHS | NIH | NHLBI | Division of Intramural Research (DIR)
- U54 HG006504 NHGRI NIH HHS
- R00HL143036-02 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 19PRE34380842 American Heart Association (AHA)
- CTSA1405 Hydrocephalus Association (HA)
- UO1 HL131003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- UO1-HL153009 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08 HL157653 NHLBI NIH HHS
- U01 HL098147 NHLBI NIH HHS
Collapse
Affiliation(s)
- Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065
| | - Sheng Chih Jin
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael C Sierant
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065
| | - Ziyu Lu
- Laboratory of Single-cell Genomics and Population Dynamics, The Rockefeller University, New York, NY 10065
| | - Boyang Li
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510
| | - Qiongshi Lu
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, WI 53706
| | - Sarah U Morton
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115
- Broad Institute of Massachusetts Institute of Technology and Harvard, Boston, MA 02142
| | - Junhui Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
| | | | | | - James R Knight
- Yale Center for Genome Analysis, Yale University, New Haven, CT 06516
| | - Hongyu Zhao
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510
| | - Junyue Cao
- Laboratory of Single-cell Genomics and Population Dynamics, The Rockefeller University, New York, NY 10065
| | - Shrikant Mane
- Yale Center for Genome Analysis, Yale University, New Haven, CT 06516
| | - Peter J Gruber
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06510
| | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
| | - Elizabeth Goldmuntz
- Division of Cardiology, Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104
| | - John Deanfield
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, United Kingdom
| | - Alessandro Giardini
- Pediatric Cardiology, Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
| | - Seema Mital
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G1X8, Canada
| | - Mark Russell
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - J William Gaynor
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - James F Cnota
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Michael Wagner
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Daniel Bernstein
- Department of Pediatrics, Cardiology, Stanford University, Stanford, CA 94304
| | - George A Porter
- Department of Pediatrics, The School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY 14642
| | - Jane Newburger
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115
| | - Amy E Roberts
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115
| | - Mark Yandell
- Department of Human Genetics, University of Utah and School of Medicine, Salt Lake City, UT 84112
| | - H Joseph Yost
- Department of Human Genetics, University of Utah and School of Medicine, Salt Lake City, UT 84112
- The Catholic University of America, Washington, DC 20064
| | | | - Richard Kim
- Pediatric Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY 10032
- Department of Medicine, Columbia University Medical Center, New York, NY 10032
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Bruce D Gelb
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Christine E Seidman
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115
- HHMI, Chevy Chase, MD 20815
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065
| | - Martina Brueckner
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
- Department of Pediatrics, Section of Cardiology, Yale School of Medicine, New Haven, CT 06510
| |
Collapse
|
9
|
Kars ME, Stein D, Stenson PD, Cooper DN, Chung WK, Gruber PJ, Seidman CE, Shen Y, Tristani-Firouzi M, Gelb BD, Itan Y. Deciphering the digenic architecture of congenital heart disease using trio exome sequencing data. Am J Hum Genet 2025; 112:583-598. [PMID: 39983722 PMCID: PMC11947165 DOI: 10.1016/j.ajhg.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
Congenital heart disease (CHD) is the most common congenital anomaly and a leading cause of infant morbidity and mortality. Despite extensive exploration of the monogenic causes of CHD over the last decades, ∼55% of cases still lack a molecular diagnosis. Investigating digenic interactions, the simplest form of oligogenic interactions, using high-throughput sequencing data can elucidate additional genetic factors contributing to the disease. Here, we conducted a comprehensive analysis of digenic interactions in CHD by utilizing a large CHD trio exome sequencing cohort, comprising 3,910 CHD and 3,644 control trios. We extracted pairs of presumably deleterious rare variants observed in CHD-affected and unaffected children but not in a single parent. Burden testing of gene pairs derived from these variant pairs revealed 29 nominally significant gene pairs. These gene pairs showed a significant enrichment for known CHD genes (p < 1.0 × 10-4) and exhibited a shorter average biological distance to known CHD genes than expected by chance (p = 3.0 × 10-4). Utilizing three complementary biological relatedness approaches including network analyses, biological distance calculations, and candidate gene prioritization methods, we prioritized 10 final gene pairs that are likely to underlie CHD. Analysis of bulk RNA-sequencing data showed that these genes are highly expressed in the developing embryonic heart (p < 1 × 10-4). In conclusion, our findings suggest the potential role of digenic interactions in CHD pathogenesis and provide insights into unresolved molecular diagnoses. We suggest that the application of the digenic approach to additional disease cohorts will significantly enhance genetic discovery rates.
Collapse
Affiliation(s)
- Meltem Ece Kars
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David Stein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter D Stenson
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter J Gruber
- Department of Surgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard University, Boston, MA 02115, USA
| | - Yufeng Shen
- Departments of Systems Biology and Biomedical Informatics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Martin Tristani-Firouzi
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84113, USA
| | - Bruce D Gelb
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
10
|
Arogbokun Knutson OC, Luben TJ, Stingone JA, Engel LS, Martin CL, Olshan AF. Racial disparities in maternal exposure to ambient air pollution during pregnancy and prevalence of congenital heart defects. Am J Epidemiol 2025; 194:709-721. [PMID: 39108168 PMCID: PMC11955996 DOI: 10.1093/aje/kwae253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/05/2024] [Accepted: 07/31/2024] [Indexed: 03/06/2025] Open
Abstract
Air pollution may be a potential cause of congenital heart defects (CHDs), but racial disparities in this association are unexplored. We conducted a statewide population-based cohort study using North Carolina birth data from 2003 to 2015 (n = 1 225 285) to investigate the relationship between air pollution and CHDs (specifically pulmonary valve atresia/stenosis, tetralogy of Fallot [TOF], and atrioventricular septal defect [AVSD]). Maternal exposure to particulate matter ≤ 2.5 μm in diameter (PM2.5) and ozone during weeks 3 to 9 of pregnancy were estimated using the Environmental Protection Agency's Downscaler Model. Single- and co-pollutant log-binomial models were created for the entire population and stratified by race to investigate disparities. Positive associations between PM2.5 and CHDs were observed. An increasing concentration-response association was found for PM2.5 and TOF in adjusted, co-pollutant models (quartile 4 prevalence ratio: 1.46; 95% CI, 1.06-2.03). Differences in the effect of PM2.5 on CHD prevalence were seen in some models stratified by race, although clear exposure-prevalence gradients were not evident. Positive associations were also seen in adjusted, co-pollutant models of ozone and AVSD. Study results suggest that prenatal PM2.5 and ozone exposure may increase the prevalence of certain CHDs. A consistent pattern of differences in association by race/ethnicity was not apparent. This article is part of a Special Collection on Environmental Epidemiology.
Collapse
Affiliation(s)
- Olufunmilayo C Arogbokun Knutson
- Department of Health and Exercise Science, Morrison Family College of Health, University of St. Thomas, St. Paul, MN, United States
| | - Thomas J Luben
- United States Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, United States
| | - Jeanette A Stingone
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Lawrence S Engel
- Department of Epidemiology, Gillings School of Global Public Health University of North Carolina, Chapel Hill, NC, United States
| | - Chantel L Martin
- Department of Epidemiology, Gillings School of Global Public Health University of North Carolina, Chapel Hill, NC, United States
| | - Andrew F Olshan
- Department of Epidemiology, Gillings School of Global Public Health University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
11
|
Saijo N, Yaoita H, Takayama J, Ota C, Kawai E, Kimura M, Ozawa A, Tamiya G, Kure S, Kikuchi A. A Prevalent TMEM260 Deletion Causes Conotruncal Heart Defects, Including Truncus Arteriosus. Am J Med Genet A 2025; 197:e63906. [PMID: 39425509 DOI: 10.1002/ajmg.a.63906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024]
Abstract
Conotruncal heart defects are severe congenital malformations of the outflow tract, including truncus arteriosus (TA) and double-outlet right ventricle (DORV). TA is a severe congenital heart disease (CHD) in which the main arterial outflow tract of the heart fails to separate. We recently reported TMEM260 (NM_017799.4), c.1617del (p.Trp539Cysfs*9), as a major cause of TA in the Japanese population (TMEM260 Keio-Tohoku variant) comparable to the prevalence of the 22q11.2 deletion syndrome, which accounts for 12%-35% of TA. However, no other major causes of TA have not been identified. Here, we report a family that included a TA patient and a DORV patient, harboring the compound heterozygous variants of TMEM260, a 7066-bp deletion encompassing exons 6-7 and c.1393C > T, p.(Gln465*). The allele frequency of the 7066-bp deletion was particularly high in the Japanese population (0.17%). Based on the allele frequency of this deletion and c.1617del (0.36%) in the Japanese population, TMEM260 variants might be associated with more than half of the Japanese patients with TA. This study showed that TMEM260 pathogenic variants might be the most common cause of TA in the Japanese population and could explain the wide spectrum of phenotypes associated with TMEM260-related CHD, including DORV, demonstrating the usefulness of genetic testing in Japanese patients with TA.
Collapse
Affiliation(s)
- Naoya Saijo
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisao Yaoita
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Takayama
- Department of AI and Innovative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chiharu Ota
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Eiichiro Kawai
- Department of Pediatric Cardiology, Miyagi Children's Hospital, Sendai, Japan
| | - Masato Kimura
- Department of Pediatrics, MHO Sendai Medical Center, Sendai, Japan
| | - Akira Ozawa
- Department of Pediatric Cardiology, Miyagi Children's Hospital, Sendai, Japan
| | - Gen Tamiya
- Department of AI and Innovative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Miyagi Children's Hospital, Sendai, Japan
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
12
|
Mansoor W, Heidari MM, Khatami M, Hadadzadeh M, Tabrizi F, Darvand Araghi MH. Rare Pathogenic NR2F2 (COUP-TFII) Variants as Potential Etiological Causes in Pediatric Patients with Congenital Heart Diseases (CHDs). Hellenic J Cardiol 2025:S1109-9666(25)00050-8. [PMID: 40015456 DOI: 10.1016/j.hjc.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/07/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Congenital heart diseases (CHDs) are complex genetic disorders, and their genetic basis is not yet fully understood. Nuclear receptor subfamily 2 group F member 2 (NR2F2 or COUP-TFII) encodes a transcription factor which is expressed at high levels during mammalian development. Few studies have identified heterozygous and rare variants in the NR2F2 gene in individuals with congenital heart disease (CHD). OBJECTIVES This study aimed to evaluate the association between pathogenic genetic alterations in NR2F2 with CHD risk. METHODS A case-control study was conducted on a group of 135 patients (83 boys and 52 girls) with non-hereditary various types of isolated congenital heart disease who were undergoing open-heart surgery. Additionally, 95 matched healthy children without syndromic or isolated heart abnormalities were selected. RESULTS Using the Sanger sequencing method, we identified five heterozygous single nucleotide variations in exons two and three of the NR2F2 gene. These variations were novel and not present in any genomic variation databases. Four of the variations were missense mutations (p.Pro159Arg, p.Ser329Phe, p.Qln338Pro, and p.Tyr348Ser) and one was a synonymous variant (p.G361=) in the coding region. Importantly, in-silico results indicated that the missense variants had pathogenic effects on protein function. Additionally, the missense variants substantially altered the predicted structure of COUP-TFII. CONCLUSION The results we obtained not only validate the correlation between NR2F2 mutations and CHDs but also have significant potential for guiding new preventive and therapeutic strategies. This could contribute to the advancement of medical interventions in the fields of cardiology and genetics.
Collapse
Affiliation(s)
| | | | | | - Mehdi Hadadzadeh
- Department of Cardiac Surgery, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | |
Collapse
|
13
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
14
|
Garand M, Huang SSY, Dineen B, Glass IA, Eghtesady P. Differential Regulation of Immune-Related Genes in the Developing Heart. Pediatr Cardiol 2025; 46:442-457. [PMID: 38480572 DOI: 10.1007/s00246-024-03441-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/02/2024] [Indexed: 02/02/2025]
Abstract
In many congenital heart defects, it can be difficult to ascertain primary pathology from secondary consequences from altered flow through the developing heart. The molecular differences between the growing right and left ventricles (RV and LV, respectively) following the completion of septation and the impact of sex on these mechanisms have not been investigated. We analyzed RNA-seq data derived from twelve RV and LVs, one with Hypoplastic Left Heart Syndrome (HLHS), to compare the transcriptomic landscape between the ventricles during development. Differential gene expression analysis revealed a large proportion of genes unique to either the RV or LV as well as sex bias. Our GO enrichment and network analysis strategy highlighted the differential role of immune functions between the RV and LV in the developing heart. Comparatively, RNA-seq analysis of data from C57Bl6/J mice hearts collected at E14 resulted in the enrichment of similar processes related to T cells and leukocyte migration and activation. Differential gene expression analysis of an HLHS case highlighted significant downregulation of chromatin organization pathways and upregulation of genes involved in muscle organ development. This analysis also identified previously unreported upregulation of genes involved in IL-17 production pathways. In conclusion, differences exist between the gene expression profiles of RV versus LV with the expression of immune-related genes being significantly different between these two chambers. The pathogenesis of HLHS may involve alterations in the expression of chromatin and muscle gene organization as well as upregulation of the IL-17 response pathway.
Collapse
Affiliation(s)
- Mathieu Garand
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Susie S Y Huang
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Dineen
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Ian A Glass
- Department of Pediatrics and Medicine, University of Washington, Seattle, WA, USA
| | - Pirooz Eghtesady
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
15
|
Al-Korashy M, Binomar H, Al-Mostafa A, Al-Mogarri I, Al-Oufi S, Al-Admawi M, Al-Jufan M, Echahidi N, Mokeem A, Alfares A, Ramzan K, Tulbah S, Al-Qahtani A, Takroni S, Maddirevula S, Al-Hassnan Z. Genetic Analysis of Heterotaxy in a Consanguineous Cohort. Clin Genet 2025; 107:224-230. [PMID: 39513328 DOI: 10.1111/cge.14641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/20/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
Heterotaxy (HTX) is a group of clinical conditions with a shared pathology of dislocation of one or more organs along the left-right axis. The etiology of HTX is tremendously heterogeneous spanning environmental factors, chromosomal aberrations, mono/oligogenic variants, and complex inheritance. However, in the vast majority of cases, the etiology of HTX remains elusive. Here, we sought to describe the yield of genetic analysis and spectrum of variants in HTX in our highly consanguineous population. Twenty-four affected individuals, from 19 unrelated families, were consecutively recruited. Genetic analysis, with exome sequencing, genome sequencing, or multigene panel, detected 9 unique variants, 7 of which were novel, in 8 genes known to be implicated in autosomal recessive form of HTX (C1orf127, CCDC39, CIROP, DNAAF3, DNAH5, DNAH9, MMP21, and MNS1) providing a yield of 42.1%. Of note, 7 of the 9 variants were homozygous, while 2 were inherited in compound heterozygosity, including a heterozygous CNV deletion. A search for candidate genes in negative cases did not reveal a plausible variant. Our work demonstrates a relatively high yield of genetic testing in HTX in a consanguineous population with an enrichment of homozygous variants. The significant genetic heterogeneity observed herewith underscores the complex developmental mechanisms implicated in the pathogenesis of HTX and supports adopting a genome-wide analysis in the diagnostic evaluation of HTX.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Amal Mokeem
- Neuroscience Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | | | | | - Sahar Tulbah
- Center for Genomic Medicine, Riyadh, Saudi Arabia
| | | | - Saud Takroni
- Center for Genomic Medicine, Riyadh, Saudi Arabia
| | | | | |
Collapse
|
16
|
Stark CM, Hughes BN, Schacht JP, Urbina TM. Decoding Hearts: Genetic Insights and Clinical Strategies in Congenital Heart Disease. Neoreviews 2025; 26:e73-e88. [PMID: 39889766 DOI: 10.1542/neo.26-2-010] [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: 07/01/2024] [Accepted: 10/02/2024] [Indexed: 02/03/2025]
Abstract
Structural congenital heart disease (CHD) represents a heterogeneous group of cardiac anomalies of variable embryologic and molecular origins. A basic understanding of the genetics implicated in nonsyndromic (isolated) and syndromic structural CHD can better inform management decisions and family counseling. When a fetus or neonate develops CHD as a result of a genetic cause, it can be due to a mutation or a monogenic, oligogenic, or polygenic pathogenic variant. In this review, we summarize basic cardiac embryology in the context of genetic signaling pathways and proteins that are commonly implicated in syndromic and nonsyndromic structural CHD. We also provide an overview of the basic genetic evaluation in infants with common syndromic structural CHD.
Collapse
Affiliation(s)
- Christopher M Stark
- Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Brian N Hughes
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - John Paul Schacht
- Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Theresa M Urbina
- Department of Pediatrics, Madigan Army Medical Center, Tacoma, Washington
| |
Collapse
|
17
|
Watanabe D, Hasebe Y, Yagasaki H, Nakato D, Yamada M, Suzuki H, Kono Y, Sunaga Y, Yoshizawa M, Narusawa H, Miya F, Takenouchi T, Inukai T, Kosaki K. Brain calcification in congenital heart defects and ectodermal dysplasia (CHDED). Eur J Med Genet 2025; 73:104992. [PMID: 39740729 DOI: 10.1016/j.ejmg.2024.104992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/02/2025]
Abstract
Congenital Heart Defect and Ectodermal Dysplasia (CHDED) is an autosomal dominant disorder caused by the PRKD1 gene. CHDED is characterized by heart defects and ectodermal dysplasia. To date, eight patients with CHDED have been described. Calcifications were present in three patients with CHDED. (two patients; renal calcifications, one patient; brain calcifications). The organ distribution of calcifications in CHDED has been unclear. We report here another patient with CHDED and brain calcifications. The patient was a 9-month-old Japanese girl. She presented with heart defects and ectodermal dysplasia. At 6 months of age, she had generalized seizures, and a CT scan revealed calcifications in the bilateral deep cerebral white matter. The seizures resolved with the administration of levetiracetam. The patient had a de novo, heterozygous pathogenic variant, c.1808G > A, p.(Arg603His), in the PRKD1 gene. Together with the previously reported patients mentioned above, we demonstrated the role of the PRKD1 variant in brain calcification. We propose that PRKD1 and two genes, ITGB2 and JAM2, which are known to be associated with brain calcification, act through a common signaling pathway abnormality. In support of our hypothesis, there are some experimental results that link PRKD1 and JAM2. PRKD1 functions with the integrin ITGB2 as a partner. JAM2, which is associated with brain calcification and is critical for maintaining of the tight junction of the endothelial cells, interacts with integrins including ITGB2. Therefore, PRKD1 could lead to the pathological phenotype of brain calcification.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan; Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Yohei Hasebe
- Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Hideaki Yagasaki
- Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Daisuke Nakato
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Mamiko Yamada
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Hisato Suzuki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Yosuke Kono
- Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Yuto Sunaga
- Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Masashi Yoshizawa
- Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Hiromune Narusawa
- Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Fuyuki Miya
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Toshiki Takenouchi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Inukai
- Department of Pediatrics, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
18
|
Zhouguo Y, Yuan Z, Abdul M, Xi S, Wei T, Yan W, Wang Y, Guo R, Xing Q, Zhou Q. CBP/CREB Regulates the Proliferation and Apoptosis of Cardiomyocytes by Interacting With SERCA. J Cell Mol Med 2025; 29:e70426. [PMID: 39969158 PMCID: PMC11837278 DOI: 10.1111/jcmm.70426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Tetralogy of Fallot (TOF) is a common congenital heart disease. In this study, we proposed that cAMP response element-binding protein (CREB)-binding protein (CBP) regulates the proliferation and apoptosis in TOF by interacting with the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA). To confirm this, we collected right ventricle tissue samples from TOF patients during surgery to correct the deformity and from the donors. We performed IHC, IF, RT-qPCR, WB and ChIP experiments. The analysis of these experiments shows that the expression of CBP is higher in TOF patients than in healthy individuals. Further, the RT-qPCR results indicated that the CBP and SERCA mRNA in TOF patients were significantly higher than in the healthy donors. Similarly, WB results suggested that the expression of CBP and SERCA was predominantly elevated in TOF patients compared to healthy individuals. Further, the AC16 cell line with CBP knockdown reveals high expression of the Edu compared to normal cells, and the percentage of the cell cycle in the M phase was elevated in the CBPi group. In addition, the CCK-8 cell viability assay showed more proliferation in the CBPi group than in the control group at different time points. Moreover, the RT-qPCR results indicated a lower expression of SERCA after the knockdown of CBP and CREB. Finally, the ChIP assay shows that CREB binds to the promoter of SERCA, and the CBP enrichment decreased after the CREB knockdown. In conclusion, these results suggest that CBP interacts with SERCA to regulate cell proliferation and apoptosis during heart development and that up-regulation of CBP leads to TOF.
Collapse
Affiliation(s)
| | | | - Mannan Abdul
- Department of Anesthesiology, EENT Hospital of Fudan UniversityShanghaiChina
| | - Shun Xi
- Xuzhou Medical UniversityXuzhouChina
| | - Tao Wei
- Xuzhou Medical UniversityXuzhouChina
| | - Wei Yan
- Xuzhou Medical UniversityXuzhouChina
| | | | - Rui Guo
- Heze Medical CollegeHezeChina
| | - Quansheng Xing
- Women & Children's HospitalQingdao UniversityQingdaoChina
| | - Qing Zhou
- The Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| |
Collapse
|
19
|
Yi K, He SE, Guo T, Wang ZQ, Zhang X, Xu JG, Zhang HY, Liu WG, You T. Association of MTHFD1 G1958A (rs2236225) gene polymorphism with the risk of congenital heart disease: a systematic review and meta-analysis. BMC Med Genomics 2025; 18:20. [PMID: 39871280 PMCID: PMC11771109 DOI: 10.1186/s12920-024-02052-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 11/19/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND We did this study to better clarify the correlations of methylenetetrahydrofolate dehydrogenase 1 (MTHFD1)-G1958A (rs2236225) gene polymorphism with the risk of congenital heart diseases (CHD) and its subgroups. METHODS Relevant articles were searched in PubMed, Web of Science, Cochrane Library, Embase, CNKI, VIP database and Wanfang DATA until October 2023. We will use odds ratios (ORs) and 95% confidence intervals (CIs) to examine the potential associations of MTHFD1- G1958A gene polymorphism with CHD and its subgroups. RESULTS We included a total of 9 eligible studies, encompassing 1917 children with CHD, 1863 healthy children, 1717 mothers of the children with CHD and 1666 mothers of healthy children. In our study, the meta-analysis of fetal group revealed no significant association between any of the five genetic models for the MTHFD1-G1958A polymorphism and the risk of CHD. Subgroup analysis showed that associations between the MTHFD1-G1958A polymorphism and Tetralogy of Fallot (TOF) risk in the homozygote model (AA vs. GG, OR = 2.82, 95%CI [1.16, 6.86], P = 0.02) and recessive model (AA vs. GG + GA, OR = 3.09, 95%CI [1.36, 7.03], P = 0.007). In addition, the MTHFD1-G1958A polymorphism was associated with the risk of CHD in racial subgroup, increasing the risk of CHD in Caucasians. In maternal analysis, 2 genetic models of MTHFD1-G1958A polymorphism increased the risk of CHD: the heterozygote model (GA vs. GG, OR = 1.22, 95%CI [1.04, 1.42], P = 0.01), and the dominance model (GA + AA vs. GG, OR = 1.17, 95%CI [1.01, 1.34], P = 0.03). CONCLUSIONS The fetal MTHFD1-G1958A (rs2236225) gene polymorphism increase their risk of TOF. The maternal MTHFD1-G1958A polymorphism has a strong correlation with the risk of CHD, and there are racial differences in this correlation. Compared with GG genotype, the GA genotype increases the risk of CHD.
Collapse
Affiliation(s)
- Kang Yi
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Donggang West Road, Lanzhou City, Gansu Province, 730000, China.
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China.
- Structural Heart Center, Gansu Provincial Hospital, Lanzhou, China.
| | - Shao-E He
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
- The First Hospital of Lanzhou University, Lanzhou, China
- Lianlu Township Health Center, Kangle County, Linxia, Gansu, China
| | - Tao Guo
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Donggang West Road, Lanzhou City, Gansu Province, 730000, China
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Lanzhou, China
| | - Zi-Qiang Wang
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Donggang West Road, Lanzhou City, Gansu Province, 730000, China
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin Zhang
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
- Department of Cardiovascular Surgery, the First Hospital of Longnan City, Longnan, China
| | - Jian-Guo Xu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hao-Yue Zhang
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
- The First School of Clinical Medicine of Lanzhou University, Lanzhou, China
| | - Wei-Guo Liu
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
- Wuwei Liangzhou Hospital, Wuwei, Gansu, China
| | - Tao You
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Donggang West Road, Lanzhou City, Gansu Province, 730000, China
- Gansu International Scientific and Technological Cooperation Base of Diagnosis and Treatment of Congenital Heart Disease, Lanzhou, China
- Structural Heart Center, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
20
|
Yang H, Li Y, Chen Q, Li S, Yang Y, Lyu G. Analyzing exosomal miRNA profiles in tetralogy of fallot fetuses' amniotic fluid. Sci Rep 2025; 15:96. [PMID: 39747948 PMCID: PMC11696619 DOI: 10.1038/s41598-024-83576-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Amniotic fluid (AF)-derived exosomal miRNA have been explored as potential contributors to the pathogenesis of Tetralogy of Fallot (TOF). This study aimed to investigate the expression profiles of AF-derived exosomal miRNAs and their potential contribution to TOF development. Exosomes were isolated from AF samples obtained from pregnant women carrying fetuses diagnosed with TOF. AF-derived exosomal miRNAs expression profiles were generated using the Agilent human miRNA Array V21.0, comparing 5 TOF samples with 5 healthy controls. Differential expression analysis identified 257 significantly dysregulated miRNAs in the TOF group. KEGG pathway enrichment analysis revealed that the predicted targets of these differentially expressed miRNAs were enriched in pathways associated with congenital disorders. Notably, 25 of these miRNAs were previously reported to be regulated by both Notch and Wnt signaling pathways, which are critical to heart development. Further investigation using mouse embryonal carcinoma P19 cells revealed that miR-10a-5p overexpression inhibited cardiomyogenic differentiation, as evidenced by the suppression of cardiomyocyte marker genes like TBX5. A dual-luciferase reporter assay confirmed TBX5 as a direct target of miR-10a-5p, suggesting a regulatory mechanism involving their interaction. In summary, our study demonstrates that miR-10a-5p may contribute to the pathogenesis of TOF by impairing cardiomyocyte differentiation through direct targeting of TBX5. These findings enhance our understanding of TOF and its molecular underpinnings.
Collapse
Affiliation(s)
- Hainan Yang
- Department of Ultrasound, First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Yadan Li
- Department of Obstetrics and Gynecology, Quanzhou Women's and Children's Hospital, Quanzhou, 362000, Fujian, China
| | - Qiuyue Chen
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Shangqing Li
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Yiru Yang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Guorong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China.
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology of Education Ministry, Quanzhou Medical College, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
21
|
Zhang K, Zhong J. Bio inspired technological performance in color Doppler ultrasonography and echocardiography for enhanced diagnostic precision in fetal congenital heart disease. SLAS Technol 2024; 29:100207. [PMID: 39396732 DOI: 10.1016/j.slast.2024.100207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/02/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
The aim of this experiment is to investigate the bioinspired diagnostic performance of color Doppler ultrasound (CDUS) and two-dimensional (2D) echocardiography (ECG) for fetal congenital heart disease (FCHD). The research subjects were 33 expectant mothers with a diagnosis of FCHD at Xiangyang No. 1 People's Hospital between January 2017 and January 2021. The accuracy, sensitivity, and specificity of the two detection techniques were computed to ascertain and compare the diagnostic efficiency after CDUS and ECG examinations of all pregnant women. According to the findings, the prenatal CDUS detection rate was 92.61% higher than the 2D ECG detection rate (64.32%). The CDUS had an accuracy of 93.94%, sensitivity of 93.55%, and specificity of 100%, detecting 29 true positives, 0 false positives, 2 false negatives, and 2 true negatives. At 84.85% accuracy, 88.89% sensitivity, and 80% specificity, the 2D ECG identified 16 true positives, 3 false positives, 2 false positives, and 12 true negatives. There was a statistically significant (P < 0.05) difference between the accuracy, sensitivity, and specificity of 2D ECG and CDUS. In summary, CDUS was more effective than 2D ECG in diagnosing prenatal FCHD, and it also had a lower rate of missed and incorrect diagnoses.
Collapse
Affiliation(s)
- Kang Zhang
- Department of Ultrasound and Imaging, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, China
| | - Jing Zhong
- Department of Ultrasound and Imaging, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, China.
| |
Collapse
|
22
|
Sarić N, Atak Z, Sade CF, Reddy N, Bell G, Tolete C, Rajtboriraks MT, Hashimoto-Torii K, Jevtović-Todorović V, Haydar TF, Ishibashi N. Ciliopathy interacts with neonatal anesthesia to cause non-apoptotic caspase-mediated motor deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.624302. [PMID: 39651246 PMCID: PMC11623571 DOI: 10.1101/2024.11.27.624302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Increasing evidence suggests that anesthesia may induce developmental neurotoxicity, yet the influence of genetic predispositions associated with congenital anomalies on this toxicity remains largely unknown. Children with congenital heart disease often exhibit mutations in cilia-related genes and ciliary dysfunction, requiring sedation for their catheter or surgical interventions during the neonatal period. Here we demonstrate that briefly exposing ciliopathic neonatal mice to ketamine causes motor skill impairments, which are associated with a baseline deficit in neocortical layer V neuron apical spine density and their altered dynamics during motor learning.. These neuromorphological changes were linked to augmented non-apoptotic neuronal caspase activation. Neonatal caspase suppression rescued the spine density and motor deficits, confirming the requirement for sublethal caspase signaling in appropriate spine formation and motor learning. Our findings suggest that ciliopathy interacts with ketamine to induce motor impairments, which is reversible through caspase inhibition. Furthermore, they underscore the potential for ketamine- induced sublethal caspase responses in shaping neurodevelopmental outcomes.
Collapse
|
23
|
Ma XR, Conley SD, Kosicki M, Bredikhin D, Cui R, Tran S, Sheth MU, Qiu WL, Chen S, Kundu S, Kang HY, Amgalan D, Munger CJ, Duan L, Dang K, Rubio OM, Kany S, Zamirpour S, DePaolo J, Padmanabhan A, Olgin J, Damrauer S, Andersson R, Gu M, Priest JR, Quertermous T, Qiu X, Rabinovitch M, Visel A, Pennacchio L, Kundaje A, Glass IA, Gifford CA, Pirruccello JP, Goodyer WR, Engreitz JM. Molecular convergence of risk variants for congenital heart defects leveraging a regulatory map of the human fetal heart. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.20.24317557. [PMID: 39606363 PMCID: PMC11601760 DOI: 10.1101/2024.11.20.24317557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Congenital heart defects (CHD) arise in part due to inherited genetic variants that alter genes and noncoding regulatory elements in the human genome. These variants are thought to act during fetal development to influence the formation of different heart structures. However, identifying the genes, pathways, and cell types that mediate these effects has been challenging due to the immense diversity of cell types involved in heart development as well as the superimposed complexities of interpreting noncoding sequences. As such, understanding the molecular functions of both noncoding and coding variants remains paramount to our fundamental understanding of cardiac development and CHD. Here, we created a gene regulation map of the healthy human fetal heart across developmental time, and applied it to interpret the functions of variants associated with CHD and quantitative cardiac traits. We collected single-cell multiomic data from 734,000 single cells sampled from 41 fetal hearts spanning post-conception weeks 6 to 22, enabling the construction of gene regulation maps in 90 cardiac cell types and states, including rare populations of cardiac conduction cells. Through an unbiased analysis of all 90 cell types, we find that both rare coding variants associated with CHD and common noncoding variants associated with valve traits converge to affect valvular interstitial cells (VICs). VICs are enriched for high expression of known CHD genes previously identified through mapping of rare coding variants. Eight CHD genes, as well as other genes in similar molecular pathways, are linked to common noncoding variants associated with other valve diseases or traits via enhancers in VICs. In addition, certain common noncoding variants impact enhancers with activities highly specific to particular subanatomic structures in the heart, illuminating how such variants can impact specific aspects of heart structure and function. Together, these results implicate new enhancers, genes, and cell types in the genetic etiology of CHD, identify molecular convergence of common noncoding and rare coding variants on VICs, and suggest a more expansive view of the cell types instrumental in genetic risk for CHD, beyond the working cardiomyocyte. This regulatory map of the human fetal heart will provide a foundational resource for understanding cardiac development, interpreting genetic variants associated with heart disease, and discovering targets for cell-type specific therapies.
Collapse
Affiliation(s)
- X Rosa Ma
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Stephanie D Conley
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Michael Kosicki
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Danila Bredikhin
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ran Cui
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven Tran
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Maya U Sheth
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Wei-Lin Qiu
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sijie Chen
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Helen Y Kang
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Current address: PhD Program in Computational and Systems Biology, MIT, Cambridge, MA, USA
| | - Dulguun Amgalan
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
| | - Chad J Munger
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lauren Duan
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Katherine Dang
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Oriane Matthys Rubio
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Siavash Zamirpour
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - John DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arun Padmanabhan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jeffrey Olgin
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Scott Damrauer
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin Andersson
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - James R Priest
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Tenaya Therapeutics, South San Francisco, CA, USA
| | - Thomas Quertermous
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Xiaojie Qiu
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Bio-X, Stanford University, Stanford, CA, USA
| | - Marlene Rabinovitch
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California, Merced, Merced, CA, USA
| | - Len Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Ian A Glass
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Department of Pediatrics and Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Casey A Gifford
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Bakar Computation Health Sciences Institute, University of California, San Francisco, CA, USA
| | - William R Goodyer
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Jesse M Engreitz
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
24
|
de Oliveira FG, Foletto JVP, Medeiros YCS, Schuler-Faccini L, Kowalski TW. Bioinformatic Multi-Strategy Profiling of Congenital Heart Defects for Molecular Mechanism Recognition. Int J Mol Sci 2024; 25:12052. [PMID: 39596121 PMCID: PMC11594028 DOI: 10.3390/ijms252212052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Congenital heart defects (CHDs) rank among the most common birth defects, presenting diverse phenotypes. Genetic and environmental factors are critical in molding the process of cardiogenesis. However, these factors' interactions are not fully comprehended. Hence, this study aimed to identify and characterize differentially expressed genes involved in CHD development through bioinformatics pipelines. We analyzed experimental datasets available in genomic databases, using transcriptome, gene enrichment, and systems biology strategies. Network analysis based on genetic and phenotypic ontologies revealed that EP300, CALM3, and EGFR genes facilitate rapid information flow, while NOTCH1, TNNI3, and SMAD4 genes are significant mediators within the network. Differential gene expression (DGE) analysis identified 2513 genes across three study types, (1) Tetralogy of Fallot (ToF); (2) Hypoplastic Left Heart Syndrome (HLHS); and (3) Trisomy 21/CHD, with LYVE1, PLA2G2A, and SDR42E1 genes found in three of the six studies. Interaction networks between genes from ontology searches and the DGE analysis were evaluated, revealing interactions in ToF and HLHS groups, but none in Trisomy 21/CHD. Through enrichment analysis, we identified immune response and energy generation as some of the relevant ontologies. This integrative approach revealed genes not previously associated with CHD, along with their interactions and underlying biological processes.
Collapse
Affiliation(s)
- Fabyanne Guimarães de Oliveira
- Laboratory of Medical Genetics and Evolution, Graduate Program in Genetics and Molecular Biology, Genetics Department, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil; (F.G.d.O.); (J.V.P.F.); (L.S.-F.)
- Teratogen Information System (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil;
| | - João Vitor Pacheco Foletto
- Laboratory of Medical Genetics and Evolution, Graduate Program in Genetics and Molecular Biology, Genetics Department, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil; (F.G.d.O.); (J.V.P.F.); (L.S.-F.)
- Teratogen Information System (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil;
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil
| | - Yasmin Chaves Scimczak Medeiros
- Teratogen Information System (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil;
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil
| | - Lavínia Schuler-Faccini
- Laboratory of Medical Genetics and Evolution, Graduate Program in Genetics and Molecular Biology, Genetics Department, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil; (F.G.d.O.); (J.V.P.F.); (L.S.-F.)
- Teratogen Information System (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil;
- National Institute on Population Medical Genetics (INAGEMP), Porto Alegre 90035-903, Brazil
- Graduate Program in Children and Adolescent Health, Medicine Faculty, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, Brazil
| | - Thayne Woycinck Kowalski
- Laboratory of Medical Genetics and Evolution, Graduate Program in Genetics and Molecular Biology, Genetics Department, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil; (F.G.d.O.); (J.V.P.F.); (L.S.-F.)
- Teratogen Information System (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil;
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil
- National Institute on Population Medical Genetics (INAGEMP), Porto Alegre 90035-903, Brazil
- Bioinformatics Core, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Brazil
- Graduate Program in Medicine: Medical Sciences, Medicine Faculty, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| |
Collapse
|
25
|
Bai Z, Han J, An J, Wang H, Du X, Yang Z, Mo X. The global, regional, and national patterns of change in the burden of congenital birth defects, 1990-2021: an analysis of the global burden of disease study 2021 and forecast to 2040. EClinicalMedicine 2024; 77:102873. [PMID: 39416384 PMCID: PMC11474384 DOI: 10.1016/j.eclinm.2024.102873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Background Congenital birth defects (CBDs) present enormous challenges to global healthcare systems. These conditions severely impact patients' health and underscore issues related to socioeconomic development and healthcare accessibility and efficiency. Previous studies have been geographically limited and lacked comprehensive global analysis. This study provides global, regional, and national disability-adjusted life years (DALYs) data for four major congenital birth defects-congenital heart defects (CHD), neural tube defects (NTDs), digestive congenital anomalies (DCAs), and Down syndrome (DS) from 1990 to 2021, emphasizing health inequalities. The goal is to offer scientific evidence for optimizing resource allocation, focusing on high-burden populations, and reducing disease burden. Methods This study systematically evaluated the global, regional, and national burden of CBDs and their changes from 1990 to 2021 using the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2021. To conduct a more focused analysis, four specific CBDs were selected: CHD, NTDs, DCAs, and DS. DALYs were used as the metric, combined with the sociodemographic index (SDI). Analyses included the slope index of inequality and concentration index to measure health inequalities, frontier analysis to estimate achievable outcomes based on development levels, decomposition analysis to identify drivers of disease burden changes, Joinpoint regression analysis to assess temporal trends, and the Bayesian age-period-cohort (BAPC) model to predict future disease burden trends. Findings Compared to 1990, the global burden of the CBDs in 2021 showed a downward trend. Males had a higher burden than females, with the highest burden observed in low-SDI regions. When examining CHD, NTDs, DCAs, and DS specifically, trends in burden changes varied across different CBDs at the global, regional, and national levels. Frontier analysis revealed potential for burden improvement in various countries and territories. Decomposition analysis highlighted differences in disease burden drivers across SDI regions, showing the greatest improvement observed in low-SDI regions. Joinpoint regression analysis indicated a downward trend in DALYs burden across SDI regions, and BAPC model predictions suggested that the burden of CBDs will continue to decline in the future. Interpretation CBDs pose a major challenge to global public health. Despite an overall decline in disease burden, health inequalities remain prominent, particularly in countries and territories with lower levels of development. Future public health interventions should focus on countries and territories with low levels of development by optimizing healthcare resource allocation, improving basic health infrastructure, enhancing health education, and reducing disease burden inequalities. Global collaboration and data sharing are essential to promote a lifecycle management model for CBDs research and treatment, advancing global health development. Funding This study was supported by the National Natural Science Foundation of China (No. 82270310) and the Jiangsu Provincial Key Research and Development Program (No. BE2023662).
Collapse
Affiliation(s)
- Zihao Bai
- Nanjing Children's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jingru Han
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, China
| | - Jia An
- Nanjing Children's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Hao Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xueying Du
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zhaocong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xuming Mo
- Nanjing Children's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| |
Collapse
|
26
|
Henderson DJ, Alqahtani A, Chaudhry B, Cook A, Eley L, Houyel L, Hughes M, Keavney B, de la Pompa JL, Sled J, Spielmann N, Teboul L, Zaffran S, Mill P, Liu KJ. Beyond genomic studies of congenital heart defects through systematic modelling and phenotyping. Dis Model Mech 2024; 17:dmm050913. [PMID: 39575509 PMCID: PMC11603121 DOI: 10.1242/dmm.050913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/29/2024] [Indexed: 12/01/2024] Open
Abstract
Congenital heart defects (CHDs), the most common congenital anomalies, are considered to have a significant genetic component. However, despite considerable efforts to identify pathogenic genes in patients with CHDs, few gene variants have been proven as causal. The complexity of the genetic architecture underlying human CHDs likely contributes to this poor genetic discovery rate. However, several other factors are likely to contribute. For example, the level of patient phenotyping required for clinical care may be insufficient for research studies focused on mechanistic discovery. Although several hundred mouse gene knockouts have been described with CHDs, these are generally not phenotyped and described in the same way as CHDs in patients, and thus are not readily comparable. Moreover, most patients with CHDs carry variants of uncertain significance of crucial cardiac genes, further complicating comparisons between humans and mouse mutants. In spite of major advances in cardiac developmental biology over the past 25 years, these advances have not been well communicated to geneticists and cardiologists. As a consequence, the latest data from developmental biology are not always used in the design and interpretation of studies aimed at discovering the genetic causes of CHDs. In this Special Article, while considering other in vitro and in vivo models, we create a coherent framework for accurately modelling and phenotyping human CHDs in mice, thereby enhancing the translation of genetic and genomic studies into the causes of CHDs in patients.
Collapse
Affiliation(s)
- Deborah J. Henderson
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Ahlam Alqahtani
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Bill Chaudhry
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Andrew Cook
- University College London, Zayed Centre for Research, London WC1N 1DZ, UK
| | - Lorraine Eley
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Lucile Houyel
- Congenital and Pediatric Cardiology Unit, M3C-Necker, Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Cité, 149 Rue de Sèvres, 75015 Paris, France
| | - Marina Hughes
- Cardiology Department, Norfolk and Norwich University Hospital, Norwich NR4 7UY, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - John Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto M5G 1XS, Canada. Department of Medical Biophysics, University of Toronto, Toronto M5G 1XS, Canada
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich (GmbH), German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Lydia Teboul
- Mary Lyon Centre, MRC Harwell, Oxfordshire OX11 0RD, UK
| | - Stephane Zaffran
- Aix Marseille Université, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France
| | - Pleasantine Mill
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- MRC Human Genetics Unit, Institute for Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karen J. Liu
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
27
|
Baird DA, Mubeen H, Doganli C, Miltenburg JB, Thomsen OK, Ali Z, Naveed T, Rehman AU, Baig SM, Christensen ST, Farooq M, Larsen LA. Rare homozygous cilia gene variants identified in consanguineous congenital heart disease patients. Hum Genet 2024; 143:1323-1339. [PMID: 39347817 PMCID: PMC11522069 DOI: 10.1007/s00439-024-02703-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024]
Abstract
Congenital heart defects (CHD) appear in almost one percent of live births. Asian countries have the highest birth prevalence of CHD in the world. Recessive genotypes may represent a CHD risk factor in Asian populations with a high degree of consanguineous marriages. Genetic analysis of consanguineous families may represent a relatively unexplored source for investigating CHD etiology. To obtain insight into the contribution of recessive genotypes in CHD we analysed a cohort of forty-nine Pakistani CHD probands, originating from consanguineous unions. The majority (82%) of patient's malformations were septal defects. We identified protein altering, rare homozygous variants (RHVs) in the patient's coding genome by whole exome sequencing. The patients had a median of seven damaging RHVs each, and our analysis revealed a total of 758 RHVs in 693 different genes. By prioritizing these genes based on variant severity, loss-of-function intolerance and specific expression in the developing heart, we identified a set of 23 candidate disease genes. These candidate genes were significantly enriched for genes known to cause heart defects in recessive mouse models (P < 2.4e-06). In addition, we found a significant enrichment of cilia genes in both the initial set of 693 genes (P < 5.4e-04) and the 23 candidate disease genes (P < 5.2e-04). Functional investigation of ADCY6 in cell- and zebrafish-models verified its role in heart development. Our results confirm a significant role for cilia genes in recessive forms of CHD and suggest important functions of cilia genes in cardiac septation.
Collapse
Affiliation(s)
- Daniel A Baird
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Hira Mubeen
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Allama Iqbal Road, Faisalabad, 38000, Pakistan
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | - Canan Doganli
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Jasmijn B Miltenburg
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | | | - Zafar Ali
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
- Centre for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Tahir Naveed
- Rawalpindi Institute of Cardiology, Rawalpindi, Pakistan
| | | | - Shahid Mahmood Baig
- Faculty of Life Sciences, Health Services Academy, Islamabad, Pakistan
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Muhammad Farooq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Allama Iqbal Road, Faisalabad, 38000, Pakistan.
| | - Lars Allan Larsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
28
|
Edwards M, Zhang X, Opotowsky AR, Brown N, Shikany AR, Weaver KN. Genetic investigation and diagnosis in adults with congenital heart disease with or without structural or neurodevelopmental comorbidity: a retrospective chart review. Front Genet 2024; 15:1412806. [PMID: 39445160 PMCID: PMC11496128 DOI: 10.3389/fgene.2024.1412806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Genetic evaluation is indicated for individuals with congenital heart disease (CHD), especially if extracardiac anomalies are also present. Timely recognition of genetic diagnoses can facilitate medical management and as well as provide assessment of reproductive risk. At least 20% of the pediatric population with CHD has a syndrome or genetic diagnosis. Further, at least 30% have extracardiac congenital malformations and/or neurodevelopmental differences (NDD), and this is known to increase the likelihood of a genetic/syndromic diagnosis. However, little is known regarding whether these statistics also apply to the current population of adults living with CHD, many of whom were born prior to currently available genetic testing. Methods The primary aim of this study was to determine the prevalence of documented genetic and syndromic diagnoses in a cohort of adults with CHD followed by a dedicated adult CHD (ACHD) clinic. The secondary aims were to describe genetic testing and genetic referral patterns in this population and identify the presence of extracardiac comorbidities which are known to be indicative of an underlying genetic diagnosis in the pediatric CHD population. To answer these questions, we performed a retrospective chart review on a sample of adults with CHD (excluding those with isolated bicuspid aortic valve) seen at Cincinnati Children's Hospital in the ACHD clinic between 2010-2021. Results Among 233 adult CHD patients, 36 (14%) had a documented genetic or syndromic diagnosis but only 29 (13.7%) had received genetic testing, while 27 (11.6%) had received genetic referrals. Furthermore, of 170 patients without any documented genetics related care (defined as genetic testing, genetic referrals, or genetic diagnosis), 35 (20%) had at least one congenital and/or neurodevelopmental comorbidity. Factors associated with individuals having received genetics related care included younger age (<40), male sex, and presence of extracardiac comorbidities. Discussion Our results indicate important gaps in genetics-related care for adults living with CHD. The subset of our cohort with congenital and/or neurodevelopmental comorbidities who received no genetic-related care, represent a population of adults with CHD who may have unrecognized genetic diagnoses.
Collapse
Affiliation(s)
- Moriah Edwards
- Cincinnati Genetic Counseling Graduate Program, Cincinnati, OH, United States
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Xue Zhang
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Alexander R. Opotowsky
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Nicole Brown
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Amy R. Shikany
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Kathryn Nicole Weaver
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
29
|
Fitzgerald-Butt SM, Schartman AF, Schmit K, Ison HE, Helm BM. Genetic counselors and congenital heart disease: Clinical roles, genetic testing practices, and perceived genetic testing utility. J Genet Couns 2024; 33:1004-1014. [PMID: 37872860 DOI: 10.1002/jgc4.1821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/26/2023] [Accepted: 10/06/2023] [Indexed: 10/25/2023]
Abstract
Genetic counseling and genetic testing are essential for individuals with congenital heart disease/defects (CHD/CHDs). However, the clinical practices of genetic counselors (GCs) and their preferences for different CHD genetic testing strategies are previously unexplored. To address these gaps, GCs (n = 112) representing diverse specialties completed an online survey regarding their counseling and testing practices for syndromic CHD and apparently isolated/non-syndromic CHDs (iCHD). We found practice variability around family screening recommendations, with prenatal respondents reporting lower prevalence of this practice for iCHDs (p = 0.0004). We found that all specialties considered chromosomal microarray (CMA) the most common prioritized genetic test for syndromic and iCHD, while more prenatal respondents considered FISH and karyotype useful for iCHDs compared to postnatal respondents (p = 0.0002 and p = 0.002, respectively). Among postnatal respondents, a higher proportion considered exome/genome sequencing as useful compared to prenatal respondents (p = 0.0159); specifically, postnatal respondents' preference for exome/genome sequencing for iCHDs was ~2.6-fold higher than prenatal respondents. We estimated participants' assessment of utility for different genetic testing modalities for iCHDs and found that prenatal respondents assigned higher mean utility to FISH (p = 0.0002), karyotype (p = 0.0006), and CMA (p < 0.0001). There were relatively moderate to decreased utility scores for gene panels and exome/genome sequencing for iCHDs compared to cytogenetic testing, across all specialties. Overall, these results provide insight into GC practices and use of various genetic testing strategies for syndromic CHDs and iCHDs. Findings may help inform and/or standardize clinical practices for CHD genetic testing, though additional studies are warranted.
Collapse
Affiliation(s)
- Sara M Fitzgerald-Butt
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Allison F Schartman
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Obstetrics & Gynecology, Division of Maternal Fetal Medicine, Indiana University Health, Indianapolis, Indiana, USA
| | - Kelly Schmit
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hannah E Ison
- Stanford Healthcare, Stanford Center for Inherited Cardiovascular Disease, Stanford, California, USA
| | - Benjamin M Helm
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Epidemiology, Indiana University Fairbanks School of Public Health, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Peterson JK, Clarke S, Gelb BD, Kasparian NA, Kazazian V, Pieciak K, Pike NA, Setty SP, Uveges MK, Rudd NA. Trisomy 21 and Congenital Heart Disease: Impact on Health and Functional Outcomes From Birth Through Adolescence: A Scientific Statement From the American Heart Association. J Am Heart Assoc 2024; 13:e036214. [PMID: 39263820 DOI: 10.1161/jaha.124.036214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 09/13/2024]
Abstract
Due to improvements in recognition and management of their multisystem disease, the long-term survival of infants, children, and adolescents with trisomy 21 and congenital heart disease now matches children with congenital heart disease and no genetic condition in many scenarios. Although this improved survival is a triumph, individuals with trisomy 21 and congenital heart disease have unique and complex care needs in the domains of physical, developmental, and psychosocial health, which affect functional status and quality of life. Pulmonary hypertension and single ventricle heart disease are 2 known cardiovascular conditions that reduce life expectancy in individuals with trisomy 21. Multisystem involvement with respiratory, endocrine, gastrointestinal, hematological, neurological, and sensory systems can interact with cardiovascular health concerns to amplify adverse effects. Neurodevelopmental, psychological, and functional challenges can also affect quality of life. A highly coordinated interdisciplinary care team model, or medical home, can help address these complex and interactive conditions from infancy through the transition to adult care settings. The purpose of this Scientific Statement is to identify ongoing cardiovascular and multisystem, developmental, and psychosocial health concerns for children with trisomy 21 and congenital heart disease from birth through adolescence and to provide a framework for monitoring and management to optimize quality of life and functional status.
Collapse
|
31
|
Duarte VE, Singh MN. Genetic syndromes associated with congenital heart disease. Heart 2024; 110:1231-1237. [PMID: 38040449 DOI: 10.1136/heartjnl-2023-323126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2023] Open
Abstract
Congenital heart defects are the most common type of birth defect, affecting 1% of live births. The underlying cause of congenital heart disease is frequently unknown. However, advances in human genetics and genome technologies have helped expand congenital heart disease pathogenesis knowledge during the last few decades. When the cardiac defects are part of a genetic syndrome, they are associated with extracardiac conditions and require multidisciplinary care and surveillance. Some genetic syndromes can have subtle clinical findings and remain undiagnosed well into adulthood. Each syndrome is associated with specific congenital and acquired comorbidities and a particular clinical risk profile. A timely diagnosis is essential for risk stratification, surveillance of associated conditions and counselling, particularly during family planning. However, genetic testing and counselling indications can be challenging to identify in clinical practice. This document intends to provide an overview of the most clinically relevant syndromes to consider, focusing on the phenotype and genotype diagnosis, outcome data, clinical guidelines and implications for care.
Collapse
Affiliation(s)
- Valeria E Duarte
- Houston Methodist Debakey Heart and Vascular Center, Houston, Texas, USA
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Michael N Singh
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
- Cardiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Marmech E, Barkallah O, Selmi I, Ben Hamida N, Guizani A, Ouerda H, Khlif S, Ben Hfaiedh J, Kanzari J, Khlayfia Z, Halioui S, Azzabi O, Siala N. Congenital heart disease: Epidemiological, genetic and evolutive profil. LA TUNISIE MEDICALE 2024; 102:576-581. [PMID: 39287351 PMCID: PMC11459234 DOI: 10.62438/tunismed.v102i9.5060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/08/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Congenital heart disease is a heterogeneous group of malformations and one of the most common causes of mortality in children. AIM The aim of this study was to investigate the clinical, genetic and evolutive characteristics of congenital heart disease. METHODS A retrospective, descriptive study was carried out between 2020 and 2023 at the pediatrics and neonatology department of Mongi Slim university hospital of Tunis. All children with confirmed congenital heart disease were included. RESULTS Forty-five patients were included, representing 5.7‰ of all admissions. The sex ratio was 1.4. A prenatal diagnosis of congenital heart disease was established in 9% of cases. The median age at the time of discovery was 18 days. The initial symptomatology was respiratory distress in 64% of cases. The main reasons for performing a cardiac ultrasound were heart murmur in 38% followed by polymalformative assessment in 27% of cases. Most of the cardiopathies were atrial septal defects (42%) and ventricular septal defects (40%). Cyanotic heart diseases represented 29% of cases, conotruncal ones 13% and ductodependent ones 16%. Congenital heart disease was associated with a genetic anomaly in 53% of patients, including 15 cases of trisomy 21 and four Di-George syndromes. The treatment was mainly medical (38%), associated with surgery in 5 cases. Death occurred in nine patients, representing a mortality rate of 20%. CONCLUSION Efforts still need to be made to improve pre- and post-natal diagnosis and ensure rapid treatment in order to reduce morbidity and mortality in our country.
Collapse
Affiliation(s)
- Emna Marmech
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Oumaima Barkallah
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Ines Selmi
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Nourzed Ben Hamida
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Amani Guizani
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Haifa Ouerda
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Syrine Khlif
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Jihen Ben Hfaiedh
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Jihed Kanzari
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Zied Khlayfia
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Sonia Halioui
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Ons Azzabi
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| | - Nadia Siala
- Department of Pediatrics and Neonatology, Mongi Slim Hospital, La Marsa, Tunis, Tunisia. University of Tunis el Manar Faculty of Medicine of Tunis
| |
Collapse
|
33
|
Nawaz K, Alifah N, Hussain T, Hameed H, Ali H, Hamayun S, Mir A, Wahab A, Naeem M, Zakria M, Pakki E, Hasan N. From genes to therapy: A comprehensive exploration of congenital heart disease through the lens of genetics and emerging technologies. Curr Probl Cardiol 2024; 49:102726. [PMID: 38944223 DOI: 10.1016/j.cpcardiol.2024.102726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Congenital heart disease (CHD) affects approximately 1 % of live births worldwide, making it the most common congenital anomaly in newborns. Recent advancements in genetics and genomics have significantly deepened our understanding of the genetics of CHDs. While the majority of CHD etiology remains unclear, evidence consistently indicates that genetics play a significant role in its development. CHD etiology holds promise for enhancing diagnosis and developing novel therapies to improve patient outcomes. In this review, we explore the contributions of both monogenic and polygenic factors of CHDs and highlight the transformative impact of emerging technologies on these fields. We also summarized the state-of-the-art techniques, including targeted next-generation sequencing (NGS), whole genome and whole exome sequencing (WGS, WES), single-cell RNA sequencing (scRNA-seq), human induced pluripotent stem cells (hiPSCs) and others, that have revolutionized our understanding of cardiovascular disease genetics both from diagnosis perspective and from disease mechanism perspective in children and young adults. These molecular diagnostic techniques have identified new genes and chromosomal regions involved in syndromic and non-syndromic CHD, enabling a more defined explanation of the underlying pathogenetic mechanisms. As our knowledge and technologies continue to evolve, they promise to enhance clinical outcomes and reduce the CHD burden worldwide.
Collapse
Affiliation(s)
- Khalid Nawaz
- Department of Medical Laboratory Technology, Khyber Medical University, Peshawar, 25100, Khyber Pakhtunkhwa, Pakistan
| | - Nur Alifah
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar, 90245, Republic of Indonesia
| | - Talib Hussain
- Women Dental College, Khyber Medical University, Abbottabad, 22080, Khyber Pakhtunkhwa, Pakistan
| | - Hamza Hameed
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485, Punjab, Pakistan
| | - Haider Ali
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Shah Hamayun
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485, Punjab, Pakistan
| | - Awal Mir
- Department of Medical Laboratory Technology, Khyber Medical University, Peshawar, 25100, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Naeem
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Mohammad Zakria
- Advanced Center for Genomic Technologies, Khyber Medical University, Peshawar, 25100, Khyber Pakhtunkhwa, Pakistan
| | - Ermina Pakki
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar, 90245, Republic of Indonesia
| | - Nurhasni Hasan
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar, 90245, Republic of Indonesia.
| |
Collapse
|
34
|
Chobufo MD, Ali S, Taha A, Duhan S, Patel N, Gonuguntla K, Ludhwani D, Thyagaturu H, Keisham B, Shaik A, Alharbi A, Sattar Y, Mamas MA, Kohli U, Balla S. Temporal Trends of Infant Mortality Secondary to Congenital Heart Disease: National CDC Cohort Analysis (1999-2020). Birth Defects Res 2024; 116:e2398. [PMID: 39219403 DOI: 10.1002/bdr2.2398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/20/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Infant mortality continues to be a significant problem for patients with congenital heart disease (CHD). Limited data exist on the recent trends of mortality in infants with CHD. METHODS The CDC WONDER (Centers for Disease Control and Prevention Wide-Ranging Online Data for Epidemiologic Research) was queried to identify deaths occurring within the United States with CHD listed as one of the causes of death between 1999 and 2020. Subsequently, trends were calculated using the Joinpoint regression program (version 4.9.1.0; National Cancer Institute). RESULTS A total of 47,015 deaths occurred in infants due to CHD at the national level from the year 1999 to 2020. The overall proportional infant mortality (compared to all deaths) declined (47.3% to 37.1%, average annual percent change [AAPC]: -1.1 [95% CI -1.6 to -0.6, p < 0.001]). There was a significant decline in proportional mortality in both Black (45.3% to 34.3%, AAPC: -0.5 [-0.8 to -0.2, p = 0.002]) and White patients (55.6% to 48.6%, AAPC: -1.2 [-1.7 to -0.7, p = 0.001]), with a steeper decline among White than Black patients. A statistically significant decline in the proportional infant mortality in both non-Hispanic (43.3% to 33.0%, AAPC: -1.3% [95% CI -1.9 to -0.7, p < 0.001]) and Hispanic (67.6% to 57.7%, AAPC: -0.7 [95% CI -0.9 to -0.4, p < 0.001]) patients was observed, with a steeper decline among non-Hispanic infant population. The proportional infant mortality decreased in males (47.5% to 53.1%, AAPC: -1.4% [-1.9 to -0.9, p < 0.001]) and females (47.1% to 39.6%, AAPC: -0.9 [-1.9 to 0.0, p = 0.05]). A steady decline in for both females and males was noted. CONCLUSION Our study showed a significant decrease in CHD-related mortality rate in infants and age-adjusted mortality rate (AAMR) between 1999 and 2020. However, sex-based, racial/ethnic disparities were noted, with female, Black, and Hispanic patients showing a lesser decline than male, White, and non-Hispanic patients.
Collapse
Affiliation(s)
- Muchi Ditah Chobufo
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Shafaqat Ali
- Department of Medicine, Louisiana State University, Shreveport, Louisiana, USA
| | - Amro Taha
- Department of Medicine, Weiss Memorial Hospital, Chicago, Illinois, USA
| | - Sanchit Duhan
- Department of Cardiology, Carle Foundation Hospital, Urbana, Illinois, USA
| | - Neel Patel
- Department of Internal Medicine, Landmark Medical Center, Woonsocket, Rhode Island, USA
| | - Karthik Gonuguntla
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Dipesh Ludhwani
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Harshith Thyagaturu
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Bijeta Keisham
- Department of Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Ayesha Shaik
- Department of Cardiology, Hartford Hospital, Hartford, Connecticut, USA
| | - Anas Alharbi
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Yasar Sattar
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Mamas A Mamas
- Keele Cardiovascular Research Group, Keele University, Stoke-on-Trent, UK
| | - Utkarsh Kohli
- Department of Paediatric Cardiology, West Virginia University, Morgantown, West Virginia, USA
| | - Sudarshan Balla
- Department of Cardiology, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
35
|
Joshi R, Goswami D, Saha P, Hole A, Mandhare P, Wadke R, Murthy PR, Borgohain S, C MK, Kapoor S. Serum Raman spectroscopy: Unearthing the snapshot of distinct metabolic profile in patients with congenital heart defects (CHDs). Heliyon 2024; 10:e34575. [PMID: 39262980 PMCID: PMC11388677 DOI: 10.1016/j.heliyon.2024.e34575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 09/13/2024] Open
Abstract
In the present study, efficacy of minimally-invasive serum Raman spectroscopy (SRS) in stratification of congenital heart diseases was explored. Blood was collected from 62 subjects [42 congenital heart defect (CHD) patients (19 with atrial septal defect, 13 with ventricular septal defect and 10 with tetralogy of fallot) and 20 controls], and serum separated. Raman spectra of sera were recorded, pre-processed and subjected to spectral and multivariate analyses. Multivariate curve resolution-alternating least squares (MCR-ALS) analyses indicated alterations in lipid and protein levels between the study groups. Principal Component Analysis (PCA) and Principal Component based Linear Discriminant Analysis (PC-LDA), cross-validated with Leave-one-out cross validation (LOOCV), were employed to study stratification between the different groups. CHD could be classified from controls with 76 % efficiency. The different CHD subtypes could be distinguished with efficiencies as high as ∼90 %. To the best of our knowledge, differentiation between controls and CHDs as well as the stratification between controls and CHDs subtypes was for the first time successfully accomplished by serum-based Raman spectroscopy.
Collapse
Affiliation(s)
- Radha Joshi
- Sri Sathya Sai Sanjeevani Research Centre, Sri Sathya Sai Sanjeevani Research Foundation, Plot No. 2, Sector 38, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| | - Debosmita Goswami
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Sector 22, Utsav Chowk - CISF Road, Owe Camp, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| | - Panchali Saha
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Sector 22, Utsav Chowk - CISF Road, Owe Camp, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Training School Complex, Homi Bhabha National Institute, Anushakti Nagar, Maharashtra, India
| | - Arti Hole
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Sector 22, Utsav Chowk - CISF Road, Owe Camp, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| | - Poonam Mandhare
- Sri Sathya Sai Sanjeevani Research Centre, Sri Sathya Sai Sanjeevani Research Foundation, Plot No. 2, Sector 38, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| | - Rishikesh Wadke
- Sri Sathya Sai Sanjeevani Centre for Child Heart Care & Training in Pediatric Cardiac Skills, Plot No. 2, Sector 38, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| | - Prabhatha Rashmi Murthy
- Sri Sathya Sai Sanjeevani Centre for Child Heart Care & Training in Pediatric Cardiac Skills, Plot No. 2, Sector 38, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| | - Shyamdeep Borgohain
- Sri Sathya Sai Sanjeevani Centre for Child Heart Care & Training in Pediatric Cardiac Skills, Plot No. 2, Sector 38, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| | - Murali Krishna C
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Sector 22, Utsav Chowk - CISF Road, Owe Camp, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Training School Complex, Homi Bhabha National Institute, Anushakti Nagar, Maharashtra, India
| | - Sudhir Kapoor
- Sri Sathya Sai Sanjeevani Research Centre, Sri Sathya Sai Sanjeevani Research Foundation, Plot No. 2, Sector 38, Kharghar, Navi Mumbai, 410210, Maharashtra, India
| |
Collapse
|
36
|
Huang YL, Luo WY, Wang XL, Zheng F, Gao JH, Chen MX, Pan YD. The effect of nutritional risk management program on the growth and development of infants and toddlers with congenital heart disease after discharge. Front Pediatr 2024; 12:1416778. [PMID: 39323510 PMCID: PMC11423420 DOI: 10.3389/fped.2024.1416778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 08/15/2024] [Indexed: 09/27/2024] Open
Abstract
Objective To evaluate the effect of nutritional risk management program on the growth and development of infants and toddlers with congenital heart disease (CHD) after discharge. Methods Infants and toddlers with CHD discharged from a children's specialized hospital in southeast China were selected as the research subjects. The subjects were divided into the intervention group and the control group. The intervention group underwent a nutritional risk management program combined with traditional follow-up after discharge, whereas the control group received traditional follow-up after discharge. The primary outcome measure were the height-for-age Z-score (HAZ), weight-for-age Z-score (WAZ), and weight-for-height Z-score (WHZ) at different time point and the percentage of growth and development curves were also recorded and analyzed. Results There were no statistically significant differences in general characteristics between the two groups. However, in the intervention group, the percentages of HAZ < -2, WAZ < -2, and WHZ < -2 were lower than those in the control group at 3rd and 6th months after discharge (P < 0.05). The percentage of growth and development curves (3%-97%) was higher than that in the control group (P < 0.05). The readmission rate within 6 months after discharge in the intervention group was lower than that in the control group (P < 0.05). Conclusion Implementing nutritional risk management program for infants and toddlers with CHD after discharge can help improve postoperative malnutrition, promote growth and development and achieve catch-up growth as soon as possible.
Collapse
Affiliation(s)
- Ya-Li Huang
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Wen-Yi Luo
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xie-Lei Wang
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Feng Zheng
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Jian-Hua Gao
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Ming-Xia Chen
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Yang-Dong Pan
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| |
Collapse
|
37
|
Gomersall JC, Moore VM, Fernandez RC, Giles LC, Grzeskowiak LE, Davies MJ, Rumbold AR. Maternal modifiable factors and risk of congenital heart defects: systematic review and causality assessment. BMJ Open 2024; 14:e082961. [PMID: 39181550 PMCID: PMC11344500 DOI: 10.1136/bmjopen-2023-082961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 07/14/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Primary prevention strategies are critical to reduce the global burden of congenital heart defects (CHDs); this requires robust knowledge of causal agents. We aimed to review associations between CHDs and maternal advanced age, obesity, diabetes, hypertension, smoking and alcohol consumption and assess the causal nature of the associations. DESIGN Systematic review of reviews with application of a Bradford Hill criteria score-based causal assessment system. DATA SOURCES We searched PubMed, Embase and Episteminokos (January 1990-April 2023). ELIGIBILITY CRITERIA Systematic reviews of original epidemiological studies reporting association (relative risk) between one or more of the above maternal factors and CHDs overall (any type) in subsequent offspring. DATA EXTRACTION AND SYNTHESIS Two independent reviewers selected eligible reviews, assessed the risk of bias and assigned the strength of evidence for causality. RESULTS There was strong evidence of a causal relationship between CHDs and maternal obesity (prepregnancy and early pregnancy) and pre-existing diabetes (six of seven Bradford Hill criteria met). For pre-existing hypertension (strength and biological gradient not met), and advanced age (strength, consistency and biological gradient not met), causal evidence was moderate. Evidence for the causal contribution of gestational diabetes, gestational hypertension, smoking and alcohol consumption was weak (strength, consistency, temporality and biological gradient not met). CONCLUSIONS CHDs can be reduced with stronger action to reduce maternal obesity and pre-existing diabetes prevalence. Investigating environmental exposures that have received limited attention, such as air pollutants and chemical exposures, is important to further inform prevention.
Collapse
Affiliation(s)
- Judith C Gomersall
- School of Public Health and Lifecourse and Intergenerational Health Research Group, The University of Adelaide, Adelaide, South Australia, Australia
| | - Vivienne M Moore
- School of Public Health and Lifecourse and Intergenerational Health Research Group, The University of Adelaide, Adelaide, South Australia, Australia
| | - Renae C Fernandez
- Robinson Research Institute, Discipline of Obstetrics and Gynaecology, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lynne C Giles
- School of Public Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Luke E Grzeskowiak
- Women and Kids Theme, South Australian Health and Medical Research Institute and College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Michael J Davies
- The Robinson Research Institute and Lifecourse and Intergenerational Health Research Group, The University of Adelaide, Adelaide, South Australia, Australia
| | - Alice R Rumbold
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
38
|
Sun S, Ji Y, Shao D, Xu Y, Yang X, Sun L, Li N, Huang H, Wu Q. Genomic insights into prenatal diagnosis of congenital heart defects: value of CNV-seq and WES in clinical practice. Front Genet 2024; 15:1448383. [PMID: 39205944 PMCID: PMC11349688 DOI: 10.3389/fgene.2024.1448383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to assess the efficiency of CNV-seq and WES in detecting genetic cause of congenital heart disease (CHDs) in prenatal diagnoses and to compare CNV detection rate between isolated and non-isolated CHD cases. We conducted a retrospective study of 118 Chinese fetuses diagnosed with CHD by prenatal ultrasound. Participants underwent CNV-seq and, if necessary, WES to detect chromosomal and single nucleotide variations. The overall detection rate for pathogenic or likely pathogenic chromosomal abnormalities was 16.9%, including 7.6% aneuploidies and 9.3% pathogenic/likely pathogenic copy number variations (CNVs), predominantly 22q11.2 deletion syndrome (54.4%). The sensitivity and specificity of CNV-Seq for detecting P/Lp CNVs were 95% and 100%, respectively. CNV-Seq offered a 6.7% improvement in detecting chromosomal abnormalities over karyotyping. WES further identified significant single nucleotide and small indel variations contributing to CHD in genes such as TMEM67, PLD1, ANKRD11, and PNKP, enhancing diagnostic yield by 14.8% in cases negative for CNVs. Non-isolated CHD cases exhibited higher rates of detectable chromosomal abnormalities compared to isolated cases (32.4% vs. 9.9%, p = 0.005), underlining the genetic complexity of these conditions. The combined use of CNV-seq and WES provides a comprehensive approach to prenatal genetic testing for CHDs, unveiling significant genetic cause that could impact clinical management and parental decision-making. This study supports the integration of these advanced genomic technologies in routine prenatal diagnostics to increase detection diagnostic yields of causal genetic variants associated with CHDs.
Collapse
Affiliation(s)
- Shiyu Sun
- Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yizhen Ji
- Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Di Shao
- BGI Genomics, Shenzhen, China
| | - Yasong Xu
- Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaomei Yang
- Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Li Sun
- Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Nan Li
- BGI Genomics, Shenzhen, China
| | | | - Qichang Wu
- Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
39
|
Luo X, Liu L, Rong H, Liu X, Yang L, Li N, Shi H. ENU-based dominant genetic screen identifies contractile and neuronal gene mutations in congenital heart disease. Genome Med 2024; 16:97. [PMID: 39135118 PMCID: PMC11318149 DOI: 10.1186/s13073-024-01372-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is the most prevalent congenital anomaly, but its underlying causes are still not fully understood. It is believed that multiple rare genetic mutations may contribute to the development of CHD. METHODS In this study, we aimed to identify novel genetic risk factors for CHD using an ENU-based dominant genetic screen in mice. We analyzed fetuses with malformed hearts and compared them to control littermates by whole exome or whole genome sequencing (WES/WGS). The differences in mutation rates between observed and expected values were tested using the Poisson and Binomial distribution. Additionally, we compared WES data from human CHD probands obtained from the Pediatric Cardiac Genomics Consortium with control subjects from the 1000 Genomes Project using Fisher's exact test to evaluate the burden of rare inherited damaging mutations in patients. RESULTS By screening 10,285 fetuses, we identified 1109 cases with various heart defects, with ventricular septal defects and bicuspid aortic valves being the most common types. WES/WGS analysis of 598 cases and 532 control littermates revealed a higher number of ENU-induced damaging mutations in cases compared to controls. GO term and KEGG pathway enrichment analysis showed that pathways related to cardiac contraction and neuronal development and functions were enriched in cases. Further analysis of 1457 human CHD probands and 2675 control subjects also revealed an enrichment of genes associated with muscle and nervous system development in patients. By combining the mice and human data, we identified a list of 101 candidate digenic genesets, from which each geneset was co-mutated in at least one mouse and two human probands with CHD but not in control mouse and control human subjects. CONCLUSIONS Our findings suggest that gene mutations affecting early hemodynamic perturbations in the developing heart may play a significant role as a genetic risk factor for CHD. Further validation of the candidate gene set identified in this study could enhance our understanding of the complex genetics underlying CHD and potentially lead to the development of new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Xiaoxi Luo
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Lifeng Liu
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Haowei Rong
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiangyang Liu
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Ling Yang
- Westlake University High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Nan Li
- Westlake University High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Hongjun Shi
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
40
|
Gao M, Shen Y, Yang P, Yuan C, Sun Y, Li Z. Transcriptomics integrated with metabolomics reveals partial molecular mechanisms of nutritional risk and neurodevelopment in children with congenital heart disease. Front Cardiovasc Med 2024; 11:1414089. [PMID: 39185136 PMCID: PMC11341388 DOI: 10.3389/fcvm.2024.1414089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Purpose To explore molecular mechanisms affecting nutritional risk and neurodevelopment in children with congenital heart disease (CHD) by combining transcriptome and metabolome analysis. Methods A total of 26 blood and serum samples from 3 groups of children with CHD low nutritional risk combined with normal neurodevelopment (group A), low nutritional risk combined with neurodevelopmental disorders (group B) and high nutritional risk combined with normal neurodevelopment (group C) were analyzed by transcriptome and metabolomics to search for differentially expressed genes (DEGs) and metabolites (DEMs). Functional analysis was conducted for DEGs and DEMs. Further, the joint pathway analysis and correlation analysis of DEGs and DEMs were performed. Results A total of 362 and 1,351 DEGs were detected in group B and C compared to A, respectively. A total of 6 and 7 DEMs were detected in group B and C compared to A in positive mode, respectively. There were 39 and 31 DEMs in group B and C compared to A in negative mode. Transcriptomic analysis indicated that neurodevelopment may be regulated by some genes such as NSUN7, SLC6A8, CXCL1 and LCN8, nutritional risk may be regulated by SLC1A3 and LCN8. Metabolome analysis and joint pathway analysis showed that tryptophan metabolism, linoleic and metabolism and glycerophospholipid metabolism may be related to neurodevelopment, and glycerophospholipid metabolism pathway may be related to nutritional risk. Conclusion By integrating transcriptome and metabolome analyses, this study revealed key genes and metabolites associated with nutritional risk and neurodevelopment in children with CHD, as well as significantly altered pathways. It has important clinical translational significance.
Collapse
Affiliation(s)
- Minglei Gao
- Heart Center, Qingdao Women and Children’s Hospital, Shandong University, Qingdao, China
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Yang Shen
- Clinical Laboratory, Dalian Women and Children’s Medical Group, Dalian, China
| | - Ping Yang
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Chang Yuan
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Yanan Sun
- Heart Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Zipu Li
- Heart Center, Qingdao Women and Children’s Hospital, Shandong University, Qingdao, China
| |
Collapse
|
41
|
Downing KF, Lin AE, Nembhard WN, Rose CE, Andrews JG, Goudie A, Klewer SE, Oster ME, Farr SL. Survival to Young Adulthood Among Individuals With Congenital Heart Defects and Genetic Syndromes: Congenital Heart Survey to Recognize Outcomes, Needs, and Well-Being. J Am Heart Assoc 2024; 13:e036049. [PMID: 39082431 PMCID: PMC11964017 DOI: 10.1161/jaha.124.036049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024]
Affiliation(s)
- Karrie F. Downing
- National Center on Birth Defects and Developmental DisabilitiesCenters for Disease Control and PreventionAtlantaGA
| | - Angela E. Lin
- Medical Genetics, Massachusetts General Hospital for ChildrenBostonMA
| | - Wendy N. Nembhard
- Department of Epidemiology, Fay W Boozman College of Public Health and the Arkansas Center for Birth Defects Research and PreventionUniversity of Arkansas for Medical SciencesLittle RockAR
| | - Charles E. Rose
- National Center on Birth Defects and Developmental DisabilitiesCenters for Disease Control and PreventionAtlantaGA
| | | | - Anthony Goudie
- Department of Health Policy and ManagementFay W Boozman College of Public Health, University of Arkansas for Medical SciencesLittle RockAR
| | | | - Matthew E. Oster
- National Center on Birth Defects and Developmental DisabilitiesCenters for Disease Control and PreventionAtlantaGA
- Children’s Healthcare of Atlanta and Emory University School of MedicineAtlantaGA
| | - Sherry L. Farr
- National Center on Birth Defects and Developmental DisabilitiesCenters for Disease Control and PreventionAtlantaGA
| |
Collapse
|
42
|
Zhang Y, Dong X, Zhang J, Zhao M, Wang J, Chu J, Yang Z, Ma S, Lin K, Sun H, Luo Z. FLT4 gene polymorphisms influence isolated ventricular septal defect predisposition in a Southwest China population. BMC Med Genomics 2024; 17:197. [PMID: 39107825 PMCID: PMC11302092 DOI: 10.1186/s12920-024-01971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Ventricular septal defect (VSD) is the most common congenital heart disease. Although a small number of genes associated with VSD have been found, the genetic factors of VSD remain unclear. In this study, we evaluated the association of 10 candidate single nucleotide polymorphisms (SNPs) with isolated VSD in a population from Southwest China. METHODS Based on the results of 34 congenital heart disease whole-exome sequencing and 1000 Genomes databases, 10 candidate SNPs were selected. A total of 618 samples were collected from the population of Southwest China, including 285 VSD samples and 333 normal samples. Ten SNPs in the case group and the control group were identified by SNaPshot genotyping. The chi-square (χ2) test was used to evaluate the relationship between VSD and each candidate SNP. The SNPs that had significant P value in the initial stage were further analysed using linkage disequilibrium, and haplotypes were assessed in 34 congenital heart disease whole-exome sequencing samples using Haploview software. The bins of SNPs that were in very strong linkage disequilibrium were further used to predict haplotypes by Arlequin software. ViennaRNA v2.5.1 predicted the haplotype mRNA secondary structure. We evaluated the correlation between mRNA secondary structure changes and ventricular septal defects. RESULTS The χ2 results showed that the allele frequency of FLT4 rs383985 (P = 0.040) was different between the control group and the case group (P < 0.05). FLT4 rs3736061 (r2 = 1), rs3736062 (r2 = 0.84), rs3736063 (r2 = 0.84) and FLT4 rs383985 were in high linkage disequilibrium (r2 > 0.8). Among them, rs3736061 and rs3736062 SNPs in the FLT4 gene led to synonymous variations of amino acids, but predicting the secondary structure of mRNA might change the secondary structure of mRNA and reduce the free energy. CONCLUSIONS These findings suggest a possible molecular pathogenesis associated with isolated VSD, which warrants investigation in future studies.
Collapse
Affiliation(s)
- Yunhan Zhang
- The Department of Ultrasound Imaging, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, China
- The Department of Ultrasound Imaging, Fuwai Yunnan Cardiovascular Hospital, Chinese Academy of Medical Sciences, 528 Shahe Road, Kunming, Yunnan, 650032, China
| | - Xiaoli Dong
- The Department of Ultrasound Imaging, Fuwai Yunnan Cardiovascular Hospital, Chinese Academy of Medical Sciences, 528 Shahe Road, Kunming, Yunnan, 650032, China
| | - Jun Zhang
- The Department of Ultrasound Imaging, Fuwai Yunnan Cardiovascular Hospital, Chinese Academy of Medical Sciences, 528 Shahe Road, Kunming, Yunnan, 650032, China
| | - Miao Zhao
- The Department of Ultrasound Imaging, Fuwai Yunnan Cardiovascular Hospital, Chinese Academy of Medical Sciences, 528 Shahe Road, Kunming, Yunnan, 650032, China
| | - Jiang Wang
- The Department of Ultrasound Imaging, Fuwai Yunnan Cardiovascular Hospital, Chinese Academy of Medical Sciences, 528 Shahe Road, Kunming, Yunnan, 650032, China
| | - Jiayou Chu
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, Yunnan, 650118, China
| | - Zhaoqing Yang
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, Yunnan, 650118, China
| | - Shaohui Ma
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, Yunnan, 650118, China
| | - Keqin Lin
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, Yunnan, 650118, China
| | - Hao Sun
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, Yunnan, 650118, China.
| | - Zhiling Luo
- The Department of Ultrasound Imaging, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, China.
- The Department of Ultrasound Imaging, Fuwai Yunnan Cardiovascular Hospital, Chinese Academy of Medical Sciences, 528 Shahe Road, Kunming, Yunnan, 650032, China.
| |
Collapse
|
43
|
Gao H, Huang X, Chen W, Feng Z, Zhao Z, Li P, Tan C, Wang J, Zhuang Q, Gao Y, Min S, Yao Q, Qian M, Ma X, Wu F, Yan W, Sheng W, Huang G. Association of copy number variation in X chromosome-linked PNPLA4 with heterotaxy and congenital heart disease. Chin Med J (Engl) 2024; 137:1823-1834. [PMID: 38973237 DOI: 10.1097/cm9.0000000000003192] [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: 10/25/2023] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Heterotaxy (HTX) is a thoracoabdominal organ anomaly syndrome and commonly accompanied by congenital heart disease (CHD). The aim of this study was to analyze rare copy number variations (CNVs) in a HTX/CHD cohort and to examine the potential mechanisms contributing to HTX/CHD. METHODS Chromosome microarray analysis was used to identify rare CNVs in a cohort of 120 unrelated HTX/CHD patients, and available samples from parents were used to confirm the inheritance pattern. Potential candidate genes in CNVs region were prioritized via the DECIPHER database, and PNPLA4 was identified as the leading candidate gene. To validate, we generated PNPLA4 -overexpressing human induced pluripotent stem cell lines as well as pnpla4 -overexpressing zebrafish model, followed by a series of transcriptomic, biochemical and cellular analyses. RESULTS Seventeen rare CNVs were identified in 15 of the 120 HTX/CHD patients (12.5%). Xp22.31 duplication was one of the inherited CNVs identified in this HTX/CHD cohort, and PNPLA4 in the Xp22.31 was a candidate gene associated with HTX/CHD. PNPLA4 is expressed in the lateral plate mesoderm, which is known to be critical for left/right embryonic patterning as well as cardiomyocyte differentiation, and in the neural crest cell lineage. Through a series of in vivo and in vitro analyses at the molecular and cellular levels, we revealed that the biological function of PNPLA4 is importantly involved in the primary cilia formation and function via its regulation of energy metabolism and mitochondria-mediated ATP production. CONCLUSIONS Our findings demonstrated a significant association between CNVs and HTX/CHD. Our data strongly suggested that an increased genetic dose of PNPLA4 due to Xp22.31 duplication is a disease-causing risk factor for HTX/CHD.
Collapse
Affiliation(s)
- Han Gao
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Xianghui Huang
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital, Xiamen, Fujian 361006, China
| | - Weicheng Chen
- Children's Hospital of Fudan University, Shanghai 201102, China
| | - Zhiyu Feng
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Zhengshan Zhao
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Ping Li
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Chaozhong Tan
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Jinxin Wang
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Quannan Zhuang
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Yuan Gao
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Shaojie Min
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Qinyu Yao
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Maoxiang Qian
- Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaojing Ma
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Feizhen Wu
- Children's Hospital of Fudan University, Shanghai 201102, China
| | - Weili Yan
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Wei Sheng
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Guoying Huang
- Children's Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| |
Collapse
|
44
|
Mead TJ, Bhutada S, Foulcer SJ, Peruzzi N, Nelson CM, Seifert DE, Larkin J, Tran-Lundmark K, Filmus J, Apte SS. Combined genetic-pharmacologic inactivation of tightly linked ADAMTS proteases in temporally specific windows uncovers distinct roles for versican proteolysis and glypican-6 in cardiac development. Matrix Biol 2024; 131:1-16. [PMID: 38750698 PMCID: PMC11526477 DOI: 10.1016/j.matbio.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Extracellular matrix remodeling mechanisms are understudied in cardiac development and congenital heart defects. We show that matrix-degrading metalloproteases ADAMTS1 and ADAMTS5, are extensively co-expressed during mouse cardiac development. The mouse mutants of each gene have mild cardiac anomalies, however, their combined genetic inactivation to elicit cooperative roles is precluded by tight gene linkage. Therefore, we coupled Adamts1 inactivation with pharmacologic ADAMTS5 blockade to uncover stage-specific cooperative roles and investigated their potential substrates in mouse cardiac development. ADAMTS5 blockade was achieved in Adamts1 null mouse embryos using an activity-blocking monoclonal antibody during distinct developmental windows spanning myocardial compaction or cardiac septation and outflow tract rotation. Synchrotron imaging, RNA in situ hybridization, immunofluorescence microscopy and electron microscopy were used to determine the impact on cardiac development and compared to Gpc6 and ADAMTS-cleavage resistant versican mutants. Mass spectrometry-based N-terminomics was used to seek relevant substrates. Combined inactivation of ADAMTS1 and ADAMTS5 prior to 12.5 days of gestation led to dramatic accumulation of versican-rich cardiac jelly and inhibited formation of compact and trabecular myocardium, which was also observed in mice with ADAMTS cleavage-resistant versican. Combined inactivation after 12.5 days impaired outflow tract development and ventricular septal closure, generating a tetralogy of Fallot-like defect. N-terminomics of combined ADAMTS knockout and control hearts identified a cleaved glypican-6 peptide only in the controls. ADAMTS1 and ADAMTS5 expression in cells was associated with specific glypican-6 cleavages. Paradoxically, combined ADAMTS1 and ADAMTS5 inactivation reduced cardiac glypican-6 and outflow tract Gpc6 transcription. Notably, Gpc6-/- hearts demonstrated similar rotational defects as combined ADAMTS inactivated hearts and both had reduced hedgehog signaling. Thus, versican proteolysis in cardiac jelly at the canonical Glu441-Ala442 site is cooperatively mediated by ADAMTS1 and ADAMTS5 and required for proper ventricular cardiomyogenesis, whereas, reduced glypican-6 after combined ADAMTS inactivation impairs hedgehog signaling, leading to outflow tract malrotation.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA; University Hospitals Rainbow Babies and Children's Hospital, Cleveland, OH, USA.
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Simon J Foulcer
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Niccolò Peruzzi
- Department of Experimental Medical Science, and Wallenberg Center for Molecular Medicine Lund University and The Pediatric Heart Center, Skane University Hospital, Lund, Sweden
| | - Courtney M Nelson
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Deborah E Seifert
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Karin Tran-Lundmark
- Department of Experimental Medical Science, and Wallenberg Center for Molecular Medicine Lund University and The Pediatric Heart Center, Skane University Hospital, Lund, Sweden
| | - Jorge Filmus
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| |
Collapse
|
45
|
Hedermann G, Hedley PL, Gadsbøll K, Thagaard IN, Krebs L, Hagen CM, Sørensen TIA, Christiansen M, Ekelund CK. Maternal obesity, interpregnancy weight changes and congenital heart defects in the offspring: a nationwide cohort study. Int J Obes (Lond) 2024; 48:1126-1132. [PMID: 38734850 PMCID: PMC11281899 DOI: 10.1038/s41366-024-01531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024]
Abstract
OBJECTIVE To evaluate the association between maternal BMI and congenital heart defects (CHDs) in the offspring when including live births, stillbirths, aborted and terminated pregnancies and to investigate if maternal interpregnancy weight changes between the first and second pregnancy influences the risk of foetal CHDs. METHODS A nationwide cohort study of all singleton pregnancies in Denmark from 2008 to 2018. Data were retrieved from the Danish Foetal Medicine Database, which included both pre- and postnatal diagnoses of CHDs. Children or foetuses with chromosomal aberrations were excluded. Odds ratios were calculated with logistic regression models for CHDs overall, severe CHDs and five of the most prevalent subtypes of CHDs. RESULTS Of the 547 105 pregnancies included in the cohort, 5 442 had CHDs (1.0%). Risk of CHDs became gradually higher with higher maternal BMI; for BMI 25-29.9 kg/m2, adjusted odds ratio (aOR) 1.17 (95% CI 1.10-1.26), for BMI 30-34.9 kg/m2, aOR 1.21 (95% CI 1.09-1.33), for BMI 35-39.9 kg/m2, aOR 1.29 (95% CI 1.11-1.50) and for BMI ≥ 40 kg/m2, aOR 1.85 (95% CI 1.54-2.21). Data was adjusted for maternal age, smoking status and year of estimated due date. The same pattern was seen for the subgroup of severe CHDs. Among the atrioventricular septal defects (n = 231), an association with maternal BMI ≥ 30 kg/m2 was seen, OR 1.67 (95% CI 1.13-2.44). 109 654 women were identified with their first and second pregnancies in the cohort. Interpregnancy BMI change was associated with the risk of CHDs in the second pregnancy (BMI 2 to < 4 kg/m2: aOR 1.29, 95% CI 1.09-1.53; BMI ≥ 4 kg/m2: aOR 1.36, 95% CI 1.08-1.68). CONCLUSION The risk of foetal CHDs became gradually higher with higher maternal BMI and interpregnancy weight increases above 2 BMI units were also associated with a higher risk of CHDs.
Collapse
Affiliation(s)
- Gitte Hedermann
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institut, Copenhagen, Denmark.
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
- Department of Obstetrics and Gynaecology, Slagelse Hospital, Slagelse, Denmark.
| | - Paula L Hedley
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
- Department of Epidemiology, School of Public Health, University of Iowa, Iowa City, IA, USA
| | - Kasper Gadsbøll
- Centre of Foetal Medicine, Department of Obstetrics and Gynaecology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Ida N Thagaard
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
- Department of Obstetrics and Gynaecology, Nordsjaellands Hospital, Farum, Denmark
| | - Lone Krebs
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Amager and Hvidovre Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian M Hagen
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
| | - Thorkild I A Sørensen
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Christiansen
- Department for Congenital Disorders, Danish National Biobank and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
- Department of Epidemiology, School of Public Health, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte K Ekelund
- Centre of Foetal Medicine, Department of Obstetrics and Gynaecology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
46
|
D'Souza EE, Findley TO, Hu R, Khazal ZSH, Signorello R, Dash C, D'Gama AM, Feldman HA, Agrawal PB, Wojcik MH, Morton SU. Genomic testing and molecular diagnosis among infants with congenital heart disease in the neonatal intensive care unit. J Perinatol 2024; 44:1196-1202. [PMID: 38499751 PMCID: PMC11300151 DOI: 10.1038/s41372-024-01935-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To evaluate patterns of genetic testing among infants with CHD at a tertiary care center. STUDY DESIGN We conducted a retrospective observational cohort study of infants in the NICU with suspicion of a genetic disorder. 1075 of 7112 infants admitted to BCH had genetic evaluation including 329 with CHD and 746 without CHD. 284 of 525 infants with CHD admitted to CMHH had genetic evaluation. Patterns of testing and diagnoses were compared. RESULTS The rate of diagnosis after testing was similar for infants with or without CHD (38% [121/318] vs. 36% [246/676], p = 0.14). In a multiple logistic regression, atrioventricular septal defects were most high associated with genetic diagnosis (odds ratio 29.99, 95% confidence interval 2.69-334.12, p < 0.001). CONCLUSIONS Infants with suspicion of a genetic disorder with CHD had similar rates of molecular diagnosis as those without CHD. These results support a role for genetic testing among NICU infants with CHD.
Collapse
Affiliation(s)
- Erica E D'Souza
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Tina O Findley
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, 77030, USA
| | - Rachel Hu
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Zahra S H Khazal
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Rachel Signorello
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Camille Dash
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Alissa M D'Gama
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Henry A Feldman
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Pankaj B Agrawal
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, 02115, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL, USA
| | - Monica H Wojcik
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, 02115, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Sarah U Morton
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, 02115, USA.
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
47
|
Zhu Z, Zhou X, Yu M, Cong R, Wang Y, Zhou X, Ji C, Luan J, Yao L, Wang W, Song N. Risk factors for distant metastasis and prognosis of the penile cancer with distant metastasis. Transl Androl Urol 2024; 13:1256-1267. [PMID: 39100830 PMCID: PMC11291400 DOI: 10.21037/tau-24-92] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/24/2024] [Indexed: 08/06/2024] Open
Abstract
Background Penile cancer (PC) is a rare malignant tumor, whose distant metastasis (DM) is associated with the poorest outcomes. The risk factors associated with DM and prognosis of the PC with DM remain elusive. This study was aimed at investigating risk factors associated with DM and constructing prediction models of PC with DM. Methods This study analyzed data from the Surveillance, Epidemiology, and End Results (SEER) database over a period of 2000-2020, including clinical characteristics such as age, marital status, tumor size, Tumor Node Metastasis (TNM) staging, and treatment information. Utilizing univariate and multivariate logistic regression, alongside cox regression analysis, we identified independent risk factors for DM and prognosis in the total cases and the cases with DM. Nomograms were developed for predicting DM and prognosis in PC patients. Results Enrolling 1,488 cases, our study identified tumor size and N stage as independent predictors of DM. The predictive nomogram for DM achieved an area under the curve (AUC) of 0.904. Notably, the 1-, 3-, and 5-year cumulative survival rates for PC with DM were 35%, 17%, and 13%, respectively, with larger tumor size associated with prognosis of PC cases with DM. This study verified a correlation between advanced age and TNM stage, as well as chemotherapy with the poor PC prognosis. The nomogram yielded 0.72, 0.69 and 0.69, in predicting 1-, 3-, and 5-year overall survivals (OS), while 0.73, 0.70 and 0.69 in predicting 1-, 3-, 5-year cancer specific survivals (CSS), respectively. Conclusions This study investigated risk factors of PC with DM. Also, nomograms for predicting DM, OS and CSS of PC patients were developed.
Collapse
Affiliation(s)
- Zheng Zhu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Zhou
- Department of Urology, The Second Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Mengchi Yu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Cong
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yichun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Zhou
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chengjian Ji
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiaochen Luan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liangyu Yao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ninghong Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
48
|
Landis BJ, Helm BM, Durbin MD, Helvaty LR, Herrmann JL, Johansen M, Geddes GC, Ware SM. Early ascertainment of genetic diagnoses clarifies impact on medium-term survival following neonatal congenital heart surgery. J Clin Invest 2024; 134:e180098. [PMID: 39078715 PMCID: PMC11405028 DOI: 10.1172/jci180098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Affiliation(s)
| | | | | | | | - Jeremy L. Herrmann
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | - Stephanie M. Ware
- Department of Pediatrics, Riley Hospital for Children
- Department of Medical and Molecular Genetics, and
| |
Collapse
|
49
|
Slavotinek AM, Thompson ML, Martin LJ, Gelb BD. Diagnostic yield after next-generation sequencing in pediatric cardiovascular disease. HGG ADVANCES 2024; 5:100286. [PMID: 38521975 PMCID: PMC11024993 DOI: 10.1016/j.xhgg.2024.100286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024] Open
Abstract
Genetic testing with exome sequencing and genome sequencing is increasingly offered to infants and children with cardiovascular diseases. However, the rates of positive diagnoses after genetic testing within the different categories of cardiac disease and phenotypic subtypes of congenital heart disease (CHD) have been little studied. We report the diagnostic yield after next-generation sequencing in 500 patients with CHD from diverse population subgroups that were enrolled at three different sites in the Clinical Sequencing Evidence-Generating Research consortium. Patients were ascertained due to a primary cardiovascular issue comprising arrhythmia, cardiomyopathy, and/or CHD, and corresponding human phenotype ontology terms were selected to describe the cardiac and extracardiac findings. We examined the diagnostic yield for patients with arrhythmia, cardiomyopathy, and/or CHD and phenotypic subtypes of CHD comprising conotruncal defects, heterotaxy, left ventricular outflow tract obstruction, septal defects, and "other" heart defects. We found a significant increase in the frequency of positive findings for patients who underwent genome sequencing compared to exome sequencing and for syndromic cardiac defects compared to isolated cardiac defects. We also found significantly higher diagnostic rates for patients who presented with isolated cardiomyopathy compared to isolated CHD. For patients with syndromic presentations who underwent genome sequencing, there were significant differences in the numbers of positive diagnoses for phenotypic subcategories of CHD, ranging from 31.7% for septal defects to 60% for "other". Despite variation in the diagnostic yield at each site, our results support genetic testing in pediatric patients with syndromic and isolated cardiovascular issues and in all subtypes of CHD.
Collapse
Affiliation(s)
- Anne M Slavotinek
- Division of Medical Genetics, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA; Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Michelle L Thompson
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa J Martin
- Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
50
|
Viswanathan S, Sandeep Oza P, Bellad A, Uttarilli A. Conotruncal Heart Defects: A Narrative Review of Molecular Genetics, Genomics Research and Innovation. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:324-346. [PMID: 38986083 DOI: 10.1089/omi.2024.0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Congenital heart defects (CHDs) are most prevalent cardiac defects that occur at birth, leading to significant neonatal mortality and morbidity, especially in the developing nations. Among the CHDs, conotruncal heart defects (CTDs) are particularly noteworthy, comprising a significant portion of congenital cardiac anomalies. While advances in imaging and surgical techniques have improved the diagnosis, prognosis, and management of CTDs, their molecular genetics and genomic substrates remain incompletely understood. This expert review covers the recent advances from January 2016 onward and examines the complexities surrounding the genetic etiologies, prevalence, embryology, diagnosis, and clinical management of CTDs. We also emphasize the known copy number variants and single nucleotide variants associated with CTDs, along with the current planetary health research efforts aimed at CTDs in large cohort studies. In all, this comprehensive narrative review of molecular genetics and genomics research and innovation on CTDs draws from and highlights selected works from around the world and offers new ideas for advances in CTD diagnosis, precision medicine interventions, and accurate assessment of prognosis and recurrence risks.
Collapse
Affiliation(s)
- Sruthi Viswanathan
- Institute of Bioinformatics, Bengaluru, Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Prachi Sandeep Oza
- Institute of Bioinformatics, Bengaluru, Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anikha Bellad
- Institute of Bioinformatics, Bengaluru, Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anusha Uttarilli
- Institute of Bioinformatics, Bengaluru, Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|