151
|
Li Q, Zhao L, Zeng Y, Kuang Y, Guan Y, Chen B, Xu S, Tang B, Wu L, Mao X, Sun X, Shi J, Xu P, Diao F, Xue S, Bao S, Meng Q, Yuan P, Wang W, Ma N, Song D, Xu B, Dong J, Mu J, Zhang Z, Fan H, Gu H, Li Q, He L, Jin L, Wang L, Sang Q. Large-scale analysis of de novo mutations identifies risk genes for female infertility characterized by oocyte and early embryo defects. Genome Biol 2023; 24:68. [PMID: 37024973 PMCID: PMC10080761 DOI: 10.1186/s13059-023-02894-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 03/01/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Oocyte maturation arrest and early embryonic arrest are important reproductive phenotypes resulting in female infertility and cause the recurrent failure of assisted reproductive technology (ART). However, the genetic etiologies of these female infertility-related phenotypes are poorly understood. Previous studies have mainly focused on inherited mutations based on large pedigrees or consanguineous patients. However, the role of de novo mutations (DNMs) in these phenotypes remains to be elucidated. RESULTS To decipher the role of DNMs in ART failure and female infertility with oocyte and embryo defects, we explore the landscape of DNMs in 473 infertile parent-child trios and identify a set of 481 confident DNMs distributed in 474 genes. Gene ontology analysis reveals that the identified genes with DNMs are enriched in signaling pathways associated with female reproductive processes such as meiosis, embryonic development, and reproductive structure development. We perform functional assays on the effects of DNMs in a representative gene Tubulin Alpha 4a (TUBA4A), which shows the most significant enrichment of DNMs in the infertile parent-child trios. DNMs in TUBA4A disrupt the normal assembly of the microtubule network in HeLa cells, and microinjection of DNM TUBA4A cRNAs causes abnormalities in mouse oocyte maturation or embryo development, suggesting the pathogenic role of these DNMs in TUBA4A. CONCLUSIONS Our findings suggest novel genetic insights that DNMs contribute to female infertility with oocyte and embryo defects. This study also provides potential genetic markers and facilitates the genetic diagnosis of recurrent ART failure and female infertility.
Collapse
Affiliation(s)
- Qun Li
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
- Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Lin Zhao
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Yang Zeng
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Yanping Kuang
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Yichun Guan
- Department of Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, 200032, China
| | - Shiru Xu
- Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, 518001, Guangdong, China
| | - Bin Tang
- Reproductive Medicine Center, The First People's Hospital of Changde City, Changde, 415000, China
| | - Ling Wu
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xiaoyan Mao
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Juanzi Shi
- Reproductive Medicine Center, Northwest Women's and Children's Hospital, Xi'an, 710000, China
| | - Peng Xu
- Hainan Jinghua Hejing Hospital for Reproductive Medicine, Haikou, 570125, China
| | - Feiyang Diao
- Reproductive Medicine Center, Jiangsu Province Hospital, Nanjing, 210036, China
| | - Songguo Xue
- Reproductive Medicine Center, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Shihua Bao
- Department of Reproductive Immunology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, China
| | - Qingxia Meng
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215000, China
| | - Ping Yuan
- IVF Center, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wenjun Wang
- IVF Center, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ning Ma
- Reproductive Medical Center, Maternal and Child Health Care Hospital of Hainan Province, Haikou, 570206, Hainan Province, China
| | - Di Song
- Naval Medical University, Changhai Hospital, Shanghai, China
| | - Bei Xu
- Reproductive Medicine Centre, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Jian Mu
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Zhihua Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Huizhen Fan
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Hao Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Qiaoli Li
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
152
|
Duan J, Ye Y, Liao J, Chen L, Zhao X, Liu C, Wen J. White-Sutton syndrome and congenital heart disease: case report and literature review. BMC Pediatr 2023; 23:158. [PMID: 37016333 PMCID: PMC10071667 DOI: 10.1186/s12887-023-03972-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/24/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND White-Sutton syndrome is an autosomal dominant neurodevelopmental disorder caused by heterozygous mutation in POGZ (Pogo Transposable Element Derived with ZNF Domain). This syndrome is characterized by delayed psychomotor development apparent in infancy and abnormal facial features. To date, 80 cases have been reported in the literature; however, the phenotypic characterizations remain incomplete. CASE PRESENTATION We herein describe a 2-year-old girl harboring a novel frameshift de novo POGZ variant: c.2746del (p.Thr916ProfsTer12). This patient presented with multisystem abnormalities affecting the digestive tract and neurological functioning, as well as congenital heart disease, which involved an atrial septal defect (18 × 23 × 22 mm) with pulmonary arterial hypertension (42 mmHg). The relationship between congenital heart disease and White-Sutton syndrome as described in both the GeneReview and OMIM databases (#616,364) remains unclear. A review of the current literature revealed 18 cases of White-Sutton syndrome with POGZ variants and congenital heart disease, and we summarize their clinical features in this study. CONCLUSIONS Our findings based on the present case and those in the literature indicate a relationship between POGZ mutation and congenital heart disease.
Collapse
Affiliation(s)
- Jing Duan
- Department of Neurology, Shenzhen Children's Hospital, 7019# Yitian Road, Futian District, Guangdong Province , 518038, Shenzhen, PR China
| | - Yuanzhen Ye
- Department of Neurology, Shenzhen Children's Hospital, 7019# Yitian Road, Futian District, Guangdong Province , 518038, Shenzhen, PR China
| | - Jianxiang Liao
- Department of Neurology, Shenzhen Children's Hospital, 7019# Yitian Road, Futian District, Guangdong Province , 518038, Shenzhen, PR China
| | - Li Chen
- Department of Neurology, Shenzhen Children's Hospital, 7019# Yitian Road, Futian District, Guangdong Province , 518038, Shenzhen, PR China
| | - Xia Zhao
- Department of Neurology, Shenzhen Children's Hospital, 7019# Yitian Road, Futian District, Guangdong Province , 518038, Shenzhen, PR China
| | - Chao Liu
- Department of Bioinformatics, Berry Genomics Co. Ltd, Beijing, China
| | - Jialun Wen
- Department of Neurology, Shenzhen Children's Hospital, 7019# Yitian Road, Futian District, Guangdong Province , 518038, Shenzhen, PR China.
| |
Collapse
|
153
|
Wang L, Owusu-Hammond C, Sievert D, Gleeson JG. Stem Cell-Based Organoid Models of Neurodevelopmental Disorders. Biol Psychiatry 2023; 93:622-631. [PMID: 36759260 PMCID: PMC10022535 DOI: 10.1016/j.biopsych.2023.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
The past decade has seen an explosion in the identification of genetic causes of neurodevelopmental disorders, including Mendelian, de novo, and somatic factors. These discoveries provide opportunities to understand cellular and molecular mechanisms as well as potential gene-gene and gene-environment interactions to support novel therapies. Stem cell-based models, particularly human brain organoids, can capture disease-associated alleles in the context of the human genome, engineered to mirror disease-relevant aspects of cellular complexity and developmental timing. These models have brought key insights into neurodevelopmental disorders as diverse as microcephaly, autism, and focal epilepsy. However, intrinsic organoid-to-organoid variability, low levels of certain brain-resident cell types, and long culture times required to reach maturity can impede progress. Several recent advances incorporate specific morphogen gradients, mixtures of diverse brain cell types, and organoid engraftment into animal models. Together with nonhuman primate organoid comparisons, mechanisms of human neurodevelopmental disorders are emerging.
Collapse
Affiliation(s)
- Lu Wang
- From the Department of Neuroscience, Rady Children's Institute for Genomic Medicine, University of California, San Diego, San Diego, California
| | - Charlotte Owusu-Hammond
- From the Department of Neuroscience, Rady Children's Institute for Genomic Medicine, University of California, San Diego, San Diego, California
| | - David Sievert
- From the Department of Neuroscience, Rady Children's Institute for Genomic Medicine, University of California, San Diego, San Diego, California
| | - Joseph G Gleeson
- From the Department of Neuroscience, Rady Children's Institute for Genomic Medicine, University of California, San Diego, San Diego, California.
| |
Collapse
|
154
|
Lewis MJ, Hsieh A, Qiao L, Tan R, Kazzi B, Channing A, Griffin EL, Jobanputra V, Su J, Shahryar C, Kochilas L, Gaynor JW, Lee T, Goldmuntz E, Russell M, Mital S, Tristani M, Brueckner M, Newburger J, Shen Y, Chung WK. Association of Predicted Damaging De Novo Variants on Ventricular Function in Individuals With Congenital Heart Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:e003900. [PMID: 36866680 PMCID: PMC10121832 DOI: 10.1161/circgen.122.003900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Affiliation(s)
- Matthew J Lewis
- Department of Medicine (M.J.L., B.K., W.K.C.), Columbia University Irving Medical Center, New York
| | - Alexander Hsieh
- Departmens of Systems Biology and Biomedical Informatics (A.H., R.T., Y.S.), Columbia University Irving Medical Center, New York
| | - Lu Qiao
- Department of Pediatrics (L.Q., A.C., E.L.G., V.J., T.L., W.K.C.), Columbia University Irving Medical Center, New York
| | - Renjie Tan
- Departmens of Systems Biology and Biomedical Informatics (A.H., R.T., Y.S.), Columbia University Irving Medical Center, New York
| | - Brigitte Kazzi
- Department of Medicine (M.J.L., B.K., W.K.C.), Columbia University Irving Medical Center, New York
| | - Alexandra Channing
- Department of Pediatrics (L.Q., A.C., E.L.G., V.J., T.L., W.K.C.), Columbia University Irving Medical Center, New York
| | - Emily L Griffin
- Department of Pediatrics (L.Q., A.C., E.L.G., V.J., T.L., W.K.C.), Columbia University Irving Medical Center, New York
- Department of Pediatrics (E.L.G.), The Children's Hospital of Philadelphia, PA
| | - Vaidehi Jobanputra
- Department of Pediatrics (L.Q., A.C., E.L.G., V.J., T.L., W.K.C.), Columbia University Irving Medical Center, New York
| | - Jennifer Su
- Department of Pediatrics, Children's Hospital Los Angles, CA (J.S.)
| | - Chowdhury Shahryar
- Department of Pediatrics, Medical University of South Carolina, Charleston (C.S.)
| | | | - J William Gaynor
- Department of Pediatric Cardiac Surgery (J.W.G.), The Children's Hospital of Philadelphia, PA
| | - Teresa Lee
- Department of Pediatrics (L.Q., A.C., E.L.G., V.J., T.L., W.K.C.), Columbia University Irving Medical Center, New York
| | | | - Mark Russell
- Division of Pediatric Cardiology, University of Michigan, Ann Arbor (M.R.)
| | - Seema Mital
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Ontario, Canada (S.M.)
| | - Martin Tristani
- Department of Pediatrics, University of Utah, Salt Lake City (M.T.)
| | | | - Jane Newburger
- Department of Cardiology, Boston Children's Hospital, MA (J.N.)
| | - Yufeng Shen
- Departmens of Systems Biology and Biomedical Informatics (A.H., R.T., Y.S.), Columbia University Irving Medical Center, New York
| | - Wendy K Chung
- Department of Medicine (M.J.L., B.K., W.K.C.), Columbia University Irving Medical Center, New York
- Department of Pediatrics (L.Q., A.C., E.L.G., V.J., T.L., W.K.C.), Columbia University Irving Medical Center, New York
| |
Collapse
|
155
|
Griffin EL, Nees SN, Morton SU, Wynn J, Patel N, Jobanputra V, Robinson S, Kochav SM, Tao A, Andrews C, Cross N, Geva J, Lanzilotta K, Ritter A, Taillie E, Thompson A, Meyer C, Akers R, King EC, Cnota JF, Kim RW, Porter GA, Brueckner M, Seidman CE, Shen Y, Gelb BD, Goldmuntz E, Newburger JW, Roberts AE, Chung WK. Evidence-Based Assessment of Congenital Heart Disease Genes to Enable Returning Results in a Genomic Study. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:e003791. [PMID: 36803080 PMCID: PMC10121846 DOI: 10.1161/circgen.122.003791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/28/2022] [Indexed: 02/23/2023]
Abstract
BACKGROUND Congenital heart disease (CHD) is the most common major congenital anomaly and causes significant morbidity and mortality. Epidemiologic evidence supports a role of genetics in the development of CHD. Genetic diagnoses can inform prognosis and clinical management. However, genetic testing is not standardized among individuals with CHD. We sought to develop a list of validated CHD genes using established methods and to evaluate the process of returning genetic results to research participants in a large genomic study. METHODS Two-hundred ninety-five candidate CHD genes were evaluated using a ClinGen framework. Sequence and copy number variants involving genes in the CHD gene list were analyzed in Pediatric Cardiac Genomics Consortium participants. Pathogenic/likely pathogenic results were confirmed on a new sample in a clinical laboratory improvement amendments-certified laboratory and disclosed to eligible participants. Adult probands and parents of probands who received results were asked to complete a post-disclosure survey. RESULTS A total of 99 genes had a strong or definitive clinical validity classification. Diagnostic yields for copy number variants and exome sequencing were 1.8% and 3.8%, respectively. Thirty-one probands completed clinical laboratory improvement amendments-confirmation and received results. Participants who completed postdisclosure surveys reported high personal utility and no decision regret after receiving genetic results. CONCLUSIONS The application of ClinGen criteria to CHD candidate genes yielded a list that can be used to interpret clinical genetic testing for CHD. Applying this gene list to one of the largest research cohorts of CHD participants provides a lower bound for the yield of genetic testing in CHD.
Collapse
Affiliation(s)
- Emily L. Griffin
- Dept of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Shannon N. Nees
- Nemours Cardiac Center, Nemours Children’s Hospital, Delaware. Wilmington, DE
| | - Sarah U. Morton
- Division of Newborn Medicine, Dept of Medicine, Boston Children’s Hospital
- Dept of Pediatrics, Harvard Medical School, Boston, MA
| | - Julia Wynn
- Dept of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Nihir Patel
- Mindich Child Health & Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Vaidehi Jobanputra
- Dept of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Scott Robinson
- Dept of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Stephanie M. Kochav
- Division of Cardiology, Dept of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY
| | - Alice Tao
- Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Carli Andrews
- Dept of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Nancy Cross
- Division of Pediatric Cardiology, Yale School of Medicine, New Haven, CT
| | - Judith Geva
- Dept of Cardiology, Boston Children’s Hospital
| | - Kristen Lanzilotta
- Division of Cardiology, Children’s Hospital of Philadelphia, Dept of Pediatrics, Perelman School of Medicine, University of Pennsylvania
| | - Alyssa Ritter
- Division of Cardiology, Children’s Hospital of Philadelphia, Dept of Pediatrics, Perelman School of Medicine, University of Pennsylvania
- Division of Human Genetics, Dept of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Eileen Taillie
- Dept of Pediatrics, Golisano Children’s Hospital, University of Rochester Medical Center, Rochester, NY
| | - Alexandra Thompson
- Division of Cardiothoracic Surgery, Children’s Hospital of Los Angeles, Los Angeles, CA
| | | | - Rachel Akers
- Division of Biostatistics & Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Eileen C. King
- Division of Biostatistics & Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - James F Cnota
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Richard W. Kim
- Pediatric Cardiac Surgery, Children's Hospital of Los Angeles, Los Angeles, CA
| | - George A. Porter
- Dept of Pediatrics, University of Rochester Medical Center, The School of Medicine & Dentistry, Rochester, NY
| | - Martina Brueckner
- Dept of Genetics & Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Christine E. Seidman
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA
- Dept of Genetics, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Chevy Chase, MD
| | - Yufeng Shen
- Depts of Systems Biology & Biomedical Informatics, Columbia University, New York, NY
| | - Bruce D. Gelb
- Mindich Child Health & Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Depts of Pediatrics and Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Elizabeth Goldmuntz
- Division of Cardiology, Children’s Hospital of Philadelphia, Dept of Pediatrics, Perelman School of Medicine, University of Pennsylvania
| | - Jane W. Newburger
- Dept of Pediatrics, Harvard Medical School, Boston, MA
- Dept of Cardiology, Boston Children’s Hospital
| | - Amy E. Roberts
- Dept of Cardiology, Boston Children’s Hospital
- Division of Genetics, Dept of Pediatrics, Boston Children’s Hospital
| | - Wendy K. Chung
- Dept of Pediatrics, Columbia University Irving Medical Center, New York, NY
- Dept of Medicine, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
156
|
Singh AK, Viviano S, Allington G, McGee S, Kiziltug E, Mekbib KY, Shohfi JP, Duy PQ, DeSpenza T, Furey CG, Reeves BC, Smith H, Ma S, Sousa AMM, Cherskov A, Allocco A, Nelson-Williams C, Haider S, Rizvi SRA, Alper SL, Sestan N, Shimelis H, Walsh LK, Lifton RP, Moreno-De-Luca A, Jin SC, Kruszka P, Deniz E, Kahle KT. A novel SMARCC1 -mutant BAFopathy implicates epigenetic dysregulation of neural progenitors in hydrocephalus. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.19.23287455. [PMID: 36993720 PMCID: PMC10055611 DOI: 10.1101/2023.03.19.23287455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Importance Hydrocephalus, characterized by cerebral ventriculomegaly, is the most common disorder requiring brain surgery. A few familial forms of congenital hydrocephalus (CH) have been identified, but the cause of most sporadic cases of CH remains elusive. Recent studies have implicated SMARCC1 , a component of the B RG1- a ssociated factor (BAF) chromatin remodeling complex, as a candidate CH gene. However, SMARCC1 variants have not been systematically examined in a large patient cohort or conclusively linked with a human syndrome. Moreover, CH-associated SMARCC1 variants have not been functionally validated or mechanistically studied in vivo . Objectives The aims of this study are to (i) assess the extent to which rare, damaging de novo mutations (DNMs) in SMARCC1 are associated with cerebral ventriculomegaly; (ii) describe the clinical and radiographic phenotypes of SMARCC1 -mutated patients; and (iii) assess the pathogenicity and mechanisms of CH-associated SMARCC1 mutations in vivo . Design setting and participants A genetic association study was conducted using whole-exome sequencing from a cohort consisting of 2,697 ventriculomegalic trios, including patients with neurosurgically-treated CH, totaling 8,091 exomes collected over 5 years (2016-2021). Data were analyzed in 2023. A comparison control cohort consisted of 1,798 exomes from unaffected siblings of patients with autism spectrum disorder and their unaffected parents sourced from the Simons simplex consortium. Main outcomes and measures Gene variants were identified and filtered using stringent, validated criteria. Enrichment tests assessed gene-level variant burden. In silico biophysical modeling estimated the likelihood and extent of the variant impact on protein structure. The effect of a CH-associated SMARCC1 mutation on the human fetal brain transcriptome was assessed by analyzing RNA-sequencing data. Smarcc1 knockdowns and a patient-specific Smarcc1 variant were tested in Xenopus and studied using optical coherence tomography imaging, in situ hybridization, and immunofluorescence microscopy. Results SMARCC1 surpassed genome-wide significance thresholds in DNM enrichment tests. Six rare protein-altering DNMs, including four loss-of-function mutations and one recurrent canonical splice site mutation (c.1571+1G>A) were detected in unrelated patients. DNMs localized to the highly conserved DNA-interacting SWIRM, Myb-DNA binding, Glu-rich, and Chromo domains of SMARCC1 . Patients exhibited developmental delay (DD), aqueductal stenosis, and other structural brain and heart defects. G0 and G1 Smarcc1 Xenopus mutants exhibited aqueductal stenosis and cardiac defects and were rescued by human wild-type SMARCC1 but not a patient-specific SMARCC1 mutant. Hydrocephalic SMARCC1 -mutant human fetal brain and Smarcc1 -mutant Xenopus brain exhibited a similarly altered expression of key genes linked to midgestational neurogenesis, including the transcription factors NEUROD2 and MAB21L2 . Conclusions SMARCC1 is a bona fide CH risk gene. DNMs in SMARCC1 cause a novel human BAFopathy we term " S MARCC1- a ssociated D evelopmental D ysgenesis S yndrome (SaDDS)", characterized by cerebral ventriculomegaly, aqueductal stenosis, DD, and a variety of structural brain or cardiac defects. These data underscore the importance of SMARCC1 and the BAF chromatin remodeling complex for human brain morphogenesis and provide evidence for a "neural stem cell" paradigm of human CH pathogenesis. These results highlight the utility of trio-based WES for identifying risk genes for congenital structural brain disorders and suggest WES may be a valuable adjunct in the clinical management of CH patients. KEY POINTS Question: What is the role of SMARCC1 , a core component of the B RG1- a ssociated factor (BAF) chromatin remodeling complex, in brain morphogenesis and congenital hydrocephalus (CH)? Findings: SMARCC1 harbored an exome-wide significant burden of rare, protein-damaging de novo mutations (DNMs) (p = 5.83 × 10 -9 ) in the largest ascertained cohort to date of patients with cerebral ventriculomegaly, including treated CH (2,697 parent-proband trios). SMARCC1 contained four loss-of-function DNMs and two identical canonical splice site DNMs in a total of six unrelated patients. Patients exhibited developmental delay, aqueductal stenosis, and other structural brain and cardiac defects. Xenopus Smarcc1 mutants recapitulated core human phenotypes and were rescued by the expression of human wild-type but not patient-mutant SMARCC1 . Hydrocephalic SMARCC1 -mutant human brain and Smarcc1 -mutant Xenopus brain exhibited similar alterationsin the expression of key transcription factors that regulate neural progenitor cell proliferation. Meaning: SMARCC1 is essential for human brain morphogenesis and is a bona fide CH risk gene. SMARCC1 mutations cause a novel human BAFopathy we term " S MARCC1- a ssociated D evelopmental D ysgenesis S yndrome (SaDDS)". These data implicate epigenetic dysregulation of fetal neural progenitors in the pathogenesis of hydrocephalus, with diagnostic and prognostic implications for patients and caregivers.
Collapse
|
157
|
Lajmi Y, Loeuillet L, Petrilli G, Egloff C, Nectoux J, Molac C, Roux N, Pannier E, Achaiaa A, Arkoub ZA, Chuon S, Coussement A, Dupont JM, Malan V, Spaggiari E, Razavi F, Amiel J, Bessières B, Grotto S, Attié-Bitach T. Two novel variations p.(Ser1275Thr) and p.(Ser1275Arg) in FLT4 causing prenatal hereditary lymphedema type 1. Birth Defects Res 2023; 115:563-571. [PMID: 36538874 DOI: 10.1002/bdr2.2141] [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: 11/22/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Hereditary lymphedema 1 is a rare congenital condition, characterized by the development of chronic swelling in body parts. It is highly variable in expression and age of onset with different presentations: from feet edema to hydrops fetalis. This affection is genetically heterogeneous with autosomal dominant inheritance and incomplete penetrance due to a mutation in the FLT4 gene in most cases. CASES In our study, we report on two fetuses harboring congenital lymphedema with FLT4 variation and review the prenatal confirmed ones of the literatures. Our cases were selected within fetuses explored by exome sequencing in a diagnosis setting. Prenatal ultrasonography showed hydrops fetalis in one case and an increased nuchal translucency with hydrothorax in the other. Comparative genomic hybridization array on amniocentesis was normal in both cases. Exome sequencing identified a variation p.(Ser1275Thr) and p.(Ser1275Arg) in fetus 1 and fetus 2 in the FLT4 gene, respectively. A de novo mutation at the same codon was reported in prenatal literature suggesting possible genotype phenotype correlation. CONCLUSION Cystic hygroma/hydrops fetalis are possible manifestations of several disorders. This study illustrates how the integration of exome sequencing in prenatal clinical practice can facilitate the diagnosis and genetic counseling of heterogeneous developmental affections.
Collapse
Affiliation(s)
- Yosra Lajmi
- Department of Genomic Medicine of System and Organs Diseases, Cochin Hospital, APHP Center, University Paris Cité, Paris, France
| | - Laurence Loeuillet
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Giulia Petrilli
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Charles Egloff
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Juliette Nectoux
- Department of Genomic Medicine of System and Organs Diseases, Cochin Hospital, APHP Center, University Paris Cité, Paris, France
| | - Clémence Molac
- Clinical Genetics, Maternity Port-Royal, APHP Center, University Paris Cité, Paris, France
| | - Nathalie Roux
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Emmanuelle Pannier
- Gynecology-Obstetrics Department, Port-Royal Hospital, APHP Center, University Paris Cité, Paris, France
| | - Amale Achaiaa
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Zaina Ait Arkoub
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Sophie Chuon
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Aurélie Coussement
- Department of Genomic Medicine of System and Organs Diseases, Cochin Hospital, APHP Center, University Paris Cité, Paris, France
| | - Jean Michel Dupont
- Department of Genomic Medicine of System and Organs Diseases, Cochin Hospital, APHP Center, University Paris Cité, Paris, France
| | - Valérie Malan
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Emmanuel Spaggiari
- Gynecology-Obstetrics Department, Necker Hospital for Children Diseases, APHP Center, University Paris Cité, Paris, France
| | - Ferechte Razavi
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Jeanne Amiel
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Bettina Bessières
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| | - Sarah Grotto
- Clinical Genetics, Maternity Port-Royal, APHP Center, University Paris Cité, Paris, France
| | - Tania Attié-Bitach
- Department of Genomic Medicine of Rare Disorders, Necker Hospital, APHP Center, University Paris Cité, Paris, France
| |
Collapse
|
158
|
Colleluori V, Khokha MK. Mink1 regulates spemann organizer cell fate in the xenopus gastrula via Hmga2. Dev Biol 2023; 495:42-53. [PMID: 36572140 PMCID: PMC10116378 DOI: 10.1016/j.ydbio.2022.11.010] [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: 05/06/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
Congenital Heart Disease (CHD) is the most common birth defect and leading cause of infant mortality, yet molecular mechanisms explaining CHD remain mostly unknown. Sequencing studies are identifying CHD candidate genes at a brisk rate including MINK1, a serine/threonine kinase. However, a plausible molecular mechanism connecting CHD and MINK1 is unknown. Here, we reveal that mink1 is required for proper heart development due to its role in left-right patterning. Mink1 regulates canonical Wnt signaling to define the cell fates of the Spemann Organizer and the Left-Right Organizer, a ciliated structure that breaks bilateral symmetry in the vertebrate embryo. To identify Mink1 targets, we applied an unbiased proteomics approach and identified the high mobility group architectural transcription factor, Hmga2. We report that Hmga2 is necessary and sufficient for regulating Spemann's Organizer. Indeed, we demonstrate that Hmga2 can induce Spemann Organizer cell fates even when β-catenin, a critical effector of the Wnt signaling pathway, is depleted. In summary, we discover a transcription factor, Hmga2, downstream of Mink1 that is critical for the regulation of Spemann's Organizer, as well as the LRO, defining a plausible mechanism for CHD.
Collapse
Affiliation(s)
- Vaughn Colleluori
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT, United States.
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
159
|
Role of calcium-sensor proteins in cell membrane repair. Biosci Rep 2023; 43:232522. [PMID: 36728029 PMCID: PMC9970828 DOI: 10.1042/bsr20220765] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/03/2023] Open
Abstract
Cell membrane repair is a critical process used to maintain cell integrity and survival from potentially lethal chemical, and mechanical membrane injury. Rapid increases in local calcium levels due to a membrane rupture have been widely accepted as a trigger for multiple membrane-resealing models that utilize exocytosis, endocytosis, patching, and shedding mechanisms. Calcium-sensor proteins, such as synaptotagmins (Syt), dysferlin, S100 proteins, and annexins, have all been identified to regulate, or participate in, multiple modes of membrane repair. Dysfunction of membrane repair from inefficiencies or genetic alterations in these proteins contributes to diseases such as muscular dystrophy (MD) and heart disease. The present review covers the role of some of the key calcium-sensor proteins and their involvement in membrane repair.
Collapse
|
160
|
Ritchie FD, Lizarraga SB. The role of histone methyltransferases in neurocognitive disorders associated with brain size abnormalities. Front Neurosci 2023; 17:989109. [PMID: 36845425 PMCID: PMC9950662 DOI: 10.3389/fnins.2023.989109] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 01/17/2023] [Indexed: 02/12/2023] Open
Abstract
Brain size is controlled by several factors during neuronal development, including neural progenitor proliferation, neuronal arborization, gliogenesis, cell death, and synaptogenesis. Multiple neurodevelopmental disorders have co-morbid brain size abnormalities, such as microcephaly and macrocephaly. Mutations in histone methyltransferases that modify histone H3 on Lysine 36 and Lysine 4 (H3K36 and H3K4) have been identified in neurodevelopmental disorders involving both microcephaly and macrocephaly. H3K36 and H3K4 methylation are both associated with transcriptional activation and are proposed to sterically hinder the repressive activity of the Polycomb Repressor Complex 2 (PRC2). During neuronal development, tri-methylation of H3K27 (H3K27me3) by PRC2 leads to genome wide transcriptional repression of genes that regulate cell fate transitions and neuronal arborization. Here we provide a review of neurodevelopmental processes and disorders associated with H3K36 and H3K4 histone methyltransferases, with emphasis on processes that contribute to brain size abnormalities. Additionally, we discuss how the counteracting activities of H3K36 and H3K4 modifying enzymes vs. PRC2 could contribute to brain size abnormalities which is an underexplored mechanism in relation to brain size control.
Collapse
|
161
|
Li W, He XD, Yang ZT, Han DM, Sun Y, Chen YX, Han XT, Guo SC, Ma YT, Jin X, Yang HM, Gao Y, Wang ZS, Li JK, He W. De Novo Mutations Contributes Approximately 7% of Pathogenicity in Inherited Eye Diseases. Invest Ophthalmol Vis Sci 2023; 64:5. [PMID: 36729443 PMCID: PMC9907368 DOI: 10.1167/iovs.64.2.5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Purpose The purpose of this study was to describe genotype-phenotype associations and novel insights into genetic characteristics in a trio-based cohort of inherited eye diseases (IEDs). Methods To determine the etiological role of de novo mutations (DNMs) and genetic profile in IEDs, we retrospectively reviewed a large cohort of proband-parent trios of Chinese origin. The patients underwent a detailed examination and was clinically diagnosed by an ophthalmologist. Panel-based targeted exome sequencing was performed on DNA extracted from blood samples, containing coding regions of 792 IED-causative genes and their flanking exons. All participants underwent genetic testing. Results All proband-parent trios were divided into 22 subgroups, the overall diagnostic yield was 48.67% (605/1243), ranging from 4% to 94.44% for each of the subgroups. A total of 108 IED-causative genes were identified, with the top 24 genes explaining 67% of the 605 genetically solved trios. The genetic etiology of 6.76% (84/1243) of the trio was attributed to disease-causative DNMs, and the top 3 subgroups with the highest incidence of DNM were aniridia (n = 40%), Marfan syndrome/ectopia lentis (n = 38.78%), and retinoblastoma (n = 37.04%). The top 10 genes have a diagnostic yield of DNM greater than 3.5% in their subgroups, including PAX6 (40.00%), FBN1 (38.78%), RB1 (37.04%), CRX (10.34%), CHM (9.09%), WFS1 (8.00%), RP1L1 (5.88%), RS1 (5.26%), PCDH15 (4.00%), and ABCA4 (3.51%). Additionally, the incidence of DNM in offspring showed a trend of correlation with paternal age at reproduction, but not statistically significant with paternal (P = 0.154) and maternal (P = 0.959) age at reproduction. Conclusions Trios-based genetic analysis has high accuracy and validity. Our study helps to quantify the burden of the full spectrum IED caused by each gene, offers novel potential for elucidating etiology, and plays a crucial role in genetic counseling and patient management.
Collapse
Affiliation(s)
- Wei Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China,BGI-Shenzhen, Shenzhen, China
| | | | - Zheng-Tao Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China,BGI-Shenzhen, Shenzhen, China
| | - Dong-Ming Han
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China,BGI-Shenzhen, Shenzhen, China
| | - Yan Sun
- He University, Shenyang, China
| | - Yan-Xian Chen
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xiao-Tong Han
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Si-Cheng Guo
- BGI-Shenzhen, Shenzhen, China,College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, China
| | - Yu-Ting Ma
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China,BGI-Shenzhen, Shenzhen, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, China
| | - Huan-Ming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China,BGI-Shenzhen, Shenzhen, China
| | - Ya Gao
- BGI-Shenzhen, Shenzhen, China
| | | | | | - Wei He
- He University, Shenyang, China
| |
Collapse
|
162
|
Wu Y, Bayrak CS, Dong B, He S, Stenson PD, Cooper DN, Itan Y, Chen L. Identifying shared genetic factors underlying epilepsy and congenital heart disease in Europeans. Hum Genet 2023; 142:275-288. [PMID: 36352240 DOI: 10.1007/s00439-022-02502-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Epilepsy (EP) and congenital heart disease (CHD) are two apparently unrelated diseases that nevertheless display substantial mutual comorbidity. Thus, while congenital heart defects are associated with an elevated risk of developing epilepsy, the incidence of epilepsy in CHD patients correlates with CHD severity. Although genetic determinants have been postulated to underlie the comorbidity of EP and CHD, the precise genetic etiology is unknown. We performed variant and gene association analyses on EP and CHD patients separately, using whole exomes of genetically identified Europeans from the UK Biobank and Mount Sinai BioMe Biobank. We prioritized biologically plausible candidate genes and investigated the enriched pathways and other identified comorbidities by biological proximity calculation, pathway analyses, and gene-level phenome-wide association studies. Our variant- and gene-level results point to the Voltage-Gated Calcium Channels (VGCC) pathway as being a unifying framework for EP and CHD comorbidity. Additionally, pathway-level analyses indicated that the functions of disease-associated genes partially overlap between the two disease entities. Finally, phenome-wide association analyses of prioritized candidate genes revealed that cerebral blood flow and ulcerative colitis constitute the two main traits associated with both EP and CHD.
Collapse
Affiliation(s)
- Yiming Wu
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Cigdem Sevim Bayrak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bosi Dong
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shixu He
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Peter D Stenson
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - David N Cooper
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - Yuval Itan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, USA.
| | - Lei Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
163
|
Peyvandi S, Rollins C. Fetal Brain Development in Congenital Heart Disease. Can J Cardiol 2023; 39:115-122. [PMID: 36174913 PMCID: PMC9905309 DOI: 10.1016/j.cjca.2022.09.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/08/2022] [Accepted: 09/18/2022] [Indexed: 02/07/2023] Open
Abstract
Neurodevelopmental impairments are the most common extracardiac morbidities among patients with complex congenital heart disease (CHD) across the lifespan. Robust clinical research in this area has revealed several cardiac, medical, and social factors that can contribute to neurodevelopmental outcome in the context of CHD. Studies using brain magnetic resonance imaging (MRI) have been instrumental in identifying quantitative and qualitative difference in brain structure and maturation in this patient population. Full-term newborns with complex CHD are known to have abnormal microstructural and metabolic brain development with patterns similar to those seen in premature infants at approximately 34 to 36 weeks' gestation. With the advent of fetal brain MRI, these brain abnormalities are now documented as they begin in utero, as early as the third trimester. Importantly, disturbed brain development in utero is now known to be independently associated with neurodevelopmental outcome in early childhood, making the prenatal period an important timeframe for potential interventions. Advances in fetal brain MRI provide a robust imaging tool to use in future neuroprotective clinical trials. The causes of abnormal fetal brain development are multifactorial and include cardiovascular physiology, genetic abnormalities, placental impairment, and other environmental and social factors. This review provides an overview of current knowledge of brain development in the context of CHD, common prenatal imaging tools to evaluate the developing fetal brain in CHD, and known risk factors contributing to brain immaturity.
Collapse
Affiliation(s)
- Shabnam Peyvandi
- University of California San Francisco Benioff Children's Hospital, San Francisco, California, USA.
| | - Caitlin Rollins
- Boston Children's Hospital and Harvard University Departments of Neurology, Boston, Massachusetts, USA
| |
Collapse
|
164
|
Patt E, Singhania A, Roberts AE, Morton SU. The Genetics of Neurodevelopment in Congenital Heart Disease. Can J Cardiol 2023; 39:97-114. [PMID: 36183910 DOI: 10.1016/j.cjca.2022.09.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/23/2022] [Accepted: 09/25/2022] [Indexed: 02/07/2023] Open
Abstract
Congenital heart disease (CHD) is the most common birth anomaly, affecting almost 1% of infants. Neurodevelopmental delay is the most common extracardiac feature in people with CHD. Many factors may contribute to neurodevelopmental risk, including genetic factors, CHD physiology, and the prenatal/postnatal environment. Damaging variants are most highly enriched among individuals with extracardiac anomalies or neurodevelopmental delay in addition to CHD, indicating that genetic factors have an impact beyond cardiac tissues in people with CHD. Potential sources of genetic risk include large deletions or duplications that affect multiple genes, such as 22q11 deletion syndrome, single genes that alter both heart and brain development, such as CHD7, and common variants that affect neurodevelopmental resiliency, such as APOE. Increased use of genome-sequencing technologies in studies of neurodevelopmental outcomes in people with CHD will improve our ability to detect relevant genes and variants. Ultimately, such knowledge can lead to improved and more timely intervention of learning support for affected children.
Collapse
Affiliation(s)
- Eli Patt
- Harvard Medical School, Boston, Massachusetts, USA
| | - Asmita Singhania
- School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Amy E Roberts
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sarah U Morton
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA; Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
165
|
Nie S. Use of Frogs as a Model to Study the Etiology of HLHS. J Cardiovasc Dev Dis 2023; 10:51. [PMID: 36826547 PMCID: PMC9965361 DOI: 10.3390/jcdd10020051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
A frog is a classical model organism used to uncover processes and regulations of early vertebrate development, including heart development. Recently, we showed that a frog also represents a useful model to study a rare human congenital heart disease, hypoplastic left heart syndrome. In this review, we first summarized the cellular events and molecular regulations of vertebrate heart development, and the benefit of using a frog model to study congenital heart diseases. Next, we described the challenges in elucidating the etiology of hypoplastic left heart syndrome and discussed how a frog model may contribute to our understanding of the molecular and cellular bases of the disease. We concluded that a frog model offers its unique advantage in uncovering the cellular mechanisms of hypoplastic left heart syndrome; however, combining multiple model organisms, including frogs, is needed to gain a comprehensive understanding of the disease.
Collapse
Affiliation(s)
- Shuyi Nie
- School of Biological Sciences, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
166
|
Suppression of RBFox2 by Multiple MiRNAs in Pressure Overload-Induced Heart Failure. Int J Mol Sci 2023; 24:ijms24021283. [PMID: 36674797 PMCID: PMC9867119 DOI: 10.3390/ijms24021283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/25/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Heart failure is the final stage of various cardiovascular diseases and seriously threatens human health. Increasing mediators have been found to be involved in the pathogenesis of heart failure, including the RNA binding protein RBFox2. It participates in multiple aspects of the regulation of cardiac function and plays a critical role in the process of heart failure. However, how RBFox2 itself is regulated remains unclear. Here, we dissected transcriptomic signatures, including mRNAs and miRNAs, in a mouse model of heart failure after TAC surgery. A global analysis showed that an asymmetric alternation in gene expression and a large-scale upregulation of miRNAs occurred in heart failure. An association analysis revealed that the latter not only contributed to the degradation of numerous mRNA transcripts, but also suppressed the translation of key proteins such as RBFox2. With the aid of Ago2 CLIP-seq data, luciferase assays verified that RBFox2 was targeted by multiple miRNAs, including Let-7, miR-16, and miR-200b, which were significantly upregulated in heart failure. The overexpression of these miRNAs suppressed the RBFox2 protein and its downstream effects in cardiomyocytes, which was evidenced by the suppressed alternative splicing of the Enah gene and impaired E-C coupling via the repression of the Jph2 protein. The inhibition of Let-7, the most abundant miRNA family targeting RBFox2, could restore the RBFox2 protein as well as its downstream effects in dysfunctional cardiomyocytes induced by ISO treatment. In all, these findings revealed the molecular mechanism leading to RBFox2 depression in heart failure, and provided an approach to rescue RBFox2 through miRNA inhibition for the treatment of heart failure.
Collapse
|
167
|
Morton SU, Norris-Brilliant A, Cunningham S, King E, Goldmuntz E, Brueckner M, Miller TA, Thomas NH, Liu C, Adams HR, Bellinger DC, Cleveland J, Cnota JF, Dale AM, Frommelt M, Gelb BD, Grant PE, Goldberg CS, Huang H, Kuperman JM, Li JS, McQuillen PS, Panigrahy A, Porter GA, Roberts AE, Russell MW, Seidman CE, Tivarus ME, Anagnoustou E, Hagler DJ, Chung WK, Newburger JW. Association of Potentially Damaging De Novo Gene Variants With Neurologic Outcomes in Congenital Heart Disease. JAMA Netw Open 2023; 6:e2253191. [PMID: 36701153 PMCID: PMC9880793 DOI: 10.1001/jamanetworkopen.2022.53191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/01/2022] [Indexed: 01/27/2023] Open
Abstract
Importance Neurodevelopmental disabilities are commonly associated with congenital heart disease (CHD), but medical and sociodemographic factors explain only one-third of the variance in outcomes. Objective To examine whether potentially damaging de novo variants (dDNVs) in genes not previously linked to neurodevelopmental disability are associated with neurologic outcomes in CHD and, post hoc, whether some dDNVs or rare putative loss-of-function variants (pLOFs) in specific gene categories are associated with outcomes. Design, Setting, and Participants This cross-sectional study was conducted from September 2017 to June 2020 in 8 US centers. Inclusion criteria were CHD, age 8 years or older, and available exome sequencing data. Individuals with pathogenic gene variants in known CHD- or neurodevelopment-related genes were excluded. Cases and controls were frequency-matched for CHD class, age group, and sex. Exposures Heterozygous for (cases) or lacking (controls) dDNVs in genes not previously associated with neurodevelopmental disability. Participants were separately stratified as heterozygous or not heterozygous for dDNVs and/or pLOFs in 4 gene categories: chromatin modifying, constrained, high level of brain expression, and neurodevelopmental risk. Main Outcomes and Measures Main outcomes were neurodevelopmental assessments of academic achievement, intelligence, fine motor skills, executive function, attention, memory, social cognition, language, adaptive functioning, and anxiety and depression, as well as 7 structural, diffusion, and functional brain magnetic resonance imaging metrics. Results The study cohort included 221 participants in the post hoc analysis and 219 in the case-control analysis (109 cases [49.8%] and 110 controls [50.2%]). Of those 219 participants (median age, 15.0 years [IQR, 10.0-21.2 years]), 120 (54.8%) were male. Cases and controls had similar primary outcomes (reading composite, spelling, and math computation on the Wide Range Achievement Test, Fourth Edition) and secondary outcomes. dDNVs and/or pLOFs in chromatin-modifying genes were associated with lower mean (SD) verbal comprehension index scores (91.4 [20.4] vs 103.4 [17.8]; P = .01), Social Responsiveness Scale, Second Edition, scores (57.3 [17.2] vs 49.4 [11.2]; P = .03), and Wechsler Adult Intelligence Scale, Fourth Edition, working memory scores (73.8 [16.4] vs 97.2 [15.7]; P = .03), as well as higher likelihood of autism spectrum disorder (28.6% vs 5.2%; P = .01). dDNVs and/or pLOFs in constrained genes were associated with lower mean (SD) scores on the Wide Range Assessment of Memory and Learning, Second Edition (immediate story memory: 9.7 [3.7] vs 10.7 [3.0]; P = .03; immediate picture memory: 7.8 [3.1] vs 9.0 [2.9]; P = .008). Adults with dDNVs and/or pLOFs in genes with a high level of brain expression had greater Conners adult attention-deficit hyperactivity disorder rating scale scores (mean [SD], 55.5 [15.4] vs 46.6 [12.3]; P = .007). Conclusions and Relevance The study findings suggest neurodevelopmental outcomes are not associated with dDNVs as a group but may be worse in individuals with dDNVs and/or pLOFs in some gene sets, such as chromatin-modifying genes. Future studies should confirm the importance of specific gene variants to brain function and structure.
Collapse
Affiliation(s)
- Sarah U. Morton
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Ami Norris-Brilliant
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sean Cunningham
- Department of Pediatrics, Division of General Pediatrics, University of Utah, Salt Lake City
| | - Eileen King
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Elizabeth Goldmuntz
- Division of Cardiology, Children’s Hospital of Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Martina Brueckner
- Departments of Genetics and Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Thomas A. Miller
- Department of Pediatrics, Primary Children’s Hospital, University of Utah, Salt Lake City
- Division of Pediatric Cardiology, Maine Medical Center, Portland
| | - Nina H. Thomas
- Department of Child and Adolescent Psychiatry and Behavioral Sciences and Center for Human Phenomic Science, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Psychiatry, University of Pennsylvania, Philadelphia
| | - Chunyan Liu
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Heather R. Adams
- Departments of Neurology and Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - David C. Bellinger
- Departments of Neurology and Psychiatry, Boston Children’s Hospital, Boston, Massachusetts
- Departments of Neurology and Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - John Cleveland
- Departments of Surgery and Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles
| | - James F. Cnota
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
- Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Anders M. Dale
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla
- Department of Radiology, School of Medicine, University of California San Diego, La Jolla
- Departments of Cognitive Science and Neuroscience, University of California San Diego, La Jolla
| | - Michele Frommelt
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee
| | - Bruce D. Gelb
- Mindich Child Health and Development Institute and Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - P. Ellen Grant
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Caren S. Goldberg
- Department of Pediatrics, C.S. Mott Children’s Hospital, University of Michigan, Ann Arbor
| | - Hao Huang
- Department of Radiology, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Joshua M. Kuperman
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla
- Department of Radiology, School of Medicine, University of California San Diego, La Jolla
- Departments of Cognitive Science and Neuroscience, University of California San Diego, La Jolla
| | - Jennifer S. Li
- Division of Pediatric Cardiology, Duke University, Durham, North Carolina
| | - Patrick S. McQuillen
- Departments of Pediatrics and Neurology, University of California, San Francisco
| | - Ashok Panigrahy
- Department of Pediatric Radiology, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - George A. Porter
- Departments of Neurology and Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Amy E. Roberts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, Massachusetts
| | - Mark W. Russell
- Department of Pediatrics, C.S. Mott Children’s Hospital, University of Michigan, Ann Arbor
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Madalina E. Tivarus
- Departments of Imaging Sciences and Neuroscience, University of Rochester Medical Center, Rochester, New York
| | - Evdokia Anagnoustou
- Department of Pediatrics, University of Toronto, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Donald J. Hagler
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla
- Department of Radiology, School of Medicine, University of California San Diego, La Jolla
- Departments of Cognitive Science and Neuroscience, University of California San Diego, La Jolla
| | - Wendy K. Chung
- Departments of Pediatrics and Medicine, Columbia University Medical Center, New York, New York
| | - Jane W. Newburger
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
| |
Collapse
|
168
|
Jia Y, Chen J, Zhong J, He X, Zeng L, Wang Y, Li J, Xia S, Ye E, Zhao J, Ke B, Li C. Novel rare mutation in a conserved site of PTPRB causes human hypoplastic left heart syndrome. Clin Genet 2023; 103:79-86. [PMID: 36148623 DOI: 10.1111/cge.14234] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 12/14/2022]
Abstract
Hypoplastic left heart syndrome (HLHS) is a rare but fatal birth defect in which the left side of the heart is underdeveloped. HLHS accounts for 2% to 4% of congenital heart anomalies. Whole genome sequencing (WGS) was conducted for a family trio consisting of a proband and his parents. A homozygous rare variant was detected in the PTPRB (Protein Tyrosine Phosphatase Receptor Type B) gene of the proband by functional annotation and co-segregation analysis. Sanger sequencing was used to confirm genotypes of the variant. The in silico prediction tools, including Mutation Taster, SpliceAI, and CADD, were used to predict the impact of the mutation. The allele frequencies across populations were compared based on multiple databases, including "1000 genomes" and "gnomAD". We used two vectors (pcMINI and pcDNA3.1) to generate a minigene construct to validate the mutational effect at the transcriptional level. Family-based WGS analyses showed that only a homozygous splice acceptor variant (NC_000012.12: g.70636068T>G, NM_001109754.4: c.56-2A>C, NG_029940.2: g.6373A>C) at the exon-intron border of PTPRB gene associates with HLHS. This variant is also within the region with the enhancer activity based on UCSC genome annotation. Genotyping and Sanger sequencing revealed that the proband's parents are heterozygous for this variant. Evolutionary conservation analysis revealed that the site (NC_000012.12: g.70636068) is extremely conserved across species, supporting the evolutionary functional constraints of the ancestral wild type (T). In silico tools universally predicted a deleterious or disease-causing impact of the mutation from T to G. The mutation was not found in the 1000 genomes and gnomAD databases, which indicates that this mutation is very rare in most human populations. A splicing assay indicated that the mutated minigene caused aberrant splicing of mRNA, in which a 3 bp missing in the second exon resulted in the deletion of one amino acid (NP_001103224.1:p.Glu19del) compared to the normal protein of PRPTB (also the VE-PTP). Structure prediction revealed that the deletion occurred within the C-region of the signal peptide of VE-PTP, suggesting signal peptide-related defects as a potential mechanism for the HLHS cellular pathogeny. We report a rare homozygous variant with splicing error in PTPRB associated with HLHS. Previous model species studies revealed conserved functions of PTPRB in cardiovascular and heart development in mice and zebrafish. Our study is the first report to show the association between PTPRB and HLHS in humans.
Collapse
Affiliation(s)
- Yangying Jia
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jianhai Chen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhong
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xuefei He
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zeng
- The Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yanmin Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Jiakun Li
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Shengqian Xia
- Department of Ecology and Evolution, The University of Chicago, Chicago, Illinois, USA
| | - Erdengqieqieke Ye
- Department of Prenatal Diagnosis, Reproductive Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jing Zhao
- Department of Prenatal Diagnosis, Reproductive Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Bin Ke
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
169
|
Cai R, Tan Y, Wang M, Yu H, Wang J, Ren Z, Dong Z, He Y, Li Z, Lin L, Gu Y. Detection of Novel Pathogenic Variants in Two Families with Recurrent Fetal Congenital Heart Defects. Pharmgenomics Pers Med 2023; 16:173-181. [PMID: 36923242 PMCID: PMC10008912 DOI: 10.2147/pgpm.s394120] [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: 10/27/2022] [Accepted: 02/17/2023] [Indexed: 03/10/2023] Open
Abstract
Background Congenital heart disease (CHD) is the most common birth defect with strong genetic heterogeneity. To date, about 400 genes have been linked to CHD, including cell signaling molecules, transcription factors, and structural proteins that are important for heart development. Genetic analysis of CHD cases is crucial for clinical management and etiological analysis. Methods Whole-exome sequencing (WES) was performed to identify the genetic variants in two independent CHD cases with DNA samples from fetuses and their parents, followed by the exclusion of aneuploidy and large copy number variations (CNVs). The WES results were verified by Sanger sequencing. Results In family A, a compound heterozygous variation in PLD1 gene consisting of c.1132dupA (p.I378fs) and c.1171C>T (p.R391C) was identified in the fetus. The two variants were inherited from the father (c.1132dupA) and the mother (c.1171C>T), respectively. In family B, a hemizygous variant ZIC3: c.861delG (p.G289Afs*119) was identified in the fetus, which was inherited from the heterozygous mother. We further confirmed that these variants PLD1: c.1132dupA and ZIC3: c.861delG were novel. Conclusion The findings in our study identified novel variants to the mutation spectrum of CHD and provided reliable evidence for the recurrent risk and reproductive care options to the affected families. Our study also demonstrates that WES has considerable prospects of clinical application in prenatal diagnosis.
Collapse
Affiliation(s)
- Rongqin Cai
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Ya Tan
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Mingming Wang
- Be Creative Lab (Beijing) Co. Ltd, Beijing, 101111, People's Republic of China
| | - Huijun Yu
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Zhuo Ren
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Zhe Dong
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Yiwen He
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Zhi Li
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Li Lin
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China
| | - Ying Gu
- Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, 102206, People's Republic of China.,Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, 222000, People's Republic of China
| |
Collapse
|
170
|
Banerjee D, Girirajan S. Pathogenic Variants and Ascertainment: Neuropsychiatric Disease Risk in a Health System Cohort. Am J Psychiatry 2023; 180:11-13. [PMID: 36587271 PMCID: PMC9995216 DOI: 10.1176/appi.ajp.20220934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Deepro Banerjee
- Bioinformatics and Genomics Program, Huck Institutes of Life Sciences, University Park, Pa. (Banerjee, Girirajan); Department of Biochemistry and Molecular Biology and Department of Anthropology, Pennsylvania State University, University Park, Pa. (Girirajan)
| | - Santhosh Girirajan
- Bioinformatics and Genomics Program, Huck Institutes of Life Sciences, University Park, Pa. (Banerjee, Girirajan); Department of Biochemistry and Molecular Biology and Department of Anthropology, Pennsylvania State University, University Park, Pa. (Girirajan)
| |
Collapse
|
171
|
Lee FT, Sun L, Freud L, Seed M. A guide to prenatal counseling regarding neurodevelopment in congenital heart disease. Prenat Diagn 2022; 43:661-673. [PMID: 36575573 DOI: 10.1002/pd.6292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
Advances in cardiac surgical techniques taking place over the past 50 years have resulted in the vast majority of children born with congenital cardiac malformations now surviving into adulthood. As the focus shifts from survival to the functional outcomes of our patients, it is increasingly being recognized that a significant proportion of patients undergoing infant cardiac repair experience adverse neurodevelopmental (ND) outcomes. The etiology of abnormal brain development in the setting of congenital heart disease is poorly understood, complex, and likely multifactorial. Furthermore, the efficacy of therapies available for the learning disabilities, attention deficit, and hyperactivity disorders and other ND deficits complicating congenital heart disease is currently uncertain. This situation presents a challenge for prenatal counseling as current antenatal testing does not usually provide prognostic information regarding the likely ND trajectories of individual patients. However, we believe it is important for parents to be informed about potential issues with child development when a new diagnosis of congenital heart disease is disclosed. Parents deserve a comprehensive and thoughtful approach to this subject, which conveys the uncertainties involved in predicting the severity of any developmental disorders encountered, while emphasizing the improvements in outcomes that have already been achieved in infants with congenital heart disease. A balanced approach to counseling should also discuss what local arrangements are in place for ND follow-up. This review presents an up-to-date overview of ND outcomes in patients with congenital heart disease, providing possible approaches to communicating this information to parents during prenatal counseling in a sensitive and accurate manner.
Collapse
Affiliation(s)
- Fu-Tsuen Lee
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Liqun Sun
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Lindsay Freud
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Mike Seed
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
172
|
Ameen M, Sundaram L, Shen M, Banerjee A, Kundu S, Nair S, Shcherbina A, Gu M, Wilson KD, Varadarajan A, Vadgama N, Balsubramani A, Wu JC, Engreitz JM, Farh K, Karakikes I, Wang KC, Quertermous T, Greenleaf WJ, Kundaje A. Integrative single-cell analysis of cardiogenesis identifies developmental trajectories and non-coding mutations in congenital heart disease. Cell 2022; 185:4937-4953.e23. [PMID: 36563664 PMCID: PMC10122433 DOI: 10.1016/j.cell.2022.11.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 09/13/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
To define the multi-cellular epigenomic and transcriptional landscape of cardiac cellular development, we generated single-cell chromatin accessibility maps of human fetal heart tissues. We identified eight major differentiation trajectories involving primary cardiac cell types, each associated with dynamic transcription factor (TF) activity signatures. We contrasted regulatory landscapes of iPSC-derived cardiac cell types and their in vivo counterparts, which enabled optimization of in vitro differentiation of epicardial cells. Further, we interpreted sequence based deep learning models of cell-type-resolved chromatin accessibility profiles to decipher underlying TF motif lexicons. De novo mutations predicted to affect chromatin accessibility in arterial endothelium were enriched in congenital heart disease (CHD) cases vs. controls. In vitro studies in iPSCs validated the functional impact of identified variation on the predicted developmental cell types. This work thus defines the cell-type-resolved cis-regulatory sequence determinants of heart development and identifies disruption of cell type-specific regulatory elements in CHD.
Collapse
Affiliation(s)
- Mohamed Ameen
- Department of Cancer Biology, Stanford University, Stanford, CA, USA; Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA
| | - Laksshman Sundaram
- Department of Computer Science, Stanford University, Stanford, CA, USA; Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA
| | - Mengcheng Shen
- Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Abhimanyu Banerjee
- Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA; Department of Physics, Stanford University, Stanford, CA, USA
| | - Soumya Kundu
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Anna Shcherbina
- Department of Biomedical Informatics, Stanford University, Stanford, CA, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Avyay Varadarajan
- Department of Computer Science, California Institute of Technology, Pasadena, CA, USA
| | - Nirmal Vadgama
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | | | - Joseph C Wu
- Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | | | - Kyle Farh
- Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, CA, USA
| | - Ioannis Karakikes
- Cardiovascular Institute, Stanford University, Stanford, CA, USA; Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA.
| | - Kevin C Wang
- Department of Cancer Biology, Stanford University, Stanford, CA, USA; Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA; Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA.
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA, USA; Department of Applied Physics, Stanford University, Stanford, CA, USA.
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
173
|
Ramond F, Dalgliesh C, Grimmel M, Wechsberg O, Vetro A, Guerrini R, FitzPatrick D, Poole RL, Lebrun M, Bayat A, Grasshoff U, Bertrand M, Witt D, Turnpenny PD, Faundes V, Santa María L, Mendoza Fuentes C, Mabe P, Hussain SA, Mullegama SV, Torti E, Oehl-Jaschkowitz B, Salmon LB, Orenstein N, Shahar NR, Hagari O, Bazak L, Hoffjan S, Prada CE, Haack T, Elliott DJ. Clustered variants in the 5' coding region of TRA2B cause a distinctive neurodevelopmental syndrome. Genet Med 2022; 25:100003. [PMID: 36549593 DOI: 10.1016/j.gim.2022.100003] [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: 09/21/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Transformer2 proteins (Tra2α and Tra2β) control splicing patterns in human cells, and no human phenotypes have been associated with germline variants in these genes. The aim of this work was to associate germline variants in the TRA2B gene to a novel neurodevelopmental disorder. METHODS A total of 12 individuals from 11 unrelated families who harbored predicted loss-of-function monoallelic variants, mostly de novo, were recruited. RNA sequencing and western blot analyses of Tra2β-1 and Tra2β-3 isoforms from patient-derived cells were performed. Tra2β1-GFP, Tra2β3-GFP and CHEK1 exon 3 plasmids were transfected into HEK-293 cells. RESULTS All variants clustered in the 5' part of TRA2B, upstream of an alternative translation start site responsible for the expression of the noncanonical Tra2β-3 isoform. All affected individuals presented intellectual disability and/or developmental delay, frequently associated with infantile spasms, microcephaly, brain anomalies, autism spectrum disorder, feeding difficulties, and short stature. Experimental studies showed that these variants decreased the expression of the canonical Tra2β-1 isoform, whereas they increased the expression of the Tra2β-3 isoform, which is shorter and lacks the N-terminal RS1 domain. Increased expression of Tra2β-3-GFP were shown to interfere with the incorporation of CHEK1 exon 3 into its mature transcript, normally incorporated by Tra2β-1. CONCLUSION Predicted loss-of-function variants clustered in the 5' portion of TRA2B cause a new neurodevelopmental syndrome through an apparently dominant negative disease mechanism involving the use of an alternative translation start site and the overexpression of a shorter, repressive Tra2β protein.
Collapse
Affiliation(s)
- Francis Ramond
- Service de Génétique, Hôpital Nord, CHU Saint-Etienne, Saint-Etienne, France.
| | - Caroline Dalgliesh
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mona Grimmel
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tübingen, Germany
| | - Oded Wechsberg
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Maccabi Healthcare Services, Tel Aviv, Israel
| | - Annalisa Vetro
- Neuroscience Department, Meyer Children's Hospital and University of Florence, Florence, Italy
| | - Renzo Guerrini
- Neuroscience Department, Meyer Children's Hospital and University of Florence, Florence, Italy
| | - David FitzPatrick
- MRC Human Genetics Unit, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Rebecca L Poole
- NHS Education for Scotland South East Region, South East of Scotland Clinical Genetics Service, Edinburgh, United Kingdom
| | - Marine Lebrun
- Service de Génétique, Hôpital Nord, CHU Saint-Etienne, Saint-Etienne, France
| | - Allan Bayat
- Institute for Regional Health Services, University of Southern Denmark, Odense, Denmark; Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Center, Dianalund, Denmark
| | - Ute Grasshoff
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tübingen, Germany
| | - Miriam Bertrand
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tübingen, Germany
| | - Dennis Witt
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tübingen, Germany
| | - Peter D Turnpenny
- Clinical Genetics, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Víctor Faundes
- Laboratorio de Genética y Enfermedades Metabólicas, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Lorena Santa María
- Laboratorio de Genética y Enfermedades Metabólicas, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Carolina Mendoza Fuentes
- Unidad de Endocrinología, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Mabe
- Unidad de Neurología, Hospital de Niños Dr. Exequiel González Cortés, Santiago, Chile
| | - Shaun A Hussain
- Division of Pediatric Neurology, University of California, Los Angeles, Los Angeles, CA
| | | | | | | | - Lina Basel Salmon
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Raphael Recanati Genetic Institute, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel; Pediatric Immunogenetics, Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Naama Orenstein
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noa Ruhrman Shahar
- The Raphael Recanati Genetic Institute, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Ofir Hagari
- The Raphael Recanati Genetic Institute, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Lily Bazak
- The Raphael Recanati Genetic Institute, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Sabine Hoffjan
- Abteilung für Humangenetik, Ruhr-Universitat Bochum, Bochum, Germany
| | - Carlos E Prada
- Division of Genetics, Birth Defects and Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Department of Pediatrics, Feinberg School of Medicine of Northwestern University, Chicago, IL
| | - Tobias Haack
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tübingen, Germany; Centre for Rare Diseases, University of Tuebingen, Tuebingen, Germany
| | - David J Elliott
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
174
|
Yi T, Hao X, Sun H, Zhang Y, Han J, Gu X, Sun L, Liu X, Zhao Y, Guo Y, Zhou X, He Y. Genetic aetiology distribution of 398 foetuses with congenital heart disease in the prenatal setting. ESC Heart Fail 2022; 10:917-930. [PMID: 36478645 PMCID: PMC10053168 DOI: 10.1002/ehf2.14209] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/24/2022] [Accepted: 10/02/2022] [Indexed: 12/12/2022] Open
Abstract
AIMS Copy number variant-sequencing (CNV-seq) and exome sequencing (ES) have been used as powerful tools in understanding the role of genetic variants in congenital heart diseases (CHDs). A few previous large cohort studies have utilized CNV-seq and ES to investigate prenatally diagnosed CHD. Here, we sought to determine the value of CNV-seq and ES for genetic evaluation of foetal CHDs. METHODS AND RESULTS We recruited 398 pregnant women diagnosed with CHDs between 8 January 2017 and 30 November 2020. CNV-seq and ES were performed on foetal and parent samples. CHD cases were classified following the guidelines of the International Paediatric and Congenital Cardiac Code and the Tenth and Eleventh Revisions of the International Classification of Diseases. Data on aneuploids (AUP), pathogenic CNVs (pCNVs), and single nucleotide variants (SNVs) were collected and compared, following appropriate procedures. We identified genetic abnormalities in 129 (32.41%) foetuses. These abnormalities included AUP (10.80%), pCNVs (13.32%), and SNVs (8.04%). ES analysis yielded higher SNVs in cases without AUP or pCNVs. Non-isolated CHDs were associated with higher genetic abnormalities than isolated CHDs, mainly due to AUP differences between the two groups. The prevalence of genetic defects was the highest in foetuses with atrioventricular septal defects (AVSD), left ventricular outflow tract obstruction (LVOTO), and conotruncal defects (CTD). AVSD and anomalies of atrioventricular junctions and valves were associated with AUP abnormalities. CTD, anomalies of extrapericardial arterial trunks, and anomalies of the ventricular outflow tracts were the most common CHD categories diagnosed using CNVs. The most common CHDs associated with single ventricle (SV) abnormalities were heterotaxy (Hex) (14.89%), LVOTO (14.58%), and ventricular septal defect (VSD) (26.67%, 4/15). Although the ES yields were higher than CNV-seq for VSD (44.4%, 4/9), LVOTO (20%, 7/35), Hex (14.89%, 7/47), and CTD (9.1%, 11/121), its diagnostic yield did not increase for SV (6.7%, 1/15), AVSD (3.8%, 1/26), or right ventricular obstruction defects (2.6%, 1/38). The most common mutations were observed in KMT2D, CHD7, and NOTCH1. CONCLUSIONS To our knowledge, this is the largest cohort study to investigate the incidence of SNVs using ES in foetal CHD. CNV-seq and ES identified genetic abnormalities in nearly 1/3 of foetal CHD cases. Thus, CNV-seq and ES can provide clinically relevant information for pregnancy management.
Collapse
Affiliation(s)
- Tong Yi
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaoyan Hao
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Hairui Sun
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Ye Zhang
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Jiancheng Han
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaoyan Gu
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Lin Sun
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaowei Liu
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Ying Zhao
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Yong Guo
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Xiaoxue Zhou
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| | - Yihua He
- Beijing Key Laboratory of Maternal‐Fetal Medicine and Fetal Heart Disease Capital Medical University Affiliated Anzhen Hospital Beijing China
| |
Collapse
|
175
|
Akerberg AA, Trembley M, Butty V, Schwertner A, Zhao L, Beerens M, Liu X, Mahamdeh M, Yuan S, Boyer L, MacRae C, Nguyen C, Pu WT, Burns CE, Burns CG. RBPMS2 Is a Myocardial-Enriched Splicing Regulator Required for Cardiac Function. Circ Res 2022; 131:980-1000. [PMID: 36367103 PMCID: PMC9770155 DOI: 10.1161/circresaha.122.321728] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND RBPs (RNA-binding proteins) perform indispensable functions in the post-transcriptional regulation of gene expression. Numerous RBPs have been implicated in cardiac development or physiology based on gene knockout studies and the identification of pathogenic RBP gene mutations in monogenic heart disorders. The discovery and characterization of additional RBPs performing indispensable functions in the heart will advance basic and translational cardiovascular research. METHODS We performed a differential expression screen in zebrafish embryos to identify genes enriched in nkx2.5-positive cardiomyocytes or cardiopharyngeal progenitors compared to nkx2.5-negative cells from the same embryos. We investigated the myocardial-enriched gene RNA-binding protein with multiple splicing (variants) 2 [RBPMS2)] by generating and characterizing rbpms2 knockout zebrafish and human cardiomyocytes derived from RBPMS2-deficient induced pluripotent stem cells. RESULTS We identified 1848 genes enriched in the nkx2.5-positive population. Among the most highly enriched genes, most with well-established functions in the heart, we discovered the ohnologs rbpms2a and rbpms2b, which encode an evolutionarily conserved RBP. Rbpms2 localizes selectively to cardiomyocytes during zebrafish heart development and strong cardiomyocyte expression persists into adulthood. Rbpms2-deficient embryos suffer from early cardiac dysfunction characterized by reduced ejection fraction. The functional deficit is accompanied by myofibril disarray, altered calcium handling, and differential alternative splicing events in mutant cardiomyocytes. These phenotypes are also observed in RBPMS2-deficient human cardiomyocytes, indicative of conserved molecular and cellular function. RNA-sequencing and comparative analysis of genes mis-spliced in RBPMS2-deficient zebrafish and human cardiomyocytes uncovered a conserved network of 29 ortholog pairs that require RBPMS2 for alternative splicing regulation, including RBFOX2, SLC8A1, and MYBPC3. CONCLUSIONS Our study identifies RBPMS2 as a conserved regulator of alternative splicing, myofibrillar organization, and calcium handling in zebrafish and human cardiomyocytes.
Collapse
Affiliation(s)
- Alexander A. Akerberg
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Michael Trembley
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Vincent Butty
- BioMicroCenter, Department of Biology (V.B.), Massachusetts Institute of Technology, Cambridge‚ MA
- Department of Biology (V.B., L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
| | - Asya Schwertner
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Long Zhao
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Manu Beerens
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (M.B., C.M.)
| | - Xujie Liu
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Mohammed Mahamdeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Shiaulou Yuan
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Laurie Boyer
- Department of Biology (V.B., L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
- Department of Biological Engineering (L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
| | - Calum MacRae
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (M.B., C.M.)
| | - Christopher Nguyen
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Innovation Research Center, Heart Vascular & Thoracic Institute, Cleveland Clinic‚ Cleveland‚ OH (C.N.)
| | - William T. Pu
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Harvard Stem Cell Institute, Cambridge, MA (W.T.P., C.E.B.)
| | - Caroline E. Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Harvard Stem Cell Institute, Cambridge, MA (W.T.P., C.E.B.)
| | - C. Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| |
Collapse
|
176
|
Liu S, Wei W, Wang P, Liu C, Jiang X, Li T, Li F, Wu Y, Chen S, Sun K, Xu R. LOF variants identifying candidate genes of laterality defects patients with congenital heart disease. PLoS Genet 2022; 18:e1010530. [PMID: 36459505 DOI: 10.1371/journal.pgen.1010530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 12/14/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Defects in laterality pattern can result in abnormal positioning of the internal organs during the early stages of embryogenesis, as manifested in heterotaxy syndrome and situs inversus, while laterality defects account for 3~7% of all congenital heart defects (CHDs). However, the pathogenic mechanism underlying most laterality defects remains unknown. In this study, we recruited 70 laterality defect patients with CHDs to identify candidate disease genes by exome sequencing. We then evaluated rare, loss-of-function (LOF) variants, identifying candidates by referring to previous literature. We chose TRIP11, DNHD1, CFAP74, and EGR4 as candidates from 776 LOF variants that met the initial screening criteria. After the variants-to-gene mapping, we performed function research on these candidate genes. The expression patterns and functions of these four candidate genes were studied by whole-mount in situ hybridization, gene knockdown, and gene rescue methods in zebrafish models. Among the four genes, trip11, dnhd1, and cfap74 morphant zebrafish displayed abnormalities in both cardiac looping and expression patterns of early signaling molecules, suggesting that these genes play important roles in the establishment of laterality patterns. Furthermore, we performed immunostaining and high-speed cilia video microscopy to investigate Kupffer's vesicle organogenesis and ciliogenesis of morphant zebrafish. Impairments of Kupffer's vesicle organogenesis or ciliogenesis were found in trip11, dnhd1, and cfap74 morphant zebrafish, which revealed the possible pathogenic mechanism of their LOF variants in laterality defects. These results highlight the importance of rare, LOF variants in identifying disease-related genes and identifying new roles for TRIP11, DNHD1, and CFAP74 in left-right patterning. Additionally, these findings are consistent with the complex genetics of laterality defects.
Collapse
Affiliation(s)
- Sijie Liu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Wei
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pengcheng Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunjie Liu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuechao Jiang
- Scientific Research Center, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Li
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fen Li
- Department of Cardiology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yurong Wu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rang Xu
- Scientific Research Center, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
177
|
Ng JK, Vats P, Fritz-Waters E, Sarkar S, Sams EI, Padhi EM, Payne ZL, Leonard S, West MA, Prince C, Trani L, Jansen M, Vacek G, Samadi M, Harkins TT, Pohl C, Turner TN. de novo variant calling identifies cancer mutation signatures in the 1000 Genomes Project. Hum Mutat 2022; 43:1979-1993. [PMID: 36054329 PMCID: PMC9771978 DOI: 10.1002/humu.24455] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 07/22/2022] [Accepted: 08/29/2022] [Indexed: 01/25/2023]
Abstract
Detection of de novo variants (DNVs) is critical for studies of disease-related variation and mutation rates. To accelerate DNV calling, we developed a graphics processing units-based workflow. We applied our workflow to whole-genome sequencing data from three parent-child sequenced cohorts including the Simons Simplex Collection (SSC), Simons Foundation Powering Autism Research (SPARK), and the 1000 Genomes Project (1000G) that were sequenced using DNA from blood, saliva, and lymphoblastoid cell lines (LCLs), respectively. The SSC and SPARK DNV callsets were within expectations for number of DNVs, percent at CpG sites, phasing to the paternal chromosome of origin, and average allele balance. However, the 1000G DNV callset was not within expectations and contained excessive DNVs that are likely cell line artifacts. Mutation signature analysis revealed 30% of 1000G DNV signatures matched B-cell lymphoma. Furthermore, we found variants in DNA repair genes and at Clinvar pathogenic or likely-pathogenic sites and significant excess of protein-coding DNVs in IGLL5; a gene known to be involved in B-cell lymphomas. Our study provides a new rapid DNV caller for the field and elucidates important implications of using sequencing data from LCLs for reference building and disease-related projects.
Collapse
Affiliation(s)
- Jeffrey K. Ng
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pankaj Vats
- NVIDIA Corporation, Santa Clara, California, USA
| | - Elyn Fritz-Waters
- Research Infrastructure Services, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephanie Sarkar
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Eleanor I. Sams
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Evin M. Padhi
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zachary L. Payne
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shawn Leonard
- Research Infrastructure Services, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marc A. West
- NVIDIA Corporation, Santa Clara, California, USA
| | - Chandler Prince
- Research Infrastructure Services, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lee Trani
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marshall Jansen
- Research Infrastructure Services, Washington University School of Medicine, St. Louis, Missouri, USA
| | - George Vacek
- NVIDIA Corporation, Santa Clara, California, USA
| | | | | | - Craig Pohl
- Research Infrastructure Services, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tychele N. Turner
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
178
|
Self- and informant-reported executive function in young adults operated for atrial or ventricular septal defects in childhood. Cardiol Young 2022; 32:1917-1924. [PMID: 34991743 DOI: 10.1017/s1047951121005151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Adults with simple congenital heart defects (CHD) have increased risk of neurodevelopmental challenges including executive dysfunction. It is unknown if the executive dysfunction is universal or if it is driven by dysfunction in specific clinical subscales and how it might affect psychosocial aspects of everyday life. METHODS The self-reported and informant-reported executive function of adults with an average age of 26 ± 5 (range 18-41) who underwent childhood surgery for atrial septal defects (n = 34) or ventricular septal defects (n = 32) and matched controls (n = 40) were evaluated using the Behavior Rating Inventory of Executive Functions - Adult version (BRIEF-A). RESULTS The CHD group reported having more executive dysfunction than controls in all BRIEF-A clinical subscales (p < 0.020) and more than their informants reported on their behalf (p < 0.006). The CHD group had received three times more special teaching (44% compared to 16%) and pedagogical psychological counselling (14% compared to none) and had a three times higher occurrence of psychiatric disorders than controls (33% compared to 11%). Lower educational levels and psychiatric disorders were associated with higher BRIEF-A scores (p < 0.03). CONCLUSIONS Adults operated for septal defects in childhood report more challenges with all aspects of the executive functions than controls and more than relatives are aware of.
Collapse
|
179
|
Gelb BD. Prospects for precision genetic medicine in congenital heart disease. Curr Opin Genet Dev 2022; 77:101983. [PMID: 36115276 PMCID: PMC9729438 DOI: 10.1016/j.gde.2022.101983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 01/27/2023]
Abstract
Precision medicine, defined as tailoring medical care individually based upon relevant factors, is primarily implemented currently through the use of genetic variation. Over the past thirty years, the possibility of determining specific genetic variants underlying congenital heart disease has increased dramatically. This has created the potential for using precision genetic approaches to improve care and outcomes for patients and families with congenital heart disease. In this review, recent advances in understanding the roles of genetic variants in various outcomes, in developing novel therapeutic approaches, and in refining clinical trials for congenital heart disease are discussed.
Collapse
Affiliation(s)
- Bruce D Gelb
- Mindich Child Health and Development Institute and the Departments of Pediatrics and Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
180
|
Congenital Heart Diseases: Genetic Risk Variants and Their Methylation Status. Genes (Basel) 2022; 13:genes13112115. [DOI: 10.3390/genes13112115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/01/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The interaction between single nucleotide variants (SNVs) associated with congenital heart diseases (CHDs) and their gene methylation status has not been well researched. The aim of the present study was to determine if there is a relationship between the methy lation status (MS) of genes and the allelic variants associated with CHDs. (2) Methods: Seven SNVs of the genes AXIN1, TBX1, TBX20, and MTHFR were selected from the literature. DNA extraction, genotyping, and a methylation analysis were performed on healthy subjects and subjects with CHDs. (3) Results: Twenty-two subjects with CHDs were selected as the case group (15 with ventricular septal defects (VSDs) and 7 with atrial septal defects (ASDs)), and 44 healthy subjects comprised the control group. The MTHFR and AXIN1 genes were hypermethylated in the control group when compared to the case group. When analyzed separately, those with atrial septum defects exhibited greater methylation, except for the gene MTHFR where there were no differences. Only the alternate alleles of MTHFR showed a significantly different methylation status in those without cardiopathy. (4) Conclusions: The MTHFR and AXIN genes were hypermethylated in the control group; however, only the alternate alleles of MTHFR (rs1801133 and rs1801131) showed a significantly different methylation status.
Collapse
|
181
|
Ma Q, Yang Y, Liu Y. Associations between NKX2-5 gene polymorphisms and congenital heart disease in the Chinese Tibetan population. Am J Transl Res 2022; 14:8407-8415. [PMID: 36505279 PMCID: PMC9730064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/14/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND The pathogenesis of congenital heart disease (CHD) has not been fully elucidated, and this study considers the interaction between inheritance and the environment as the main cause of CHD. Previous studies have found that the incidence of CHD in the Tibetan plateau population is significantly higher than in low-altitude populations. Numerous reports have confirmed that NKX2-5 gene mutations can lead to coronary heart disease, but the relationship between NKX2-5 and Tibetan nationality has not yet been reported. OBJECTIVE To explore the relationship between NKX2-5 gene polymorphisms and CHD in Tibetan people. METHODS Blood samples were collected retrospectively from Tibetan patients diagnosed with CHD as well as healthy Tibetans, and the exons of NKX2-5 were sequenced. The MassARRAY technique was used to detect and genotype candidate tag single nucleotide polymorphisms (SNPs) in the non-coding regions of NKX2-5. RESULTS Exon sequencing revealed no difference in the coding regions of the NKX2-5 gene between the CHD and control groups. In the non-coding regions of NKX2-5, rs6882776 and rs2546741 differed significantly between the two groups. Strong linkage disequilibrium was found between the selected sites of NKX2-5. CONCLUSIONS The NKX2-5 exons do not associate with CHD in Tibetans. Rs6882776 and rs2546741 in the non-coding regions of NKX2-5 may protect against CHD in Tibetans. The NKX2-5 haplotype associated with CHD occurrence in the Tibetan population.
Collapse
Affiliation(s)
- Qiang Ma
- High Altitude Medical Research Center, Medical College of Qinghai University16 Kunlun Road, Xining 810001, Qinghai, China,Department of Pathology, Sunshine Union HospitalYingqian Road, Weifang 261000, Shandong, China
| | - Yingzhong Yang
- High Altitude Medical Research Center, Medical College of Qinghai University16 Kunlun Road, Xining 810001, Qinghai, China
| | - Yongnian Liu
- High Altitude Medical Research Center, Medical College of Qinghai University16 Kunlun Road, Xining 810001, Qinghai, China
| |
Collapse
|
182
|
Ortinau CM, Smyser CD, Arthur L, Gordon EE, Heydarian HC, Wolovits J, Nedrelow J, Marino BS, Levy VY. Optimizing Neurodevelopmental Outcomes in Neonates With Congenital Heart Disease. Pediatrics 2022; 150:e2022056415L. [PMID: 36317967 PMCID: PMC10435013 DOI: 10.1542/peds.2022-056415l] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 11/05/2022] Open
Abstract
Neurodevelopmental impairment is a common and important long-term morbidity among infants with congenital heart disease (CHD). More than half of those with complex CHD will demonstrate some form of neurodevelopmental, neurocognitive, and/or psychosocial dysfunction requiring specialized care and impacting long-term quality of life. Preventing brain injury and treating long-term neurologic sequelae in this high-risk clinical population is imperative for improving neurodevelopmental and psychosocial outcomes. Thus, cardiac neurodevelopmental care is now at the forefront of clinical and research efforts. Initial research primarily focused on neurocritical care and operative strategies to mitigate brain injury. As the field has evolved, investigations have shifted to understanding the prenatal, genetic, and environmental contributions to impaired neurodevelopment. This article summarizes the recent literature detailing the brain abnormalities affecting neurodevelopment in children with CHD, the impact of genetics on neurodevelopmental outcomes, and the best practices for neonatal neurocritical care, focusing on developmental care and parental support as new areas of importance. A framework is also provided for the infrastructure and resources needed to support CHD families across the continuum of care settings.
Collapse
Affiliation(s)
- Cynthia M. Ortinau
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri
| | - Christopher D. Smyser
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri
| | - Lindsay Arthur
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Erin E. Gordon
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Haleh C. Heydarian
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Cardiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Joshua Wolovits
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jonathan Nedrelow
- Department of Neonatology, Cook Children’s Medical Center, Fort Worth, Texas
| | - Bradley S. Marino
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Divisions of Cardiology and Critical Care Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago
| | - Victor Y. Levy
- Department of Pediatrics, Stanford University School of Medicine, Lucile Packard Children’s Hospital, Palo Alto, California
| |
Collapse
|
183
|
Pfitzer C, Sievers LK, Hütter A, Khaliq HA, Poryo M, Berger F, Bauer UMM, Helm PC, Schmitt KRL. Microcephaly is associated with impaired educational development in children with congenital heart disease. Front Cardiovasc Med 2022; 9:917507. [PMID: 36277771 PMCID: PMC9584804 DOI: 10.3389/fcvm.2022.917507] [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: 04/11/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives This study aims to evaluate the school careers of patients with congenital heart disease (CHD) and microcephaly. Methods An exploratory online survey was conducted on patients from a previous study on somatic development in children with CHD in 2018 (n = 2818). A total of 750 patients participated in the online survey (26.6%). This publication focuses on 91 patients (12.1%) diagnosed with CHD and microcephaly who participated in the new online survey. Results Microcephaly was significantly associated with CHD severity (p < 0.001). Microcephalic patients suffered from psychiatric comorbidity two times as often (67.0%) as non-microcephalic patients (29.8%). In particular, the percentage of patients with developmental delay, intellectual debility, social disability, learning disorder, or language disorder was significantly increased in microcephalic CHD patients (p < 0.001). A total of 85.7% of microcephalic patients and 47.6% of non-microcephalic patients received early interventions to foster their development. The school enrollment of both groups was similar at approximately six years of age. However, 89.9% of non-microcephalic but only 51.6% of microcephalic patients were enrolled in a regular elementary school. Regarding secondary school, only half as many microcephalic patients (14.3%) went to grammar school, while the proportion of pupils at special schools was eight times higher. Supportive interventions, e.g., for specific learning disabilities, were used by 52.7% of microcephalic patients and 21.6% of non-microcephalic patients. Conclusion Patients with CHD and microcephaly are at high risk for impaired educational development. Early identification should alert clinicians to provide targeted interventions to optimize the developmental potential.
Collapse
Affiliation(s)
- Constanze Pfitzer
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany,Berlin Institute of Health (BIH), Berlin, Germany,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Laura K. Sievers
- Department of Internal Medicine I., Christian-Albrechts-University and University Hospital Schleswig-Holstein, Kiel, Germany,*Correspondence: Laura K. Sievers
| | - Alina Hütter
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Hashim-Abdul Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Martin Poryo
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Felix Berger
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany,Department of Pediatric Cardiology, Charite – Universitaetsmedizin Berlin, Berlin, Germany
| | - Ulrike M. M. Bauer
- National Register for Congenital Heart Defects, Berlin, Germany,Competence Network for Congenital Heart Defects, Berlin, Germany
| | - Paul C. Helm
- National Register for Congenital Heart Defects, Berlin, Germany,Competence Network for Congenital Heart Defects, Berlin, Germany
| | - Katharina R. L. Schmitt
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
184
|
Althali NJ, Hentges KE. Genetic insights into non-syndromic Tetralogy of Fallot. Front Physiol 2022; 13:1012665. [PMID: 36277185 PMCID: PMC9582763 DOI: 10.3389/fphys.2022.1012665] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/13/2022] [Indexed: 10/17/2023] Open
Abstract
Congenital heart defects (CHD) include structural abnormalities of the heart or/and great vessels that are present at birth. CHD affects around 1% of all newborns worldwide. Tetralogy of Fallot (TOF) is the most prevalent cyanotic congenital cardiac abnormality, affecting three out of every 10,000 live infants with a prevalence rate of 5-10% of all congenital cardiac defects. The four hallmark characteristics of TOF are: right ventricular hypertrophy, pulmonary stenosis, ventricular septal defect, and overriding aorta. Approximately 20% of cases of TOF are associated with a known disease or chromosomal abnormality, with the remaining 80% of TOF cases being non-syndromic, with no known aetiology. Relatively few TOF patients have been studied, and little is known about critical causative genes for non-syndromic TOF. However, rare genetic variants have been identified as significant risk factors for CHD, and are likely to cause some cases of TOF. Therefore, this review aims to provide an update on well-characterized genes and the most recent variants identified for non-syndromic TOF.
Collapse
Affiliation(s)
- Nouf J. Althali
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Biology Department, Science College, King Khalid University, Abha, Saudi Arabia
| | - Kathryn E. Hentges
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
185
|
Intrinsic myocardial defects underlie an Rbfox-deficient zebrafish model of hypoplastic left heart syndrome. Nat Commun 2022; 13:5877. [PMID: 36198703 PMCID: PMC9534849 DOI: 10.1038/s41467-022-32982-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/18/2022] [Indexed: 02/03/2023] Open
Abstract
Hypoplastic left heart syndrome (HLHS) is characterized by underdevelopment of left sided structures including the ventricle, valves, and aorta. Prevailing paradigm suggests that HLHS is a multigenic disease of co-occurring phenotypes. Here, we report that zebrafish lacking two orthologs of the RNA binding protein RBFOX2, a gene linked to HLHS in humans, display cardiovascular defects overlapping those in HLHS patients including ventricular, valve, and aortic deficiencies. In contrast to current models, we demonstrate that these structural deficits arise secondary to impaired pump function as these phenotypes are rescued when Rbfox is specifically expressed in the myocardium. Mechanistically, we find diminished expression and alternative splicing of sarcomere and mitochondrial components that compromise sarcomere assembly and mitochondrial respiration, respectively. Injection of human RBFOX2 mRNA restores cardiovascular development in rbfox mutant zebrafish, while HLHS-linked RBFOX2 variants fail to rescue. This work supports an emerging paradigm for HLHS pathogenesis that centers on myocardial intrinsic defects.
Collapse
|
186
|
Huang W, Zhu JY, Fu Y, van de Leemput J, Han Z. Lpt, trr, and Hcf regulate histone mono- and dimethylation that are essential for Drosophila heart development. Dev Biol 2022; 490:53-65. [PMID: 35853502 PMCID: PMC10728806 DOI: 10.1016/j.ydbio.2022.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 11/22/2022]
Abstract
Mammalian KMT2C, KMT2D, and HCFC1 are expressed during heart development and have been associated with congenital heart disease, but their roles in heart development remain elusive. We found that the Drosophila Lpt and trr genes encode the N-terminal and C-terminal homologs, respectively, of mammalian KMT2C or KMT2D. Lpt and trr mutant embryos showed reduced cardiac progenitor cells. Silencing of Lpt, trr, or both simultaneously in the heart led to similar abnormal cardiac morphology, tissue fibrosis, and cardiac functional defects. Like KMT2D, Lpt and trr were found to modulate histone H3K4 mono- and dimethylation, but not trimethylation. Investigation of downstream genes regulated by mouse KMT2D in the heart showed that their fly homologs are similarly regulated by Lpt or trr in the fly heart, suggesting that Lpt and trr regulate an evolutionarily conserved transcriptional network for heart development. Moreover, we showed that cardiac silencing of Hcf, the fly homolog of mammalian HCFC1, leads to heart defects similar to those observed in Lpt and trr silencing, as well as reduced H3K4 monomethylation. Our findings suggest that Lpt and trr function together to execute the conserved function of mammalian KMT2C and KMT2D in histone H3 lysine K4 mono- and dimethylation required for heart development. Possibly aided by Hcf, which we show plays a related role in H3K4 methylation during fly heart development.
Collapse
Affiliation(s)
- Wen Huang
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jun-Yi Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yulong Fu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Genomic Diagnostics and Bioinformatics, Department of Pathology, The University of Alabama at Birmingham, Alabama, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
187
|
Zhong G, Shen Y. Statistical models of the genetic etiology of congenital heart disease. Curr Opin Genet Dev 2022; 76:101967. [PMID: 35939966 PMCID: PMC10586490 DOI: 10.1016/j.gde.2022.101967] [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: 02/28/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 11/03/2022]
Abstract
Congenital heart disease (CHD) is a collection of anatomically and clinically heterogeneous structure anomalies of heart at birth. Finding genetic causes of CHD can not only shed light on developmental biology of heart, but also provide basis for improving clinical care and interventions. The optimal study design and analytical approaches to identify genetic causes depend on the underlying genetic architecture. A few well-known syndromes with CHD as core conditions, such as Noonan and CHARGE, have known monogenic causes. The genetic causes of most of CHD patients, however, are unknown and likely to be complex. In this review, we highlight recent studies that assume a complex genetic architecture of CHD with two main approaches. One is genomic sequencing studies aiming for identifying rare or de novo risk variants with large genetic effect. The other is genome-wide association studies optimized for common variants with moderate genetic effect.
Collapse
Affiliation(s)
- Guojie Zhong
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA; Integrated Program in Cellular, Molecular, and Biological Studies, Columbia University Irving Medical Center, New York, NY, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA; Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA; JP Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
188
|
Chung HL, Rump P, Lu D, Glassford MR, Mok JW, Fatih J, Basal A, Marcogliese PC, Kanca O, Rapp M, Fock JM, Kamsteeg EJ, Lupski JR, Larson A, Haninbal MC, Bellen H, Harel T. De novo variants in EMC1 lead to neurodevelopmental delay and cerebellar degeneration and affect glial function in Drosophila. Hum Mol Genet 2022; 31:3231-3244. [PMID: 35234901 PMCID: PMC9523557 DOI: 10.1093/hmg/ddac053] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The endoplasmic reticulum (ER)-membrane protein complex (EMC) is a multi-protein transmembrane complex composed of 10 subunits that functions as a membrane-protein chaperone. Variants in EMC1 lead to neurodevelopmental delay and cerebellar degeneration. Multiple families with biallelic variants have been published, yet to date, only a single report of a monoallelic variant has been described, and functional evidence is sparse. METHODS Exome sequencing was used to investigate the genetic cause underlying severe developmental delay in three unrelated children. EMC1 variants were modeled in Drosophila, using loss-of-function (LoF) and overexpression studies. Glial-specific and neuronal-specific assays were used to determine whether the dysfunction was specific to one cell type. RESULTS Exome sequencing identified de novo variants in EMC1 in three individuals affected by global developmental delay, hypotonia, seizures, visual impairment and cerebellar atrophy. All variants were located at Pro582 or Pro584. Drosophila studies indicated that imbalance of EMC1-either overexpression or knockdown-results in pupal lethality and suggest that the tested homologous variants are LoF alleles. In addition, glia-specific gene dosage, overexpression or knockdown, of EMC1 led to lethality, whereas neuron-specific alterations were tolerated. DISCUSSION We establish de novo monoallelic EMC1 variants as causative of a neurological disease trait by providing functional evidence in a Drosophila model. The identified variants failed to rescue the lethality of a null allele. Variations in dosage of the wild-type EMC1, specifically in glia, lead to pupal lethality, which we hypothesize results from the altered stoichiometry of the multi-subunit protein complex EMC.
Collapse
Affiliation(s)
- Hyung-Lok Chung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Patrick Rump
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen 9700 RB, The Netherlands
| | - Di Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Megan R Glassford
- Division of Pediatric Genetics, Metabolism & Genomic Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jung-Wan Mok
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Jawid Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adily Basal
- Department of Genetics, Hadassah Medical Organization, Jerusalem 9112001, Israel
| | - Paul C Marcogliese
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Michele Rapp
- University of Colorado Anschutz Medical Campus, Aurora, CO 60045, USA
| | - Johanna M Fock
- University of Groningen, University Medical Centre Groningen, Department of Neurology, Groningen 9700 RB, The Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen 6500 HB, The Netherlands
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Texas Children's Hospital, Houston, TX 77030, USA
| | - Austin Larson
- University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 60045, United States
| | - Mark C Haninbal
- Division of Pediatric Genetics, Metabolism & Genomic Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hugo Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tamar Harel
- Department of Genetics, Hadassah Medical Organization, Jerusalem 9112001, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| |
Collapse
|
189
|
Considering the Genetic Architecture of Hypoplastic Left Heart Syndrome. J Cardiovasc Dev Dis 2022; 9:jcdd9100315. [PMID: 36286267 PMCID: PMC9604382 DOI: 10.3390/jcdd9100315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Hypoplastic left heart syndrome (HLHS) is among the most severe cardiovascular malformations and understanding its causes is crucial to making progress in prevention and treatment. Genetic analysis is a broadly useful tool for dissecting complex causal mechanisms and it is playing a significant role in HLHS research. However, unlike classical Mendelian disorders where a relatively small number of genes are largely determinative of the occurrence and severity of the disease, the picture in HLHS is complex. De novo single-gene and copy number variant (CNV) disorders make an important contribution, but there is emerging evidence for causal contributions from lower penetrance and common variation. Integrating this emerging knowledge into clinical diagnostics and translating the findings into effective prevention and treatment remain challenges for the future.
Collapse
|
190
|
Zhu L, Choudhary K, Gonzalez-Teran B, Ang YS, Thomas R, Stone NR, Liu L, Zhou P, Zhu C, Ruan H, Huang Y, Jin S, Pelonero A, Koback F, Padmanabhan A, Sadagopan N, Hsu A, Costa MW, Gifford CA, van Bemmel J, Hüttenhain R, Vedantham V, Conklin BR, Black BL, Bruneau BG, Steinmetz L, Krogan NJ, Pollard KS, Srivastava D. Transcription Factor GATA4 Regulates Cell Type-Specific Splicing Through Direct Interaction With RNA in Human Induced Pluripotent Stem Cell-Derived Cardiac Progenitors. Circulation 2022; 146:770-787. [PMID: 35938400 PMCID: PMC9452483 DOI: 10.1161/circulationaha.121.057620] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND GATA4 (GATA-binding protein 4), a zinc finger-containing, DNA-binding transcription factor, is essential for normal cardiac development and homeostasis in mice and humans, and mutations in this gene have been reported in human heart defects. Defects in alternative splicing are associated with many heart diseases, yet relatively little is known about how cell type- or cell state-specific alternative splicing is achieved in the heart. Here, we show that GATA4 regulates cell type-specific splicing through direct interaction with RNA and the spliceosome in human induced pluripotent stem cell-derived cardiac progenitors. METHODS We leveraged a combination of unbiased approaches including affinity purification of GATA4 and mass spectrometry, enhanced cross-linking with immunoprecipitation, electrophoretic mobility shift assays, in vitro splicing assays, and unbiased transcriptomic analysis to uncover GATA4's novel function as a splicing regulator in human induced pluripotent stem cell-derived cardiac progenitors. RESULTS We found that GATA4 interacts with many members of the spliceosome complex in human induced pluripotent stem cell-derived cardiac progenitors. Enhanced cross-linking with immunoprecipitation demonstrated that GATA4 also directly binds to a large number of mRNAs through defined RNA motifs in a sequence-specific manner. In vitro splicing assays indicated that GATA4 regulates alternative splicing through direct RNA binding, resulting in functionally distinct protein products. Correspondingly, knockdown of GATA4 in human induced pluripotent stem cell-derived cardiac progenitors resulted in differential alternative splicing of genes involved in cytoskeleton organization and calcium ion import, with functional consequences associated with the protein isoforms. CONCLUSIONS This study shows that in addition to its well described transcriptional function, GATA4 interacts with members of the spliceosome complex and regulates cell type-specific alternative splicing via sequence-specific interactions with RNA. Several genes that have splicing regulated by GATA4 have functional consequences and many are associated with dilated cardiomyopathy, suggesting a novel role for GATA4 in achieving the necessary cardiac proteome in normal and stress-responsive conditions.
Collapse
Affiliation(s)
- Lili Zhu
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | | | - Barbara Gonzalez-Teran
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Yen-Sin Ang
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | | | - Nicole R. Stone
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Lei Liu
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Ping Zhou
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Palo Alto, CA, USA
| | - Hongmei Ruan
- Department of Medicine, University of California, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Yu Huang
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Shibo Jin
- Division of Cellular and Developmental Biology, Molecular and Cell Biology Department, University of California at Berkeley, Berkeley, CA, USA
| | - Angelo Pelonero
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Frances Koback
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Arun Padmanabhan
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Nandhini Sadagopan
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Austin Hsu
- Gladstone Institutes, San Francisco, CA, USA
| | - Mauro W. Costa
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Casey A. Gifford
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Joke van Bemmel
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Ruth Hüttenhain
- Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA, USA
| | - Vasanth Vedantham
- Department of Medicine, University of California, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Bruce R. Conklin
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Brian L. Black
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Benoit G. Bruneau
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Lars Steinmetz
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Palo Alto, CA, USA
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Nevan J. Krogan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA, USA
| | - Katherine S. Pollard
- Gladstone Institutes, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, Institute for Computational Health Sciences, and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Deepak Srivastava
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| |
Collapse
|
191
|
Gabriel GC, Yagi H, Xu X, Lo CW. Novel Insights into the Etiology, Genetics, and Embryology of Hypoplastic Left Heart Syndrome. World J Pediatr Congenit Heart Surg 2022; 13:565-570. [PMID: 36053093 PMCID: PMC10010598 DOI: 10.1177/21501351221102961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hypoplastic left heart syndrome (HLHS) is a relatively rare severe congenital heart defect (CHD) closely linked to other left ventricular outflow tract (LVOT) lesions including bicuspid aortic valve (BAV), one of the most common heart defects. While HLHS, BAV, and other LVOT lesions have a strong genetic underpinning, their genetic etiology remains poorly understood. Findings from a large-scale mouse mutagenesis screen showed HLHS has a multigenic etiology and is genetically heterogenous, explaining difficulties in identifying the genetic causes of HLHS. In Ohia mice, HLHS shows incomplete penetrance. Some mice exhibited small LV with normal aorta, and others a normal LV with hypoplastic aorta, indicating the LV hypoplasia is not hemodynamically driven. In Ohia mutants, HLHS was found to have a digenic modular construction, with mutation in a chromatin modifier causing the small LV phenotype and mutation in Pcdha9 causing the aorta/aortic valve hypoplasia. The Pcdha9 mutation alone can cause BAV, and in the human genome two common deletion copy number variants spanning PCDHA7-10 are associated with BAV. Hence the digenic etiology of HLHS can account for the close association of HLHS, a rare CHD, with BAV, one of the most common CHD. Functional analysis of Ohia HLHS heart tissue showed severe mitochondrial dysfunction in the small LV, while the normal size RV is also affected but milder, suggesting possible role in vulnerability of surgically palliated HLHS patients to heart failure. These findings suggest insights into the genetics of HLHS may yield new therapies for improving outcome for patients with HLHS.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, 6614University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hisato Yagi
- Department of Developmental Biology, 6614University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xinxiu Xu
- Department of Developmental Biology, 6614University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cecilia W Lo
- Department of Developmental Biology, 6614University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
192
|
Destici E, Zhu F, Tran S, Preissl S, Farah EN, Zhang Y, Hou X, Poirion OB, Lee AY, Grinstein JD, Bloomekatz J, Kim HS, Hu R, Evans SM, Ren B, Benner C, Chi NC. Human-gained heart enhancers are associated with species-specific cardiac attributes. NATURE CARDIOVASCULAR RESEARCH 2022; 1:830-843. [PMID: 36817700 PMCID: PMC9937543 DOI: 10.1038/s44161-022-00124-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/26/2022] [Indexed: 11/09/2022]
Abstract
The heart, a vital organ which is first to develop, has adapted its size, structure and function in order to accommodate the circulatory demands for a broad range of animals. Although heart development is controlled by a relatively conserved network of transcriptional/chromatin regulators, how the human heart has evolved species-specific features to maintain adequate cardiac output and function remains to be defined. Here, we show through comparative epigenomic analysis the identification of enhancers and promoters that have gained activity in humans during cardiogenesis. These cis-regulatory elements (CREs) are associated with genes involved in heart development and function, and may account for species-specific differences between human and mouse hearts. Supporting these findings, genetic variants that are associated with human cardiac phenotypic/disease traits, particularly those differing between human and mouse, are enriched in human-gained CREs. During early stages of human cardiogenesis, these CREs are also gained within genomic loci of transcriptional regulators, potentially expanding their role in human heart development. In particular, we discovered that gained enhancers in the locus of the early human developmental regulator ZIC3 are selectively accessible within a subpopulation of mesoderm cells which exhibits cardiogenic potential, thus possibly extending the function of ZIC3 beyond its conserved left-right asymmetry role. Genetic deletion of these enhancers identified a human gained enhancer that was required for not only ZIC3 and early cardiac gene expression at the mesoderm stage but also cardiomyocyte differentiation. Overall, our results illuminate how human gained CREs may contribute to human-specific cardiac attributes, and provide insight into how transcriptional regulators may gain cardiac developmental roles through the evolutionary acquisition of enhancers.
Collapse
Affiliation(s)
- Eugin Destici
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Fugui Zhu
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Shaina Tran
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sebastian Preissl
- Ludwig Institute for Cancer Research, La Jolla, CA, 92093, USA
- Center for Epigenomics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Elie N. Farah
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yanxiao Zhang
- Ludwig Institute for Cancer Research, La Jolla, CA, 92093, USA
| | - Xiameng Hou
- Center for Epigenomics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Olivier B. Poirion
- Center for Epigenomics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ah Young Lee
- Ludwig Institute for Cancer Research, La Jolla, CA, 92093, USA
| | - Jonathan D. Grinstein
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - Hong Sook Kim
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Robert Hu
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sylvia M. Evans
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Bing Ren
- Ludwig Institute for Cancer Research, La Jolla, CA, 92093, USA
- Center for Epigenomics, University of California, San Diego, La Jolla, CA, 92093, USA
- Institute of Genomic Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Chris Benner
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Neil C. Chi
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
- Institute of Genomic Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
193
|
Wilde AAM, Semsarian C, Márquez MF, Shamloo AS, Ackerman MJ, Ashley EA, Sternick EB, Barajas-Martinez H, Behr ER, Bezzina CR, Breckpot J, Charron P, Chockalingam P, Crotti L, Gollob MH, Lubitz S, Makita N, Ohno S, Ortiz-Genga M, Sacilotto L, Schulze-Bahr E, Shimizu W, Sotoodehnia N, Tadros R, Ware JS, Winlaw DS, Kaufman ES. European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the state of genetic testing for cardiac diseases. Europace 2022; 24:1307-1367. [PMID: 35373836 PMCID: PMC9435643 DOI: 10.1093/europace/euac030] [Citation(s) in RCA: 188] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Arthur A M Wilde
- Heart Centre, Department of Cardiology, Amsterdam Universitair Medische
Centra, Amsterdam, location AMC, The Netherlands
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute,
University of Sydney, Sydney, Australia
| | - Manlio F Márquez
- Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de
México, Mexico
- Member of the Latin American Heart Rhythm Society (LAHRS)
| | | | - Michael J Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine,
and Molecular Pharmacology & Experimental Therapeutics; Divisions of Heart Rhythm
Services and Pediatric Cardiology; Windland Smith Rice Genetic Heart Rhythm Clinic and
Windland Smith Rice Sudden Death Genomics Laboratory, Mayo
Clinic, Rochester, MN, USA
| | - Euan A Ashley
- Department of Cardiovascular Medicine, Stanford University,
Stanford, California, USA
| | - Eduardo Back Sternick
- Arrhythmia and Electrophysiology Unit, Biocor Institute,
Minas Gerais, Brazil; and
Member of the Latin American Heart Rhythm Society (LAHRS)
| | - Héctor Barajas-Martinez
- Cardiovascular Research, Lankenau Institute of Medical
Research, Wynnewood, PA, USA; and Member of the Latin American Heart Rhythm Society (LAHRS)
| | - Elijah R Behr
- Cardiovascular Clinical Academic Group, Institute of Molecular and Clinical
Sciences, St. George’s, University of London; St. George’s University Hospitals NHS
Foundation Trust, London, UK; Mayo Clinic Healthcare, London
| | - Connie R Bezzina
- Amsterdam UMC Heart Center, Department of Experimental
Cardiology, Amsterdam, The
Netherlands
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospitals Leuven,
Leuven, Belgium
| | - Philippe Charron
- Sorbonne Université, APHP, Centre de Référence des Maladies Cardiaques
Héréditaires, ICAN, Inserm UMR1166, Hôpital
Pitié-Salpêtrière, Paris, France
| | | | - Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin,
Istituto Auxologico Italiano, IRCCS, Milan, Italy
- Cardiomyopathy Unit and Cardiac Rehabilitation Unit, San Luca Hospital,
Istituto Auxologico Italiano, IRCCS, Milan,
Italy
- Department of Medicine and Surgery, University of
Milano-Bicocca, Milan, Italy
| | - Michael H Gollob
- Inherited Arrhythmia and Cardiomyopathy Program, Division of Cardiology,
University of Toronto, Toronto, ON, Canada
| | - Steven Lubitz
- Cardiac Arrhythmia Service, Massachusetts General Hospital and Harvard
Medical School, Boston, MA, USA
| | - Naomasa Makita
- National Cerebral and Cardiovascular Center, Research
Institute, Suita, Japan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular
Center, Suita, Japan
| | - Martín Ortiz-Genga
- Clinical Department, Health in Code, A
Coruña, Spain; and Member of the Latin
American Heart Rhythm Society (LAHRS)
| | - Luciana Sacilotto
- Arrhythmia Unit, Instituto do Coracao, Hospital das Clinicas HCFMUSP,
Faculdade de Medicina, Universidade de Sao Paulo, Sao
Paulo, Brazil; and Member of the Latin
American Heart Rhythm Society (LAHRS)
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases, University Hospital
Münster, Münster, Germany
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Graduate School of Medicine, Nippon
Medical School, Bunkyo-ku, Tokyo, Japan
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of
Medicine, University of Washington, Seattle, WA,
USA
| | - Rafik Tadros
- Cardiovascular Genetics Center, Department of Medicine, Montreal Heart
Institute, Université de Montréal, Montreal,
Canada
| | - James S Ware
- National Heart and Lung Institute and MRC London Institute of Medical
Sciences, Imperial College London, London,
UK
- Royal Brompton & Harefield Hospitals, Guy’s
and St. Thomas’ NHS Foundation Trust, London, UK
| | - David S Winlaw
- Cincinnati Children's Hospital Medical Centre, University of
Cincinnati, Cincinnati, OH, USA
| | - Elizabeth S Kaufman
- Metrohealth Medical Center, Case Western Reserve University,
Cleveland, OH, USA
| |
Collapse
|
194
|
Zheng S, Huang H, Ma L, Zhu T. RASopathies due to de novo pathogenic variants: clinical features, genetic findings and outcomes in nine neonates born with congenital heart defects. BMC Med Genomics 2022; 15:184. [PMID: 36002837 PMCID: PMC9400306 DOI: 10.1186/s12920-022-01336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/11/2022] [Indexed: 12/04/2022] Open
Abstract
Background There are limited information available related to neonatal characteristics of RASopathies, a group of autosomal dominant syndromes with considerable phenotypic overlap. Methods The retrospective review revealed 9 neonates born with congenital heart defects (CHDs) and diagnosed as RASopathies due to de novo mutations (DNMs) by trio-based exome sequencing (ES) between January 2017 and December 2020. We report in details of the neonatal course, molecular analysis and 180-days of age follow-up in affected individuals. Results The early clinical spectrum included various types of CHDs, less noticeable multiple extracardiac anomalies and unspecific symptoms like poor feeding. Of the 8 variants identified from 6 genes, 2 in RASA1 were novel: (NM_002890.2: c.2828 T > C (p.Leu943Pro)) and (NM_002890.2: c.2001del (p.Pro668Leufs*10)), which functionally impaired the protein structure. There was a relatively high mortality rate of 33.33% (3/9) for all the defects combined. A RAF1-deficient male and a RASA1-deficient male survived from severe heart failure by surgical interventions in early life. Conclusions Our results revealed that family-based ES was useful in identifying DNMs and causal genes for sporadic diseases and screening Rasopathies shortly after birth. We recommended a family-based ES and a full phenotypic evaluation including echocardiogram, magnetic resonance imaging, ultrasonography and coagulation screening in neonates with CHDs and a suspected genetic etiology.
Collapse
Affiliation(s)
- Simin Zheng
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huanyang Huang
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Ma
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianwen Zhu
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
195
|
Hulen J, Kenny D, Black R, Hallgren J, Hammond KG, Bredahl EC, Wickramasekara RN, Abel PW, Stessman HAF. KMT5B is required for early motor development. Front Genet 2022; 13:901228. [PMID: 36035149 PMCID: PMC9411648 DOI: 10.3389/fgene.2022.901228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Disruptive variants in lysine methyl transferase 5B (KMT5B/SUV4-20H1) have been identified as likely-pathogenic among humans with neurodevelopmental phenotypes including motor deficits (i.e., hypotonia and motor delay). However, the role that this enzyme plays in early motor development is largely unknown. Using a Kmt5b gene trap mouse model, we assessed neuromuscular strength, skeletal muscle weight (i.e., muscle mass), neuromuscular junction (NMJ) structure, and myofiber type, size, and distribution. Tests were performed over developmental time (postnatal days 17 and 44) to represent postnatal versus adult structures in slow- and fast-twitch muscle types. Prior to the onset of puberty, slow-twitch muscle weight was significantly reduced in heterozygous compared to wild-type males but not females. At the young adult stage, we identified decreased neuromuscular strength, decreased skeletal muscle weights (both slow- and fast-twitch), increased NMJ fragmentation (in slow-twitch muscle), and smaller myofibers in both sexes. We conclude that Kmt5b haploinsufficiency results in a skeletal muscle developmental deficit causing reduced muscle mass and body weight.
Collapse
Affiliation(s)
- Jason Hulen
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - Dorothy Kenny
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - Rebecca Black
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - Jodi Hallgren
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - Kelley G. Hammond
- Department of Exercise Science, College of Arts and Sciences, Creighton University, Omaha, NE, United States
| | - Eric C. Bredahl
- Department of Exercise Science, College of Arts and Sciences, Creighton University, Omaha, NE, United States
| | - Rochelle N. Wickramasekara
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
- Molecular Diagnostic Laboratory, Boys Town National Research Hospital, Omaha, NE, United States
| | - Peter W. Abel
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - Holly A. F. Stessman
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| |
Collapse
|
196
|
Ziegler A, Chung WK. Recent advances in understanding neuro. Curr Opin Genet Dev 2022; 75:101938. [DOI: 10.1016/j.gde.2022.101938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/18/2022] [Accepted: 05/27/2022] [Indexed: 11/26/2022]
|
197
|
Weaver KN, Chen J, Shikany A, White PS, Prada CE, Gelb BD, Cnota JF. Prevalence of Genetic Diagnoses in a Cohort With Valvar Pulmonary Stenosis. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003635. [PMID: 35666834 PMCID: PMC9388589 DOI: 10.1161/circgen.121.003635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Valvar pulmonary stenosis (vPS) accounts for 8% to 12% of congenital heart disease cases. Multiple genetic syndromes are associated with vPS, most commonly Noonan syndrome, but the cause is unknown in most cases. We analyzed genomic data from a large cohort with vPS to determine the prevalence of genetic diagnosis. METHODS The Pediatric Cardiac Genomics Consortium database was queried to identify probands with vPS without complex congenital heart disease or aneuploidy and with existing whole exome or genome sequencing. A custom analysis workflow was used to identify likely pathogenic or pathogenic variants in disease-associated genes. Demographic and phenotypic characteristics were compared between groups with and without molecular diagnoses. RESULTS Data from 119 probands (105 trios) were included. A molecular diagnosis was identified in 22 (18%); 17 (14%) had Noonan syndrome or a related disorder. Extracardiac and neurodevelopmental comorbidities were seen in 67/119 (56%) of probands. Molecular diagnosis was more common in those with extracardiac and neurodevelopmental phenotypes than those without (18/67 versus 4/52, P=0.0086). CONCLUSIONS Clinicians should have high suspicion for a genetic diagnosis in individuals with vPS, particularly if additional phenotypes are present. Our results suggest that clinicians should consider offering sequencing of at least the known congenital heart disease and RASopathy genes to all individuals with vPS, regardless of whether that individual has extracardiac or neurodevelopmental phenotypes present.
Collapse
Affiliation(s)
- K Nicole Weaver
- Division of Human Genetics (K.N.W.), Cincinnati Children's Hospital Medical Center
- Heart Institute (K.N.W., A.S., J.F.C.), Cincinnati Children's Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH (K.N.W., J.C., P.S.W., J.F.C.)
| | - Jing Chen
- Division of Biomedical Informatics (J.C.), Cincinnati Children's Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH (K.N.W., J.C., P.S.W., J.F.C.)
| | - Amy Shikany
- Heart Institute (K.N.W., A.S., J.F.C.), Cincinnati Children's Hospital Medical Center
| | - Pete S White
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH (K.N.W., J.C., P.S.W., J.F.C.)
| | - Carlos E Prada
- Division of Genetics, Birth Defects & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago (C.E.P.)
- Department of Pediatrics, Feinberg School of Medicine of Northwestern University, Chicago, IL (C.E.P.)
| | - Bruce D Gelb
- Mindich Child Health and Development Institute, Departments of Pediatrics and Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY (B.D.G.)
| | - James F Cnota
- Heart Institute (K.N.W., A.S., J.F.C.), Cincinnati Children's Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH (K.N.W., J.C., P.S.W., J.F.C.)
| |
Collapse
|
198
|
Wilde AAM, Semsarian C, Márquez MF, Sepehri Shamloo A, Ackerman MJ, Ashley EA, Sternick Eduardo B, Barajas‐Martinez H, Behr ER, Bezzina CR, Breckpot J, Charron P, Chockalingam P, Crotti L, Gollob MH, Lubitz S, Makita N, Ohno S, Ortiz‐Genga M, Sacilotto L, Schulze‐Bahr E, Shimizu W, Sotoodehnia N, Tadros R, Ware JS, Winlaw DS, Kaufman ES, Aiba T, Bollmann A, Choi J, Dalal A, Darrieux F, Giudicessi J, Guerchicoff M, Hong K, Krahn AD, Mac Intyre C, Mackall JA, Mont L, Napolitano C, Ochoa Juan P, Peichl P, Pereira AC, Schwartz PJ, Skinner J, Stellbrink C, Tfelt‐Hansen J, Deneke T. European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the state of genetic testing for cardiac diseases. J Arrhythm 2022; 38:491-553. [PMID: 35936045 PMCID: PMC9347209 DOI: 10.1002/joa3.12717] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Arthur A. M. Wilde
- Heart Centre, Department of Cardiology, Amsterdam Universitair Medische CentraAmsterdamThe Netherlands
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary InstituteUniversity of SydneySydneyAustralia
| | - Manlio F. Márquez
- Instituto Nacional de Cardiología Ignacio ChávezCiudad de MéxicoMexico
| | | | - Michael J. Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics; Divisions of Heart Rhythm Services and Pediatric Cardiology; Windland Smith Rice Genetic Heart Rhythm Clinic and Windland Smith Rice Sudden Death Genomics Laboratory, Mayo ClinicRochesterMNUSA
| | - Euan A. Ashley
- Department of Cardiovascular MedicineStanford UniversityStanfordCAUSA
| | | | | | - Elijah R. Behr
- Cardiovascular Clinical Academic Group, Institute of Molecular and Clinical Sciences, St. George’sUniversity of London; St. George’s University Hospitals NHS Foundation TrustLondonUKMayo Clinic HealthcareLondon
| | - Connie R. Bezzina
- Amsterdam UMC Heart Center, Department of Experimental CardiologyAmsterdamThe Netherlands
| | - Jeroen Breckpot
- Center for Human GeneticsUniversity Hospitals LeuvenLeuvenBelgium
| | | | | | - Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCSMilanItaly
- Cardiomyopathy Unit and Cardiac Rehabilitation Unit, San Luca Hospital, Istituto Auxologico Italiano, IRCCSMilanItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMilanItaly
| | - Michael H. Gollob
- Inherited Arrhythmia and Cardiomyopathy Program, Division of CardiologyUniversity of TorontoTorontoONCanada
| | - Steven Lubitz
- Cardiac Arrhythmia ServiceMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Naomasa Makita
- National Cerebral and Cardiovascular CenterResearch InstituteSuitaJapan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular CenterSuitaJapan
| | | | - Luciana Sacilotto
- Arrhythmia Unit, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao PauloBrazil
| | - Eric Schulze‐Bahr
- Institute for Genetics of Heart DiseasesUniversity Hospital MünsterMünsterGermany
| | - Wataru Shimizu
- Department of Cardiovascular MedicineGraduate School of MedicineTokyoJapan
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of MedicineUniversity of WashingtonSeattleWAUSA
| | - Rafik Tadros
- Cardiovascular Genetics Center, Department of Medicine, Montreal Heart InstituteUniversité de MontréalMontrealCanada
| | - James S. Ware
- National Heart and Lung Institute and MRC London Institute of Medical SciencesImperial College LondonLondonUK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation TrustLondonUK
| | - David S. Winlaw
- Cincinnati Children's Hospital Medical CentreUniversity of CincinnatiCincinnatiOHUSA
| | | | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and Genetics, National Cerebral and Cardiovascular Center, SuitaOsakaJapan
| | - Andreas Bollmann
- Department of ElectrophysiologyHeart Center Leipzig at University of LeipzigLeipzigGermany
- Leipzig Heart InstituteLeipzigGermany
| | - Jong‐Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Anam HospitalKorea University College of MedicineSeoulRepublic of Korea
| | - Aarti Dalal
- Department of Pediatrics, Division of CardiologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Francisco Darrieux
- Arrhythmia Unit, Instituto do Coração, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São PauloSão PauloBrazil
| | - John Giudicessi
- Department of Cardiovascular Medicine (Divisions of Heart Rhythm Services and Circulatory Failure and the Windland Smith Rice Genetic Heart Rhythm Clinic), Mayo ClinicRochesterMNUSA
| | - Mariana Guerchicoff
- Division of Pediatric Arrhythmia and Electrophysiology, Italian Hospital of Buenos AiresBuenos AiresArgentina
| | - Kui Hong
- Department of Cardiovascular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Andrew D. Krahn
- Division of CardiologyUniversity of British ColumbiaVancouverCanada
| | - Ciorsti Mac Intyre
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo ClinicRochesterMNUSA
| | - Judith A. Mackall
- Center for Cardiac Electrophysiology and Pacing, University Hospitals Cleveland Medical CenterCase Western Reserve University School of MedicineClevelandOHUSA
| | - Lluís Mont
- Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS). Barcelona, Spain; Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), MadridSpain
| | - Carlo Napolitano
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCSPaviaItaly
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
| | - Pablo Ochoa Juan
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), MadridSpain
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de HierroMadridSpain
- Centro de Investigacion Biomedica en Red en Enfermedades Cariovasculares (CIBERCV), MadridSpain
| | - Petr Peichl
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Alexandre C. Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart InstituteUniversity of São Paulo Medical SchoolSão PauloBrazil
- Hipercol Brasil ProgramSão PauloBrazil
| | - Peter J. Schwartz
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCSMilanItaly
| | - Jon Skinner
- Sydney Childrens Hospital NetworkUniversity of SydneySydneyAustralia
| | - Christoph Stellbrink
- Department of Cardiology and Intensive Care MedicineUniversity Hospital Campus Klinikum BielefeldBielefeldGermany
| | - Jacob Tfelt‐Hansen
- The Department of Cardiology, the Heart Centre, Copenhagen University Hospital, Rigshopitalet, Copenhagen, Denmark; Section of genetics, Department of Forensic Medicine, Faculty of Medical SciencesUniversity of CopenhagenDenmark
| | - Thomas Deneke
- Heart Center Bad NeustadtBad Neustadt a.d. SaaleGermany
| |
Collapse
|
199
|
Rashkin SR, Cleves M, Shaw GM, Nembhard WN, Nestoridi E, Jenkins MM, Romitti PA, Lou XY, Browne ML, Mitchell LE, Olshan AF, Lomangino K, Bhattacharyya S, Witte JS, Hobbs CA, National Birth Defects Prevention Study. A genome-wide association study of obstructive heart defects among participants in the National Birth Defects Prevention Study. Am J Med Genet A 2022; 188:2303-2314. [PMID: 35451555 PMCID: PMC9283270 DOI: 10.1002/ajmg.a.62759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 01/19/2023]
Abstract
Obstructive heart defects (OHDs) share common structural lesions in arteries and cardiac valves, accounting for ~25% of all congenital heart defects. OHDs are highly heritable, resulting from interplay among maternal exposures, genetic susceptibilities, and epigenetic phenomena. A genome-wide association study was conducted in National Birth Defects Prevention Study participants (Ndiscovery = 3978; Nreplication = 2507), investigating the genetic architecture of OHDs using transmission/disequilibrium tests (TDT) in complete case-parental trios (Ndiscovery_TDT = 440; Nreplication_TDT = 275) and case-control analyses separately in infants (Ndiscovery_CCI = 1635; Nreplication_CCI = 990) and mothers (case status defined by infant; Ndiscovery_CCM = 1703; Nreplication_CCM = 1078). In the TDT analysis, the SLC44A2 single nucleotide polymorphism (SNP) rs2360743 was significantly associated with OHD (pdiscovery = 4.08 × 10-9 ; preplication = 2.44 × 10-4 ). A CAPN11 SNP (rs55877192) was suggestively associated with OHD (pdiscovery = 1.61 × 10-7 ; preplication = 0.0016). Two other SNPs were suggestively associated (p < 1 × 10-6 ) with OHD in only the discovery sample. In the case-control analyses, no SNPs were genome-wide significant, and, even with relaxed thresholds ( × discovery < 1 × 10-5 and preplication < 0.05), only one SNP (rs188255766) in the infant analysis was associated with OHDs (pdiscovery = 1.42 × 10-6 ; preplication = 0.04). Additional SNPs with pdiscovery < 1 × 10-5 were in loci supporting previous findings but did not replicate. Overall, there was modest evidence of an association between rs2360743 and rs55877192 and OHD and some evidence validating previously published findings.
Collapse
Affiliation(s)
- Sara R. Rashkin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, US
| | - Mario Cleves
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Health Informatics Institute, Tampa, FL 33612, US
| | - Gary M. Shaw
- Dept of Pediatrics, Stanford University, Stanford, CA 94305, US
| | - Wendy N. Nembhard
- University of Arkansas for Medical Sciences, Department of Epidemiology and Arkansas Center for Birth Defects and Prevention, University of Arkansas for Medical Sciences, Little Rock, AR 72205, US
| | - Eirini Nestoridi
- Massachusetts Center for Birth Defects Research and Prevention, Massachusetts Department of Public Health, Boston, MA 02108, US
| | - Mary M. Jenkins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30333, US
| | - Paul A. Romitti
- Department of Epidemiology, University of Iowa, Iowa City, IA 52242, US
| | - Xiang-Yang Lou
- Department of Biostatistics, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, FL 32603, US
| | - Marilyn L. Browne
- Birth Defects Research Section, New York State Department of Health, Albany, NY 12203, US; Department of Epidemiology and Biostatistics, School of Public Health, University at Albany, Rensselaer, NY 12114, US
| | - Laura E. Mitchell
- Department of Epidemiology, Human Genetics, and Environmental Sciences, UTHealth School of Public Health, Houston, TX 77030, US
| | - Andrew F. Olshan
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, US
| | | | - Sudeepa Bhattacharyya
- Bioinformatics and Data Science at University of Arkansas, Little Rock, AR 72204, US
| | - John S. Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, US
- These authors contributed equally to this work
| | - Charlotte A. Hobbs
- Rady Children’s Institute for Genomic Medicine, San Diego, CA 92123, US
- These authors contributed equally to this work
| | | |
Collapse
|
200
|
Spendlove SJ, Bondhus L, Lluri G, Sul JH, Arboleda VA. Polygenic risk scores of endo-phenotypes identify the effect of genetic background in congenital heart disease. HGG ADVANCES 2022; 3:100112. [PMID: 35599848 PMCID: PMC9118152 DOI: 10.1016/j.xhgg.2022.100112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/19/2022] [Indexed: 01/28/2023] Open
Abstract
Congenital heart disease (CHD) is a rare structural defect that occurs in ∼1% of live births. Studies on CHD genetic architecture have identified pathogenic single-gene mutations in less than 30% of cases. Single-gene mutations often show incomplete penetrance and variable expressivity. Therefore, we hypothesize that genetic background may play a role in modulating disease expression. Polygenic risk scores (PRSs) aggregate effects of common genetic variants to investigate whether, cumulatively, these variants are associated with disease penetrance or severity. However, the major limitations in this field have been in generating sufficient sample sizes for these studies. Here we used CHD-phenotype matched genome-wide association study (GWAS) summary statistics from the UK Biobank (UKBB) as our base study and whole-genome sequencing data from the CHD cohort (n1 = 711 trios, n2 = 362 European trios) of the Gabriella Miller Kids First dataset as our target study to develop PRSs for CHD. PRSs estimated using a GWAS for heart valve problems and heart murmur explain 2.5% of the variance in case-control status of CHD (all SNVs, p = 7.90 × 10-3; fetal cardiac SNVs, p = 8.00 × 10-3) and 1.8% of the variance in severity of CHD (fetal cardiac SNVs, p = 6.20 × 10-3; all SNVs, p = 0.015). These results show that common variants captured in CHD phenotype-matched GWASs have a modest but significant contribution to phenotypic expression of CHD. Further exploration of the cumulative effect of common variants is necessary for understanding the complex genetic etiology of CHD and other rare diseases.
Collapse
Affiliation(s)
- Sarah J. Spendlove
- Interdepartmental Bioinformatics Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Leroy Bondhus
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gentian Lluri
- Ahmanson/UCLA Adult Congenital Heart Disease Center, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jae Hoon Sul
- Interdepartmental Bioinformatics Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Valerie A. Arboleda
- Interdepartmental Bioinformatics Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|