1
|
Li L, Zhang C, Qin Y. AHDC1/Gibbin: a master regular of chromatin structure and gene transcription. Front Neurol 2025; 15:1388029. [PMID: 39835152 PMCID: PMC11743279 DOI: 10.3389/fneur.2024.1388029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 10/21/2024] [Indexed: 01/22/2025] Open
Affiliation(s)
- Linyi Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Chao Zhang
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai, China
| | - Yanwen Qin
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| |
Collapse
|
2
|
Jiang N, Zhang L, Zheng Z, Du H, Chen S, Pan H. Phenotypic subtypes of Xia-Gibbs syndrome: a latent class analysis. Eur J Hum Genet 2024:10.1038/s41431-024-01754-0. [PMID: 39648204 DOI: 10.1038/s41431-024-01754-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/17/2024] [Accepted: 11/22/2024] [Indexed: 12/10/2024] Open
Abstract
Xia-Gibbs syndrome (XGS) is a rare neurodevelopmental disorder with considerable clinical heterogeneity. To further characterize the syndrome's heterogeneity, we applied latent class analysis (LCA) on reported cases to identify phenotypic subtypes. By searching PubMed, Embase, China National Knowledge Infrastructure and Wanfang databases from inception to February 2024, we enrolled 97 cases with nonsense, frameshift or missense variants in the AHDC1 gene. LCA was based on the following 6 phenotypes with moderate occurrence and low missingness: ataxia, seizure, autism, sleep apnea, short stature and scoliosis. After excluding cases with missing data on all LCA variables or with unmatched phenotype-genotype information, a total of 85 cases were selected for LCA. Models with 1-5 classes were compared based on Akaike Information Criterion, Bayesian Information Criterion, Sample-Size Adjusted BIC and entropy. We used multinomial logistic regression (MLR) analyses to investigate the phenotype-genotype association and potential predictors for class membership. LCA revealed 3 distinct classes labeled as Ataxia subtype (n = 11 [12.9%]), Sleep apnea & short stature subtype (n = 23 [27.1%]) and Neuropsychological subtype (n = 51 [60.0%]). The commonest Neuropsychological subtype was characterized by high estimated probabilities of seizure, ataxia and autism. By adjusting for sex, age and variant type, MLR showed no significant association between phenotypic subtype and variant position. Age and variant type were identified as predictors of class membership. The findings of this review offer novel insights for different presentations of XGS. It is possible to deliver targeted monitoring and treatment for each subtype in the early stage.
Collapse
Affiliation(s)
- Nan Jiang
- 4+4 Medical Doctor Program, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Liyuan Zhang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Zeyan Zheng
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Hanze Du
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Shi Chen
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
| |
Collapse
|
3
|
Bertrand M, Shah G, Pedersen BS, Schulz A, Weise A, Liehr T, Huppke P, DiTroia S, Quinlan AR, Haack TB, Husain RA. De novo AHDC1 Deletions Identified by Genome Sequencing in Two Individuals with Xia-Gibbs Syndrome. Mol Syndromol 2024; 15:389-397. [PMID: 39359946 PMCID: PMC11444710 DOI: 10.1159/000538918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/13/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Xia-Gibbs syndrome (XGS) is a rare syndromic disorder characterized by developmental delay with intellectual disability, muscular hypotonia, brain anomalies, and nonspecific dysmorphic features. Different heterozygous variants in AHDC1 have been reported as causal for XGS, comprising mainly de novo stop-gain and frameshift events, but also missense variants, deletions, and a duplication of the locus. Case Presentation We hereby report 2 patients with clinical features of XGS. In the first patient, a de novo interstitial deletion in 1p36.11p35.3 encompassing the entire coding region of AHDC1 was initially suspected by trio exome sequencing and subsequently confirmed by shallow genome sequencing. In the second patient, a de novo deletion comprising most of the 5' untranslated region of AHDC1 was detected by genome sequencing. Conclusion We identified the smallest deletion comprising AHDC1 reported so far by shallow genome sequencing as well as another small AHDC1 deletion by genome sequencing. These methods represent useful techniques for the identification and confirmation of small deletions and structural variants. Furthermore, our data provide additional evidence of AHDC1 haploinsufficiency as a disease mechanism in XGS. Clinically, foot deformity, skin and connective tissue abnormalities observed in one of the patients are consistent with other reported cases of XGS. These findings suggest that these manifestations could be considered as more prevalent characteristics, underscoring the importance of in-depth phenotyping.
Collapse
Affiliation(s)
- Miriam Bertrand
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Gulalai Shah
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Brent S. Pedersen
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Alexander Schulz
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Anja Weise
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Center for Rare Diseases, Jena University Hospital, Jena, Germany
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Center for Rare Diseases, Jena University Hospital, Jena, Germany
| | - Peter Huppke
- Center for Rare Diseases, Jena University Hospital, Jena, Germany
- Department of Neuropediatrics, Jena University Hospital, Jena, Germany
| | - Stephanie DiTroia
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron R. Quinlan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
- Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT, USA
| | - Tobias B. Haack
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Ralf A. Husain
- Center for Rare Diseases, Jena University Hospital, Jena, Germany
- Department of Neuropediatrics, Jena University Hospital, Jena, Germany
| |
Collapse
|
4
|
Cheng X, Baki VB, Moran M, Liu B, Yu J, Zhao M, Li Q, Riethoven JJ, Gurumurth CB, Harris EN, Sun X. Liver matrin-3 protects mice against hepatic steatosis and stress response via constitutive androstane receptor. Mol Metab 2024; 86:101977. [PMID: 38936659 PMCID: PMC11267048 DOI: 10.1016/j.molmet.2024.101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024] Open
Abstract
OBJECTIVE The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise with the increasing obesity epidemic. Rezdiffra as an activator of a thyroid hormone receptor-beta is the only Food and Drug Administration approved therapy. As such, there is a critical need to improve our understanding of gene expression regulation and signaling transduction in MASLD to develop new therapies. Matrin-3 is a DNA- and RNA-binding protein involved in the pathogenesis of human diseases. Here we examined its previously uncharacterized role in limiting hepatic steatosis and stress response via the constitutive androstane receptor (CAR). METHODS Matrin-3 floxed and liver-specific knockout mice were fed either a chow diet or 60 kcal% high-fat diet (HFD) for up to 16 weeks. The mice were euthanized for different analysis including liver histology, lipid levels, and gene expression. Bulk RNA-seq, bulk ATAC-seq, and single-nucleus Multiome were used to examine changes of transcriptome and chromatin accessibility in the liver. Integrative bioinformatics analysis of our data and publicly available datasets and different biochemical assays were performed to identify underlying the molecular mechanisms mediating matrin-3's effects. Liver-tropic adeno-associated virus was used to restore the expression of CAR for lipid, acute phase genes, and histological analysis. RESULTS Matrin-3 expression is induced in the steatotic livers of mice. Liver-specific matrin-3 deletion exacerbated HFD-induced steatosis, acute phase response, and inflammation in the liver of female mice. The transcriptome and chromatin accessibility were re-programmed in the liver of these mice with signatures indicating that CAR signaling is dysregulated. Mechanistically, matrin-3 interacts with CAR mRNA, and matrin-3 deficiency promotes CAR mRNA degradation. Consequently, matrin-3 deletion impaired CAR signaling by reducing CAR expression. Matrin-3 levels positively correlate with CAR expression in human livers. Ces2a and Il1r1 were identified as new target genes of CAR. Interestingly, we found that CAR discords with the expression of its target genes including Cyp2b10 and Ces2a in response to HFD, indicating CAR signaling is dysregulated by HFD despite increased CAR expression. Dysregulated CAR signaling upon matrin-3 deficiency reduced Ces2a and de-repressed Il1r1 expression. CAR restoration partially abrogated the dysregulated gene expression, exacerbated hepatic steatosis, acute phase response, and inflammation in liver-specific matrin-3 knockout mice fed a HFD. CONCLUSIONS Our findings demonstrate that matrin-3 is a key upstream regulator maintaining CAR signaling upon metabolic stress, and the matrin-3-CAR axis limits hepatic steatosis and stress response signaling that may give insights for therapeutic intervention.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA
| | - Vijaya Bhaskar Baki
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA
| | - Matthew Moran
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA
| | - Baolong Liu
- Department of Nutrition and Health Sciences, University of Nebraska - Lincoln, 230 Filley Hall, Lincoln, NE 68583-0922, USA
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences, University of Nebraska - Lincoln, 230 Filley Hall, Lincoln, NE 68583-0922, USA
| | - Miaoyun Zhao
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska - Lincoln, Lincoln, NE, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska - Lincoln, Lincoln, NE, USA
| | - Jean-Jack Riethoven
- Nebraska Center for Biotechnology, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA; Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska - Lincoln, Lincoln, NE 68588, USA
| | | | - Edward N Harris
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA; Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska - Lincoln, Lincoln, NE 68588, USA; Nebraska Center for the Prevention of Obesity Diseases through Dietary Molecules, University of Nebraska - Lincoln, USA
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE 68588, USA; Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska - Lincoln, Lincoln, NE 68588, USA; Nebraska Center for the Prevention of Obesity Diseases through Dietary Molecules, University of Nebraska - Lincoln, USA.
| |
Collapse
|
5
|
Sprunger ML, Jackrel ME. The role of Matrin-3 in physiology and its dysregulation in disease. Biochem Soc Trans 2024; 52:961-972. [PMID: 38813817 PMCID: PMC11209761 DOI: 10.1042/bst20220585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
The dysfunction of many RNA-binding proteins (RBPs) that are heavily disordered, including TDP-43 and FUS, are implicated in amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD). These proteins serve many important roles in the cell, and their capacity to form biomolecular condensates (BMCs) is key to their function, but also a vulnerability that can lead to misregulation and disease. Matrin-3 (MATR3) is an intrinsically disordered RBP implicated both genetically and pathologically in ALS/FTD, though it is relatively understudied as compared with TDP-43 and FUS. In addition to binding RNA, MATR3 also binds DNA and is implicated in many cellular processes including the DNA damage response, transcription, splicing, and cell differentiation. It is unclear if MATR3 localizes to BMCs under physiological conditions, which is brought further into question due to its lack of a prion-like domain. Here, we review recent studies regarding MATR3 and its roles in numerous physiological processes, as well as its implication in a range of diseases.
Collapse
Affiliation(s)
- Macy L Sprunger
- Department of Chemistry, Washington University, St. Louis, MO 63130, U.S.A
| | - Meredith E Jackrel
- Department of Chemistry, Washington University, St. Louis, MO 63130, U.S.A
| |
Collapse
|
6
|
Santos JR, Park J. MATR3's Role beyond the Nuclear Matrix: From Gene Regulation to Its Implications in Amyotrophic Lateral Sclerosis and Other Diseases. Cells 2024; 13:980. [PMID: 38891112 PMCID: PMC11171696 DOI: 10.3390/cells13110980] [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: 05/13/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Matrin-3 (MATR3) was initially discovered as a component of the nuclear matrix about thirty years ago. Since then, accumulating studies have provided evidence that MATR3 not only plays a structural role in the nucleus, but that it is also an active protein involved in regulating gene expression at multiple levels, including chromatin organization, DNA transcription, RNA metabolism, and protein translation in the nucleus and cytoplasm. Furthermore, MATR3 may play a critical role in various cellular processes, including DNA damage response, cell proliferation, differentiation, and survival. In addition to the revelation of its biological role, recent studies have reported MATR3's involvement in the context of various diseases, including neurodegenerative and neurodevelopmental diseases, as well as cancer. Moreover, sequencing studies of patients revealed a handful of disease-associated mutations in MATR3 linked to amyotrophic lateral sclerosis (ALS), which further elevated the gene's importance as a topic of study. In this review, we synthesize the current knowledge regarding the diverse functions of MATR3 in DNA- and RNA-related processes, as well as its involvement in various diseases, with a particular emphasis on ALS.
Collapse
Affiliation(s)
- Jhune Rizsan Santos
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jeehye Park
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
7
|
Islam Z, Polash A, Suzawa M, Chim B, Kuhn S, Sultana S, Cutrona N, Smith PT, Kabat J, Ganesan S, Foroushani A, Hafner M, Muljo SA. MATRIN3 deficiency triggers autoinflammation via cGAS-STING activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587645. [PMID: 38712171 PMCID: PMC11071297 DOI: 10.1101/2024.04.01.587645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Interferon-stimulated genes (ISGs) comprise a program of immune effectors important for host immune defense. When uncontrolled, ISGs play a central role in interferonopathies and other inflammatory diseases. The mechanisms responsible for turning on ISGs are not completely known. By investigating MATRIN3 (MATR3), a nuclear RNA-binding protein mutated in familial ALS, we found that perturbing MATR3 results in elevated expression of ISGs. Using an integrative approach, we elucidate a pathway that leads to activation of cGAS-STING. This outlines a plausible mechanism for pathogenesis in a subset of ALS, and suggests new diagnostic and therapeutic approaches for this fatal disease.
Collapse
Affiliation(s)
- Zohirul Islam
- Integrative Immunobiology Section, Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, Maryland 20892, USA
| | - Ahsan Polash
- RNA Molecular Biology Laboratory, National Institute for Arthritis and Musculoskeletal and Skin Disease (NIAMS), NIH; Bethesda, Maryland 20892, USA
| | - Masataka Suzawa
- RNA Molecular Biology Laboratory, National Institute for Arthritis and Musculoskeletal and Skin Disease (NIAMS), NIH; Bethesda, Maryland 20892, USA
| | - Bryan Chim
- Integrative Immunobiology Section, Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, Maryland 20892, USA
| | - Skyler Kuhn
- Integrated Data Sciences Section, Research Technologies Branch (RTB), NIAID, NIH; Bethesda, Maryland 20892, USA
| | - Sabrina Sultana
- Integrative Immunobiology Section, Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, Maryland 20892, USA
| | - Nicholas Cutrona
- Integrative Immunobiology Section, Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, Maryland 20892, USA
| | - Patrick T. Smith
- Integrative Immunobiology Section, Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, Maryland 20892, USA
| | - Juraj Kabat
- Biological Imaging Section, RTB, NIAID, NIH; Bethesda, Maryland 20892, USA
| | - Sundar Ganesan
- Biological Imaging Section, RTB, NIAID, NIH; Bethesda, Maryland 20892, USA
| | - Amir Foroushani
- Integrative Immunobiology Section, Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, Maryland 20892, USA
| | - Markus Hafner
- RNA Molecular Biology Laboratory, National Institute for Arthritis and Musculoskeletal and Skin Disease (NIAMS), NIH; Bethesda, Maryland 20892, USA
| | - Stefan A. Muljo
- Integrative Immunobiology Section, Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH); Bethesda, Maryland 20892, USA
| |
Collapse
|
8
|
Lin Y, Yang Q, Lin X, Liu X, Qian Y, Xu D, Cao N, Han X, Zhu Y, Hu W, He X, Yu Z, Kong X, Zhu L, Zhong Z, Liu K, Zhou B, Wang Y, Peng J, Zhu W, Wang J. Extracellular Matrix Disorganization Caused by ADAMTS16 Deficiency Leads to Bicuspid Aortic Valve With Raphe Formation. Circulation 2024; 149:605-626. [PMID: 38018454 DOI: 10.1161/circulationaha.123.065458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND A better understanding of the molecular mechanism of aortic valve development and bicuspid aortic valve (BAV) formation would significantly improve and optimize the therapeutic strategy for BAV treatment. Over the past decade, the genes involved in aortic valve development and BAV formation have been increasingly recognized. On the other hand, ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) gene family members have been reported to be able to modulate cardiovascular development and diseases. The present study aimed to further investigate the roles of ADAMTS family members in aortic valve development and BAV formation. METHODS Morpholino-based ADAMTS family gene-targeted screening for zebrafish heart outflow tract phenotypes combined with DNA sequencing in a 304 cohort BAV patient registry study was initially carried out to identify potentially related genes. Both ADAMTS gene-specific fluorescence in situ hybridization assay and genetic tracing experiments were performed to evaluate the expression pattern in the aortic valve. Accordingly, related genetic mouse models (both knockout and knockin) were generated using the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) method to further study the roles of ADAMTS family genes. The lineage-tracing technique was used again to evaluate how the cellular activity of specific progenitor cells was regulated by ADAMTS genes. Bulk RNA sequencing was used to investigate the signaling pathways involved. Inducible pluripotent stem cells derived from both BAV patients and genetic mouse tissue were used to study the molecular mechanism of ADAMTS. Immunohistochemistry was performed to examine the phenotype of cardiac valve anomalies, especially in the extracellular matrix components. RESULTS ADAMTS genes targeting and phenotype screening in zebrafish and targeted DNA sequencing on a cohort of patients with BAV identified ADAMTS16 (a disintegrin and metalloproteinase with thrombospondin motifs 16) as a BAV-causing gene and found the ADAMTS16 p. H357Q variant in an inherited BAV family. Both in situ hybridization and genetic tracing studies described a unique spatiotemporal pattern of ADAMTS16 expression during aortic valve development. Adamts16+/- and Adamts16+/H355Q mouse models both exhibited a right coronary cusp-noncoronary cusp fusion-type BAV phenotype, with progressive aortic valve thickening associated with raphe formation (fusion of the commissure). Further, ADAMTS16 deficiency in Tie2 lineage cells recapitulated the BAV phenotype. This was confirmed in lineage-tracing mouse models in which Adamts16 deficiency affected endothelial and second heart field cells, not the neural crest cells. Accordingly, the changes were mainly detected in the noncoronary and right coronary leaflets. Bulk RNA sequencing using inducible pluripotent stem cells-derived endothelial cells and genetic mouse embryonic heart tissue unveiled enhanced FAK (focal adhesion kinase) signaling, which was accompanied by elevated fibronectin levels. Both in vitro inducible pluripotent stem cells-derived endothelial cells culture and ex vivo embryonic outflow tract explant studies validated the altered FAK signaling. CONCLUSIONS Our present study identified a novel BAV-causing ADAMTS16 p. H357Q variant. ADAMTS16 deficiency led to BAV formation.
Collapse
Affiliation(s)
- Ying Lin
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Qifan Yang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiaoping Lin
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xianbao Liu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Yi Qian
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Dilin Xu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Naifang Cao
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (X.H.)
| | - Yanqing Zhu
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Wangxing Hu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiaopeng He
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Zhengyang Yu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiangmin Kong
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Lianlian Zhu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Zhiwei Zhong
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Kai Liu
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Bin Zhou
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences (B.Z.)
| | - Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University Health Science Center, China (Y.W.)
| | - Jinrong Peng
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Wei Zhu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Jian'an Wang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- Research Center for Life Science and Human Health, Binjiang Institute (J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| |
Collapse
|
9
|
Liu T, Zhu Q, Kai Y, Bingham T, Wang S, Cha HJ, Mehta S, Schlaeger TM, Yuan GC, Orkin SH. Matrin3 mediates differentiation through stabilizing chromatin loop-domain interactions and YY1 mediated enhancer-promoter interactions. Nat Commun 2024; 15:1274. [PMID: 38341433 PMCID: PMC10858947 DOI: 10.1038/s41467-024-45386-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Although emerging evidence indicates that alterations in proteins within nuclear compartments elicit changes in chromosomal architecture and differentiation, the underlying mechanisms are not well understood. Here we investigate the direct role of the abundant nuclear complex protein Matrin3 (Matr3) in chromatin architecture and development in the context of myogenesis. Using an acute targeted protein degradation platform (dTAG-Matr3), we reveal the dynamics of development-related chromatin reorganization. High-throughput chromosome conformation capture (Hi-C) experiments revealed substantial chromatin loop rearrangements soon after Matr3 depletion. Notably, YY1 binding was detected, accompanied by the emergence of novel YY1-mediated enhancer-promoter loops, which occurred concurrently with changes in histone modifications and chromatin-level binding patterns. Changes in chromatin occupancy by Matr3 also correlated with these alterations. Overall, our results suggest that Matr3 mediates differentiation through stabilizing chromatin accessibility and chromatin loop-domain interactions, and highlight a conserved and direct role for Matr3 in maintenance of chromosomal architecture.
Collapse
Affiliation(s)
- Tianxin Liu
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Qian Zhu
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- Lester Sue Smith Breast Center, Department of Human Molecular Genetics, Baylor College of Medicine, 1 Moursund St, Houston, TX, 77030, USA
| | - Yan Kai
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Trevor Bingham
- Stem Cell Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Stacy Wang
- Lester Sue Smith Breast Center, Department of Human Molecular Genetics, Baylor College of Medicine, 1 Moursund St, Houston, TX, 77030, USA
| | - Hye Ji Cha
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Biomedical Science & Engineering, Dankook University, Cheonan, 31116, South Korea
| | - Stuti Mehta
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Guo-Cheng Yuan
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stuart H Orkin
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, 02115, USA.
| |
Collapse
|
10
|
CLYMAN RONALDI. Patent Ductus Arteriosus in the Preterm Infant. AVERY'S DISEASES OF THE NEWBORN 2024:716-726.e11. [DOI: 10.1016/b978-0-323-82823-9.00048-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Nascimento MS, de Paula SG, Lago Alves TC, Noronha BG, Medeiros H. Latex-Free Anesthesia for Craniosynostosis Surgery Associated With Xia-Gibbs Syndrome: A Case Report. Cureus 2023; 15:e46544. [PMID: 37927632 PMCID: PMC10625484 DOI: 10.7759/cureus.46544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
This case report describes the anesthetic management of a one-year-old patient with Xia-Gibbs syndrome, which is a rare genetic condition caused by a mutation in the AHDC1 gene. The procedure involved calvarial vault remodeling and fronto-orbital advancement to correct a left coronal craniosynostosis. In addition, the patient had a history of seizures and latex-fruit syndrome, which necessitated careful preoperative planning and management. Despite the difficulties provided by the patient's cranial abnormalities and the paucity of literature on anesthetic experiences with the condition, the treatment was completed successfully and without complications. Insights are offered about the anesthetic approach for this syndromic pediatric patient undergoing neurosurgery with a high risk of bleeding. It is important to understand and prepare for the perioperative implications of this disease in order to achieve a safe outcome.
Collapse
Affiliation(s)
| | | | - Thiago C Lago Alves
- Department of Anaesthesiology, Hospital Universitário Onofre Lopes, Natal, BRA
| | - Bruna G Noronha
- Department of Anaesthesiology, Secretaria Municipal da Saúde (SMS) de Riberão Preto, Ribeirão Preto, BRA
| | - Heitor Medeiros
- Department of Anaesthesiology, Hospital Universitário Onofre Lopes, Natal, BRA
| |
Collapse
|
12
|
Schweizer L, Schaller T, Zwiebel M, Karayel Ö, Müller‐Reif JB, Zeng W, Dintner S, Nordmann TM, Hirschbühl K, Märkl B, Claus R, Mann M. Quantitative multiorgan proteomics of fatal COVID-19 uncovers tissue-specific effects beyond inflammation. EMBO Mol Med 2023; 15:e17459. [PMID: 37519267 PMCID: PMC10493576 DOI: 10.15252/emmm.202317459] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023] Open
Abstract
SARS-CoV-2 may directly and indirectly damage lung tissue and other host organs, but there are few system-wide, untargeted studies of these effects on the human body. Here, we developed a parallelized mass spectrometry (MS) proteomics workflow enabling the rapid, quantitative analysis of hundreds of virus-infected FFPE tissues. The first layer of response to SARS-CoV-2 in all tissues was dominated by circulating inflammatory molecules. Beyond systemic inflammation, we differentiated between systemic and true tissue-specific effects to reflect distinct COVID-19-associated damage patterns. Proteomic changes in the lungs resembled those of diffuse alveolar damage (DAD) in non-COVID-19 patients. Extensive organ-specific changes were also evident in the kidneys, liver, and lymphatic and vascular systems. Secondary inflammatory effects in the brain were related to rearrangements in neurotransmitter receptors and myelin degradation. These MS-proteomics-derived results contribute substantially to our understanding of COVID-19 pathomechanisms and suggest strategies for organ-specific therapeutic interventions.
Collapse
Affiliation(s)
- Lisa Schweizer
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Tina Schaller
- Pathology, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Maximilian Zwiebel
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Özge Karayel
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
Department of Physiological ChemistryGenentechSouth San FranciscoUSA
| | | | - Wen‐Feng Zeng
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | | | - Thierry M Nordmann
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Klaus Hirschbühl
- Hematology and Oncology, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Bruno Märkl
- Pathology, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Rainer Claus
- Pathology, Medical FacultyUniversity of AugsburgAugsburgGermany
- Hematology and Oncology, Medical FacultyUniversity of AugsburgAugsburgGermany
| | - Matthias Mann
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
13
|
Tessler I, Albuisson J, Piñeiro-Sabarís R, Verstraeten A, Kamber Kaya HE, Siguero-Álvarez M, Goudot G, MacGrogan D, Luyckx I, Shpitzen S, Levin G, Kelman G, Reshef N, Mananet H, Holdcraft J, Muehlschlegel JD, Peloso GM, Oppenheim O, Cheng C, Mazzella JM, Andelfinger G, Mital S, Eriksson P, Billon C, Heydarpour M, Dietz HC, Jeunemaitre X, Leitersdorf E, Sprinzak D, Blacklow SC, Body SC, Carmi S, Loeys B, de la Pompa JL, Gilon D, Messas E, Durst R. Novel Association of the NOTCH Pathway Regulator MIB1 Gene With the Development of Bicuspid Aortic Valve. JAMA Cardiol 2023; 8:721-731. [PMID: 37405741 PMCID: PMC10323766 DOI: 10.1001/jamacardio.2023.1469] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 04/21/2023] [Indexed: 07/06/2023]
Abstract
Importance Nonsyndromic bicuspid aortic valve (nsBAV) is the most common congenital heart valve malformation. BAV has a heritable component, yet only a few causative genes have been identified; understanding BAV genetics is a key point in developing personalized medicine. Objective To identify a new gene for nsBAV. Design, Setting, and Participants This was a comprehensive, multicenter, genetic association study based on candidate gene prioritization in a familial cohort followed by rare and common association studies in replication cohorts. Further validation was done using in vivo mice models. Study data were analyzed from October 2019 to October 2022. Three cohorts of patients with BAV were included in the study: (1) the discovery cohort was a large cohort of inherited cases from 29 pedigrees of French and Israeli origin; (2) the replication cohort 1 for rare variants included unrelated sporadic cases from various European ancestries; and (3) replication cohort 2 was a second validation cohort for common variants in unrelated sporadic cases from Europe and the US. Main Outcomes and Measures To identify a candidate gene for nsBAV through analysis of familial cases exome sequencing and gene prioritization tools. Replication cohort 1 was searched for rare and predicted deleterious variants and genetic association. Replication cohort 2 was used to investigate the association of common variants with BAV. Results A total of 938 patients with BAV were included in this study: 69 (7.4%) in the discovery cohort, 417 (44.5%) in replication cohort 1, and 452 (48.2%) in replication cohort 2. A novel human nsBAV gene, MINDBOMB1 homologue MIB1, was identified. MINDBOMB1 homologue (MIB1) is an E3-ubiquitin ligase essential for NOTCH-signal activation during heart development. In approximately 2% of nsBAV index cases from the discovery and replication 1 cohorts, rare MIB1 variants were detected, predicted to be damaging, and were significantly enriched compared with population-based controls (2% cases vs 0.9% controls; P = .03). In replication cohort 2, MIB1 risk haplotypes significantly associated with nsBAV were identified (permutation test, 1000 repeats; P = .02). Two genetically modified mice models carrying Mib1 variants identified in our cohort showed BAV on a NOTCH1-sensitized genetic background. Conclusions and Relevance This genetic association study identified the MIB1 gene as associated with nsBAV. This underscores the crucial role of the NOTCH pathway in the pathophysiology of BAV and its potential as a target for future diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Idit Tessler
- Cardiology Department, Hadassah Medical Center, Jerusalem, Israel
- Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, the Hebrew University, Jerusalem, Israel
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Juliette Albuisson
- Genetics Department, Assistance Publique–Hȏpitaux de Paris, Hôpital Européen Georges Pompidou, National Referral Center for Rare Vascular Diseases, VASCERN MSA European Reference Center, Paris, France
- Université Paris Cité, INSERM, U970 PARCC, Paris, France
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer –UNICANCER, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon, France
| | - Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Aline Verstraeten
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Edegem, Belgium
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hatem Elif Kamber Kaya
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Marcos Siguero-Álvarez
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Guillaume Goudot
- Université Paris Cité, INSERM, U970 PARCC, Paris, France
- Vascular Medicine Department, Assistance Publique–Hȏpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- French Research Consortium RHU STOP-AS, Rouen, France
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Ilse Luyckx
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Edegem, Belgium
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Shoshana Shpitzen
- Cardiology Department, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, the Hebrew University, Jerusalem, Israel
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Galina Levin
- Cardiology Department, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, the Hebrew University, Jerusalem, Israel
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Guy Kelman
- Faculty of Medicine, the Hebrew University, Jerusalem, Israel
- The Jerusalem Center for Personalized Computational Medicine, Jerusalem, Israel
| | - Noga Reshef
- Faculty of Medicine, the Hebrew University, Jerusalem, Israel
- The Jerusalem Center for Personalized Computational Medicine, Jerusalem, Israel
| | - Hugo Mananet
- Platform of Transfer in Cancer Biology, Georges François Leclerc Cancer –UNICANCER, Dijon, France
- Genomic and Immunotherapy Medical Institute, Dijon, France
| | - Jake Holdcraft
- Department of Anesthesiology, Boston University School of Medicine, Boston, Massachusetts
| | | | - Gina M. Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Olya Oppenheim
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Charles Cheng
- Université Paris Cité, INSERM, U970 PARCC, Paris, France
- Vascular Medicine Department, Assistance Publique–Hȏpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- French Research Consortium RHU STOP-AS, Rouen, France
| | - Jean-Michael Mazzella
- Université Paris Cité, INSERM, U970 PARCC, Paris, France
- Vascular Medicine Department, Assistance Publique–Hȏpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Gregor Andelfinger
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Université de Montreal, Montreal, Quebec, Canada
| | - Seema Mital
- Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Per Eriksson
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Solna, Sweden
| | - Clarisse Billon
- Genetics Department, Assistance Publique–Hȏpitaux de Paris, Hôpital Européen Georges Pompidou, National Referral Center for Rare Vascular Diseases, VASCERN MSA European Reference Center, Paris, France
- Université Paris Cité, INSERM, U970 PARCC, Paris, France
| | - Mahyar Heydarpour
- Department of Medicine, Division of Endocrinology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Harry C. Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xavier Jeunemaitre
- Université Paris Cité, INSERM, U970 PARCC, Paris, France
- Vascular Medicine Department, Assistance Publique–Hȏpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Eran Leitersdorf
- Cardiology Department, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, the Hebrew University, Jerusalem, Israel
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Sprinzak
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Stephen C. Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Simon C. Body
- Department of Anesthesiology, Boston University School of Medicine, Boston, Massachusetts
| | - Shai Carmi
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Bart Loeys
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Edegem, Belgium
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Dan Gilon
- Cardiology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Emmanuel Messas
- Université Paris Cité, INSERM, U970 PARCC, Paris, France
- Vascular Medicine Department, Assistance Publique–Hȏpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- French Research Consortium RHU STOP-AS, Rouen, France
| | - Ronen Durst
- Cardiology Department, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, the Hebrew University, Jerusalem, Israel
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
14
|
Muys BR, Shrestha RL, Anastasakis DG, Pongor L, Li XL, Grammatikakis I, Polash A, Chari R, Gorospe M, Harris CC, Aladjem MI, Basrai MA, Hafner M, Lal A. Matrin3 regulates mitotic spindle dynamics by controlling alternative splicing of CDC14B. Cell Rep 2023; 42:112260. [PMID: 36924503 PMCID: PMC10132239 DOI: 10.1016/j.celrep.2023.112260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/03/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
Matrin3 is an RNA-binding protein that regulates diverse RNA-related processes, including mRNA splicing. Although Matrin3 has been intensively studied in neurodegenerative diseases, its function in cancer remains unclear. Here, we report Matrin3-mediated regulation of mitotic spindle dynamics in colorectal cancer (CRC) cells. We comprehensively identified RNAs bound and regulated by Matrin3 in CRC cells and focused on CDC14B, one of the top Matrin3 targets. Matrin3 knockdown results in increased inclusion of an exon containing a premature termination codon in the CDC14B transcript and simultaneous down-regulation of the standard CDC14B transcript. Knockdown of CDC14B phenocopies the defects in mitotic spindle dynamics upon Matrin3 knockdown, and the elongated and misoriented mitotic spindle observed upon Matrin3 knockdown are rescued upon overexpression of CDC14B, suggesting that CDC14B is a key downstream effector of Matrin3. Collectively, these data reveal a role for the Matrin3/CDC14B axis in control of mitotic spindle dynamics.
Collapse
Affiliation(s)
- Bruna R Muys
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | | | - Dimitrios G Anastasakis
- RNA Molecular Biology Laboratory, National Institute for Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD 20892, USA
| | - Lorinc Pongor
- Developmental Therapeutics Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Xiao Ling Li
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Ioannis Grammatikakis
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Ahsan Polash
- RNA Molecular Biology Laboratory, National Institute for Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD 20892, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Lab for Cancer Research, Frederick, MD 21701, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD 21224, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, MD 20892, USA
| | - Mirit I Aladjem
- Developmental Therapeutics Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | | | - Markus Hafner
- RNA Molecular Biology Laboratory, National Institute for Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD 20892, USA.
| | - Ashish Lal
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD 20892, USA.
| |
Collapse
|
15
|
Taliani V, Buonaiuto G, Desideri F, Setti A, Santini T, Galfrè S, Schirone L, Mariani D, Frati G, Valenti V, Sciarretta S, Perlas E, Nicoletti C, Musarò A, Ballarino M. The long noncoding RNA Charme supervises cardiomyocyte maturation by controlling cell differentiation programs in the developing heart. eLife 2023; 12:81360. [PMID: 36877136 PMCID: PMC10023161 DOI: 10.7554/elife.81360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/03/2023] [Indexed: 03/07/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are emerging as critical regulators of heart physiology and disease, although the studies unveiling their modes of action are still limited to few examples. We recently identified pCharme, a chromatin-associated lncRNA whose functional knockout in mice results in defective myogenesis and morphological remodeling of the cardiac muscle. Here, we combined Cap-Analysis of Gene Expression (CAGE), single-cell (sc)RNA sequencing, and whole-mount in situ hybridization analyses to study pCharme cardiac expression. Since the early steps of cardiomyogenesis, we found the lncRNA being specifically restricted to cardiomyocytes, where it assists the formation of specific nuclear condensates containing MATR3, as well as important RNAs for cardiac development. In line with the functional significance of these activities, pCharme ablation in mice results in a delayed maturation of cardiomyocytes, which ultimately leads to morphological alterations of the ventricular myocardium. Since congenital anomalies in myocardium are clinically relevant in humans and predispose patients to major complications, the identification of novel genes controlling cardiac morphology becomes crucial. Our study offers unique insights into a novel lncRNA-mediated regulatory mechanism promoting cardiomyocyte maturation and bears relevance to Charme locus for future theranostic applications.
Collapse
Affiliation(s)
- Valeria Taliani
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of RomeRomeItaly
| | - Giulia Buonaiuto
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of RomeRomeItaly
| | - Fabio Desideri
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia (IIT)RomeItaly
| | - Adriano Setti
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of RomeRomeItaly
| | - Tiziana Santini
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of RomeRomeItaly
| | - Silvia Galfrè
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia (IIT)RomeItaly
| | - Leonardo Schirone
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeLatinaItaly
| | - Davide Mariani
- Center for Human Technologies, Istituto Italiano di TecnologiaGenovaItaly
| | - Giacomo Frati
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeLatinaItaly
| | - Valentina Valenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeLatinaItaly
| | - Sebastiano Sciarretta
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of RomeLatinaItaly
| | - Emerald Perlas
- Epigenetics and Neurobiology Unit, EMBL-RomeMonterotondoItaly
| | - Carmine Nicoletti
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of RomeRomeItaly
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of RomeRomeItaly
| | - Monica Ballarino
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of RomeRomeItaly
| |
Collapse
|
16
|
Huang K, Fan X, Jiang Y, Jin S, Huang J, Pang L, Wang Y, Wu Y, Sun X. Integrative identification of hub genes in development of atrial fibrillation related stroke. PLoS One 2023; 18:e0283617. [PMID: 36952494 PMCID: PMC10035830 DOI: 10.1371/journal.pone.0283617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/13/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND As the most common arrhythmia, atrial fibrillation (AF) is associated with a significantly increased risk of stroke, which causes high disability and mortality. To date, the underlying mechanism of stroke occurring after AF remains unclear. Herein, we studied hub genes and regulatory pathways involved in AF and secondary stroke and aimed to reveal biomarkers and therapeutic targets of AF-related stroke. METHODS The GSE79768 and GSE58294 datasets were used to analyze AF- and stroke-related differentially expressed genes (DEGs) to obtain a DEG1 dataset. Weighted correlation network analysis (WGCNA) was used to identify modules associated with AF-related stroke in GSE66724 (DEG2). DEG1 and DEG2 were merged, and hub genes were identified based on protein-protein interaction networks. Gene Ontology terms were used to analyze the enriched pathways. The GSE129409 and GSE70887 were applied to construct a circRNA-miRNA-mRNA network in AF-related stroke. Hub genes were verified in patients using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS We identified 3,132 DEGs in blood samples and 253 DEGs in left atrial specimens. Co-expressed hub genes of EIF4E3, ZNF595, ZNF700, MATR3, ACKR4, ANXA3, SEPSECS-AS1, and RNF166 were significantly associated with AF-related stroke. The hsa_circ_0018657/hsa-miR-198/EIF4E3 pathway was explored as the regulating axis in AF-related stroke. The qRT-PCR results were consistent with the bioinformatic analysis. CONCLUSIONS Hub genes EIF4E3, ZNF595, ZNF700, MATR3, ACKR4, ANXA3, SEPSECS-AS1, and RNF166 have potential as novel biomarkers and therapeutic targets in AF-related stroke. The hsa_circ_0018657/hsa-miR-198/EIF4E3 axis could play an important role regulating the development of AF-related stroke.
Collapse
Affiliation(s)
- Kai Huang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Xi Fan
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Yuwen Jiang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Jiechun Huang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Liewen Pang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Yiqing Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Xiaotian Sun
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
17
|
Yasuhara J, Schultz K, Bigelow AM, Garg V. Congenital aortic valve stenosis: from pathophysiology to molecular genetics and the need for novel therapeutics. Front Cardiovasc Med 2023; 10:1142707. [PMID: 37187784 PMCID: PMC10175644 DOI: 10.3389/fcvm.2023.1142707] [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: 01/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Congenital aortic valve stenosis (AVS) is one of the most common valve anomalies and accounts for 3%-6% of cardiac malformations. As congenital AVS is often progressive, many patients, both children and adults, require transcatheter or surgical intervention throughout their lives. While the mechanisms of degenerative aortic valve disease in the adult population are partially described, the pathophysiology of adult AVS is different from congenital AVS in children as epigenetic and environmental risk factors play a significant role in manifestations of aortic valve disease in adults. Despite increased understanding of genetic basis of congenital aortic valve disease such as bicuspid aortic valve, the etiology and underlying mechanisms of congenital AVS in infants and children remain unknown. Herein, we review the pathophysiology of congenitally stenotic aortic valves and their natural history and disease course along with current management strategies. With the rapid expansion of knowledge of genetic origins of congenital heart defects, we also summarize the literature on the genetic contributors to congenital AVS. Further, this increased molecular understanding has led to the expansion of animal models with congenital aortic valve anomalies. Finally, we discuss the potential to develop novel therapeutics for congenital AVS that expand on integration of these molecular and genetic advances.
Collapse
Affiliation(s)
- Jun Yasuhara
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Correspondence: Jun Yasuhara Vidu Garg
| | - Karlee Schultz
- Medical Student Research Program, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Amee M. Bigelow
- Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Vidu Garg
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States
- Correspondence: Jun Yasuhara Vidu Garg
| |
Collapse
|
18
|
Romano F, Falco M, Cappuccio G, Brunetti-Pierri N, Lonardo F, Torella A, Digilio MC, Dentici ML, Alfieri P, Agolini E, Novelli A, Garavelli L, Accogli A, Striano P, Scarano G, Nigro V, Scala M, Capra V. Genotype-phenotype spectrum and correlations in Xia-Gibbs syndrome: Report of five novel cases and literature review. Birth Defects Res 2022; 114:759-767. [PMID: 35716097 PMCID: PMC9545659 DOI: 10.1002/bdr2.2058] [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: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/08/2022] [Indexed: 11/06/2022]
Abstract
Background Xia‐Gibbs syndrome (XGS) is a rare neurodevelopmental disorder caused by pathogenic variants in the AT‐hook DNA‐binding motif‐containing 1 gene (AHDC1), encoding a protein with a crucial role in transcription and epigenetic regulation, axonogenesis, brain function, and neurodevelopment. AHDC1 variants possibly act through a dominant‐negative mechanism and may interfere with DNA repair processes, leading to genome instability and impaired DNA translesion repair. Variants affecting residues closer to the N‐terminal are thought to determine a milder phenotype with better cognitive performances. However, clean‐cut genotype–phenotype correlations are still lacking. Cases In this study, we investigated five subjects with XGS in whom exome sequencing led to the identification of five novel de novo pathogenic variants in AHDC1. All variants were extremely rare and predicted to cause a loss of protein function. The phenotype of the reported patients included developmental delay, hypotonia, and distinctive facial dysmorphisms. Additionally, uncommon clinical features were observed, including congenital hypothyroidism and peculiar skeletal abnormalities. Conclusions In this study, we report uncommon XGS features associated with five novel truncating variants in AHDC, thus expanding the genotype and phenotypic spectrum of this complex condition. We also compared our cases to previously reported cases, discussing the current status of genotype–phenotype correlations in XGS.
Collapse
Affiliation(s)
- Ferruccio Romano
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | - Annalaura Torella
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Cristina Digilio
- Medical Genetics Unit, Medical Genetics and Rare Disease Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Lisa Dentici
- Medical Genetics Unit, Medical Genetics and Rare Disease Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paolo Alfieri
- Neuropsichiatric Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Emanuele Agolini
- Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Andrea Accogli
- Division of Medical Genetics, Department of Specialized Medicine, McGill University, Quebec, Canada.,Department of Human Genetics, McGill University, Quebec, Canada
| | -
- Telethon Foundation, Rome, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.,Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Vincenzo Nigro
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.,Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Valeria Capra
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
19
|
Gordon DM, Cunningham D, Zender G, Lawrence PJ, Penaloza JS, Lin H, Fitzgerald-Butt SM, Myers K, Duong T, Corsmeier DJ, Gaither JB, Kuck HC, Wijeratne S, Moreland B, Kelly BJ, Baylor-Johns Hopkins Center for Mendelian Genomics, Garg V, White P, McBride KL. Exome sequencing in multiplex families with left-sided cardiac defects has high yield for disease gene discovery. PLoS Genet 2022; 18:e1010236. [PMID: 35737725 PMCID: PMC9258875 DOI: 10.1371/journal.pgen.1010236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/06/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022] Open
Abstract
Congenital heart disease (CHD) is a common group of birth defects with a strong genetic contribution to their etiology, but historically the diagnostic yield from exome studies of isolated CHD has been low. Pleiotropy, variable expressivity, and the difficulty of accurately phenotyping newborns contribute to this problem. We hypothesized that performing exome sequencing on selected individuals in families with multiple members affected by left-sided CHD, then filtering variants by population frequency, in silico predictive algorithms, and phenotypic annotations from publicly available databases would increase this yield and generate a list of candidate disease-causing variants that would show a high validation rate. In eight of the nineteen families in our study (42%), we established a well-known gene/phenotype link for a candidate variant or performed confirmation of a candidate variant’s effect on protein function, including variants in genes not previously described or firmly established as disease genes in the body of CHD literature: BMP10, CASZ1, ROCK1 and SMYD1. Two plausible variants in different genes were found to segregate in the same family in two instances suggesting oligogenic inheritance. These results highlight the need for functional validation and demonstrate that in the era of next-generation sequencing, multiplex families with isolated CHD can still bring high yield to the discovery of novel disease genes. Congenital heart disease is a common group of birth defects that are a leading cause of death in children under one year of age. There is strong evidence that genetics plays a role in causing congenital heart disease. While studies using individual cases have identified causative genes for those with a heart defect when accompanied by other birth defects or intellectual disabilities, for individuals who have only a heart defect without other problems, a genetic cause can be found in fewer than 10%. In this study, we enrolled families where there was more than one individual with a heart defect. This allowed us to take advantage of inheritance by searching for potential disease-causing genetic variants in common among all affected individuals in the family. Among 19 families studied, we were able to find a plausible disease-causing variant in eight of them and identified new genes that may cause or contribute to the presence of a heart defect. Two families had potential disease-causing variants in two different genes. We designed assays to test if the variants led to altered function of the protein coded by the gene, demonstrating a functional consequence that support the gene and variant as contributing to the heart defect. These findings show that studying families may be more effective than using individuals to find causes of heart defects. In addition, this family-based method suggests that changes in more than one gene may be required for a heart defect to occur.
Collapse
Affiliation(s)
- David M. Gordon
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - David Cunningham
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Gloria Zender
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Patrick J. Lawrence
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Jacqueline S. Penaloza
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Hui Lin
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Sara M. Fitzgerald-Butt
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Katherine Myers
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Tiffany Duong
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Donald J. Corsmeier
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Jeffrey B. Gaither
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Harkness C. Kuck
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Saranga Wijeratne
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Blythe Moreland
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Benjamin J. Kelly
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | | | - Vidu Garg
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (VG); (PW); (KLM)
| | - Peter White
- Computational Genomics Group, The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (VG); (PW); (KLM)
| | - Kim L. McBride
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (VG); (PW); (KLM)
| |
Collapse
|
20
|
Proteomic profiling of concurrently isolated primary microvascular endothelial cells, pericytes, and vascular smooth muscle cells from adult mouse heart. Sci Rep 2022; 12:8835. [PMID: 35614104 PMCID: PMC9132906 DOI: 10.1038/s41598-022-12749-6] [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: 12/16/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
The microcirculation serves crucial functions in adult heart, distinct from those carried out by epicardial vessels. Microvessels are governed by unique regulatory mechanisms, impairment of which leads to microvessel-specific pathology. There are few treatment options for patients with microvascular heart disease, primarily due to limited understanding of underlying pathology. High throughput mRNA sequencing and protein expression profiling in specific cells can improve our understanding of microvessel biology and disease at the molecular level. Understanding responses of individual microvascular cells to the same physiological or pathophysiological stimuli requires the ability to isolate the specific cell types that comprise the functional units of the microcirculation in the heart, preferably from the same heart, to ensure that different cells have been exposed to the same in-vivo conditions. We developed an integrated process for simultaneous isolation and culture of the main cell types comprising the microcirculation in adult mouse heart: endothelial cells, pericytes, and vascular smooth muscle cells. These cell types were characterized with isobaric labeling quantitative proteomics and mRNA sequencing. We defined microvascular cell proteomes, identified novel protein markers, and confirmed established cell-specific markers. Our results allow identification of unique markers and regulatory proteins that govern microvascular physiology and pathology.
Collapse
|
21
|
Negishi K, Aizawa K, Shindo T, Suzuki T, Sakurai T, Saito Y, Miyakawa T, Tanokura M, Kataoka Y, Maeda M, Tomida S, Morita H, Takeda N, Komuro I, Kario K, Nagai R, Imai Y. An Myh11 single lysine deletion causes aortic dissection by reducing aortic structural integrity and contractility. Sci Rep 2022; 12:8844. [PMID: 35614093 PMCID: PMC9133116 DOI: 10.1038/s41598-022-12418-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/10/2022] [Indexed: 11/12/2022] Open
Abstract
Pathogenic variants in myosin heavy chain (Myh11) cause familial thoracic aortic aneurysms and dissections (FTAAD). However, the underlying pathological mechanisms remain unclear because of a lack of animal models. In this study, we established a mouse model with Myh11 K1256del, the pathogenic variant we found previously in two FTAAD families. The Myh11∆K/∆K aorta showed increased wall thickness and ultrastructural abnormalities, including weakened cell adhesion. Notably, the Myh11∆K/+ mice developed aortic dissections and intramural haematomas when stimulated with angiotensin II. Mechanistically, integrin subunit alpha2 (Itga2) was downregulated in the Myh11∆K/∆K aortas, and the smooth muscle cell lineage cells that differentiated from Myh11∆K/∆K induced pluripotent stem cells. The contractility of the Myh11∆K/∆K aortas in response to phenylephrine was also reduced. These results imply that the suboptimal cell adhesion indicated by Itga2 downregulation causes a defect in the contraction of the aorta. Consequently, the defective contraction may increase the haemodynamic stress underlying the aortic dissections.
Collapse
Affiliation(s)
- Keita Negishi
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kenichi Aizawa
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Yuichiro Saito
- System Integration Center, Gunma University Hospital, Gunma, Japan
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yosky Kataoka
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- RIKEN-JEOL Collaboration Center, Kobe, Japan
| | - Mitsuyo Maeda
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- RIKEN-JEOL Collaboration Center, Kobe, Japan
| | - Shota Tomida
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Ryozo Nagai
- Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Yasushi Imai
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
22
|
Anderson RH, Bamforth SD. Morphogenesis of the Mammalian Aortic Arch Arteries. Front Cell Dev Biol 2022; 10:892900. [PMID: 35620058 PMCID: PMC9127140 DOI: 10.3389/fcell.2022.892900] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/20/2022] [Indexed: 12/02/2022] Open
Abstract
The major vessels in mammals that take blood away from the heart and deliver it to the arms and the head take their origin from the aortic arch and are derived from the arteries formed within the embryonic pharyngeal arches. These pharyngeal arch arteries, initially symmetrical, form in a cranial to caudal sequence within the pharyngeal mesenchyme. They then undergo a complex process of remodeling to produce the asymmetrical brachiocephalic arteries as seen in the adult. A complex interaction between the tissues of the pharyngeal arches and the genes they express is required to ensure that arterial formation and remodeling is able to proceed normally. If this process is disrupted, life-threatening congenital cardiovascular malformations can occur, such as interruption of the aortic arch, isolation of individual arteries, or so-called vascular rings. Here, using state-of-the-art imaging techniques, we describe the morphogenesis of the arteries in humans and mice and the cardiovascular defects in the Tbx1 mutant mouse model. We provide details of the process of remodeling, clarifying also the morphogenesis of the external carotid artery and the so-called "migration" of the left subclavian artery.
Collapse
|
23
|
Sprunger ML, Lee K, Sohn BS, Jackrel ME. Molecular determinants and modifiers of Matrin-3 toxicity, condensate dynamics, and droplet morphology. iScience 2022; 25:103900. [PMID: 35252808 PMCID: PMC8889142 DOI: 10.1016/j.isci.2022.103900] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/04/2022] [Accepted: 02/04/2022] [Indexed: 11/17/2022] Open
Abstract
Matrin-3 (MATR3) is a DNA- and RNA-binding protein implicated in amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and distal myopathy. Here, we report the development of a yeast model of MATR3 proteotoxicity and aggregation. MATR3 is toxic and forms dynamic shell-like nuclear condensates in yeast. Disease-associated mutations in MATR3 impair condensate dynamics and disrupt condensate morphology. MATR3 toxicity is largely driven by its RNA-recognitions motifs (RRMs). Further, deletion of one or both RRMs drives coalescence of these condensates. Aberrant phase separation of several different RBPs underpins ALS/FTD, and we have engineered Hsp104 variants to reverse this misfolding. Here, we demonstrate that these same variants also counter MATR3 toxicity. We suggest that these Hsp104 variants which rescue MATR3, TDP-43, and FUS toxicity might be employed against a range of ALS/FTD-associated proteins. We anticipate that our yeast model could be a useful platform to screen for modulators of MATR3 misfolding.
Collapse
Affiliation(s)
- Macy L. Sprunger
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Ken Lee
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Brian S. Sohn
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | | |
Collapse
|
24
|
Todd TW, Petrucelli L. Modelling amyotrophic lateral sclerosis in rodents. Nat Rev Neurosci 2022; 23:231-251. [PMID: 35260846 DOI: 10.1038/s41583-022-00564-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
The efficient study of human disease requires the proper tools, one of the most crucial of which is an accurate animal model that faithfully recapitulates the human condition. The study of amyotrophic lateral sclerosis (ALS) is no exception. Although the majority of ALS cases are considered sporadic, most animal models of this disease rely on genetic mutations identified in familial cases. Over the past decade, the number of genes associated with ALS has risen dramatically and, with each new genetic variant, there is a drive to develop associated animal models. Rodent models are of particular importance as they allow for the study of ALS in the context of a living mammal with a comparable CNS. Such models not only help to verify the pathogenicity of novel mutations but also provide critical insight into disease mechanisms and are crucial for the testing of new therapeutics. In this Review, we aim to summarize the full spectrum of ALS rodent models developed to date.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA.
| |
Collapse
|
25
|
Yarboro MT, Gopal SH, Su RL, Morgan TM, Reese J. Mouse models of patent ductus arteriosus (PDA) and their relevance for human PDA. Dev Dyn 2022; 251:424-443. [PMID: 34350653 PMCID: PMC8814064 DOI: 10.1002/dvdy.408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/16/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
The ductus arteriosus (DA) is a unique fetal vascular shunt, which allows blood to bypass the developing lungs in utero. After birth, changes in complex signaling pathways lead to constriction and permanent closure of the DA. The persistent patency of the DA (PDA) is a common disorder in preterm infants, yet the underlying causes of PDA are not fully defined. Although limits on the availability of human DA tissues prevent comprehensive studies on the mechanisms of DA function, mouse models have been developed that reveal critical pathways in DA regulation. Over 20 different transgenic models of PDA in mice have been described, with implications for human DA biology. Similarly, we enumerate 224 human single-gene syndromes that are associated with PDA, including a small subset that consistently feature PDA as a prominent phenotype. Comparison and functional analyses of these genes provide insight into DA development and identify key regulatory pathways that may serve as potential therapeutic targets for the management of PDA.
Collapse
Affiliation(s)
- Michael T Yarboro
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Srirupa H Gopal
- Department of Pediatrics, Erlanger Health System, Chattanooga, Tennessee, USA
| | - Rachel L Su
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Thomas M Morgan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeff Reese
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA.,Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
26
|
In heart failure reactivation of RNA-binding proteins is associated with the expression of 1,523 fetal-specific isoforms. PLoS Comput Biol 2022; 18:e1009918. [PMID: 35226669 PMCID: PMC8912908 DOI: 10.1371/journal.pcbi.1009918] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/10/2022] [Accepted: 02/10/2022] [Indexed: 01/03/2023] Open
Abstract
Reactivation of fetal-specific genes and isoforms occurs during heart failure. However, the underlying molecular mechanisms and the extent to which the fetal program switch occurs remains unclear. Limitations hindering transcriptome-wide analyses of alternative splicing differences (i.e. isoform switching) in cardiovascular system (CVS) tissues between fetal, healthy adult and heart failure have included both cellular heterogeneity across bulk RNA-seq samples and limited availability of fetal tissue for research. To overcome these limitations, we have deconvoluted the cellular compositions of 996 RNA-seq samples representing heart failure, healthy adult (heart and arteria), and fetal-like (iPSC-derived cardiovascular progenitor cells) CVS tissues. Comparison of the expression profiles revealed that reactivation of fetal-specific RNA-binding proteins (RBPs), and the accompanied re-expression of 1,523 fetal-specific isoforms, contribute to the transcriptome differences between heart failure and healthy adult heart. Of note, isoforms for 20 different RBPs were among those that reverted in heart failure to the fetal-like expression pattern. We determined that, compared with adult-specific isoforms, fetal-specific isoforms encode proteins that tend to have more functions, are more likely to harbor RBP binding sites, have canonical sequences at their splice sites, and contain typical upstream polypyrimidine tracts. Our study suggests that compared with healthy adult, fetal cardiac tissue requires stricter transcriptional regulation, and that during heart failure reversion to this stricter transcriptional regulation occurs. Furthermore, we provide a resource of cardiac developmental stage-specific and heart failure-associated genes and isoforms, which are largely unexplored and can be exploited to investigate novel therapeutics for heart failure. Heart failure is a chronic condition in which the heart does not pump enough blood. It has been shown that in heart failure, the adult heart reverts to a fetal-like metabolic state and oxygen consumption. Additionally, genes and isoforms that are expressed in the heart only during fetal development (i.e. not in the healthy adult heart) are turned on in heart failure. However, the underlying molecular mechanisms and the extent to which the switch to a fetal gene program occurs remains unclear. In this study, we initially characterized the differences between the fetal and adult heart transcriptomes (entire set of expressed genes and isoforms). We found that RNA binding proteins (RBPs), a family of genes that regulate multiple aspects of a transcript’s maturation, including transcription, splicing and post-transcriptional modifications, play a central role in the differences between fetal and adult heart tissues. We observed that many RBPs that are only expressed in the heart during fetal development become reactivated in heart failure, resulting in the expression of 1,523 fetal-specific isoforms. These findings suggest that reactivation of fetal-specific RBPs in heart failure drives a transcriptome-wide switch to expression of fetal-specific isoforms; and hence that RBPs could potentially serve as novel therapeutic targets.
Collapse
|
27
|
Khayat MM, Hu J, Jiang Y, Li H, Chander V, Dawood M, Hansen AW, Li S, Friedman J, Cross L, Bijlsma EK, Ruivenkamp CA, Sansbury FH, Innis JW, Omark O’Shea J, Meng Q, Rosenfeld JA, McWalter K, Wangler MF, Lupski JR, Posey JE, Murdock D, Gibbs RA. AHDC1 missense mutations in Xia-Gibbs syndrome. HGG ADVANCES 2021; 2:100049. [PMID: 34950897 PMCID: PMC8694554 DOI: 10.1016/j.xhgg.2021.100049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/04/2021] [Indexed: 12/19/2022] Open
Abstract
Xia-Gibbs syndrome (XGS; MIM: 615829) is a phenotypically heterogeneous neurodevelopmental disorder (NDD) caused by newly arising mutations in the AT-Hook DNA-Binding Motif-Containing 1 (AHDC1) gene that are predicted to lead to truncated AHDC1 protein synthesis. More than 270 individuals have been diagnosed with XGS worldwide. Despite the absence of an independent assay for AHDC1 protein function to corroborate potential functional consequences of rare variant genetic findings, there are also reports of individuals with XGS-like trait manifestations who have de novo missense AHDC1 mutations and who have been provided a molecular diagnosis of the disorder. To investigate a potential contribution of missense mutations to XGS, we mapped the missense mutations from 10 such individuals to the AHDC1 conserved protein domain structure and detailed the observed phenotypes. Five newly identified individuals were ascertained from a local XGS Registry, and an additional five were taken from external reports or databases, including one publication. Where clinical data were available, individuals with missense mutations all displayed phenotypes consistent with those observed in individuals with AHDC1 truncating mutations, including delayed motor milestones, intellectual disability (ID), hypotonia, and speech delay. A subset of the 10 reported missense mutations cluster in two regions of the AHDC1 protein with known conserved domains, likely representing functional motifs. Variants outside the clustered regions score lower for computational prediction of their likely damaging effects. Overall, de novo missense variants in AHDC1 are likely diagnostic of XGS when in silico analysis of their position relative to conserved regions is considered together with disease trait manifestations.
Collapse
Affiliation(s)
- Michael M. Khayat
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jianhong Hu
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Yunyun Jiang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - He Li
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Varuna Chander
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Moez Dawood
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Adam W. Hansen
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shoudong Li
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer Friedman
- UCSD Departments of Neuroscience and Pediatrics, Rady Children’s Hospital Division of Neurology, Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
| | - Laura Cross
- Department of Pediatrics and Genetics, Children’s Mercy Hospitals, Kansas City, MO, USA
| | - Emilia K. Bijlsma
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Francis H. Sansbury
- All Wales Medical Genomics Service, NHS Wales Cardiff and Vale University Health Board, Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - Jeffrey W. Innis
- Departments of Human Genetics, Pediatrics, and Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Qingchang Meng
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Michael F. Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Neurological Research Institute, Houston, TX, USA
| | - James R. Lupski
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer E. Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - David Murdock
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
28
|
Kern CB. Excess Provisional Extracellular Matrix: A Common Factor in Bicuspid Aortic Valve Formation. J Cardiovasc Dev Dis 2021; 8:92. [PMID: 34436234 PMCID: PMC8396938 DOI: 10.3390/jcdd8080092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
A bicuspid aortic valve (BAV) is the most common cardiac malformation, found in 0.5% to 2% of the population. BAVs are present in approximately 50% of patients with severe aortic stenosis and are an independent risk factor for aortic aneurysms. Currently, there are no therapeutics to treat BAV, and the human mutations identified to date represent a relatively small number of BAV patients. However, the discovery of BAV in an increasing number of genetically modified mice is advancing our understanding of molecular pathways that contribute to BAV formation. In this study, we utilized the comparison of BAV phenotypic characteristics between murine models as a tool to advance our understanding of BAV formation. The collation of murine BAV data indicated that excess versican within the provisional extracellular matrix (P-ECM) is a common factor in BAV development. While the percentage of BAVs is low in many of the murine BAV models, the remaining mutant mice exhibit larger and more amorphous tricuspid AoVs, also with excess P-ECM compared to littermates. The identification of common molecular characteristics among murine BAV models may lead to BAV therapeutic targets and biomarkers of disease progression for this highly prevalent and heterogeneous cardiovascular malformation.
Collapse
Affiliation(s)
- Christine B Kern
- Department of Regenerative Medicine and Cell Biology, 171 Ashley Avenue, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
29
|
Abstract
Bicuspid aortic valve (BAV) is the most common valvular congenital heart disease, with a prevalence of 0.5 to 2% in the general population. Patients with BAV are at risk for developing cardiovascular complications, some of which are life-threatening. BAV has a wide spectrum of clinical presentations, ranging from silent malformation to severe and even fatal cardiac events. Despite the significant burden on both the patients and the health systems, data are limited regarding pathophysiology, risk factors, and genetics. Family studies indicate that BAV is highly heritable, with autosomal dominant inheritance, incomplete penetrance, variable expressivity, and male predominance. Owing to its complex genetic model, including high genetic heterogenicity, only a few genes were identified in association with BAV, while the majority of BAV genetics remains obscure. Here, we review the different forms of BAV and the current data regarding its genetics. Given the clear heritably of BAV with the potential high impact on clinical outcome, the clinical value and cost effectiveness of cascade screening are discussed.
Collapse
|
30
|
Abstract
Aortic stenosis (AS) remains one of the most common forms of valve disease, with significant impact on patient survival. The disease is characterized by left ventricular outflow obstruction and encompasses a series of stenotic lesions starting from the left ventricular outflow tract to the descending aorta. Obstructions may be subvalvar, valvar, or supravalvar and can be present at birth (congenital) or acquired later in life. Bicuspid aortic valve, whereby the aortic valve forms with two instead of three cusps, is the most common cause of AS in younger patients due to primary anatomic narrowing of the valve. In addition, the secondary onset of premature calcification, likely induced by altered hemodynamics, further obstructs left ventricular outflow in bicuspid aortic valve patients. In adults, degenerative AS involves progressive calcification of an anatomically normal, tricuspid aortic valve and is attributed to lifelong exposure to multifactoral risk factors and physiological wear-and-tear that negatively impacts valve structure-function relationships. AS continues to be the most frequent valvular disease that requires intervention, and aortic valve replacement is the standard treatment for patients with severe or symptomatic AS. While the positive impacts of surgical interventions are well documented, the financial burden, the potential need for repeated procedures, and operative risks are substantial. In addition, the clinical management of asymptomatic patients remains controversial. Therefore, there is a critical need to develop alternative approaches to prevent the progression of left ventricular outflow obstruction, especially in valvar lesions. This review summarizes our current understandings of AS cause; beginning with developmental origins of congenital valve disease, and leading into the multifactorial nature of AS in the adult population.
Collapse
Affiliation(s)
- Punashi Dutta
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| | - Jeanne F James
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| | - Hail Kazik
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Biomedical Engineering, Marquette University & Medical College of Wisconsin, Milwaukee (H.K.)
| | - Joy Lincoln
- The Herma Heart Institute, Section of Pediatric Cardiology, Children's Wisconsin, Milwaukee, WI (P.D., J.F.J., H.K., J.L.).,Department of Pediatrics, Medical College of Wisconsin, Milwaukee (P.D., J.F.J., J.L.)
| |
Collapse
|
31
|
Majumdar U, Yasuhara J, Garg V. In Vivo and In Vitro Genetic Models of Congenital Heart Disease. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a036764. [PMID: 31818859 DOI: 10.1101/cshperspect.a036764] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Congenital cardiovascular malformations represent the most common type of birth defect and encompass a spectrum of anomalies that range from mild to severe. The etiology of congenital heart disease (CHD) is becoming increasingly defined based on prior epidemiologic studies that supported the importance of genetic contributors and technological advances in human genome analysis. These have led to the discovery of a growing number of disease-contributing genetic abnormalities in individuals affected by CHD. The ever-growing population of adult CHD survivors, which are the result of reductions in mortality from CHD during childhood, and this newfound genetic knowledge have led to important questions regarding recurrence risks, the mechanisms by which these defects occur, the potential for novel approaches for prevention, and the prediction of long-term cardiovascular morbidity in adult CHD survivors. Here, we will review the current status of genetic models that accurately model human CHD as they provide an important tool to answer these questions and test novel therapeutic strategies.
Collapse
Affiliation(s)
- Uddalak Majumdar
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Jun Yasuhara
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Vidu Garg
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, Ohio 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio 43205, USA.,Department of Molecular Genetics, The Ohio State University, Columbus, Ohio 43205, USA
| |
Collapse
|
32
|
Abstract
Congenital heart disease is the most common congenital defect observed in newborns. Within the spectrum of congenital heart disease are left‐sided obstructive lesions (LSOLs), which include hypoplastic left heart syndrome, aortic stenosis, bicuspid aortic valve, coarctation of the aorta, and interrupted aortic arch. These defects can arise in isolation or as a component of a defined syndrome; however, nonsyndromic defects are often observed in multiple family members and associated with high sibling recurrence risk. This clear evidence for a heritable basis has driven a lengthy search for disease‐causing variants that has uncovered both rare and common variants in genes that, when perturbed in cardiac development, can result in LSOLs. Despite advancements in genetic sequencing platforms and broadening use of exome sequencing, the currently accepted LSOL‐associated genes explain only 10% to 20% of patients. Further, the combinatorial effects of common and rare variants as a cause of LSOLs are emerging. In this review, we highlight the genes and variants associated with the different LSOLs and discuss the strengths and weaknesses of the present genetic associations. Furthermore, we discuss the research avenues needed to bridge the gaps in our current understanding of the genetic basis of nonsyndromic congenital heart disease.
Collapse
Affiliation(s)
- Lauren E Parker
- Division of Cardiology Department of Pediatrics Duke University School of Medicine Durham NC
| | - Andrew P Landstrom
- Division of Cardiology Department of Pediatrics Duke University School of Medicine Durham NC.,Department of Cell Biology Duke University School of Medicine Durham NC
| |
Collapse
|
33
|
Abstract
RNA-binding proteins (RBPs) are essential factors required for the physiological function of neurons, muscle, and other tissue types. In keeping with this, a growing body of genetic, clinical, and pathological evidence indicates that RBP dysfunction and/or gene mutation leads to neurodegeneration and myopathy. Here, we summarize the current understanding of matrin 3 (MATR3), a poorly understood RBP implicated not only in ALS and frontotemporal dementia but also in distal myopathy. We begin by reviewing MATR3's functions, its regulation, and how it may be involved in both sporadic and familial neuromuscular disease. We also discuss insights gleaned from cellular and animal models of MATR3 pathogenesis, the links between MATR3 and other disease-associated RBPs, and the mechanisms underlying RBP-mediated disorders.
Collapse
Affiliation(s)
- Ahmed M. Malik
- Medical Scientist Training Program
- Neuroscience Graduate Program, and
| | - Sami J. Barmada
- Neuroscience Graduate Program, and
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
34
|
Abstract
Xia-Gibbs syndrome (XGS) is a rare genetic disorder that has been discovered as a distinct clinical entity in the recent past. The occurrence has been attributed to the mutation of AT Hook DNA binding motif Containing 1 (AHDC1) gene that is carried on chromosome 1p36. The concerned gene participates in deoxyribonucleic acid (DNA) repair apart from other crucial functions. The mutation results in dysfunction that leads to neurodevelopmental delay. The spectrum of manifestations constitutes intellectual disabilities, hypotonia, expressive language delay, sleep difficulties, and short stature. Dysmorphic facial features include depressed nasal bridge, hypertelorism, down-slanting or up-slanting palpebral fissures, horizontal eyebrows, dysplastic dentition, thin upper lip vermilion, and micrognathia. The phenotype is still expanding. The condition may range from mild to severe dysfunction depending on the area and site of genetic aberration but variation is evident. Thus, the correlation between genotype and phenotype is largely unclear. XGS should be considered as a differential diagnosis for patients presenting with intellectual as well as developmental disabilities. Whole-exome sequencing (WES) is the genetic test that is largely used for the confirmation of diagnosis. Less is known about the natural history as only a few adults with XGS have been documented in the literature. Age-appropriate cancer screening is recommended for patients with XGS as the gene mutation alters DNA repair mechanisms that may trigger tumour formation. The management of patients diagnosed with XGS is an area that needs investigation. Though use of growth hormone replacement therapy and physiotherapy intervention have been reported as effective in previous studies, research on effective means of care of these patients is warranted on a larger number of patients. We present a review of current literature on what is known about XGS that would facilitate to identify knowledge gaps for paving a way for further studies. This, in turn, will help in provision of early and effective rehabilitation services for patients with XGS.
Collapse
Affiliation(s)
- Chanan Goyal
- Physiotherapy, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Waqar M Naqvi
- Physiotherapy, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Arti Sahu
- Physiotherapy, Datta Meghe Institute of Medical Sciences, Wardha, IND
| | - Ashish S Aujla
- Paediatric Neurology, Kids Care Paediatric Neurology Center, Raipur, IND
| |
Collapse
|
35
|
Desideri F, Cipriano A, Petrezselyova S, Buonaiuto G, Santini T, Kasparek P, Prochazka J, Janson G, Paiardini A, Calicchio A, Colantoni A, Sedlacek R, Bozzoni I, Ballarino M. Intronic Determinants Coordinate Charme lncRNA Nuclear Activity through the Interaction with MATR3 and PTBP1. Cell Rep 2020; 33:108548. [PMID: 33357424 PMCID: PMC7773549 DOI: 10.1016/j.celrep.2020.108548] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/27/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
Chromatin architect of muscle expression (Charme) is a muscle-restricted long noncoding RNA (lncRNA) that plays an important role in myogenesis. Earlier evidence indicates that the nuclear Charme isoform, named pCharme, acts on the chromatin by assisting the formation of chromatin domains where myogenic transcription occurs. By combining RNA antisense purification (RAP) with mass spectrometry and loss-of-function analyses, we have now identified the proteins that assist these chromatin activities. These proteins—which include a sub-set of splicing regulators, principally PTBP1 and the multifunctional RNA/DNA binding protein MATR3—bind to sequences located within the alternatively spliced intron-1 to form nuclear aggregates. Consistent with the functional importance of pCharme interactome in vivo, a targeted deletion of the intron-1 by a CRISPR-Cas9 approach in mouse causes the release of pCharme from the chromatin and results in cardiac defects similar to what was observed upon knockout of the full-length transcript. pCharme is the chromatin-retained isoform of the muscle-specific Charme lncRNA Intronic signals coordinate the association of pCharme with MATR3 and PTBP1 The particle assembly prompts pCharme intron-1 chromatin retention Deletion of the intron-1 by CRISPR-Cas9 leads to heart defects in mouse
Collapse
Affiliation(s)
- Fabio Desideri
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Andrea Cipriano
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Silvia Petrezselyova
- Czech Centre of Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Giulia Buonaiuto
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Tiziana Santini
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Petr Kasparek
- Czech Centre of Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Jan Prochazka
- Czech Centre of Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Giacomo Janson
- Department of Biochemical Sciences "A. Rossi Fanelli," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences "A. Rossi Fanelli," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Alessandro Calicchio
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Alessio Colantoni
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Radislav Sedlacek
- Czech Centre of Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Irene Bozzoni
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy.
| | - Monica Ballarino
- Department of Biology and Biotechnology "Charles Darwin," Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
36
|
Vigneswaran TV, Zidere V, Chivers S, Charakida M, Akolekar R, Simpson JM. Impact of prospective measurement of outflow tracts in prediction of coarctation of the aorta. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2020; 56:850-856. [PMID: 31875324 DOI: 10.1002/uog.21957] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/12/2019] [Accepted: 12/13/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVES Prenatal diagnosis of coarctation of the aorta (CoA) is associated with reduced mortality and morbidity, however, accurate prenatal prediction remains challenging. To date, studies have used retrospective measurements of the outflow tracts to evaluate their potential to predict CoA. Our primary objective was to evaluate prospectively acquired measurements of the outflow tracts in fetuses with prenatally suspected CoA. A secondary aim was to report the postnatal prevalence of bicuspid aortic valve in this cohort. METHODS Pregnancies with suspicion of isolated CoA and with a minimum of 6 months' postnatal follow-up available were identified from the cardiac database of a tertiary fetal cardiology center in the UK, between January 2002 and December 2017. Measurement of the aortic valve, pulmonary valve, distal transverse aortic arch (DTAA) and arterial duct (AD) diameters were undertaken routinely in fetuses with suspected CoA during the study period. Z-scores were computed using published reference ranges based on > 7000 fetuses from our own unit. RESULTS Of 149 pregnancies with prenatally suspected CoA included in the study, CoA was confirmed within 6 months after birth in 77/149 (51.7%) cases. DTAA diameter Z-score and the Z-score of second-trimester DTAA/AD diameter ratio were smaller in fetuses with postnatally confirmed CoA than those in false-positive cases (-2.8 vs -1.9; P = 0.039 and -3.13 vs -2.61; P = 0.005, respectively). Multiple regression analysis demonstrated that the Z-scores of DTAA and AD diameters were the only significant predictors of postnatal CoA (P = 0.001). Bicuspid aortic valve was identified in 30% of the false-positive cases. CONCLUSIONS Measurement of DTAA and AD diameter Z-scores can be used to ascertain risk for postnatal CoA in a selected cohort. The high incidence of bicuspid aortic valve in false-positive cases merits further study with respect to both etiology and longer-term significance. Copyright © 2019 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- T V Vigneswaran
- Harris Birthright Centre, Fetal Medicine Research Institute, King's College Hospital, London, UK
- Department of Congenital Heart Disease, Evelina London Children's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- Department of Biomedical Engineering, King's College Hospital, London, UK
| | - V Zidere
- Harris Birthright Centre, Fetal Medicine Research Institute, King's College Hospital, London, UK
- Department of Congenital Heart Disease, Evelina London Children's Hospital, Guy's & St Thomas' NHS Trust, London, UK
| | - S Chivers
- Department of Congenital Heart Disease, Evelina London Children's Hospital, Guy's & St Thomas' NHS Trust, London, UK
| | - M Charakida
- Harris Birthright Centre, Fetal Medicine Research Institute, King's College Hospital, London, UK
- Department of Congenital Heart Disease, Evelina London Children's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- Department of Biomedical Engineering, King's College Hospital, London, UK
| | - R Akolekar
- Harris Birthright Centre, Fetal Medicine Research Institute, King's College Hospital, London, UK
- Department of Obstetrics & Gynaecology, Medway Maritime Hospital, Gillingham, Kent, UK
| | - J M Simpson
- Harris Birthright Centre, Fetal Medicine Research Institute, King's College Hospital, London, UK
- Department of Congenital Heart Disease, Evelina London Children's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- Department of Biomedical Engineering, King's College Hospital, London, UK
| |
Collapse
|
37
|
Ramesh N, Kour S, Anderson EN, Rajasundaram D, Pandey UB. RNA-recognition motif in Matrin-3 mediates neurodegeneration through interaction with hnRNPM. Acta Neuropathol Commun 2020; 8:138. [PMID: 32811564 PMCID: PMC7437177 DOI: 10.1186/s40478-020-01021-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is an adult-onset, fatal neurodegenerative disease characterized by progressive loss of upper and lower motor neurons. While pathogenic mutations in the DNA/RNA-binding protein Matrin-3 (MATR3) are linked to ALS and distal myopathy, the molecular mechanisms underlying MATR3-mediated neuromuscular degeneration remain unclear. Methods We generated Drosophila lines with transgenic insertion of human MATR3 wildtype, disease-associated variants F115C and S85C, and deletion variants in functional domains, ΔRRM1, ΔRRM2, ΔZNF1 and ΔZNF2. We utilized genetic, behavioral and biochemical tools for comprehensive characterization of our models in vivo and in vitro. Additionally, we employed in silico approaches to find transcriptomic targets of MATR3 and hnRNPM from publicly available eCLIP datasets. Results We found that targeted expression of MATR3 in Drosophila muscles or motor neurons shorten lifespan and produces progressive motor defects, muscle degeneration and atrophy. Strikingly, deletion of its RNA-recognition motif (RRM2) mitigates MATR3 toxicity. We identified rump, the Drosophila homolog of human RNA-binding protein hnRNPM, as a modifier of mutant MATR3 toxicity in vivo. Interestingly, hnRNPM physically and functionally interacts with MATR3 in an RNA-dependent manner in mammalian cells. Furthermore, common RNA targets of MATR3 and hnRNPM converge in biological processes important for neuronal health and survival. Conclusions We propose a model of MATR3-mediated neuromuscular degeneration governed by its RNA-binding domains and modulated by interaction with splicing factor hnRNPM. Electronic supplementary material The online version of this article (10.1186/s40478-020-01021-5) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Fernández B, Soto-Navarrete MT, López-García A, López-Unzu MÁ, Durán AC, Fernández MC. Bicuspid Aortic Valve in 2 Model Species and Review of the Literature. Vet Pathol 2020; 57:321-331. [DOI: 10.1177/0300985819900018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bicuspid aortic valve (BAV) is the most common human congenital cardiac malformation. Although the etiology is unknown for most patients, formation of the 2 main BAV anatomic types (A and B) has been shown to rely on distinct morphogenetic mechanisms. Animal models of BAV include 2 spontaneous hamster strains and 27 genetically modified mouse strains. To assess the value of these models for extrapolation to humans, we examined the aortic valve anatomy of 4340 hamsters and 1823 mice from 8 and 7 unmodified strains, respectively. In addition, we reviewed the literature describing BAV in nonhuman mammals. The incidences of BAV types A and B were 2.3% and 0.03% in control hamsters and 0% and 0.3% in control mice, respectively. Hamsters from the spontaneous model had BAV type A only, whereas mice from 2 of 27 genetically modified strains had BAV type A, 23 of 27 had BAV type B, and 2 of 27 had both BAV types. In both species, BAV incidence was dependent on genetic background. Unlike mice, hamsters had a wide spectrum of aortic valve morphologies. We showed interspecific differences in the occurrence of BAV between humans, hamsters, and mice that should be considered when studying aortic valve disease using animal models. Our results suggest that genetic modifiers play a significant role in both the morphology and incidence of BAV. We propose that mutations causing anomalies in specific cardiac morphogenetic processes or cell lineages may lead to BAV types A, B, or both, depending on additional genetic, environmental, and epigenetic factors.
Collapse
Affiliation(s)
- Borja Fernández
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
- CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - María Teresa Soto-Navarrete
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Alejandro López-García
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Miguel Ángel López-Unzu
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Ana Carmen Durán
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - M. Carmen Fernández
- Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| |
Collapse
|
39
|
Cardoso-Dos-Santos AC, Oliveira Silva T, Silveira Faccini A, Woycinck Kowalski T, Bertoli-Avella A, Morales Saute JA, Schuler-Faccini L, de Oliveira Poswar F. Novel AHDC1 Gene Mutation in a Brazilian Individual: Implications of Xia-Gibbs Syndrome. Mol Syndromol 2020; 11:24-29. [PMID: 32256298 DOI: 10.1159/000505843] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2019] [Indexed: 12/18/2022] Open
Abstract
Xia-Gibbs syndrome (XGS) is a rare neurological disorder characterized by global developmental delay, hypotonia, intellectual disability, seizures, and sleep apnea. XGS is defined by monoallelic pathogenic variants in AHDC1. In this study, we identified a Brazilian patient carrying a likely de novo AHDC1 nonsense mutation (c.451C>T; p.Arg151*) which was absent in both parents. All disease-causative variants already associated with XGS have been reviewed and the mutation described here corresponds to the closest one to the N-terminal region. Our findings were discussed based on the suggested genotype-phenotype correlation of the disease.
Collapse
Affiliation(s)
| | - Thiago Oliveira Silva
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | | | | | - Jonas A Morales Saute
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Internal Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lavinia Schuler-Faccini
- Genetics Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | |
Collapse
|
40
|
Microdeletion and microduplication of 1p36.11p35.3 involving AHDC1 contribute to neurodevelopmental disorder. Eur J Med Genet 2020; 63:103611. [DOI: 10.1016/j.ejmg.2019.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 11/16/2018] [Accepted: 01/03/2019] [Indexed: 12/18/2022]
|
41
|
Gumus E. Extending the phenotype of Xia-Gibbs syndrome in a two-year-old patient with craniosynostosis with a novel de novo AHDC1 missense mutation. Eur J Med Genet 2020; 63:103637. [DOI: 10.1016/j.ejmg.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 02/08/2023]
|
42
|
Coarctation of the aorta in twins with severe hypertension. J Geriatr Cardiol 2019; 16:894-897. [PMID: 31911794 PMCID: PMC6938739 DOI: 10.11909/j.issn.1671-5411.2019.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
43
|
Bjornsson T, Thorolfsdottir RB, Sveinbjornsson G, Sulem P, Norddahl GL, Helgadottir A, Gretarsdottir S, Magnusdottir A, Danielsen R, Sigurdsson EL, Adalsteinsdottir B, Gunnarsson SI, Jonsdottir I, Arnar DO, Helgason H, Gudbjartsson T, Gudbjartsson DF, Thorsteinsdottir U, Holm H, Stefansson K. A rare missense mutation in MYH6 associates with non-syndromic coarctation of the aorta. Eur Heart J 2019; 39:3243-3249. [PMID: 29590334 PMCID: PMC6127890 DOI: 10.1093/eurheartj/ehy142] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/02/2018] [Indexed: 12/13/2022] Open
Abstract
Aims Coarctation of the aorta (CoA) accounts for 4–8% of congenital heart defects (CHDs) and confers substantial morbidity despite treatment. It is increasingly recognized as a highly heritable condition. The aim of the study was to search for sequence variants that affect the risk of CoA. Methods and results We performed a genome-wide association study of CoA among Icelanders (120 cases and 355 166 controls) based on imputed variants identified through whole-genome sequencing. We found association with a rare (frequency = 0.34%) missense mutation p.Arg721Trp in MYH6 (odds ratio = 44.2, P = 5.0 × 10−22), encoding the alpha-heavy chain subunit of cardiac myosin, an essential sarcomere protein. Approximately 20% of individuals with CoA in Iceland carry this mutation. We show that p.Arg721Trp also associates with other CHDs, in particular bicuspid aortic valve. We have previously reported broad effects of p.Arg721Trp on cardiac electrical function and strong association with sick sinus syndrome and atrial fibrillation. Conclusion Through a population approach, we found that a rare missense mutation p.Arg721Trp in the sarcomere gene MYH6 has a strong effect on the risk of CoA and explains a substantial fraction of the Icelanders with CoA. This is the first mutation associated with non-familial or sporadic form of CoA at a population level. The p.Arg721Trp in MYH6 causes a cardiac syndrome with highly variable expressivity and emphasizes the importance of sarcomere integrity for cardiac development and function.
Collapse
Affiliation(s)
| | | | | | - Patrick Sulem
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik, Iceland
| | | | | | | | | | - Ragnar Danielsen
- Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik, Iceland
| | - Emil L Sigurdsson
- Department of Family Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik, Iceland.,Department of Development, Primary Health Care of the Capital Area, Alfabakki 16, Reykjavik, Iceland
| | - Berglind Adalsteinsdottir
- Department of Cardiology, Haukeland University Hospital, Jonas Lies vei 83, Bergen, Norway.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik, Iceland
| | - Sverrir I Gunnarsson
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, 600 Highland Ave, Madison, WI, USA
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik, Iceland.,Department of Immunology, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik, Iceland
| | - David O Arnar
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik, Iceland
| | - Hrodmar Helgason
- Children's Hospital, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik, Iceland
| | - Tomas Gudbjartsson
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik, Iceland.,Department of Cardiothoracic Surgery, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Hjardarhagi 4, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik, Iceland
| |
Collapse
|
44
|
Rare Mutations in AHDC1 in Patients with Obstructive Sleep Apnea. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5907361. [PMID: 31737670 PMCID: PMC6815587 DOI: 10.1155/2019/5907361] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/12/2019] [Accepted: 09/06/2019] [Indexed: 12/18/2022]
Abstract
Objectives Obstructive sleep apnea (OSA) is a common disorder influenced by genetic and environmental factors. Mutations of AT-hook DNA-binding motif containing 1 (AHDC1) gene have been implicated which could cause rare syndromes presenting OSA. This study aims to investigate some rare mutations of AHDC1 in Chinese Han individuals with OSA. Patients and Methods Three hundred and seventy-five patients with OSA and one hundred and nine control individuals underwent polysomnography. A targeted sequencing experiment was taken in 100 patients with moderate-to-severe OSA, and genotyping was taken in 157 moderate-to-severe OSA and 100 control individuals. The effect of mutations was validated by the luciferase reporter assay. Results One rare missense mutation (AHDC1: p.G1484D) and two mutations (c.-88C>T; c.-781C>G) in 5′-untranslated region (UTR) of AHDC1 were identified. The rare mutation (c.-781C>G) in 5′-UTR that was identified in several patients presenting more severe clinical manifestations affects expression of AHDC1. Conclusions. Our results revealed three rare mutations of AHDC1 in patients with OSA in Chinese Hanindividuals.
Collapse
|
45
|
Gharibeh L, Komati H, Bossé Y, Boodhwani M, Heydarpour M, Fortier M, Hassanzadeh R, Ngu J, Mathieu P, Body S, Nemer M. GATA6 Regulates Aortic Valve Remodeling, and Its Haploinsufficiency Leads to Right-Left Type Bicuspid Aortic Valve. Circulation 2019; 138:1025-1038. [PMID: 29567669 DOI: 10.1161/circulationaha.117.029506] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Bicuspid aortic valve (BAV), the most common congenital heart defect affecting 1% to 2% of the population, is a major risk factor for premature aortic valve disease and accounts for the majority of valve replacement. The genetic basis and mechanisms of BAV etiology and pathogenesis remain largely undefined. METHODS Cardiac structure and function was assessed in mice lacking a Gata6 allele. Human GATA6 gene variants were analyzed in 452 BAV cases from the BAV consortium and 1849 controls from the Framingham GWAS (Genome Wide Association Study). GATA6 expression was determined in mice and human tissues using quantitative real-time polymerase chain reaction and immunohistochemistry. Mechanistic studies were carried out in cultured cells. RESULTS Gata6 heterozygous mice have highly penetrant right-left (RL)-type BAV, the most frequent type in humans. GATA6 transcript levels are lower in human BAV compared with normal tricuspid valves. Mechanistically, Gata6 haploinsufficiency disrupts valve remodeling and extracellular matrix composition through dysregulation of important signaling molecules, including matrix metalloproteinase 9. Cell-specific inactivation of Gata6 reveals an essential role for GATA6 in secondary heart field myocytes because loss of 1 Gata6 allele from Isl- 1-positive cells-but not from endothelial or neural crest cells-recapitulates the phenotype of Gata6 heterozygous mice. CONCLUSIONS The data identify a new cellular and molecular mechanism underlying BAV. The availability of an animal model for the most frequent human BAV opens the way for the elucidation of BAV pathogenesis and the development of much needed therapies.
Collapse
Affiliation(s)
- Lara Gharibeh
- Department of Biochemistry, Microbiology, and Immunology, Molecular Genetics and Cardiac Regeneration Laboratory, University of Ottawa, Ontario, Canada (L.G., H.K., R.H., M.T., M.N.)
| | - Hiba Komati
- Department of Biochemistry, Microbiology, and Immunology, Molecular Genetics and Cardiac Regeneration Laboratory, University of Ottawa, Ontario, Canada (L.G., H.K., R.H., M.T., M.N.)
| | - Yohan Bossé
- Department of Molecular Medicine, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Canada (Y.B., P.M.)
| | - Munir Boodhwani
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ontario, Canada (M.B., J.N.)
| | - Mahyar Heydarpour
- Department of Biochemistry, Microbiology, and Immunology, Molecular Genetics and Cardiac Regeneration Laboratory, University of Ottawa, Ontario, Canada (L.G., H.K., R.H., M.T., M.N.)
| | | | - Romina Hassanzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.H., S.B.)
| | - Janet Ngu
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ontario, Canada (M.B., J.N.)
| | - Patrick Mathieu
- Department of Molecular Medicine, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Canada (Y.B., P.M.)
| | - Simon Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.H., S.B.)
| | - Mona Nemer
- Department of Biochemistry, Microbiology, and Immunology, Molecular Genetics and Cardiac Regeneration Laboratory, University of Ottawa, Ontario, Canada (L.G., H.K., R.H., M.T., M.N.)
| | | |
Collapse
|
46
|
Zhang X, Yamashita S, Hara K, Doki T, Tawara N, Ikeda T, Misumi Y, Zhang Z, Matsuo Y, Nagai M, Kurashige T, Maruyama H, Ando Y. A mutantMATR3mouse model to explain multisystem proteinopathy. J Pathol 2019; 249:182-192. [DOI: 10.1002/path.5289] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/08/2019] [Accepted: 04/28/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Xiao Zhang
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Satoshi Yamashita
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Kentaro Hara
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Tsukasa Doki
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Nozomu Tawara
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Tokunori Ikeda
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
- Department of Clinical InvestigationKumamoto University Hospital Kumamoto Japan
| | - Yohei Misumi
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Ziwei Zhang
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Yoshimasa Matsuo
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| | - Makiko Nagai
- Department of NeurologyKitasato University School of Medicine Sagamihara Japan
| | - Takashi Kurashige
- Department of NeurologyNational Hospital Organization Kure Medical Centre Kure Hiroshima Japan
- Department of Clinical Neuroscience and TherapeuticsHiroshima University Graduate School of Biomedical and Health Sciences Hiroshima Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and TherapeuticsHiroshima University Graduate School of Biomedical and Health Sciences Hiroshima Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical SciencesKumamoto University Kumamoto Japan
| |
Collapse
|
47
|
Díaz-Ordoñez L, Ramirez-Montaño D, Candelo E, Cruz S, Pachajoa H. Syndromic Intellectual Disability Caused by a Novel Truncating Variant in AHDC1: A Case Report. IRANIAN JOURNAL OF MEDICAL SCIENCES 2019; 44:257-261. [PMID: 31182893 PMCID: PMC6525729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mutations in the AHDC1 gene are associated with the Xia-Gibbs syndrome (XGS), a sporadic genetic disorder characterised by developmental delay, intellectual disability, hypotonia, obstructive sleep apnoea, dysmorphic facial features, and cerebral malformations with plagiocephaly. Here we report the case of a 13-year-old Colombian female patient with a history of developmental delay, speech delay, sleep disturbances, and dysmorphic craniofacial features. The whole exome sequencing (WES) test revealed a novel de novo heterozygous frameshift mutation in AHDC1. The present case report describes the second case of mutations in AHDC1 in a Latin American patient. A literature review showed that the clinical features were similar in all reported patients. The WES test enabled the identification of the causality of this disorder characterised by high clinical and genetic heterogeneity.
Collapse
Affiliation(s)
- Lorena Díaz-Ordoñez
- Center for Research on Congenital Anomalies and Rare Diseases (CIACER), Universidad Icesi, Cali, Colombia;
| | - Diana Ramirez-Montaño
- Center for Research on Congenital Anomalies and Rare Diseases (CIACER), Universidad Icesi, Cali, Colombia;
| | - Estephania Candelo
- Center for Research on Congenital Anomalies and Rare Diseases (CIACER), Universidad Icesi, Cali, Colombia;
| | - Santiago Cruz
- Department of Genetics, Fundación Valle del Lili, Cali, Colombia
| | - Harry Pachajoa
- Center for Research on Congenital Anomalies and Rare Diseases (CIACER), Universidad Icesi, Cali, Colombia;
,Department of Genetics, Fundación Valle del Lili, Cali, Colombia
| |
Collapse
|
48
|
Fritz AJ, Sehgal N, Pliss A, Xu J, Berezney R. Chromosome territories and the global regulation of the genome. Genes Chromosomes Cancer 2019; 58:407-426. [PMID: 30664301 DOI: 10.1002/gcc.22732] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 12/29/2022] Open
Abstract
Spatial positioning is a fundamental principle governing nuclear processes. Chromatin is organized as a hierarchy from nucleosomes to Mbp chromatin domains (CD) or topologically associating domains (TADs) to higher level compartments culminating in chromosome territories (CT). Microscopic and sequencing techniques have substantiated chromatin organization as a critical factor regulating gene expression. For example, enhancers loop back to interact with their target genes almost exclusively within TADs, distally located coregulated genes reposition into common transcription factories upon activation, and Mbp CDs exhibit dynamic motion and configurational changes in vivo. A longstanding question in the nucleus field is whether an interactive nuclear matrix provides a direct link between structure and function. The findings of nonrandom radial positioning of CT within the nucleus suggest the possibility of preferential interaction patterns among populations of CT. Sequential labeling up to 10 CT followed by application of computer imaging and geometric graph mining algorithms revealed cell-type specific interchromosomal networks (ICN) of CT that are altered during the cell cycle, differentiation, and cancer progression. It is proposed that the ICN correlate with the global level of genome regulation. These approaches also demonstrated that the large scale 3-D topology of CT is specific for each CT. The cell-type specific proximity of certain chromosomal regions in normal cells may explain the propensity of distinct translocations in cancer subtypes. Understanding how genes are dysregulated upon disruption of the normal "wiring" of the nucleus by translocations, deletions, and amplifications that are hallmarks of cancer, should enable more targeted therapeutic strategies.
Collapse
Affiliation(s)
- Andrew J Fritz
- Department of Biochemistry and University of Vermont Cancer Center, The University of Vermont Larner College of Medicine, Burlington, Vermont
| | - Nitasha Sehgal
- Department of Biological Sciences, University at Buffalo, Buffalo, New York
| | - Artem Pliss
- Institute for Lasers, Photonics and Biophotonics and the Department of Chemistry, University at Buffalo, Buffalo, New York
| | - Jinhui Xu
- Department of Computer Science and Engineering, University at Buffalo, Buffalo, New York
| | - Ronald Berezney
- Department of Biological Sciences, University at Buffalo, Buffalo, New York
| |
Collapse
|
49
|
Manno G, Bentivegna R, Morreale P, Nobile D, Santangelo A, Novo S, Novo G. Chronic inflammation: A key role in degeneration of bicuspid aortic valve. J Mol Cell Cardiol 2019; 130:59-64. [PMID: 30885747 DOI: 10.1016/j.yjmcc.2019.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/25/2019] [Accepted: 03/14/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Bicuspid aortic valve (BAV) is the most common congenital valvular heart defect resulting from abnormal aortic cusp formation during heart development, where two of the three normal and equal sized cusps fuse into a single large cusp resulting in a two cusps aortic valve. Over the past years, much interest has been given in understanding the pathogenesis of BAV and its complications. In this review, we focused on the role of inflammation, involved in the degeneration of BAV and the development of its complications. ROLE OF INFLAMMATION From a pathophysiological point of view, BAV may rapidly progress into aortic stenosis (AS) and is related to aortopathy. Several histopathologic studies have demonstrated that the development and progression of alterations in bicuspid aortic valve are related to an active process that includes: oxidative stress, shear stress, endothelial dysfunction, disorganized tissue architecture, inflammatory cells and cytokines. These factors are closely related one to each other, constituting the basis of the structural and functional alterations of the BAV. CONCLUSION Chronic inflammation plays a key role in the degeneration of BAV. Severe aortic stenosis in bicuspid aortic valves is associated with a more aggressive inflammatory process, increased inflammatory cells infiltration and neovascularization when compared to tricuspid AS. These findings might help to explain the more frequent onset and rapid progression of AS and the heavy aortic valve calcification seen in patients with BAV.
Collapse
Affiliation(s)
- G Manno
- Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (PROMISE) "G. D'Alessandro", Italy; Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy.
| | - R Bentivegna
- Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (PROMISE) "G. D'Alessandro", Italy; Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - P Morreale
- Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (PROMISE) "G. D'Alessandro", Italy; Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - D Nobile
- Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (PROMISE) "G. D'Alessandro", Italy; Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - A Santangelo
- Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (PROMISE) "G. D'Alessandro", Italy; Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - S Novo
- Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (PROMISE) "G. D'Alessandro", Italy; Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - G Novo
- Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (PROMISE) "G. D'Alessandro", Italy; Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy.
| |
Collapse
|
50
|
Moloney C, Rayaprolu S, Howard J, Fromholt S, Brown H, Collins M, Cabrera M, Duffy C, Siemienski Z, Miller D, Borchelt DR, Lewis J. Analysis of spinal and muscle pathology in transgenic mice overexpressing wild-type and ALS-linked mutant MATR3. Acta Neuropathol Commun 2018; 6:137. [PMID: 30563574 PMCID: PMC6299607 DOI: 10.1186/s40478-018-0631-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/03/2018] [Indexed: 12/12/2022] Open
Abstract
Mutations in MATR3 have been associated with amyotrophic lateral sclerosis (ALS) as well as a form of distal myopathy termed vocal cord pharyngeal distal myopathy (VCPDM). To begin to understand how mutations in MATR3 may cause disease, here we provide initial characterization of transgenic (Tg) mice expressing human wild-type (WT) MATR3 (MATR3WT) and ALS-mutant F115C MATR3 (MATR3F115C) proteins under the control of the mouse prion promoter (MoPrP). For each construct, we established multiple independent lines of mice that stably transmitted the transgene. Unexpectedly, for all stably-transmitting lines examined, MATR3 transgenic mRNA expression was more robust in muscle, with minimal expression in spinal cord. The levels of transgenic mRNA in muscle did not differ between mice from our lead MATR3F115C line and lead MATR3WT line, but mice from the lead MATR3F115C line had significantly higher levels of MATR3 protein in muscle over the lead MATR3WT line. Mice from the three independent, established lines of MATR3F115C mice developed weakness in both fore- and hind-limbs as early as < 1 months of age; whereas, MATR3WT mice aged to > 20 months were not overtly distinguishable from non-transgenic (NT) littermates based on basic motor phenotype. Muscle of both MATR3WT and MATR3F115C mice showed vacuoles by 2 months of age which worsened by ~ 10 months, but vacuolation was noticeably more severe in MATR3F115C mice. Overall, our results indicate that increasing the levels of MATR3 in muscle can cause pathologic changes associated with myopathy, with MATR3F115C expression causing overt muscle atrophy and a profound motor phenotype. The findings suggest that analysis of muscle pathology in individuals harboring ALS-linked MATR3 mutations should be routinely considered.
Collapse
Affiliation(s)
- Christina Moloney
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Sruti Rayaprolu
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - John Howard
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Susan Fromholt
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Hilda Brown
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Matt Collins
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Mariela Cabrera
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Colin Duffy
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Zoe Siemienski
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Dave Miller
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - David R Borchelt
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA.
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA.
- McKnight Brain Institute, Department of Neuroscience, University of Florida, Gainesville, Florida, USA.
| | - Jada Lewis
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA.
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA.
- McKnight Brain Institute, Department of Neuroscience, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|