1
|
Rao VG, Subramanianbalachandar VA, Magaj MM, Redemann S, Kulkarni SS. Mechanisms of cilia regeneration in Xenopus multiciliated epithelium in vivo. EMBO Rep 2025; 26:2192-2220. [PMID: 40087471 PMCID: PMC12019409 DOI: 10.1038/s44319-025-00414-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 03/17/2025] Open
Abstract
Cilia regeneration is a physiological event, and while studied extensively in unicellular organisms, it remains poorly understood in vertebrates. In this study, using Xenopus multiciliated cells (MCCs), we demonstrate that, unlike unicellular organisms, deciliation removes the transition zone (TZ) and the ciliary axoneme. While MCCs immediately begin regenerating the axoneme, surprisingly, the TZ assembly is delayed. However, ciliary tip proteins, Sentan and Clamp, localize to regenerating cilia without delay. Using cycloheximide (CHX) to block protein synthesis, we show that the TZ protein B9d1 is not present in the cilia precursor pool and requires new transcription/translation, providing insights into the delayed repair of TZ. Moreover, MCCs in CHX treatment assemble fewer but near wild-type length cilia by gradually concentrating ciliogenesis proteins like IFTs at a few basal bodies. Using mathematical modeling, we show that cilia length, compared to cilia number, has a larger influence on the force generated by MCCs. Our results question the requirement of TZ in motile cilia assembly and provide insights into the fundamental question of how cells determine organelle size and number.
Collapse
Affiliation(s)
- Venkatramanan G Rao
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22903, USA
| | | | - Magdalena M Magaj
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22903, USA
- Center for Membrane & Cell Physiology, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Stefanie Redemann
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22903, USA
- Center for Membrane & Cell Physiology, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Saurabh S Kulkarni
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22903, USA.
- Department of Biology, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
2
|
Yang X, Wang Q, Li T, Zhou Y, Gao J, Ma W, Zhao N, Liu X, Ai Z, Cheng SY, Gu Y, Zhao B, Yue S, Hu Z. A splicing variant in EFCAB7 hinders ciliary transport and disrupts cardiac development. J Biol Chem 2025; 301:108249. [PMID: 39894222 PMCID: PMC11889548 DOI: 10.1016/j.jbc.2025.108249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
The Tetralogy of Fallot (TOF), the most prevalent form of cyanotic congenital heart disease, stems from abnormal development of the outflow tract during embryogenesis. Despite the crucial role played by primary cilia in heart development, there is currently insufficient evidence to establish a causal relationship between defects in genes related to primary cilia and non-syndromic TOF. Here, we performed Sanger sequencing on 131 Chinese patients diagnosed with TOF and identified a splicing variant (c.683-1G > C) in the EFCAB7 gene. This splicing variant triggered exon skipping, leading to the production of a non-functional protein both in vitro and in vivo. Mice carrying this variant exhibited abnormal cardiac development, impaired ciliogenesis, disrupted Hedgehog signaling, and hindered Shh/Gli pathway activity. Through the integration of CUT&Tag data on Glis and bulk RNA-seq profiles of embryonic hearts at E10.5, we found that transcriptional downregulation of Gli target genes, including Myh6, Zfpm1, and Nkx2-5, is a consequence of Shh signaling inhibition. Our findings implicate EFCAB7 as a potential causative gene for TOF, underscoring the indispensable function of primary cilia in the intricate process of cardiac septation during heart development.
Collapse
Affiliation(s)
- Xin Yang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Medical Genetics, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Qiuye Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tianyuan Li
- Department of Medical Genetics, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Yan Zhou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jimiao Gao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wanting Ma
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Na Zhao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinyue Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zihe Ai
- Department of Medical Genetics, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Steven Y Cheng
- Department of Medical Genetics, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Yayun Gu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bijun Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Shen Yue
- Department of Medical Genetics, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China; School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Ansari SS, Dillard ME, Ghonim M, Zhang Y, Stewart DP, Canac R, Moskowitz IP, Wright WC, Daly CA, Pruett-Miller SM, Steinberg J, Wang YD, Chen T, Thomas PG, Bridges JP, Ogden SK. Receptor Allostery Promotes Context-Dependent Sonic Hedgehog Signaling During Embryonic Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635336. [PMID: 39975106 PMCID: PMC11838287 DOI: 10.1101/2025.01.28.635336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Sonic Hedgehog (SHH) signaling functions in temporal- and context-dependent manners to pattern diverse tissues during embryogenesis. The signal transducer Smoothened (SMO) is activated by sterols, oxysterols, and arachidonic acid (AA) through binding pockets in its extracellular cysteine-rich domain (CRD) and 7-transmembrane (7TM) bundle. In vitro analyses suggest SMO signaling is allosterically enhanced by combinatorial ligand binding to these pockets but in vivo evidence of SMO allostery is lacking. Herein, we map an AA binding pocket at the top of the 7TM bundle and show that its disruption attenuates SHH and sterol-stimulated SMO induction. A knockin mouse model of compromised AA binding reveals that homozygous mutant mice are cyanotic, exhibit high perinatal lethality, and show congenital heart disease. Surviving mutants demonstrate pulmonary maldevelopment and fail to thrive. Neurodevelopment is unaltered in these mice, suggesting that context-dependent allosteric regulation of SMO signaling allows for precise tuning of pathway activity during cardiopulmonary development.
Collapse
Affiliation(s)
- Shariq S. Ansari
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Miriam E. Dillard
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Mohamed Ghonim
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yan Zhang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Daniel P. Stewart
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Robin Canac
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | - Ivan P. Moskowitz
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | - William C. Wright
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Christina A. Daly
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Shondra M. Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jeffrey Steinberg
- Center for In Vivo Imaging and Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Paul G. Thomas
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - James P. Bridges
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, 80206, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Stacey K. Ogden
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
4
|
Kostyanovskaya E, Lasser MC, Wang B, Schmidt J, Bader E, Buteo C, Arbelaez J, Sindledecker AR, McCluskey KE, Castillo O, Wang S, Dea J, Helde KA, Graglia JM, Brimble E, Kastner DB, Ehrlich AT, State MW, Willsey AJ, Willsey HR. Convergence of autism proteins at the cilium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.05.626924. [PMID: 39677731 PMCID: PMC11643032 DOI: 10.1101/2024.12.05.626924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Hundreds of high-confidence autism genes have been identified, yet the relevant etiological mechanisms remain unclear. Gene ontology analyses have repeatedly identified enrichment of proteins with annotated functions in gene expression regulation and neuronal communication. However, proteins are often pleiotropic and these annotations are inherently incomplete. Our recent autism functional genetics work has suggested that these genes may share a common mechanism at the cilium, a membrane-bound organelle critical for neurogenesis, brain patterning, and neuronal activity-all processes strongly implicated in autism. Moreover, autism commonly co-occurs with conditions that are known to involve ciliary-related pathologies, including congenital heart disease, hydrocephalus, and blindness. However, the role of autism genes at the cilium has not been systematically investigated. Here we demonstrate that autism proteins spanning disparate functional annotations converge in expression, localization, and function at cilia, and that patients with pathogenic variants in these genes have cilia-related co-occurring conditions and biomarkers of disrupted ciliary function. This degree of convergence among genes spanning diverse functional annotations strongly suggests that cilia are relevant to autism, as well as to commonly co-occurring conditions, and that this organelle should be explored further for therapeutic potential.
Collapse
Affiliation(s)
- Elina Kostyanovskaya
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Micaela C. Lasser
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Belinda Wang
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - James Schmidt
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Ethel Bader
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Chad Buteo
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Juan Arbelaez
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Aria Rani Sindledecker
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Kate E. McCluskey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Octavio Castillo
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Sheng Wang
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Jeanselle Dea
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | | | | | | | - David B. Kastner
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Aliza T. Ehrlich
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Matthew W. State
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - A. Jeremy Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub – San Francisco, San Francisco, CA
| |
Collapse
|
5
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Sarić N, Atak Z, Sade CF, Reddy N, Bell G, Tolete C, Rajtboriraks MT, Hashimoto-Torii K, Jevtović-Todorović V, Haydar TF, Ishibashi N. Ciliopathy interacts with neonatal anesthesia to cause non-apoptotic caspase-mediated motor deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.624302. [PMID: 39651246 PMCID: PMC11623571 DOI: 10.1101/2024.11.27.624302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Increasing evidence suggests that anesthesia may induce developmental neurotoxicity, yet the influence of genetic predispositions associated with congenital anomalies on this toxicity remains largely unknown. Children with congenital heart disease often exhibit mutations in cilia-related genes and ciliary dysfunction, requiring sedation for their catheter or surgical interventions during the neonatal period. Here we demonstrate that briefly exposing ciliopathic neonatal mice to ketamine causes motor skill impairments, which are associated with a baseline deficit in neocortical layer V neuron apical spine density and their altered dynamics during motor learning.. These neuromorphological changes were linked to augmented non-apoptotic neuronal caspase activation. Neonatal caspase suppression rescued the spine density and motor deficits, confirming the requirement for sublethal caspase signaling in appropriate spine formation and motor learning. Our findings suggest that ciliopathy interacts with ketamine to induce motor impairments, which is reversible through caspase inhibition. Furthermore, they underscore the potential for ketamine- induced sublethal caspase responses in shaping neurodevelopmental outcomes.
Collapse
|
7
|
Barbelanne M, Lu Y, Kumar K, Zhang X, Li C, Park K, Warner A, Xu XZS, Shaham S, Leroux MR. C. elegans PPEF-type phosphatase (Retinal degeneration C ortholog) functions in diverse classes of cilia to regulate nematode behaviors. Sci Rep 2024; 14:28347. [PMID: 39550471 PMCID: PMC11569196 DOI: 10.1038/s41598-024-79057-z] [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: 04/25/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024] Open
Abstract
Primary (non-motile) cilia represent structurally and functionally diverse organelles whose roles as specialized cellular antenna are central to animal cell signaling pathways, sensory physiology and development. An ever-growing number of ciliary proteins, including those found in vertebrate photoreceptors, have been uncovered and linked to human disorders termed ciliopathies. Here, we demonstrate that an evolutionarily-conserved PPEF-family serine-threonine phosphatase, not functionally linked to cilia in any organism but associated with rhabdomeric (non-ciliary) photoreceptor degeneration in the Drosophila rdgC (retinal degeneration C) mutant, is a bona fide ciliary protein in C. elegans. The nematode protein, PEF-1, depends on transition zone proteins, which make up a 'ciliary gate' in the proximal-most region of the cilium, for its compartmentalization within cilia. Animals lacking PEF-1 protein function display structural defects to several types of cilia, including potential degeneration of microtubules. They also exhibit anomalies to cilium-dependent behaviors, including impaired responses to chemical, temperature, light, and noxious CO2 stimuli. Lastly, we demonstrate that PEF-1 function depends on conserved myristoylation and palmitoylation signals. Collectively, our findings broaden the role of PPEF proteins to include cilia, and suggest that the poorly-characterized mammalian PPEF1 and PPEF2 orthologs may also have ciliary functions and thus represent ciliopathy candidates.
Collapse
Affiliation(s)
- Marine Barbelanne
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Yun Lu
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Keerthana Kumar
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Xinxing Zhang
- Life Sciences Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chunmei Li
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Kwangjin Park
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Adam Warner
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - X Z Shawn Xu
- Life Sciences Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Michel R Leroux
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
8
|
Gao H, Huang X, Chen W, Feng Z, Zhao Z, Li P, Tan C, Wang J, Zhuang Q, Gao Y, Min S, Yao Q, Qian M, Ma X, Wu F, Yan W, Sheng W, Huang G. Association of copy number variation in X chromosome-linked PNPLA4 with heterotaxy and congenital heart disease. Chin Med J (Engl) 2024; 137:1823-1834. [PMID: 38973237 PMCID: PMC12077557 DOI: 10.1097/cm9.0000000000003192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Heterotaxy (HTX) is a thoracoabdominal organ anomaly syndrome and commonly accompanied by congenital heart disease (CHD). The aim of this study was to analyze rare copy number variations (CNVs) in a HTX/CHD cohort and to examine the potential mechanisms contributing to HTX/CHD. METHODS Chromosome microarray analysis was used to identify rare CNVs in a cohort of 120 unrelated HTX/CHD patients, and available samples from parents were used to confirm the inheritance pattern. Potential candidate genes in CNVs region were prioritized via the DECIPHER database, and PNPLA4 was identified as the leading candidate gene. To validate, we generated PNPLA4 -overexpressing human induced pluripotent stem cell lines as well as pnpla4 -overexpressing zebrafish model, followed by a series of transcriptomic, biochemical and cellular analyses. RESULTS Seventeen rare CNVs were identified in 15 of the 120 HTX/CHD patients (12.5%). Xp22.31 duplication was one of the inherited CNVs identified in this HTX/CHD cohort, and PNPLA4 in the Xp22.31 was a candidate gene associated with HTX/CHD. PNPLA4 is expressed in the lateral plate mesoderm, which is known to be critical for left/right embryonic patterning as well as cardiomyocyte differentiation, and in the neural crest cell lineage. Through a series of in vivo and in vitro analyses at the molecular and cellular levels, we revealed that the biological function of PNPLA4 is importantly involved in the primary cilia formation and function via its regulation of energy metabolism and mitochondria-mediated ATP production. CONCLUSIONS Our findings demonstrated a significant association between CNVs and HTX/CHD. Our data strongly suggested that an increased genetic dose of PNPLA4 due to Xp22.31 duplication is a disease-causing risk factor for HTX/CHD.
Collapse
Affiliation(s)
- Han Gao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Xianghui Huang
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
| | - Weicheng Chen
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Zhiyu Feng
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Zhengshan Zhao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Ping Li
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Chaozhong Tan
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Jinxin Wang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Quannan Zhuang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Yuan Gao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Shaojie Min
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Qinyu Yao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Maoxiang Qian
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Xiaojing Ma
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Feizhen Wu
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Weili Yan
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Wei Sheng
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Guoying Huang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| |
Collapse
|
9
|
Heywood WE, Searle J, Collis R, Doykov I, Ashworth M, Sebire N, Bamber A, Gautel M, Eaton S, Coats CJ, Elliott PM, Mills K. A Proof of Principle 2D Spatial Proteome Mapping Analysis Reveals Distinct Regional Differences in the Cardiac Proteome. Life (Basel) 2024; 14:970. [PMID: 39202712 PMCID: PMC11355120 DOI: 10.3390/life14080970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Proteomics studies often explore phenotypic differences between whole organs and systems. Within the heart, more subtle variation exists. To date, differences in the underlying proteome are only described between whole cardiac chambers. This study, using the bovine heart as a model, investigates inter-regional differences and assesses the feasibility of measuring detailed, cross-tissue variance in the cardiac proteome. Using a bovine heart, we created a two-dimensional section through a plane going through two chambers. This plane was further sectioned into 4 × 4 mm cubes and analysed using label-free proteomics. We identified three distinct proteomes. When mapped to the extracted sections, the proteomes corresponded largely to the outer wall of the right ventricle and secondly to the outer wall of the left ventricle, right atrial appendage, tricuspid and mitral valves, modulator band, and parts of the left atrium. The third separate proteome corresponded to the inner walls of the left and right ventricles, septum, and left atrial appendage. Differential protein abundancies indicated differences in energy metabolism between regions. Data analyses of the mitochondrial proteins revealed a variable pattern of abundances of complexes I-V between the proteomes, indicating differences in the bioenergetics of the different cardiac sub-proteomes. Mapping of disease-associated proteins interestingly showed desmoglein-2, for which defects in this protein are known to cause Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy, which was present predominantly in the outer wall of the left ventricle. This study highlights that organs can have variable proteomes that do not necessarily correspond to anatomical features.
Collapse
Affiliation(s)
- Wendy E. Heywood
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Jon Searle
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Richard Collis
- Institute of Cardiovascular Science, University College London, Gower Street, London WC1E 6BT, UK; (R.C.); (P.M.E.)
| | - Ivan Doykov
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Michael Ashworth
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK (N.S.)
| | - Neil Sebire
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK (N.S.)
| | - Andrew Bamber
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK (N.S.)
| | - Mathias Gautel
- Randall Division of Cell and Molecular Biophysics, Muscle Signalling Section, King’s College, London WC2E 2LS, UK
| | - Simon Eaton
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Caroline J. Coats
- Institute of Cardiovascular Science, University College London, Gower Street, London WC1E 6BT, UK; (R.C.); (P.M.E.)
| | - Perry M. Elliott
- Institute of Cardiovascular Science, University College London, Gower Street, London WC1E 6BT, UK; (R.C.); (P.M.E.)
- Barts Heart Centre, and the Inherited Cardiovascular Diseases Unit, St Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
| | - Kevin Mills
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| |
Collapse
|
10
|
Teerikorpi N, Lasser MC, Wang S, Kostyanovskaya E, Bader E, Sun N, Dea J, Nowakowski TJ, Willsey AJ, Willsey HR. Ciliary biology intersects autism and congenital heart disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.602578. [PMID: 39131273 PMCID: PMC11312554 DOI: 10.1101/2024.07.30.602578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Autism spectrum disorder (ASD) commonly co-occurs with congenital heart disease (CHD), but the molecular mechanisms underlying this comorbidity remain unknown. Given that children with CHD come to clinical attention by the newborn period, understanding which CHD variants carry ASD risk could provide an opportunity to identify and treat individuals at high risk for developing ASD far before the typical age of diagnosis. Therefore, it is critical to delineate the subset of CHD genes most likely to increase the risk of ASD. However, to date there is relatively limited overlap between high confidence ASD and CHD genes, suggesting that alternative strategies for prioritizing CHD genes are necessary. Recent studies have shown that ASD gene perturbations commonly dysregulate neural progenitor cell (NPC) biology. Thus, we hypothesized that CHD genes that disrupt neurogenesis are more likely to carry risk for ASD. Hence, we performed an in vitro pooled CRISPR interference (CRISPRi) screen to identify CHD genes that disrupt NPC biology similarly to ASD genes. Overall, we identified 45 CHD genes that strongly impact proliferation and/or survival of NPCs. Moreover, we observed that a cluster of physically interacting ASD and CHD genes are enriched for ciliary biology. Studying seven of these genes with evidence of shared risk (CEP290, CHD4, KMT2E, NSD1, OFD1, RFX3, TAOK1), we observe that perturbation significantly impacts primary cilia formation in vitro. While in vivo investigation of TAOK1 reveals a previously unappreciated role for the gene in motile cilia formation and heart development, supporting its prediction as a CHD risk gene. Together, our findings highlight a set of CHD risk genes that may carry risk for ASD and underscore the role of cilia in shared ASD and CHD biology.
Collapse
Affiliation(s)
- Nia Teerikorpi
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Micaela C. Lasser
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sheng Wang
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elina Kostyanovskaya
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ethel Bader
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nawei Sun
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeanselle Dea
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tomasz J. Nowakowski
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco CA 94158, USA
- Department of Anatomy, University of California, San Francisco, San Francisco CA 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research University of California, San Francisco, San Francisco CA 94158, USA
| | - A. Jeremy Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub – San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
11
|
Yuan Z, Zhu X, Xie X, Wang C, Gu H, Yang J, Fan L, Xiang R, Yang Y, Tan Z. Identification of a novel MYO1D variant associated with laterality defects, congenital heart diseases, and sperm defects in humans. Front Med 2024; 18:558-564. [PMID: 38684630 DOI: 10.1007/s11684-023-1042-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/15/2023] [Indexed: 05/02/2024]
Abstract
The establishment of left-right asymmetry is a fundamental process in animal development. Interference with this process leads to a range of disorders collectively known as laterality defects, which manifest as abnormal arrangements of visceral organs. Among patients with laterality defects, congenital heart diseases (CHD) are prevalent. Through multiple model organisms, extant research has established that myosin-Id (MYO1D) deficiency causes laterality defects. This study investigated over a hundred cases and identified a novel biallelic variant of MYO1D (NM_015194: c.1531G>A; p.D511N) in a consanguineous family with complex CHD and laterality defects. Further examination of the proband revealed asthenoteratozoospermia and shortened sperm. Afterward, the effects of the D511N variant and another known MYO1D variant (NM_015194: c.2293C>T; p.P765S) were assessed. The assessment showed that both enhance the interaction with β-actin and SPAG6. Overall, this study revealed the genetic heterogeneity of this rare disease and found that MYO1D variants are correlated with laterality defects and CHD in humans. Furthermore, this research established a connection between sperm defects and MYO1D variants. It offers guidance for exploring infertility and reproductive health concerns. The findings provide a critical basis for advancing personalized medicine and genetic counseling.
Collapse
Affiliation(s)
- Zhuangzhuang Yuan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Xin Zhu
- Department of Gynecology and Obstetrics, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Xiaohui Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Chenyu Wang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Heng Gu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Junlin Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Liangliang Fan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Zhiping Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
12
|
Rao VG, Subramanianbalachandar V, Magaj MM, Redemann S, Kulkarni SS. Mechanisms of cilia regeneration in Xenopus multiciliated epithelium in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.14.544972. [PMID: 37398226 PMCID: PMC10312767 DOI: 10.1101/2023.06.14.544972] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Cilia regeneration is a physiological event, and while studied extensively in unicellular organisms, it remains poorly understood in vertebrates. In this study, using Xenopus multiciliated cells (MCCs) as a model, we demonstrate that, unlike unicellular organisms, deciliation removes the transition zone (TZ) and the ciliary axoneme. While MCCs immediately begin the regeneration of the ciliary axoneme, surprisingly, the assembly of TZ is delayed. However, ciliary tip proteins, Sentan and Clamp, localize to regenerating cilia without delay. Using cycloheximide (CHX) to block new protein synthesis, we show that the TZ protein B9d1 is not a component of the cilia precursor pool and requires new transcription/translation, providing insights into the delayed repair of TZ. Moreover, MCCs in CHX treatment assemble fewer (∼ 10 vs. ∼150 in controls) but near wild-type length (ranging between 60 to 90%) cilia by gradually concentrating ciliogenesis proteins like IFTs at a select few basal bodies. Using mathematical modeling, we show that cilia length compared to cilia number influences the force generated by MCCs more. In summary, our results question the requirement of TZ in motile cilia assembly and provide insights into how cells determine organelle size and number.
Collapse
|
13
|
Verrillo G, Obeid AM, Genco A, Scrofani J, Orange F, Hanache S, Mignon J, Leyder T, Michaux C, Kempeneers C, Bricmont N, Herkenne S, Vernos I, Martin M, Mottet D. Non-canonical role for the BAF complex subunit DPF3 in mitosis and ciliogenesis. J Cell Sci 2024; 137:jcs261744. [PMID: 38661008 PMCID: PMC11166463 DOI: 10.1242/jcs.261744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/04/2024] [Indexed: 04/26/2024] Open
Abstract
DPF3, along with other subunits, is a well-known component of the BAF chromatin remodeling complex, which plays a key role in regulating chromatin remodeling activity and gene expression. Here, we elucidated a non-canonical localization and role for DPF3. We showed that DPF3 dynamically localizes to the centriolar satellites in interphase and to the centrosome, spindle midzone and bridging fiber area, and midbodies during mitosis. Loss of DPF3 causes kinetochore fiber instability, unstable kinetochore-microtubule attachment and defects in chromosome alignment, resulting in altered mitotic progression, cell death and genomic instability. In addition, we also demonstrated that DPF3 localizes to centriolar satellites at the base of primary cilia and is required for ciliogenesis by regulating axoneme extension. Taken together, these findings uncover a moonlighting dual function for DPF3 during mitosis and ciliogenesis.
Collapse
Affiliation(s)
- Giulia Verrillo
- University of Liege, GIGA – Research Institute, Molecular Analysis of Gene Expression (MAGE) Laboratory, B34, Avenue de l'Hôpital, B-4000 Liège, Belgium
| | - Anna Maria Obeid
- University of Liege, GIGA – Research Institute, Molecular Analysis of Gene Expression (MAGE) Laboratory, B34, Avenue de l'Hôpital, B-4000 Liège, Belgium
| | - Alexia Genco
- University of Liege, GIGA – Research Institute, Molecular Analysis of Gene Expression (MAGE) Laboratory, B34, Avenue de l'Hôpital, B-4000 Liège, Belgium
| | - Jacopo Scrofani
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain
| | - François Orange
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée (CCMA), 06100 Nice, France
| | - Sarah Hanache
- University of Liege, GIGA – Research Institute, Molecular Analysis of Gene Expression (MAGE) Laboratory, B34, Avenue de l'Hôpital, B-4000 Liège, Belgium
| | - Julien Mignon
- University of Namur, Laboratory of Physical Chemistry of Biomolecules, Unité de Chimie Physique Théorique et Structurale (UCPTS), Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Tanguy Leyder
- University of Namur, Laboratory of Physical Chemistry of Biomolecules, Unité de Chimie Physique Théorique et Structurale (UCPTS), Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Catherine Michaux
- University of Namur, Laboratory of Physical Chemistry of Biomolecules, Unité de Chimie Physique Théorique et Structurale (UCPTS), Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Céline Kempeneers
- University of Liege, Pneumology Laboratory, I3 Group, GIGA Research Center, B-4000 Liège, Belgium
- Division of Respirology, Department of Pediatrics, University Hospital Liège, B-4000 Liège, Belgium
| | - Noëmie Bricmont
- University of Liege, Pneumology Laboratory, I3 Group, GIGA Research Center, B-4000 Liège, Belgium
- Division of Respirology, Department of Pediatrics, University Hospital Liège, B-4000 Liège, Belgium
| | - Stephanie Herkenne
- University of Liege, GIGA-Cancer, Laboratory of Mitochondria and Cell Communication, B34, Avenue de l'Hôpital, B-4000 Liège, Belgium
| | - Isabelle Vernos
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona 08002, Spain
- ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain
| | - Maud Martin
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, B-6041 Gosselies, Belgium
| | - Denis Mottet
- University of Liege, GIGA – Research Institute, Molecular Analysis of Gene Expression (MAGE) Laboratory, B34, Avenue de l'Hôpital, B-4000 Liège, Belgium
| |
Collapse
|
14
|
Jin Y, Zhao M, Guo Q, Zhao W, Lei M, Zhang Y, Zhang Y, Shen Y, Lin K, Yang Z, Chu J, Sun H, Luo Z. Association study of FLT4 and HYDIN single nucleotide polymorphisms with atrial septal defect susceptibility in the Han Chinese population of Southwest China. Ital J Pediatr 2024; 50:62. [PMID: 38581027 PMCID: PMC10998412 DOI: 10.1186/s13052-024-01630-z] [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: 11/13/2023] [Accepted: 03/18/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Atrial septal defect (ASD) is a common form of congenital heart disease. Although several genes related to ASD have been found, the genetic factors of ASD remain unclear. This study aimed to evaluate the correlation between 10 candidate single nucleotide polymorphisms (SNPs) and sporadic atrial septal defects. METHODS Based on the results of 34 individual whole exome sequences, 10 candidate SNPs were selected. In total, 489 ASD samples and 420 normal samples were collected. The 10 SNPs in the case group and the control group were identified through Snapshot genotyping technology. The χ2-test and unconditional regression model were used to evaluate the relationship between ASD and each candidate SNP. Haploview software was used to perform linkage disequilibrium and haplotype analysis. RESULTS The χ2 results showed that the FLT4 rs383985 (P = 0.003, OR = 1.115-1.773), HYDIN rs7198975 (P = 0.04621, OR = 1.003-1.461), and HYDIN rs1774266 (P = 0.04621, OR = 1.003-1.461) alleles were significantly different between the control group and the case group (P < 0.05). Only the association with the FLT4 polymorphism was statistically significant after adjustment for multiple comparisons. CONCLUSION These findings suggest that a possible molecular pathogenesis associated with sporadic ASD is worth exploring in future studies.
Collapse
Affiliation(s)
- Ye Jin
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Miao Zhao
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Qiuzhe Guo
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Wanyu Zhao
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Min Lei
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Yifei Zhang
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Yunhan Zhang
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Yan Shen
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China
| | - Keqin Lin
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China
| | - Zhaoqing Yang
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China
| | - Jiayou Chu
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China
| | - Hao Sun
- The Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiao ling Road, 650118, Kunming, Yunnan, China.
| | - Zhiling Luo
- Yunnan Fuwai Cardiovascular Hospital, 528 Shahe Road, 650032, Kunming, Yunnan, China.
| |
Collapse
|
15
|
Accogli A, Shakya S, Yang T, Insinna C, Kim SY, Bell D, Butov KR, Severino M, Niceta M, Scala M, Lee HS, Yoo T, Stauffer J, Zhao H, Fiorillo C, Pedemonte M, Diana MC, Baldassari S, Zakharova V, Shcherbina A, Rodina Y, Fagerberg C, Roos LS, Wierzba J, Dobosz A, Gerard A, Potocki L, Rosenfeld JA, Lalani SR, Scott TM, Scott D, Azamian MS, Louie R, Moore HW, Champaigne NL, Hollingsworth G, Torella A, Nigro V, Ploski R, Salpietro V, Zara F, Pizzi S, Chillemi G, Ognibene M, Cooney E, Do J, Linnemann A, Larsen MJ, Specht S, Walters KJ, Choi HJ, Choi M, Tartaglia M, Youkharibache P, Chae JH, Capra V, Park SG, Westlake CJ. Variants in the WDR44 WD40-repeat domain cause a spectrum of ciliopathy by impairing ciliogenesis initiation. Nat Commun 2024; 15:365. [PMID: 38191484 PMCID: PMC10774338 DOI: 10.1038/s41467-023-44611-2] [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: 11/09/2022] [Accepted: 12/14/2023] [Indexed: 01/10/2024] Open
Abstract
WDR44 prevents ciliogenesis initiation by regulating RAB11-dependent vesicle trafficking. Here, we describe male patients with missense and nonsense variants within the WD40 repeats (WDR) of WDR44, an X-linked gene product, who display ciliopathy-related developmental phenotypes that we can model in zebrafish. The patient phenotypic spectrum includes developmental delay/intellectual disability, hypotonia, distinct craniofacial features and variable presence of brain, renal, cardiac and musculoskeletal abnormalities. We demonstrate that WDR44 variants associated with more severe disease impair ciliogenesis initiation and ciliary signaling. Because WDR44 negatively regulates ciliogenesis, it was surprising that pathogenic missense variants showed reduced abundance, which we link to misfolding of WDR autonomous repeats and degradation by the proteasome. We discover that disease severity correlates with increased RAB11 binding, which we propose drives ciliogenesis initiation dysregulation. Finally, we discover interdomain interactions between the WDR and NH2-terminal region that contains the RAB11 binding domain (RBD) and show patient variants disrupt this association. This study provides new insights into WDR44 WDR structure and characterizes a new syndrome that could result from impaired ciliogenesis.
Collapse
Affiliation(s)
- Andrea Accogli
- Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre (MUHC), Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Saurabh Shakya
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Taewoo Yang
- Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 08826, Seoul, Republic of Korea
| | - Christine Insinna
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Soo Yeon Kim
- Department of Genomic Medicine, Seoul National University Hospital, 03080, Seoul, Republic of Korea
| | - David Bell
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kirill R Butov
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | | | - Marcello Niceta
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | - Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Hyun Sik Lee
- School of Biological Sciences, Seoul National University, 08826, Seoul, Republic of Korea
| | - Taekyeong Yoo
- Department of Biomedical Sciences, Seoul National University College of Medicine, 03080, Seoul, Republic of Korea
| | - Jimmy Stauffer
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Huijie Zhao
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Chiara Fiorillo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Child Neuropsychiatry, IRCCS Istituto G.Gaslini, DINOGMI University of Genova, Largo Gaslini 5, Genoa, Italy
| | - Marina Pedemonte
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Maria C Diana
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Simona Baldassari
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Viktoria Zakharova
- National Medical Research Center for Endocrinology, Clinical data analysis department, Moscow, Russian Federation, Russia
| | - Anna Shcherbina
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
| | - Yulia Rodina
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
| | - Christina Fagerberg
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Laura Sønderberg Roos
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, København, Denmark
| | - Jolanta Wierzba
- Department of Pediatrics and Internal Medicine Nursing, Department of Rare Disorders, Medical University of Gdansk, Gdansk, Poland
| | - Artur Dobosz
- Department of Medical Genetics, Faculty of Medicine, Jagiellonian University Medical College, 30-663, Krakow, Poland
| | - Amanda Gerard
- Texas Children's Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lorraine Potocki
- Texas Children's Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Baylor Genetics Laboratories, Houston, TX, USA
| | - Seema R Lalani
- Texas Children's Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Tiana M Scott
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Daryl Scott
- Baylor Genetics Laboratories, Houston, TX, USA
| | | | | | | | | | | | - Annalaura Torella
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vincenzo Nigro
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rafal Ploski
- Department of Medical Genetics, Medical University of Warsaw, Pawińskiego 3C, 02-106, Warsaw, Poland
| | - Vincenzo Salpietro
- Department of Neuromuscular Disorders, Queen Square Institute of Neurology, University. College London, London, WC1N 3BG, UK
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-food and Forest systems, DIBAF, University of Tuscia, Via S. Camillo de Lellis s.n.c, 01100, Viterbo, Italy
| | - Marzia Ognibene
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Erin Cooney
- Division of Medical Genetics and Metabolism, Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Jenny Do
- Division of Medical Genetics and Metabolism, Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Anders Linnemann
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Martin J Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Suzanne Specht
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Kylie J Walters
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Hee-Jung Choi
- School of Biological Sciences, Seoul National University, 08826, Seoul, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, 03080, Seoul, Republic of Korea
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | - Phillippe Youkharibache
- Cancer Science Data Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jong-Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, 03080, Seoul, Republic of Korea
| | - Valeria Capra
- Child Neuropsychiatry, IRCCS Istituto G.Gaslini, DINOGMI University of Genova, Largo Gaslini 5, Genoa, Italy
| | - Sung-Gyoo Park
- Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 08826, Seoul, Republic of Korea.
| | - Christopher J Westlake
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
16
|
Rochon ER, Xue J, Mohammed MS, Smith C, Hay-Schmidt A, DeMartino AW, Clark A, Xu Q, Lo CW, Tsang M, Tejero J, Gladwin MT, Corti P. Cytoglobin regulates NO-dependent cilia motility and organ laterality during development. Nat Commun 2023; 14:8333. [PMID: 38097556 PMCID: PMC10721929 DOI: 10.1038/s41467-023-43544-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023] Open
Abstract
Cytoglobin is a heme protein with unresolved physiological function. Genetic deletion of zebrafish cytoglobin (cygb2) causes developmental defects in left-right cardiac determination, which in humans is associated with defects in ciliary function and low airway epithelial nitric oxide production. Here we show that Cygb2 co-localizes with cilia and with the nitric oxide synthase Nos2b in the zebrafish Kupffer's vesicle, and that cilia structure and function are disrupted in cygb2 mutants. Abnormal ciliary function and organ laterality defects are phenocopied by depletion of nos2b and of gucy1a, the soluble guanylate cyclase homolog in fish. The defects are rescued by exposing cygb2 mutant embryos to a nitric oxide donor or a soluble guanylate cyclase stimulator, or with over-expression of nos2b. Cytoglobin knockout mice also show impaired airway epithelial cilia structure and reduced nitric oxide levels. Altogether, our data suggest that cytoglobin is a positive regulator of a signaling axis composed of nitric oxide synthase-soluble guanylate cyclase-cyclic GMP that is necessary for normal cilia motility and left-right patterning.
Collapse
Affiliation(s)
- Elizabeth R Rochon
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jianmin Xue
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Manush Sayd Mohammed
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Caroline Smith
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Anders Hay-Schmidt
- Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anthony W DeMartino
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Adam Clark
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Qinzi Xu
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Cecilia W Lo
- Department of Developmental Biology, Rangos Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15201, USA
| | - Michael Tsang
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Jesus Tejero
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, 15260, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Mark T Gladwin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Paola Corti
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
17
|
Kalbfuss N, Gönczy P. Towards understanding centriole elimination. Open Biol 2023; 13:230222. [PMID: 37963546 PMCID: PMC10645514 DOI: 10.1098/rsob.230222] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/14/2023] [Indexed: 11/16/2023] Open
Abstract
Centrioles are microtubule-based structures crucial for forming flagella, cilia and centrosomes. Through these roles, centrioles are critical notably for proper cell motility, signalling and division. Recent years have advanced significantly our understanding of the mechanisms governing centriole assembly and architecture. Although centrioles are typically very stable organelles, persisting over many cell cycles, they can also be eliminated in some cases. Here, we review instances of centriole elimination in a range of species and cell types. Moreover, we discuss potential mechanisms that enable the switch from a stable organelle to a vanishing one. Further work is expected to provide novel insights into centriole elimination mechanisms in health and disease, thereby also enabling scientists to readily manipulate organelle fate.
Collapse
Affiliation(s)
- Nils Kalbfuss
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
18
|
Huang M, Lyu C, Liu N, Nembhard WN, Witte JS, Hobbs CA, Li M. A gene-based association test of interactions for maternal-fetal genotypes identifies genes associated with nonsyndromic congenital heart defects. Genet Epidemiol 2023; 47:475-495. [PMID: 37341229 PMCID: PMC11781787 DOI: 10.1002/gepi.22533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/13/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023]
Abstract
The risk of congenital heart defects (CHDs) may be influenced by maternal genes, fetal genes, and their interactions. Existing methods commonly test the effects of maternal and fetal variants one-at-a-time and may have reduced statistical power to detect genetic variants with low minor allele frequencies. In this article, we propose a gene-based association test of interactions for maternal-fetal genotypes (GATI-MFG) using a case-mother and control-mother design. GATI-MFG can integrate the effects of multiple variants within a gene or genomic region and evaluate the joint effect of maternal and fetal genotypes while allowing for their interactions. In simulation studies, GATI-MFG had improved statistical power over alternative methods, such as the single-variant test and functional data analysis (FDA) under various disease scenarios. We further applied GATI-MFG to a two-phase genome-wide association study of CHDs for the testing of both common variants and rare variants using 947 CHD case mother-infant pairs and 1306 control mother-infant pairs from the National Birth Defects Prevention Study (NBDPS). After Bonferroni adjustment for 23,035 genes, two genes on chromosome 17, TMEM107 (p = 1.64e-06) and CTC1 (p = 2.0e-06), were identified for significant association with CHD in common variants analysis. Gene TMEM107 regulates ciliogenesis and ciliary protein composition and was found to be associated with heterotaxy. Gene CTC1 plays an essential role in protecting telomeres from degradation, which was suggested to be associated with cardiogenesis. Overall, GATI-MFG outperformed the single-variant test and FDA in the simulations, and the results of application to NBDPS samples are consistent with existing literature supporting the association of TMEM107 and CTC1 with CHDs.
Collapse
Affiliation(s)
- Manyan Huang
- Department of Epidemiology and Biostatistics, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Chen Lyu
- Department of Population Health, New York University Grossman School of Medicine, New York City, New York, USA
| | - Nianjun Liu
- Department of Epidemiology and Biostatistics, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Wendy N. Nembhard
- Department of Epidemiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - John S. Witte
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
- Department of Biomedical Data Sciences, Stanford University, Stanford, California, USA
| | - Charlotte A. Hobbs
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Ming Li
- Department of Epidemiology and Biostatistics, Indiana University Bloomington, Bloomington, Indiana, USA
| | | |
Collapse
|
19
|
Chen Z, Gao Y, Lu L, Li N, Liu P, Peng R, Liu L, Huang H, Fu Q, Hong H, Zhang J, Wang H. Rare loss-of-function variants reveal threshold and multifactorial inheritance of dextrocardia. Sci Bull (Beijing) 2023; 68:1993-1998. [PMID: 37573248 DOI: 10.1016/j.scib.2023.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/30/2023] [Accepted: 06/30/2023] [Indexed: 08/14/2023]
Affiliation(s)
- Zhongzhong Chen
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China
| | - Yunqian Gao
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lei Lu
- Shanghai Key Laboratory of Metabolic Remodelling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Nan Li
- BGI Genomics, Shenzhen 518083, China
| | - Pei Liu
- Shanghai Key Laboratory of Metabolic Remodelling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Rui Peng
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China
| | - Lingling Liu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Qihua Fu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Haifa Hong
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | | | - Hongyan Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Metabolic Remodelling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China; Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
20
|
Mill P, Christensen ST, Pedersen LB. Primary cilia as dynamic and diverse signalling hubs in development and disease. Nat Rev Genet 2023; 24:421-441. [PMID: 37072495 PMCID: PMC7615029 DOI: 10.1038/s41576-023-00587-9] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 04/20/2023]
Abstract
Primary cilia, antenna-like sensory organelles protruding from the surface of most vertebrate cell types, are essential for regulating signalling pathways during development and adult homeostasis. Mutations in genes affecting cilia cause an overlapping spectrum of >30 human diseases and syndromes, the ciliopathies. Given the immense structural and functional diversity of the mammalian cilia repertoire, there is a growing disconnect between patient genotype and associated phenotypes, with variable severity and expressivity characteristic of the ciliopathies as a group. Recent technological developments are rapidly advancing our understanding of the complex mechanisms that control biogenesis and function of primary cilia across a range of cell types and are starting to tackle this diversity. Here, we examine the structural and functional diversity of primary cilia, their dynamic regulation in different cellular and developmental contexts and their disruption in disease.
Collapse
Affiliation(s)
- Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | | | - Lotte B Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
Li-Villarreal N, Rasmussen TL, Christiansen AE, Dickinson ME, Hsu CW. Three-dimensional microCT imaging of mouse heart development from early post-implantation to late fetal stages. Mamm Genome 2023; 34:156-165. [PMID: 36595063 PMCID: PMC10290591 DOI: 10.1007/s00335-022-09976-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023]
Abstract
Comprehensive detailed characterization of new mouse models can be challenging due to the individual focus involved in developing these models. Often models are engineered to test a specific hypothesis in a limited number of tissues, stages, and/or other contexts. Whether or not the model produces the desired phenotypes, phenotyping beyond the desired context can be extremely work intensive and these studies are often not undertaken. However, the general information resulting from broader phenotyping can be invaluable to the wider scientific community. The International Mouse Phenotyping Consortium (IMPC) and its subsidiaries, like the Knockout Mouse Project (KOMP), has made great strides in streamlining this process. In particular, the use of microCT has been an invaluable resource in examining internal organ systems throughout fetal/developmental stages. Here, we provide several novel vignettes demonstrating the utility of microCT in uncovering cardiac phenotypes both based on human disease correlations and those that are unpredicted.
Collapse
Affiliation(s)
- Nanbing Li-Villarreal
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Tara L Rasmussen
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Audrey E Christiansen
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Mary E Dickinson
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chih-Wei Hsu
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Education, Innovation and Technology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
Suo MJ, Chen WC, Xu ZQ, Tian GX, Li T, Li P, Sheng W, Huang GY, Ma XJ. X-linked BCOR variants identified in Chinese Han patients with congenital heart disease. J Gene Med 2023; 25:e3461. [PMID: 36314054 DOI: 10.1002/jgm.3461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/08/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Congenital heart disease (CHD) frequently manifests as a complex phenotype and approximately one-third of cases may be caused by genetic factors. BCOR, an X-linked gene encoding the corepressor of BCL6, has been demonstrated to be closely involved in human heart development. However, whether BCOR variants represent the genetic etiology underlying CHD needs further investigation. METHODS We performed whole exome sequencing on CHD nuclear families and identified a candidate gene, BCOR, by robust bioinformatic analysis and medical literature searches. Targeted DNA sequencing of the candidate gene was conducted and then the association between variants and the risk of developing CHD was analyzed. The effects of BCOR mutations on gene expression, localization, protein interaction, and signaling pathways were evaluated in vitro. RESULTS We identified a BCOR hemizygous missense variant (c.1448C>T, p.Pro483Leu) in a male proband presented with CHD/heterotaxy. Sanger sequencing confirmed that this variant was inherited from his asymptomatic mother. Interestingly, through literature searches, we observed another novel BCOR hemizygous missense variant (c.1619G>A, p.Arg540Gln) in a CHD patient with heterotaxy, supporting the pathogenic evidence of BCOR variants. Functional experiments conducted in vitro revealed that the variant p.Pro483Leu altered the subcellular localization of BCOR protein, disrupted its interaction with BCL6, and significantly promoted cell proliferation, whereas the variant p.Arg540Gln displayed no obvious effects. Nevertheless, transcriptional analysis revealed that down-regulation of BCOR substantially enhanced the activities of mitogen-activated protein and phosphoinositide 3-kinase-AKT signaling pathways, which are closely attributed to heart development. Targeted sequencing of 932 sporadic CHD patients enriched nine variants of BCOR predicted as likely rare and damaging and a septal defect was present in 81.8% (9/11) of them, including the two probands, which was consistent with the possible phenotype caused by BCOR defects. CONCLUSIONS The findings of the present study indicate that variants in BCOR may predispose individuals to CHD in the Chinese Han population.
Collapse
Affiliation(s)
- Mei-Jiao Suo
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei-Cheng Chen
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Zi-Qing Xu
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Gui-Xiang Tian
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Ting Li
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Ping Li
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei Sheng
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Guo-Ying Huang
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| | - Xiao-Jing Ma
- Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases (2018RU002), Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
23
|
Ernault AC, Kawasaki M, Fabrizi B, Montañés-Agudo P, Amersfoorth SCM, Al-Shama RFM, Coronel R, De Groot JR. Knockdown of Ift88 in fibroblasts causes extracellular matrix remodeling and decreases conduction velocity in cardiomyocyte monolayers. Front Physiol 2022; 13:1057200. [PMID: 36467697 PMCID: PMC9713696 DOI: 10.3389/fphys.2022.1057200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 12/22/2024] Open
Abstract
Background: Atrial fibrosis plays an important role in the development and persistence of atrial fibrillation by promoting reentry. Primary cilia have been identified as a regulator of fibroblasts (FB) activation and extracellular matrix (ECM) deposition. We hypothesized that selective reduction of primary cilia causes increased fibrosis and facilitates reentry. Aim: The aim of this study was to disrupt the formation of primary cilia in FB and examine its consequences on ECM and conduction in a co-culture system of cardiomyocytes (CM) and FB. Materials: Using short interfering RNA (siRNA), we removed primary cilia in neonatal rat ventricular FB by reducing the expression of Ift88 gene required for ciliary assembly. We co-cultured neonatal rat ventricular cardiomyocytes (CM) with FB previously transfected with Ift88 siRNA (siIft88) or negative control siRNA (siNC) for 48 h. We examined the consequences of ciliated fibroblasts reduction on conduction and tissue remodeling by performing electrical mapping, microelectrode, and gene expression measurements. Results: Transfection of FB with siIft88 resulted in a significant 60% and 30% reduction of relative Ift88 expression in FB and CM-FB co-cultures, respectively, compared to siNC. Knockdown of Ift88 significantly increased the expression of ECM genes Fn1, Col1a1 and Ctgf by 38%, 30% and 18%, respectively, in comparison to transfection with siNC. Conduction velocity (CV) was significantly decreased in the siIft88 group in comparison to siNC [11.12 ± 4.27 cm/s (n = 10) vs. 17.00 ± 6.20 (n = 10) respectively, p < 0.05]. The fraction of sites with interelectrode activation block was larger in the siIft88 group than in the siNC group (6.59 × 10-2 ± 8.01 × 10-2 vs. 1.18 × 10-2 ± 3.72 × 10-2 respectively, p < 0.05). We documented spontaneous reentrant arrhythmias in two cultures in the siIft88 group and in none of the siNC group. Action potentials were not significantly different between siNC and siIft88 groups. Conclusion: Disruption of cilia formation by siIft88 causes ECM remodeling and conduction abnormalities. Prevention of cilia loss could be a target for prevention of arrhythmias.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ruben Coronel
- *Correspondence: Ruben Coronel, ; Joris R. De Groot,
| | | |
Collapse
|
24
|
Chen W, Wang F, Zeng W, Zhang X, Shen L, Zhang Y, Zhou X. Biallelic mutations of TTC12 and TTC21B were identified in Chinese patients with multisystem ciliopathy syndromes. Hum Genomics 2022; 16:48. [PMID: 36273201 PMCID: PMC9587637 DOI: 10.1186/s40246-022-00421-z] [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: 07/04/2022] [Accepted: 10/12/2022] [Indexed: 12/04/2022] Open
Abstract
Background Abnormalities in cilia ultrastructure and function lead to a range of human phenotypes termed ciliopathies. Many tetratricopeptide repeat domain (TTC) family members have been reported to play critical roles in cilium organization and function.
Results Here, we describe five unrelated family trios with multisystem ciliopathy syndromes, including situs abnormality, complex congenital heart disease, nephronophthisis or neonatal cholestasis. Through whole-exome sequencing and Sanger sequencing confirmation, we identified compound heterozygous mutations of TTC12 and TTC21B in six affected individuals of Chinese origin. These nonsynonymous mutations affected highly conserved residues and were consistently predicted to be pathogenic. Furthermore, ex vivo cDNA amplification demonstrated that homozygous c.1464 + 2 T > C of TTC12 would cause a whole exon 16 skipping. Both mRNA and protein levels of TTC12 were significantly downregulated in the cells derived from the patient carrying TTC12 mutation c.1464 + 2 T > C by real-time qPCR and immunofluorescence assays when compared with two healthy controls. Transmission electron microscopy analysis further identified ultrastructural defects of the inner dynein arms in this patient. Finally, the effect of TTC12 deficiency on cardiac LR patterning was recapitulated by employing a morpholino-mediated knockdown of ttc12 in zebrafish. Conclusions To the best of our knowledge, this is the first study reporting the association between TTC12 variants and ciliopathies in a Chinese population. In addition to nephronophthisis and laterality defects, our findings demonstrated that TTC21B should also be considered a candidate gene for biliary ciliopathy, such as TTC26, which further expands the phenotypic spectrum of TTC21B deficiency in humans. Supplementary Information The online version contains supplementary material available at 10.1186/s40246-022-00421-z.
Collapse
Affiliation(s)
- Weicheng Chen
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China
| | - Feifei Wang
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China
| | - Weijia Zeng
- State Key Lab of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xinyan Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China
| | - Libing Shen
- International Human Phenome Institutes (IHPI), Shanghai, 200433, China
| | - Yuan Zhang
- Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, China. .,, Shanghai, China.
| | - Xiangyu Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Pediatric Cardiovascular Center at Children's Hospital of Fudan University, Fudan University Shanghai Medical College, Shanghai, 200011, China. .,Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, China. .,, Shanghai, China.
| |
Collapse
|
25
|
Hedgehog Morphogens Act as Growth Factors Critical to Pre- and Postnatal Cardiac Development and Maturation: How Primary Cilia Mediate Their Signal Transduction. Cells 2022; 11:cells11121879. [PMID: 35741008 PMCID: PMC9221318 DOI: 10.3390/cells11121879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 02/06/2023] Open
Abstract
Primary cilia are crucial for normal cardiac organogenesis via the formation of cyto-architectural, anatomical, and physiological boundaries in the developing heart and outflow tract. These tiny, plasma membrane-bound organelles function in a sensory-integrative capacity, interpreting both the intra- and extra-cellular environments and directing changes in gene expression responses to promote, prevent, and modify cellular proliferation and differentiation. One distinct feature of this organelle is its involvement in the propagation of a variety of signaling cascades, most notably, the Hedgehog cascade. Three ligands, Sonic, Indian, and Desert hedgehog, function as growth factors that are most commonly dependent on the presence of intact primary cilia, where the Hedgehog receptors Patched-1 and Smoothened localize directly within or at the base of the ciliary axoneme. Hedgehog signaling functions to mediate many cell behaviors that are critical for normal embryonic tissue/organ development. However, inappropriate activation and/or upregulation of Hedgehog signaling in postnatal and adult tissue is known to initiate oncogenesis, as well as the pathogenesis of other diseases. The focus of this review is to provide an overview describing the role of Hedgehog signaling and its dependence upon the primary cilium in the cell types that are most essential for mammalian heart development. We outline the breadth of developmental defects and the consequential pathologies resulting from inappropriate changes to Hedgehog signaling, as it pertains to congenital heart disease and general cardiac pathophysiology.
Collapse
|
26
|
Song T, Yang Y, Zhou P, Ran J, Zhang L, Wu X, Xie W, Zhong T, Liu H, Liu M, Li D, Zhao H, Zhou J. ENKD1 promotes CP110 removal through competing with CEP97 to initiate ciliogenesis. EMBO Rep 2022; 23:e54090. [PMID: 35301795 PMCID: PMC9066061 DOI: 10.15252/embr.202154090] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Despite the importance of cilia in cell signaling and motility, the molecular mechanisms regulating cilium formation remain incompletely understood. Herein, we characterize enkurin domain-containing protein 1 (ENKD1) as a novel centrosomal protein that mediates the removal of centriolar coiled-coil protein 110 (CP110) from the mother centriole to promote ciliogenesis. We show that Enkd1 knockout mice possess ciliogenesis defects in multiple organs. Super-resolution microscopy reveals that ENKD1 is a stable component of the centrosome throughout the ciliogenesis process. Simultaneous knockdown of ENKD1 and CP110 significantly reverses the ciliogenesis defects induced by ENKD1 depletion. Protein interaction analysis shows that ENKD1 competes with centrosomal protein 97 (CEP97) in binding to CP110. Depletion of ENKD1 enhances the CP110-CEP97 interaction and detains CP110 at the mother centriole. These findings thus identify ENKD1 as a centrosomal protein and uncover a novel mechanism controlling CP110 removal from the mother centriole for the initiation of ciliogenesis.
Collapse
Affiliation(s)
- Ting Song
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Yunfan Yang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peng Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Jie Ran
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Liang Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Xiaofan Wu
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecology, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Wei Xie
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Tao Zhong
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Hongbin Liu
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecology, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Huijie Zhao
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, China.,State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecology, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
27
|
Djenoune L, Berg K, Brueckner M, Yuan S. A change of heart: new roles for cilia in cardiac development and disease. Nat Rev Cardiol 2022; 19:211-227. [PMID: 34862511 PMCID: PMC10161238 DOI: 10.1038/s41569-021-00635-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/27/2022]
Abstract
Although cardiac abnormalities have been observed in a growing class of human disorders caused by defective primary cilia, the function of cilia in the heart remains an underexplored area. The primary function of cilia in the heart was long thought to be restricted to left-right axis patterning during embryogenesis. However, new findings have revealed broad roles for cilia in congenital heart disease, valvulogenesis, myocardial fibrosis and regeneration, and mechanosensation. In this Review, we describe advances in our understanding of the mechanisms by which cilia function contributes to cardiac left-right axis development and discuss the latest findings that highlight a broader role for cilia in cardiac development. Specifically, we examine the growing line of evidence connecting cilia function to the pathogenesis of congenital heart disease. Furthermore, we also highlight research from the past 10 years demonstrating the role of cilia function in common cardiac valve disorders, including mitral valve prolapse and aortic valve disease, and describe findings that implicate cardiac cilia in mechanosensation potentially linking haemodynamic and contractile forces with genetic regulation of cardiac development and function. Finally, given the presence of cilia on cardiac fibroblasts, we also explore the potential role of cilia in fibrotic growth and summarize the evidence implicating cardiac cilia in heart regeneration.
Collapse
Affiliation(s)
- Lydia Djenoune
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn Berg
- Department of Paediatrics, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Martina Brueckner
- Department of Paediatrics, Yale University School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| | - Shiaulou Yuan
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Whole genome sequencing in transposition of the great arteries and associations with clinically relevant heart, brain and laterality genes. Am Heart J 2022; 244:1-13. [PMID: 34670123 DOI: 10.1016/j.ahj.2021.10.185] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The most common cyanotic congenital heart disease (CHD) requiring management as a neonate is transposition of great arteries (TGA). Clinically, up to 50% of TGA patients develop some form of neurodevelopmental disability (NDD), thought to have a significant genetic component. A "ciliopathy" and links with laterality disorders have been proposed. This first report of whole genome sequencing in TGA, sought to identify clinically relevant variants contributing to heart, brain and laterality defects. METHODS Initial whole genome sequencing analyses on 100 TGA patients focussed on established disease genes related to CHD (n = 107), NDD (n = 659) and heterotaxy (n = 74). Single variant as well as copy number variant analyses were conducted. Variant pathogenicity was assessed using the American College of Medical Genetics and Genomics-Association for Molecular Pathology guidelines. RESULTS Fifty-five putatively damaging variants were identified in established disease genes associated with CHD, NDD and heterotaxy; however, no clinically relevant variants could be attributed to disease. Notably, case-control analyses identified significantly more predicted-damaging, silent and total variants in TGA cases than healthy controls in established CHD genes (P < .001), NDD genes (P < .001) as well as across the three gene panels (P < .001). CONCLUSION We present compelling evidence that the majority of TGA is not caused by monogenic rare variants and is most likely oligogenic and/or polygenic in nature, highlighting the complex genetic architecture and multifactorial influences on this CHD sub-type and its long-term sequelae. Assessment of variant burden in key heart, brain and/or laterality genes may be required to unravel the genetic contributions to TGA and related disabilities.
Collapse
|
29
|
Biallelic DNAH9 mutations are identified in Chinese patients with defective left-right patterning and cilia-related complex congenital heart disease. Hum Genet 2022; 141:1339-1353. [PMID: 35050399 DOI: 10.1007/s00439-021-02426-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022]
Abstract
Defective left-right (LR) pattering results in a spectrum of laterality disorders including situs inversus totalis (SIT) and heterotaxy syndrome (Htx). Approximately, 50% of patients with primary ciliary dyskinesia (PCD) displayed SIT. Recessive variants in DNAH9 have recently been implicated in patients with situs inversus. Here, we describe six unrelated family trios and 2 sporadic patients with laterality defects and complex congenital heart disease (CHD). Through whole exome sequencing (WES), we identified compound heterozygous mutations in DNAH9 in the affected individuals of these family trios. Ex vivo cDNA amplification revealed that DNAH9 mRNA expression was significantly downregulated in these patients carrying biallelic DNAH9 mutations, which cause a premature stop codon or exon skipping. Transmission electron microscopy (TEM) analysis identified ultrastructural defects of the outer dynein arms in these affected individuals. dnah9 knockdown in zebrafish lead to the disturbance of cardiac left-right patterning without affecting ciliogenesis in Kupffer's vesicle (KV). By generating a Dnah9 knockout (KO) C57BL/6n mouse model, we found that Dnah9 loss leads to compromised cardiac function. In this study, we identified recessive DNAH9 mutations in Chinese patients with cardiac abnormalities and defective LR pattering.
Collapse
|
30
|
Sheng X, Gao S, Sheng Y, Xie X, Wang J, He Y. Vangl2 participates in the primary ciliary assembly under low fluid shear stress in hUVECs. Cell Tissue Res 2021; 387:95-109. [PMID: 34738156 DOI: 10.1007/s00441-021-03546-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 10/13/2021] [Indexed: 11/30/2022]
Abstract
The pattern of blood fluid shear stress (FSS) is considered the main factor that affects ciliogenesis in human umbilical vein endothelial cells (hUVECs), the underlying mechanism is unclear. Microfluidic chamber experiments were carried out to load hUVECs with low fluid shear stress (LSS, 0.1 dynes/cm2) or high fluid shear stress (HSS, 15 dynes/cm2). Van Gogh2 (Vangl2), a core protein in the planar cell polarity (PCP) pathway, was silenced and overexpressed in hUVECs. Immunofluorescence analysis showed that primary cilia assemble under LSS while disassembling under HSS. Vangl2 expression was consistent with cilia assembly, and its localization showed a polar distribution under LSS. Furthermore, the average number of ciliated cells and primary cilia length were increased in the Vangl2 overexpressing cell lines (the OE group) but decreased in the Vangl2 silenced cell lines (the SH group). When these cells were loaded with different FSS, more ciliated cells with longest primary cilia were observed in the LSS loaded OE group compared with those in the other groups. Immunoprecipitation showed that the interaction between Bardet-Biedl syndrome 8 (BBS8) and Vangl2 was enhanced following LSS loading compared to that under HSS. However, the interactions between phosphorylated dishevelled segment polarity protein 2 (pDvl2), kinesin family member 2a (Kif2a), and polo-like kinase 1 (Plk1) and Vangl2 were restrained following LSS loading. Overall, the results indicated that Vangl2 played a significant role during LSS-induced primary cilia assembly by recruiting BBS to promote the apical docking of basal bodies and by restraining Dvl2 phosphorylation from reducing primary cilia disassembly.
Collapse
Affiliation(s)
- Xin Sheng
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China.
| | - Shuanglin Gao
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Yan Sheng
- Laboratory of Basic Medical Morphology, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Xiadan Xie
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Junhua Wang
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Yan He
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| |
Collapse
|
31
|
Bornhorst D, Abdelilah-Seyfried S. Strong as a Hippo's Heart: Biomechanical Hippo Signaling During Zebrafish Cardiac Development. Front Cell Dev Biol 2021; 9:731101. [PMID: 34422841 PMCID: PMC8375320 DOI: 10.3389/fcell.2021.731101] [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: 06/26/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
The heart is comprised of multiple tissues that contribute to its physiological functions. During development, the growth of myocardium and endocardium is coupled and morphogenetic processes within these separate tissue layers are integrated. Here, we discuss the roles of mechanosensitive Hippo signaling in growth and morphogenesis of the zebrafish heart. Hippo signaling is involved in defining numbers of cardiac progenitor cells derived from the secondary heart field, in restricting the growth of the epicardium, and in guiding trabeculation and outflow tract formation. Recent work also shows that myocardial chamber dimensions serve as a blueprint for Hippo signaling-dependent growth of the endocardium. Evidently, Hippo pathway components act at the crossroads of various signaling pathways involved in embryonic zebrafish heart development. Elucidating how biomechanical Hippo signaling guides heart morphogenesis has direct implications for our understanding of cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Dorothee Bornhorst
- Stem Cell Program, Division of Hematology and Oncology, Boston Children's Hospital, Boston, MA, United States.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
32
|
Zhao Y, Wang LK, Eskin A, Kang X, Fajardo VM, Mehta Z, Pineles S, Schmidt RJ, Nagiel A, Satou G, Garg M, Federman M, Reardon LC, Lee SL, Biniwale R, Grody WW, Halnon N, Khanlou N, Quintero-Rivera F, Alejos JC, Nakano A, Fishbein GA, Van Arsdell GS, Nelson SF, Touma M. Recessive ciliopathy mutations in primary endocardial fibroelastosis: a rare neonatal cardiomyopathy in a case of Alstrom syndrome. J Mol Med (Berl) 2021; 99:1623-1638. [PMID: 34387706 PMCID: PMC8541947 DOI: 10.1007/s00109-021-02112-z] [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: 12/01/2020] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 11/09/2022]
Abstract
Abstract Among neonatal cardiomyopathies, primary endocardial fibroelastosis (pEFE) remains a mysterious disease of the endomyocardium that is poorly genetically characterized, affecting 1/5000 live births and accounting for 25% of the entire pediatric dilated cardiomyopathy (DCM) with a devastating course and grave prognosis. To investigate the potential genetic contribution to pEFE, we performed integrative genomic analysis, using whole exome sequencing (WES) and RNA-seq in a female infant with confirmed pathological diagnosis of pEFE. Within regions of homozygosity in the proband genome, WES analysis revealed novel parent-transmitted homozygous mutations affecting three genes with known roles in cilia assembly or function. Among them, a novel homozygous variant [c.1943delA] of uncertain significance in ALMS1 was prioritized for functional genomic and mechanistic analysis. Loss of function mutations of ALMS1 have been implicated in Alstrom syndrome (AS) [OMIM 203800], a rare recessive ciliopathy that has been associated with cardiomyopathy. The variant of interest results in a frameshift introducing a premature stop codon. RNA-seq of the proband’s dermal fibroblasts confirmed the impact of the novel ALMS1 variant on RNA-seq reads and revealed dysregulated cellular signaling and function, including the induction of epithelial mesenchymal transition (EMT) and activation of TGFβ signaling. ALMS1 loss enhanced cellular migration in patient fibroblasts as well as neonatal cardiac fibroblasts, while ALMS1-depleted cardiomyocytes exhibited enhanced proliferation activity. Herein, we present the unique pathological features of pEFE compared to DCM and utilize integrated genomic analysis to elucidate the molecular impact of a novel mutation in ALMS1 gene in an AS case. Our report provides insights into pEFE etiology and suggests, for the first time to our knowledge, ciliopathy as a potential underlying mechanism for this poorly understood and incurable form of neonatal cardiomyopathy. Key message Primary endocardial fibroelastosis (pEFE) is a rare form of neonatal cardiomyopathy that occurs in 1/5000 live births with significant consequences but unknown etiology. Integrated genomics analysis (whole exome sequencing and RNA sequencing) elucidates novel genetic contribution to pEFE etiology. In this case, the cardiac manifestation in Alstrom syndrome is pEFE. To our knowledge, this report provides the first evidence linking ciliopathy to pEFE etiology. Infants with pEFE should be examined for syndromic features of Alstrom syndrome. Our findings lead to a better understanding of the molecular mechanisms of pEFE, paving the way to potential diagnostic and therapeutic applications.
Supplementary information The online version contains supplementary material available at 10.1007/s00109-021-02112-z.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Lee-Kai Wang
- Institute for Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ascia Eskin
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Xuedong Kang
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Viviana M Fajardo
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Zubin Mehta
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stacy Pineles
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ryan J Schmidt
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Aaron Nagiel
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Gary Satou
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Meena Garg
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Myke Federman
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Leigh C Reardon
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Ahmanson/UCLA Adult Congenital Heart Disease Center, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Steven L Lee
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Reshma Biniwale
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Wayne W Grody
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nancy Halnon
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Negar Khanlou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Fabiola Quintero-Rivera
- Department of Pathology and Laboratory Medicine and Department of Pediatrics, University of California Irvine, CA, Irvine, USA
| | - Juan C Alejos
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA
| | - Atsushi Nakano
- Eli and Edythe Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gregory A Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Glen S Van Arsdell
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stanley F Nelson
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA.,Institute for Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Marlin Touma
- Department of Pediatrics, 3762 MacDonald Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E. Young Dr S, CA, 90095, Los Angeles, USA. .,Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,Eli and Edythe Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,The Molecular Biology Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
33
|
Diab NS, Barish S, Dong W, Zhao S, Allington G, Yu X, Kahle KT, Brueckner M, Jin SC. Molecular Genetics and Complex Inheritance of Congenital Heart Disease. Genes (Basel) 2021; 12:1020. [PMID: 34209044 PMCID: PMC8307500 DOI: 10.3390/genes12071020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/09/2023] Open
Abstract
Congenital heart disease (CHD) is the most common congenital malformation and the leading cause of mortality therein. Genetic etiologies contribute to an estimated 90% of CHD cases, but so far, a molecular diagnosis remains unsolved in up to 55% of patients. Copy number variations and aneuploidy account for ~23% of cases overall, and high-throughput genomic technologies have revealed additional types of genetic variation in CHD. The first CHD risk genotypes identified through high-throughput sequencing were de novo mutations, many of which occur in chromatin modifying genes. Murine models of cardiogenesis further support the damaging nature of chromatin modifying CHD mutations. Transmitted mutations have also been identified through sequencing of population scale CHD cohorts, and many transmitted mutations are enriched in cilia genes and Notch or VEGF pathway genes. While we have come a long way in identifying the causes of CHD, more work is required to end the diagnostic odyssey for all CHD families. Complex genetic explanations of CHD are emerging but will require increasingly sophisticated analysis strategies applied to very large CHD cohorts before they can come to fruition in providing molecular diagnoses to genetically unsolved patients. In this review, we discuss the genetic architecture of CHD and biological pathways involved in its pathogenesis.
Collapse
Affiliation(s)
- Nicholas S. Diab
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
| | - Syndi Barish
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
| | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065, USA
| | - Shujuan Zhao
- Department of Genetics, School of Medicine, Washington University, St. Louis, MO 63110, USA; (S.Z.); (X.Y.)
| | - Garrett Allington
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Xiaobing Yu
- Department of Genetics, School of Medicine, Washington University, St. Louis, MO 63110, USA; (S.Z.); (X.Y.)
- Department of Computer Science & Engineering, Washington University, St. Louis, MO 63130, USA
| | - Kristopher T. Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA;
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Martina Brueckner
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; (N.S.D.); (S.B.); (W.D.)
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sheng Chih Jin
- Department of Genetics, School of Medicine, Washington University, St. Louis, MO 63110, USA; (S.Z.); (X.Y.)
- Department of Pediatrics, School of Medicine, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
34
|
Trevino CE, Holleman AM, Corbitt H, Maslen CL, Rosser TC, Cutler DJ, Johnston HR, Rambo-Martin BL, Oberoi J, Dooley KJ, Capone GT, Reeves RH, Cordell HJ, Keavney BD, Agopian AJ, Goldmuntz E, Gruber PJ, O'Brien JE, Bittel DC, Wadhwa L, Cua CL, Moskowitz IP, Mulle JG, Epstein MP, Sherman SL, Zwick ME. Identifying genetic factors that contribute to the increased risk of congenital heart defects in infants with Down syndrome. Sci Rep 2020; 10:18051. [PMID: 33093519 PMCID: PMC7582922 DOI: 10.1038/s41598-020-74650-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/05/2020] [Indexed: 01/16/2023] Open
Abstract
Atrioventricular septal defects (AVSD) are a severe congenital heart defect present in individuals with Down syndrome (DS) at a > 2000-fold increased prevalence compared to the general population. This study aimed to identify risk-associated genes and pathways and to examine a potential polygenic contribution to AVSD in DS. We analyzed a total cohort of 702 individuals with DS with or without AVSD, with genomic data from whole exome sequencing, whole genome sequencing, and/or array-based imputation. We utilized sequence kernel association testing and polygenic risk score (PRS) methods to examine rare and common variants. Our findings suggest that the Notch pathway, particularly NOTCH4, as well as genes involved in the ciliome including CEP290 may play a role in AVSD in DS. These pathways have also been implicated in DS-associated AVSD in prior studies. A polygenic component for AVSD in DS has not been examined previously. Using weights based on the largest genome-wide association study of congenital heart defects available (2594 cases and 5159 controls; all general population samples), we found PRS to be associated with AVSD with odds ratios ranging from 1.2 to 1.3 per standard deviation increase in PRS and corresponding liability r2 values of approximately 1%, suggesting at least a small polygenic contribution to DS-associated AVSD. Future studies with larger sample sizes will improve identification and quantification of genetic contributions to AVSD in DS.
Collapse
Affiliation(s)
- Cristina E Trevino
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - Aaron M Holleman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Holly Corbitt
- Division of Cardiovascular Medicine and the Heart Research Center, Oregon Health and Science University, Portland, OR, USA
| | - Cheryl L Maslen
- Division of Cardiovascular Medicine and the Heart Research Center, Oregon Health and Science University, Portland, OR, USA
| | - Tracie C Rosser
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - David J Cutler
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - H Richard Johnston
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - Benjamin L Rambo-Martin
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - Jai Oberoi
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - Kenneth J Dooley
- Sibley Heart Center Cardiology, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | | | - Roger H Reeves
- Department of Physiology and the Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Heather J Cordell
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A J Agopian
- Human Genetics Center; Department of Epidemiology, Human Genetics, and Environmental Sciences, UTHealth School of Public Health, Houston, TX, USA
| | - Elizabeth Goldmuntz
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter J Gruber
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - James E O'Brien
- The Ward Family Heart Center, Section of Cardiac Surgery, Children's Mercy Hospital, Kansas City, MO, USA
| | - Douglas C Bittel
- College of Biosciences, Kansas City University of Medicine and Biosciences, Kansas City, MO, USA
| | | | - Clifford L Cua
- Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Ivan P Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Jennifer G Mulle
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
| | - Stephanie L Sherman
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael E Zwick
- Department of Human Genetics, Emory University School of Medicine, 300 Whitehead Biomedical Research Building, 615 Michael St., Atlanta, GA, 30322, USA.
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
35
|
Sherman F, Wodrich M, Zampi JD, Lee J, McCaffery H, Saba TG. Phenotypic features of ciliary dyskinesia among patients with congenital cardiovascular malformations. Pediatr Pulmonol 2020; 55:2674-2682. [PMID: 32662935 DOI: 10.1002/ppul.24959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/11/2020] [Indexed: 11/06/2022]
Abstract
BACKGROUND Cilia are cell membrane-bound organelles responsible for airway mucus clearance, establishment of left-right organ asymmetry, cardiogenesis, and many other functions in utero. Phenotypic features suggestive of respiratory ciliary dyskinesia among patients with heterotaxy syndrome, defined as complex cardiovascular malformations (CVM) and situs ambiguus (SA), has not been adequately explored. OBJECTIVES We hypothesized that there is a greater incidence of phenotypic features consistent with ciliary dyskinesia among patients with heterotaxy syndrome compared to patients with other CVM and laterality defects without heterotaxy syndrome. METHODS Thirty six subjects were identified by medical record search and divided into four groups based on situs status and type of CVM as follows: SA and complex CVM (group 1); SA and simple CVM (group 2); situs solitus and complex CVM (group 3); and situs solitus and simple CVM (group 4). Phenotype was assessed with a clinical questionnaire, nasal nitric oxide (NO) level, and pulmonary function testing. Those with complex CVM underwent additional testing for variants in genes involved in ciliary structure and function. RESULTS The mean nasal NO level was significantly lower among all subjects with complex CVM regardless of situs anomalies (groups 1 and 3). There was no significant difference in respiratory symptoms or lung function among the four groups. No bi-allelic genetic mutations were detected among patients with complex CVM. CONCLUSIONS This study identified a relatively lower mean nasal NO level, suggestive of relative ciliary dyskinesia, among subjects with complex CVM. Pulmonary function and clinical symptoms did not reflect significant pulmonary disease among those with complex CVM.
Collapse
Affiliation(s)
- Forrest Sherman
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Mitchel Wodrich
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Jeffrey D Zampi
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Julie Lee
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Harlan McCaffery
- Center for Human Growth and Development, University of Michigan, Ann Arbor, Michigan
| | - Thomas G Saba
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
36
|
Abstract
Congenital heart disease (CHD) is the most common major congenital anomaly with an incidence of ∼1% of live births and is a significant cause of birth defect-related mortality. The genetic mechanisms underlying the development of CHD are complex and remain incompletely understood. Known genetic causes include all classes of genetic variation including chromosomal aneuploidies, copy number variants, and rare and common single-nucleotide variants, which can be either de novo or inherited. Among patients with CHD, ∼8%-12% have a chromosomal abnormality or aneuploidy, between 3% and 25% have a copy number variation, and 3%-5% have a single-gene defect in an established CHD gene with higher likelihood of identifying a genetic cause in patients with nonisolated CHD. These genetic variants disrupt or alter genes that play an important role in normal cardiac development and in some cases have pleiotropic effects on other organs. This work reviews some of the most common genetic causes of CHD as well as what is currently known about the underlying mechanisms.
Collapse
Affiliation(s)
| | - Wendy K Chung
- Department of Pediatrics
- Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, USA
| |
Collapse
|
37
|
Yu T, Matsuda M. Epb41l5 interacts with Iqcb1 and regulates ciliary function in zebrafish embryos. J Cell Sci 2020; 133:jcs240648. [PMID: 32501287 PMCID: PMC7338265 DOI: 10.1242/jcs.240648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 05/13/2020] [Indexed: 11/20/2022] Open
Abstract
Erythrocyte protein band 4.1 like 5 (EPB41L5) is an adaptor protein beneath the plasma membrane that functions to control epithelial morphogenesis. Here we report a previously uncharacterized role of EPB41L5 in controlling ciliary function. We found that EPB41L5 forms a complex with IQCB1 (previously known as NPHP5), a ciliopathy protein. Overexpression of EPB41L5 reduced IQCB1 localization at the ciliary base in cultured mammalian epithelial cells. Conversely, epb41l5 knockdown increased IQCB1 localization at the ciliary base. epb41l5-deficient zebrafish embryos or embryos expressing C-terminally modified forms of Epb41l5 developed cilia with reduced motility and exhibited left-right patterning defects, an outcome of abnormal ciliary function. We observed genetic synergy between epb41l5 and iqcb1. Moreover, EPB41L5 decreased IQCB1 interaction with CEP290, another ciliopathy protein and a component of the ciliary base and centrosome. Together, these observations suggest that EPB41L5 regulates the composition of the ciliary base and centrosome through IQCB1 and CEP290.
Collapse
Affiliation(s)
- Tiffany Yu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07302, USA
| | - Miho Matsuda
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07302, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
38
|
Bozal-Basterra L, Gonzalez-Santamarta M, Muratore V, Bermejo-Arteagabeitia A, Da Fonseca C, Barroso-Gomila O, Azkargorta M, Iloro I, Pampliega O, Andrade R, Martín-Martín N, Branon TC, Ting AY, Rodríguez JA, Carracedo A, Elortza F, Sutherland JD, Barrio R. LUZP1, a novel regulator of primary cilia and the actin cytoskeleton, is a contributing factor in Townes-Brocks Syndrome. eLife 2020; 9:e55957. [PMID: 32553112 PMCID: PMC7363444 DOI: 10.7554/elife.55957] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/18/2020] [Indexed: 12/20/2022] Open
Abstract
Primary cilia are sensory organelles crucial for cell signaling during development and organ homeostasis. Cilia arise from centrosomes and their formation and function is governed by numerous factors. Through our studies on Townes-Brocks Syndrome (TBS), a rare disease linked to abnormal cilia formation in human fibroblasts, we uncovered the leucine-zipper protein LUZP1 as an interactor of truncated SALL1, a dominantly-acting protein causing the disease. Using TurboID proximity labeling and pulldowns, we show that LUZP1 associates with factors linked to centrosome and actin filaments. Here, we show that LUZP1 is a cilia regulator. It localizes around the centrioles and to actin cytoskeleton. Loss of LUZP1 reduces F-actin levels, facilitates ciliogenesis and alters Sonic Hedgehog signaling, pointing to a key role in cytoskeleton-cilia interdependency. Truncated SALL1 increases the ubiquitin proteasome-mediated degradation of LUZP1. Together with other factors, alterations in LUZP1 may be contributing to TBS etiology.
Collapse
Affiliation(s)
- Laura Bozal-Basterra
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - María Gonzalez-Santamarta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Veronica Muratore
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Aitor Bermejo-Arteagabeitia
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Carolina Da Fonseca
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Orhi Barroso-Gomila
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERehd, Instituto de Salud Carlos IIIMadridSpain
- ProteoRed-ISCIII, Instituto de Salud Carlos IIIMadridSpain
| | - Ibon Iloro
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERehd, Instituto de Salud Carlos IIIMadridSpain
- ProteoRed-ISCIII, Instituto de Salud Carlos IIIMadridSpain
| | - Olatz Pampliega
- Department of Neurosciences, University of the Basque Country, Achucarro Basque Center for Neuroscience-UPV/EHULeioaSpain
| | - Ricardo Andrade
- Analytical & High Resolution Biomedical Microscopy Core Facility, University of the Basque Country (UPV/EHU)LeioaSpain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Tess C Branon
- Department of Chemistry, Massachusetts Institute of TechnologyCambridgeUnited States
- Departments of Genetics, Chemistry and Biology, Stanford UniversityStanfordUnited States
| | - Alice Y Ting
- Department of Chemistry, Massachusetts Institute of TechnologyCambridgeUnited States
- Departments of Genetics, Chemistry and Biology, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Jose A Rodríguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU)LeioaSpain
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERONC, Instituto de Salud Carlos IIIMadridSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU)BilbaoSpain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
- CIBERehd, Instituto de Salud Carlos IIIMadridSpain
- ProteoRed-ISCIII, Instituto de Salud Carlos IIIMadridSpain
| | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| |
Collapse
|
39
|
Affiliation(s)
- Barry London
- From the Division of Cardiovascular Medicine, University of Iowa
| |
Collapse
|
40
|
Liu X, Chen W, Li W, Priest JR, Fu Y, Pang K, Ma B, Han B, Liu X, Hu S, Zhou Z. Exome-Based Case-Control Analysis Highlights the Pathogenic Role of Ciliary Genes in Transposition of the Great Arteries. Circ Res 2020; 126:811-821. [PMID: 32078439 DOI: 10.1161/circresaha.119.315821] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Transposition of the great arteries (TGA) is one of the most severe types of congenital heart diseases. Understanding the clinical characteristics and pathogenesis of TGA is, therefore, urgently needed for patient management of this severe disease. However, the clinical characteristics and genetic cause underlying TGA remain largely unexplored. OBJECTIVE We sought to systematically examine the clinical characteristics and genetic cause for isolated nonsyndromic TGA. METHODS AND RESULTS We recruited 249 patients with TGA (66 family trios) and performed whole-exome sequencing. The incidence of patent ductus arteriosus in dextro-TGA (52.7%) and dextrocardia/mesocardia in congenitally corrected TGA (32.8%) were significantly higher than that in other subtypes. A high prevalence of bicuspid pulmonic valve (9.6%) was observed in patients with TGA. Similar results were observed in a replication group of TGA (n=132). Through a series of bioinformatics filtering steps, we obtained 82 candidate genes harboring potentially damaging de novo, loss of function, compound heterozygous, or X-linked recessive variants. Established congenital heart disease-causing genes, such as FOXH1, were found among the list of candidate genes. A total of 19 ciliary genes harboring rare potentially damaging variants were also found; for example, DYNC2LI1 with a de novo putatively damaging variant. The enrichment of ciliary genes supports the roles of cilia in the pathogenesis of TGA. In total, 33% of the TGA probands had >1 candidate gene hit by putatively deleterious variants, suggesting that a portion of the TGA cases were probably affected by oligogenic or polygenic inheritance. CONCLUSIONS The findings of clinical characteristic analyses have important implications for TGA patient stratification. The results of genetic analyses highlight the pathogenic role of ciliary genes and a complex genetic architecture underlying TGA.
Collapse
Affiliation(s)
- Xuanyu Liu
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.).,Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, China (Xuanyu Liu, W.C., W.L., Y.F., Z.Z.)
| | - Wen Chen
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.).,Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, China (Xuanyu Liu, W.C., W.L., Y.F., Z.Z.)
| | - Wenke Li
- Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, China (Xuanyu Liu, W.C., W.L., Y.F., Z.Z.)
| | - James R Priest
- Department of Pediatrics, Stanford University School of Medicine, CA (J.R.P.)
| | - Yuanyuan Fu
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.).,Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, China (Xuanyu Liu, W.C., W.L., Y.F., Z.Z.)
| | - Kunjing Pang
- Department of Echocardiography, Fuwai Hospital, Beijing, China (K.P.)
| | - Baihui Ma
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.)
| | - Bianmei Han
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.)
| | - Xuewen Liu
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.)
| | - Shengshou Hu
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.)
| | - Zhou Zhou
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Xuanyu Liu, W.C., W.L., Y.F., B.M., B.H., Xuewen Liu, S.H., Z.Z.).,Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, China (Xuanyu Liu, W.C., W.L., Y.F., Z.Z.)
| |
Collapse
|
41
|
Tugging at the Heart Strings: The Septin Cytoskeleton in Heart Development and Disease. J Cardiovasc Dev Dis 2020; 7:jcdd7010003. [PMID: 31936541 PMCID: PMC7151155 DOI: 10.3390/jcdd7010003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/26/2019] [Accepted: 12/31/2019] [Indexed: 12/25/2022] Open
Abstract
Septin genes were originally identified in budding yeast in 1971. Since their original discovery, at least 13 mammalian genes have now been found, which give rise to a vast array of alternatively spliced proteins that display unique spatial-temporal function across organs systems. Septin’s are now recognized as the 4th major component of the cytoskeleton. Their role in regulating ciliogenesis, actin and microtubule organization and their involvement in mechanotransduction clearly solidify their place as both a responder and driver of cellular activity. Although work on septin’s has escalated over the past decades, knowledge of septin function in the heart remains rudimentary. Whereas many cardiovascular diseases have been associated with genetic loci that include septin genes, new and additional concerted efforts will likely uncover previously unrecognized mechanisms by which the septin class of proteins contribute to clinical cardiac phenotypes. In this review, we place known function of septin proteins in the context of heart development and disease and provide perspectives on how increased knowledge of these proteins can mechanistically inform cardiac pathologies.
Collapse
|
42
|
Yang Y, Hao H, Wu X, Guo S, Liu Y, Ran J, Li T, Li D, Liu M, Zhou J. Mixed-lineage leukemia protein 2 suppresses ciliary assembly by the modulation of actin dynamics and vesicle transport. Cell Discov 2019; 5:33. [PMID: 31263570 PMCID: PMC6591415 DOI: 10.1038/s41421-019-0100-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 01/27/2023] Open
Abstract
Primary cilia are critically involved in the coordination of diverse signaling pathways and ciliary defects are associated with a variety of human diseases. The past decades have witnessed great progress in the core machinery orchestrating ciliary assembly. However, the upstream epigenetic cues that direct ciliogenesis remain elusive. Herein, we demonstrate that mixed-lineage leukemia protein 2 (MLL2), a histone methyltransferase, plays a negative role in ciliogenesis. RNA-sequencing analysis reveals that the expression of five actin-associated proteins is significantly downregulated in MLL2-depleted cells. Overexpression of these proteins partially rescues ciliary abnormality elicited by MLL2 depletion. Our data also show that actin dynamics is remarkably changed in MLL2-depleted cells, resulting in the impairment of cell adhesion, spreading, and motility. In addition, MLL2 depletion promotes ciliary vesicle trafficking to the basal body in an actin-related manner. Together, these results reveal that MLL2 inhibits ciliogenesis by modulating actin dynamics and vesicle transport, and suggest that alteration of MLL2 may contribute to the pathogenesis of cilium-associated diseases.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Huijie Hao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Xiaofan Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Song Guo
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Jie Ran
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014 China
| | - Te Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Min Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014 China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071 China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014 China
| |
Collapse
|
43
|
DNAH11 variants and its association with congenital heart disease and heterotaxy syndrome. Sci Rep 2019; 9:6683. [PMID: 31040315 PMCID: PMC6491566 DOI: 10.1038/s41598-019-43109-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/16/2019] [Indexed: 01/09/2023] Open
Abstract
Congenital heart diseases (CHDs) are the most common types of birth defects, affecting approximately 1% of live births and remaining the leading cause of mortality. CHD patients often show a higher incidence of heterotaxy syndrome. However, the exact aetiology of CHD and heterotaxy syndrome remains unclear. In this study, targeted sequencing and Sanger sequencing were performed to analyze the exonic regions of 37 primary ciliary dysfunction (PCD)- related candidate genes in 42 CHD patients with heterotaxy syndrome. Variants affecting protein-coding regions were filtered according to databases of known variants and predicted in silico using functional prediction program. Thirty-four potential disease-causing heterozygous variants in 11 genes were identified in the 19 CHD patients with heterotaxy syndrome (45.2%, 19/42). The DNAH11 gene showed the highest mutation rate (16.7%; 14 of 84 alleles) among the CHD patients with heterotaxy. Fisher’s exact test revealed a significant association of DNAH11 variants with CHD and heterotaxy (P = 0.0001). In families, six different compound heterozygous variants of DNAH11 were validated in family 1-5031 (p.W802X/p.M282I), family 2-5045 (p.T3460K/p.G4425S), family 3-5065 (p.G447R/p.L1157R), family 4-5130 (p.I2262T/p.D3800H), family 5-5707 (p.S1823fs/p.F2759L/p.R4395X) and family 6-5062 (p.D3610V/p.I243V). These findings suggest that the DNAH11 variants are significantly associated with CHD and heterotaxy syndrome and that compound heterozygous DNAH11 variants may be the common genetic cause of the development of familial CHD and heterotaxy syndrome.
Collapse
|
44
|
Grubb S, Vestergaard ML, Andersen AS, Rasmussen KK, Mamsen LS, Tuckute G, Grunnet-Lauridsen K, Møllgård K, Ernst E, Christensen ST, Calloe K, Andersen CY. Comparison of Cultured Human Cardiomyocyte Clusters Obtained from Embryos/Fetuses or Derived from Human Embryonic Stem Cells. Stem Cells Dev 2019; 28:608-619. [PMID: 30755084 DOI: 10.1089/scd.2018.0231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cardiomyocytes (CMs) derived from human embryonic stem cells (hESCs) or induced pluripotent stem cells (iPSCs) are used to study cardiogenesis and mechanisms of heart disease, and are being used in methods for toxiological screening of drugs. The phenotype of stem-cell-derived CMs should ideally resemble native CMs. Here, we compare embryonic/fetal CMs with hESC-derived CMs according to function and morphology. CM clusters were obtained from human embryonic/fetal hearts from elective terminated pregnancies before gestational week 12, and separated into atrial and ventricular tissues. Specific markers for embryonic CMs and primary cilia were visualized using immunofluorescence microscopy analysis. Contracting human embryonic cardiomyocyte (hECM) clusters morphologically and phenotypically resemble CMs in the embryonic/fetal heart. In addition, the contracting hECM clusters expressed primary cilia similar to that of cells in the embryonic/fetal heart. The electrophysiological characteristics of atrial and ventricular CMs were established by recording action potentials (APs) using sharp electrodes. In contrast to ventricular APs, atrial APs displayed a marked early repolarization followed by a plateau phase. hESC-CMs displayed a continuum of AP shapes. In all embryonic/fetal clusters, both atrial and ventricular, AP duration was prolonged by exposure to the KV11.1 channel inhibitor dofetilide (50 nM); however, the prolongation was not significant, possibly due to the relatively small number of experiments. This study provides novel information on APs and functional characteristics of atrial and ventricular CMs in first trimester hearts, and demonstrates that Kv11.1 channels play a functional role already at these early stages. These results provide information needed to validate methods being developed on the basis of in vitro-derived CMs from either hESC or iPSC, and although there was a good correlation between the morphology of the two types of CMs, differences in electrophysiological characteristics exist.
Collapse
Affiliation(s)
- Søren Grubb
- 1 Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maj Linea Vestergaard
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Astrid Sten Andersen
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Karen Koefoed Rasmussen
- 3 Section of Cell and Developmental Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Linn Salto Mamsen
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Greta Tuckute
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| | | | - Kjeld Møllgård
- 4 Institute for Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Erik Ernst
- 5 The Department of Gynecology and Obstetrics, University Hospital of Aarhus, Aarhus, Denmark
| | - Søren Tvorup Christensen
- 3 Section of Cell and Developmental Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Calloe
- 1 Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Yding Andersen
- 2 Laboratory of Reproductive Biology, University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Maerz LD, Burkhalter MD, Schilpp C, Wittekindt OH, Frick M, Philipp M. Pharmacological cholesterol depletion disturbs ciliogenesis and ciliary function in developing zebrafish. Commun Biol 2019; 2:31. [PMID: 30729178 PMCID: PMC6351647 DOI: 10.1038/s42003-018-0272-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022] Open
Abstract
Patients with an inherited inability to synthesize sufficient amounts of cholesterol develop congenital malformations of the skull, toes, kidney and heart. As development of these structures depends on functional cilia we investigated whether cholesterol regulates ciliogenesis through inhibition of hydroxymethylglutaryl-Coenzyme A reductase (HMG-CoA-R), the rate-limiting enzyme in cholesterol synthesis. HMG-CoA-R is efficiently inhibited by statins, a standard medication for hyperlipidemia. When zebrafish embryos are treated with statins cilia dysfunction phenotypes including heart defects, left-right asymmetry defects and malformation of ciliated organs develop, which are ameliorated by cholesterol replenishment. HMG-CoA-R inhibition and other means of cholesterol reduction lowered ciliation frequency and cilia length in zebrafish as well as several mammalian cell types. Cholesterol depletion further triggers an inability for ciliary signalling. Because of a reduction of the transition zone component Pi(4,5)P2 we propose that cholesterol governs crucial steps of cilium extension. Taken together, we report that cholesterol abrogation provokes cilia defects.
Collapse
Affiliation(s)
- Lars D. Maerz
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Martin D. Burkhalter
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Carolin Schilpp
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Oliver H. Wittekindt
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Melanie Philipp
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
46
|
Abstract
The centriole is an ancient microtubule-based organelle with a conserved nine-fold symmetry. Centrioles form the core of centrosomes, which organize the interphase microtubule cytoskeleton of most animal cells and form the poles of the mitotic spindle. Centrioles can also be modified to form basal bodies, which template the formation of cilia and play central roles in cellular signaling, fluid movement, and locomotion. In this review, we discuss developments in our understanding of the biogenesis of centrioles and cilia and the regulatory controls that govern their structure and number. We also discuss how defects in these processes contribute to a spectrum of human diseases and how new technologies have expanded our understanding of centriole and cilium biology, revealing exciting avenues for future exploration.
Collapse
Affiliation(s)
- David K Breslow
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06511, USA;
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| |
Collapse
|
47
|
Burns TA, Deepe RN, Bullard J, Phelps AL, Toomer KA, Hiriart E, Norris RA, Haycraft CJ, Wessels A. A Novel Mouse Model for Cilia-Associated Cardiovascular Anomalies with a High Penetrance of Total Anomalous Pulmonary Venous Return. Anat Rec (Hoboken) 2019; 302:136-145. [PMID: 30289203 PMCID: PMC6312498 DOI: 10.1002/ar.23909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/25/2018] [Accepted: 03/12/2018] [Indexed: 01/15/2023]
Abstract
Primary cilia are small organelles projecting from the cell surface of many cell types. They play a crucial role in the regulation of various signaling pathway. In this study, we investigated the importance of cilia for heart development by conditionally deleting intraflagellar transport protein Ift88 using the col3.6-cre mouse. Analysis of col3.6;Ift88 offspring showed a wide spectrum of cardiovascular defects including double outlet right ventricle and atrioventricular septal defects. In addition, we found that in the majority of specimens the pulmonary veins did not properly connect to the developing left atrium. The abnormal connections found resemble those seen in patients with total anomalous pulmonary venous return. Analysis of mutant hearts at early stages of development revealed abnormal development of the dorsal mesocardium, a second heart field-derived structure at the venous pole intrinsically related to the development of the pulmonary veins. Data presented support a crucial role for primary cilia in outflow tract development and atrioventricular septation and their significance for the formation of the second heart field-derived tissues at the venous pole including the dorsal mesocardium. Furthermore, the results of this study indicate that proper formation of the dorsal mesocardium is critically important for the development of the pulmonary veins. Anat Rec, 302:136-145, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tara A. Burns
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - Raymond N. Deepe
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - John Bullard
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - Aimee L. Phelps
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - Katelynn A. Toomer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - Emilye Hiriart
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - Courtney J. Haycraft
- Department of Biological Sciences, Mississippi College, 200 S Capitol St, Clinton, Mississippi 39058, USA
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW The aim of this study is to review genetics of congenital heart disease (CHD) with a focus on clinical applications, genetic testing and clinical challenges. RECENT FINDINGS With improved clinical care, there is a rapidly expanding population of adults, especially women, with CHD who have not undergone contemporary genetic assessment and do not understand their risk for having a child with CHD. Many patients have never undergone assessment or had genetic testing. A major barrier is medical geneticist availability, resulting in this burden of care shifting to providers outside of genetics. Even with current understanding, the cause for the majority of cases of CHD is still not known. There are significant gaps in knowledge in the realms of more complex causes such as noncoding variants, multigenic contribution and small structural chromosomal anomalies. SUMMARY Standard assessment of patients with CHD, including adult survivors, is indicated. The best first-line genetic assessment for most patients with CHD is a chromosomal microarray, and this will soon evolve to be genomic sequencing with copy number variant analysis. Due to lack of medical geneticists, creative solutions to maximize the number of patients with CHD who undergo assessment with standard protocols and plans for support with result interpretation need to be explored.
Collapse
|
49
|
Loomba RS, Bhushan A, Afolayan AJ. A pilot study exploring the impact of cardiac medications on ciliary beat frequency: possible implications for clinical management. J Basic Clin Physiol Pharmacol 2018; 29:565-571. [PMID: 29723155 DOI: 10.1515/jbcpp-2017-0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 03/13/2018] [Indexed: 11/15/2022]
Abstract
Abstract
Background
Cilia are involved in several physiologic processes, and at least a single primary cilium can be found on nearly every cell in the human body. Various factors, such as pH, temperature, exposure to medications and toxins can impact ciliary function as is manifested by changes in the ciliary beat frequency. Those with ciliary dyskinesia may also have congenital cardiac malformations and may require care in a cardiac intensive care unit. This study investigates the effect on the ciliary beat frequency of medications frequently used in a cardiac intensive care unit.
Methods
The ciliated epithelial cells were obtained via nasal swab from a relatively healthy individual. These cells were cultured for 24 h. Video microscopy was then employed to determine the ciliary beat frequency at baseline and then at 15, 30, 60 and 90 min after exposure to either normal saline (control) or one of several medications. The ciliary beat frequency at each time point was then compared to the ciliary beat frequency at the same time point in the control sample as well as the baseline value for that particular sample.
Results
Epinephrine increased the ciliary beat frequency compared to the baseline and the controls up to 30 min and then subsequently led to a significant decrease in ciliary beat frequency at 90 min. On the one hand, norepinephrine, dexmedetomidine, procainamide, propranolol and enalapril all decreased ciliary beat frequency significantly throughout the 90-min observation period. On the other hand, Milrinone significantly increased the ciliary beat frequency throughout the observation period, while heparin had no impact on ciliary beat frequency.
Conclusions
The medications frequently used in cardiac intensive care unit impact ciliary function, with most being ciliodepressant. Further investigation is needed to determine the clinical impacts and whether these effects are exaggerated in those with ciliary dyskinesia.
Collapse
Affiliation(s)
- Rohit S Loomba
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 53229, USA, Phone: +630-8818-8342
| | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60622-3086, USA
| | - Adeleye J Afolayan
- Division of Neonatology, Children's Hospital of Wisconsin/Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
50
|
A novel ZIC3 gene mutation identified in patients with heterotaxy and congenital heart disease. Sci Rep 2018; 8:12386. [PMID: 30120289 PMCID: PMC6098004 DOI: 10.1038/s41598-018-30204-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 07/19/2018] [Indexed: 12/12/2022] Open
Abstract
Heterotaxy syndrome (HTX) is characterized by left-right (LR) asymmetry disturbances associated with severe heart malformations. However, the exact genetic cause of HTX pathogenesis remains unclear. The aim of this study was to investigate the pathogenic mechanism underlying heterotaxy syndrome. Targeted next-generation sequencing (NGS) was performed for twenty-two candidate genes correlated with LR axis development in sixty-six HTX patients from unrelated families. Variants were filtered from databases and predicted in silico using prediction programs. A total of twenty-one potential disease-causing variants were identified in seven genes. Next, we used Sanger sequencing to confirm the identified variants in the family pedigree and found a novel hemizygous mutation (c.890G > T, p.C297F) in the ZIC3 gene in a male patient that was inherited from his mother, who was a carrier. The results of functional indicated that this ZIC3 mutation decreases transcriptional activity, affects the affinity of the GLI-binding site and results in aberrant cellular localization in transfected cells. Moreover, morpholino-knockdown experiments in zebrafish demonstrated that zic3 mutant mRNA failed to rescue the abnormal phenotype, suggesting a role for the novel ZIC3 mutation in heterotaxy syndrome.
Collapse
|