1
|
Guo Q, Zhong X, Dang Z, Zhang B, Yang Z. Identification of GBN5 as a molecular biomarker of pan-cancer species by integrated multi-omics analysis. Discov Oncol 2025; 16:85. [PMID: 39862327 PMCID: PMC11762033 DOI: 10.1007/s12672-025-01840-9] [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: 11/24/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
INTRODUCTION We conducted a panoramic analysis of GBN5 expression and prognosis in 33 cancers, aiming to deepen the systematic understanding of GBN5 in cancer. MATERIALS AND METHODS We employed a multi-omics approach, including transcriptomic, genomic, proteomic, single-cell cytomic, spatial transcriptomic, and genomic data, to explore the prognostic value and potential oncogenic mechanisms of GBN5 across pan-cancers from multiple perspectives. RESULTS We found that GBN5 was differentially expressed in multiple tumors and showed early diagnostic value. Mutations, somatic copy number alterations, and DNA methylation lead to its aberrant expression. GBN5 expression is associated with many clinical features. GBN5 expression has been validated to be associated with many metabolic, metastatic, and immune-related pathways. We also demonstrated that GBN5 expression was significantly associated with immunomodulatory molecules and biomarkers of lymphocyte subpopulation infiltration. Methylation levels of GBN5 expression were significantly negatively correlated in a variety of tumors, and GBN5 missense mutations were the predominant SNP type in pan-cancer. In addition, GBN5 was positively correlated with multiple genomic scores, implying that higher GBN5 expression tends to imply that patients have higher chromosomal instability. More importantly, GBN5 has an important role in predicting drug sensitivity. We have also developed effective targeted drugs against GBN5. CONCLUSION GBN5 plays an important role in the genesis and progression of various tumors and is a potential tumor diagnostic and prognostic biomarker as well as an anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinxin Zhong
- School of Integrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing, Jiangsu, China
| | - Zihan Dang
- Department of Health Studies and Applied Educational Psychology, Columbia University, New York, NY, USA
| | - Baiquan Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zixin Yang
- Second Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
2
|
Siri-Angkul N, Kamp TJ. Cardiac L-type calcium channel regulation by Leucine-Rich Repeat-Containing Protein 10. Channels (Austin) 2024; 18:2355121. [PMID: 38762910 PMCID: PMC11110685 DOI: 10.1080/19336950.2024.2355121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024] Open
Abstract
L-type calcium channels (LTCCs), the major portal for Ca2+ entry into cardiomyocytes, are essential for excitation-contraction coupling and thus play a central role in regulating overall cardiac function. LTCC function is finely tuned by multiple signaling pathways and accessory proteins. Leucine-rich repeat-containing protein 10 (LRRC10) is a little studied cardiomyocyte-specific protein recently identified as a modulator of LTCCs. LRRC10 exerts a remarkable effect on LTCC function, more than doubling L-type Ca2+ current (ICa,L) amplitude in a heterologous expression system by altering the gating of the channels without changing their surface membrane expression. Genetic ablation of LRRC10 expression in mouse and zebrafish hearts leads to a significant reduction in ICa,L density and a slowly progressive dilated cardiomyopathy in mice. Rare sequence variants of LRRC10 have been identified in dilated cardiomyopathy and sudden unexplained nocturnal cardiac death syndrome, but these variants have not been clearly linked to disease. Nevertheless, the DCM-associated variant, I195T, converted LRRC10 from a ICa,L potentiator to a ICa,L suppressor, thus illustrating the wide dynamic range of LRRC10-mediated ICa,L regulation. This review focuses on the contemporary knowledge of LTCC modulation by LRRC10 and discusses potential directions for future investigations.
Collapse
Affiliation(s)
- Natthaphat Siri-Angkul
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Timothy J Kamp
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - Madison, Madison, WI, USA
| |
Collapse
|
3
|
Zhang J, Pandey M, Awe A, Lue N, Kittock C, Fikse E, Degner K, Staples J, Mokhasi N, Chen W, Yang Y, Adikaram P, Jacob N, Greenfest-Allen E, Thomas R, Bomeny L, Zhang Y, Petros TJ, Wang X, Li Y, Simonds WF. The association of GNB5 with Alzheimer disease revealed by genomic analysis restricted to variants impacting gene function. Am J Hum Genet 2024; 111:473-486. [PMID: 38354736 PMCID: PMC10940018 DOI: 10.1016/j.ajhg.2024.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Disease-associated variants identified from genome-wide association studies (GWASs) frequently map to non-coding areas of the genome such as introns and intergenic regions. An exclusive reliance on gene-agnostic methods of genomic investigation could limit the identification of relevant genes associated with polygenic diseases such as Alzheimer disease (AD). To overcome such potential restriction, we developed a gene-constrained analytical method that considers only moderate- and high-risk variants that affect gene coding sequences. We report here the application of this approach to publicly available datasets containing 181,388 individuals without and with AD and the resulting identification of 660 genes potentially linked to the higher AD prevalence among Africans/African Americans. By integration with transcriptome analysis of 23 brain regions from 2,728 AD case-control samples, we concentrated on nine genes that potentially enhance the risk of AD: AACS, GNB5, GNS, HIPK3, MED13, SHC2, SLC22A5, VPS35, and ZNF398. GNB5, the fifth member of the heterotrimeric G protein beta family encoding Gβ5, is primarily expressed in neurons and is essential for normal neuronal development in mouse brain. Homozygous or compound heterozygous loss of function of GNB5 in humans has previously been associated with a syndrome of developmental delay, cognitive impairment, and cardiac arrhythmia. In validation experiments, we confirmed that Gnb5 heterozygosity enhanced the formation of both amyloid plaques and neurofibrillary tangles in the brains of AD model mice. These results suggest that gene-constrained analysis can complement the power of GWASs in the identification of AD-associated genes and may be more broadly applicable to other polygenic diseases.
Collapse
Affiliation(s)
- Jianhua Zhang
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Mritunjay Pandey
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adam Awe
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Lue
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Claire Kittock
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emma Fikse
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine Degner
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jenna Staples
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neha Mokhasi
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiping Chen
- Genomic Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bldg. 8/Rm 1A11, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yanqin Yang
- Laboratory of Transplantation Genomics, National Heart Lung and Blood Institute, Bldg. 10/Rm 7S261, National Institutes of Health, Bethesda, MD 20892, USA
| | - Poorni Adikaram
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nirmal Jacob
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emily Greenfest-Allen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachel Thomas
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura Bomeny
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yajun Zhang
- Unit on Cellular and Molecular Neurodevelopment, Bldg. 35/Rm 3B 1002, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, Bldg. 35/Rm 3B 1002, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaowen Wang
- Partek Incorporated, 12747 Olive Boulevard, St. Louis, MO 63141, USA
| | - Yulong Li
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - William F Simonds
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
4
|
Padda I, Sebastian SA, Khehra N, Mahtani A, Sethi Y, Panthangi V, Fulton M, Bandyopadhyay D, Johal G. Tachy-brady syndrome: Electrophysiology and evolving principles of management. Dis Mon 2024; 70:101637. [PMID: 37690863 DOI: 10.1016/j.disamonth.2023.101637] [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] [Indexed: 09/12/2023]
Abstract
Sudden alterations in the heart rate may be associated with diverse symptoms. Sinus node dysfunction (SND), also known as sick sinus syndrome, is a sinoatrial (SA) node disorder. SND is primarily caused by the dysfunction of the pacemaker, as well as impaired impulse transmission resulting in a multitude of abnormalities in the heart rhythms, such as bradycardia-tachycardia, atrial bradyarrhythmias, and atrial tachyarrhythmias. The transition from bradycardia to tachycardia is generally referred to as "tachy-brady syndrome" (TBS). Although TBS is etiologically variable, the manifestations remain consistent throughout. Abnormal heart rhythms have the propensity to limit tissue perfusion resulting in palpitations, fatigue, lightheadedness, presyncope, and syncope. In this review, we examine the physiology of tachy-brady syndrome, the practical approach to its diagnosis and management, and the role of adenosine in treating SND.
Collapse
Affiliation(s)
- Inderbir Padda
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, NY, USA.
| | | | - Nimrat Khehra
- Saint James School of Medicine, Arnos Vale, Saint Vincent and the Grenadines
| | - Arun Mahtani
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, NY, USA
| | - Yashendra Sethi
- Department of Internal Medicine, Government Doon Medical College, Dehradun, India
| | | | - Matthew Fulton
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, NY, USA
| | | | - Gurpreet Johal
- Department of Cardiology, University of Washington, Valley Medical Center, Seattle, WA, USA
| |
Collapse
|
5
|
Peña-Martínez EG, Pomales-Matos DA, Rivera-Madera A, Messon-Bird JL, Medina-Feliciano JG, Sanabria-Alberto L, Barreiro-Rosario AC, Rivera-Del Valle J, Rodríguez-Ríos JM, Rodríguez-Martínez JA. Prioritizing cardiovascular disease-associated variants altering NKX2-5 and TBX5 binding through an integrative computational approach. J Biol Chem 2023; 299:105423. [PMID: 37926287 PMCID: PMC10750078 DOI: 10.1016/j.jbc.2023.105423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide and are heavily influenced by genetic factors. Genome-wide association studies have mapped >90% of CVD-associated variants within the noncoding genome, which can alter the function of regulatory proteins, such as transcription factors (TFs). However, due to the overwhelming number of single-nucleotide polymorphisms (SNPs) (>500,000) in genome-wide association studies, prioritizing variants for in vitro analysis remains challenging. In this work, we implemented a computational approach that considers support vector machine (SVM)-based TF binding site classification and cardiac expression quantitative trait loci (eQTL) analysis to identify and prioritize potential CVD-causing SNPs. We identified 1535 CVD-associated SNPs within TF footprints and putative cardiac enhancers plus 14,218 variants in linkage disequilibrium with genotype-dependent gene expression in cardiac tissues. Using ChIP-seq data from two cardiac TFs (NKX2-5 and TBX5) in human-induced pluripotent stem cell-derived cardiomyocytes, we trained a large-scale gapped k-mer SVM model to identify CVD-associated SNPs that altered NKX2-5 and TBX5 binding. The model was tested by scoring human heart TF genomic footprints within putative enhancers and measuring in vitro binding through electrophoretic mobility shift assay. Five variants predicted to alter NKX2-5 (rs59310144, rs6715570, and rs61872084) and TBX5 (rs7612445 and rs7790964) binding were prioritized for in vitro validation based on the magnitude of the predicted change in binding and are in cardiac tissue eQTLs. All five variants altered NKX2-5 and TBX5 DNA binding. We present a bioinformatic approach that considers tissue-specific eQTL analysis and SVM-based TF binding site classification to prioritize CVD-associated variants for in vitro analysis.
Collapse
Affiliation(s)
- Edwin G Peña-Martínez
- Department of Biology, University of Puerto Rico Río Piedras Campus, San Juan, Puerto Rico
| | - Diego A Pomales-Matos
- Department of Biology, University of Puerto Rico Río Piedras Campus, San Juan, Puerto Rico
| | | | - Jean L Messon-Bird
- Department of Biology, University of Puerto Rico Río Piedras Campus, San Juan, Puerto Rico
| | | | | | | | | | | | | |
Collapse
|
6
|
Wang S, Sass MI, Kwon Y, Ludlam WG, Smith TM, Carter EJ, Gladden NE, Riggi M, Iwasa JH, Willardson BM, Shen PS. Visualizing the chaperone-mediated folding trajectory of the G protein β5 β-propeller. Mol Cell 2023; 83:3852-3868.e6. [PMID: 37852256 PMCID: PMC10841713 DOI: 10.1016/j.molcel.2023.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/24/2023] [Accepted: 09/26/2023] [Indexed: 10/20/2023]
Abstract
The Chaperonin Containing Tailless polypeptide 1 (CCT) complex is an essential protein folding machine with a diverse clientele of substrates, including many proteins with β-propeller domains. Here, we determine the structures of human CCT in complex with its accessory co-chaperone, phosducin-like protein 1 (PhLP1), in the process of folding Gβ5, a component of Regulator of G protein Signaling (RGS) complexes. Cryoelectron microscopy (cryo-EM) and image processing reveal an ensemble of distinct snapshots that represent the folding trajectory of Gβ5 from an unfolded molten globule to a fully folded β-propeller. These structures reveal the mechanism by which CCT directs Gβ5 folding through initiating specific intermolecular contacts that facilitate the sequential folding of individual β sheets until the propeller closes into its native structure. This work directly visualizes chaperone-mediated protein folding and establishes that CCT orchestrates folding by stabilizing intermediates through interactions with surface residues that permit the hydrophobic core to coalesce into its folded state.
Collapse
Affiliation(s)
- Shuxin Wang
- Department of Biochemistry, School of Medicine, University of Utah, 15 N. Medical Drive East, Salt Lake City, UT 84112, USA
| | - Mikaila I Sass
- Department of Chemistry and Biochemistry, Brigham Young University, C100 BNSN, Provo, UT 84602, USA
| | - Yujin Kwon
- Department of Chemistry and Biochemistry, Brigham Young University, C100 BNSN, Provo, UT 84602, USA
| | - W Grant Ludlam
- Department of Chemistry and Biochemistry, Brigham Young University, C100 BNSN, Provo, UT 84602, USA
| | - Theresa M Smith
- Department of Chemistry and Biochemistry, Brigham Young University, C100 BNSN, Provo, UT 84602, USA
| | - Ethan J Carter
- Department of Chemistry and Biochemistry, Brigham Young University, C100 BNSN, Provo, UT 84602, USA
| | - Nathan E Gladden
- Department of Chemistry and Biochemistry, Brigham Young University, C100 BNSN, Provo, UT 84602, USA
| | - Margot Riggi
- Department of Biochemistry, School of Medicine, University of Utah, 15 N. Medical Drive East, Salt Lake City, UT 84112, USA
| | - Janet H Iwasa
- Department of Biochemistry, School of Medicine, University of Utah, 15 N. Medical Drive East, Salt Lake City, UT 84112, USA
| | - Barry M Willardson
- Department of Chemistry and Biochemistry, Brigham Young University, C100 BNSN, Provo, UT 84602, USA.
| | - Peter S Shen
- Department of Biochemistry, School of Medicine, University of Utah, 15 N. Medical Drive East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
7
|
Yuan J, Wang G, Zhao L, Kitchener AC, Sun T, Chen W, Huang C, Wang C, Xu X, Wang J, Lu H, Xu L, Jiangzuo Q, Murphy WJ, Wu D, Li G. How genomic insights into the evolutionary history of clouded leopards inform their conservation. SCIENCE ADVANCES 2023; 9:eadh9143. [PMID: 37801506 PMCID: PMC10558132 DOI: 10.1126/sciadv.adh9143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/06/2023] [Indexed: 10/08/2023]
Abstract
Clouded leopards (Neofelis spp.), a morphologically and ecologically distinct lineage of big cats, are severely threatened by habitat loss and fragmentation, targeted hunting, and other human activities. The long-held poor understanding of their genetics and evolution has undermined the effectiveness of conservation actions. Here, we report a comprehensive investigation of the whole genomes, population genetics, and adaptive evolution of Neofelis. Our results indicate the genus Neofelis arose during the Pleistocene, coinciding with glacial-induced climate changes to the distributions of savannas and rainforests, and signatures of natural selection associated with genes functioning in tooth, pigmentation, and tail development, associated with clouded leopards' unique adaptations. Our study highlights high-altitude adaptation as the main factor driving nontaxonomic population differentiation in Neofelis nebulosa. Population declines and inbreeding have led to reduced genetic diversity and the accumulation of deleterious variation that likely affect reproduction of clouded leopards, highlighting the urgent need for effective conservation efforts.
Collapse
Affiliation(s)
- Jiaqing Yuan
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Guiqiang Wang
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Le Zhao
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
- QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C., School of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Andrew C. Kitchener
- Department of Natural Sciences, National Museums Scotland, Chambers Street, Edinburgh EH1 1JF, UK
- School of Geosciences, University of Edinburgh, Drummond Street, Edinburgh EH9 3PX, UK
| | - Ting Sun
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Wu Chen
- Guangzhou Zoo, Guangzhou Wildlife Research Center, Guangzhou, China
| | - Chen Huang
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Chen Wang
- Guangzhou Zoo, Guangzhou Wildlife Research Center, Guangzhou, China
| | - Xiao Xu
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Jinhong Wang
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Huimeng Lu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Lulu Xu
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Qigao Jiangzuo
- Key Laboratory of Vertebrate Evolution and Human Origins of Chinese Academy of Sciences, Institute of Vertebrate Paleontology and Paleoanthropology, Chinese Academy of Sciences, Beijing, China
| | - William J. Murphy
- Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Dongdong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Natural History Museum of Zoology Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Gang Li
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
- Guangzhou Zoo, Guangzhou Wildlife Research Center, Guangzhou, China
| |
Collapse
|
8
|
Wang S, Sass MI, Kwon Y, Ludlam WG, Smith TM, Carter EJ, Gladden NE, Riggi M, Iwasa JH, Willardson BM, Shen PS. Visualizing the chaperone-mediated folding trajectory of the G protein β5 β-propeller. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.539424. [PMID: 37205387 PMCID: PMC10187262 DOI: 10.1101/2023.05.04.539424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The cytosolic Chaperonin Containing Tailless polypeptide 1 (CCT) complex is an essential protein folding machine with a diverse clientele of substrates, including many proteins with β-propeller domains. Here, we determined structures of CCT in complex with its accessory co-chaperone, phosducin-like protein 1 (PhLP1), in the process of folding Gβ5, a component of Regulator of G protein Signaling (RGS) complexes. Cryo-EM and image processing revealed an ensemble of distinct snapshots that represent the folding trajectory of Gβ5 from an unfolded molten globule to a fully folded β-propeller. These structures reveal the mechanism by which CCT directs Gβ5 folding through initiating specific intermolecular contacts that facilitate the sequential folding of individual β-sheets until the propeller closes into its native structure. This work directly visualizes chaperone-mediated protein folding and establishes that CCT directs folding by stabilizing intermediates through interactions with surface residues that permit the hydrophobic core to coalesce into its folded state.
Collapse
Affiliation(s)
- Shuxin Wang
- Department of Biochemistry, 15 N. Medical Drive East, University of Utah, Salt Lake City, UT, 84112, USA
| | - Mikaila I. Sass
- Department of Chemistry and Biochemistry, C100 BNSN, Brigham Young University, Provo, UT, 84602, USA
| | - Yujin Kwon
- Department of Chemistry and Biochemistry, C100 BNSN, Brigham Young University, Provo, UT, 84602, USA
| | - W. Grant Ludlam
- Department of Chemistry and Biochemistry, C100 BNSN, Brigham Young University, Provo, UT, 84602, USA
| | - Theresa M. Smith
- Department of Chemistry and Biochemistry, C100 BNSN, Brigham Young University, Provo, UT, 84602, USA
| | - Ethan J. Carter
- Department of Chemistry and Biochemistry, C100 BNSN, Brigham Young University, Provo, UT, 84602, USA
| | - Nathan E. Gladden
- Department of Chemistry and Biochemistry, C100 BNSN, Brigham Young University, Provo, UT, 84602, USA
| | - Margot Riggi
- Department of Biochemistry, 15 N. Medical Drive East, University of Utah, Salt Lake City, UT, 84112, USA
| | - Janet H. Iwasa
- Department of Biochemistry, 15 N. Medical Drive East, University of Utah, Salt Lake City, UT, 84112, USA
| | - Barry M. Willardson
- Department of Chemistry and Biochemistry, C100 BNSN, Brigham Young University, Provo, UT, 84602, USA
| | - Peter S. Shen
- Department of Biochemistry, 15 N. Medical Drive East, University of Utah, Salt Lake City, UT, 84112, USA
| |
Collapse
|
9
|
Coppola A, Lombari P, Mazzella E, Capolongo G, Simeoni M, Perna AF, Ingrosso D, Borriello M. Zebrafish as a Model of Cardiac Pathology and Toxicity: Spotlight on Uremic Toxins. Int J Mol Sci 2023; 24:ijms24065656. [PMID: 36982730 PMCID: PMC10052014 DOI: 10.3390/ijms24065656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health care problem. About 10% of the general population is affected by CKD, representing the sixth cause of death in the world. Cardiovascular events are the main mortality cause in CKD, with a cardiovascular risk 10 times higher in these patients than the rate observed in healthy subjects. The gradual decline of the kidney leads to the accumulation of uremic solutes with a negative effect on every organ, especially on the cardiovascular system. Mammalian models, sharing structural and functional similarities with humans, have been widely used to study cardiovascular disease mechanisms and test new therapies, but many of them are rather expensive and difficult to manipulate. Over the last few decades, zebrafish has become a powerful non-mammalian model to study alterations associated with human disease. The high conservation of gene function, low cost, small size, rapid growth, and easiness of genetic manipulation are just some of the features of this experimental model. More specifically, embryonic cardiac development and physiological responses to exposure to numerous toxin substances are similar to those observed in mammals, making zebrafish an ideal model to study cardiac development, toxicity, and cardiovascular disease.
Collapse
Affiliation(s)
- Annapaola Coppola
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Patrizia Lombari
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Elvira Mazzella
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Giovanna Capolongo
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Mariadelina Simeoni
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alessandra F. Perna
- Department of Translational Medical Science, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Diego Ingrosso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Margherita Borriello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence:
| |
Collapse
|
10
|
Manoj P, Kim JA, Kim S, Li T, Sewani M, Chelu MG, Li N. Sinus node dysfunction: current understanding and future directions. Am J Physiol Heart Circ Physiol 2023; 324:H259-H278. [PMID: 36563014 PMCID: PMC9886352 DOI: 10.1152/ajpheart.00618.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
The sinoatrial node (SAN) is the primary pacemaker of the heart. Normal SAN function is crucial in maintaining proper cardiac rhythm and contraction. Sinus node dysfunction (SND) is due to abnormalities within the SAN, which can affect the heartbeat frequency, regularity, and the propagation of electrical pulses through the cardiac conduction system. As a result, SND often increases the risk of cardiac arrhythmias. SND is most commonly seen as a disease of the elderly given the role of degenerative fibrosis as well as other age-dependent changes in its pathogenesis. Despite the prevalence of SND, current treatment is limited to pacemaker implantation, which is associated with substantial medical costs and complications. Emerging evidence has identified various genetic abnormalities that can cause SND, shedding light on the molecular underpinnings of SND. Identification of these molecular mechanisms and pathways implicated in the pathogenesis of SND is hoped to identify novel therapeutic targets for the development of more effective therapies for this disease. In this review article, we examine the anatomy of the SAN and the pathophysiology and epidemiology of SND. We then discuss in detail the most common genetic mutations correlated with SND and provide our perspectives on future research and therapeutic opportunities in this field.
Collapse
Affiliation(s)
- Pavan Manoj
- School of Public Health, Texas A&M University, College Station, Texas
| | - Jitae A Kim
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Stephanie Kim
- Department of BioSciences, Rice University, Houston, Texas
| | - Tingting Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Maham Sewani
- Department of BioSciences, Rice University, Houston, Texas
| | - Mihail G Chelu
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Na Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
11
|
Yu C, Deng XJ, Xu D. Gene mutations in comorbidity of epilepsy and arrhythmia. J Neurol 2023; 270:1229-1248. [PMID: 36376730 DOI: 10.1007/s00415-022-11430-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022]
Abstract
Epilepsy is one of the most common neurological disorders, and sudden unexpected death in epilepsy (SUDEP) is the most severe outcome of refractory epilepsy. Arrhythmia is one of the heterogeneous factors in the pathophysiological mechanism of SUDEP with a high incidence in patients with refractory epilepsy, increasing the risk of premature death. The gene co-expressed in the brain and heart is supposed to be the genetic basis between epilepsy and arrhythmia, among which the gene encoding ion channel contributes to the prevalence of "cardiocerebral channelopathy" theory. Nevertheless, this theory could only explain the molecular mechanism of comorbid arrhythmia in part of patients with epilepsy (PWE). Therefore, we summarized the mutant genes that can induce comorbidity of epilepsy and arrhythmia and the possible corresponding treatments. These variants involved the genes encoding sodium, potassium, calcium and HCN channels, as well as some non-ion channel coding genes such as CHD4, PKP2, FHF1, GNB5, and mitochondrial genes. The relationship between genotype and clinical phenotype was not simple linear. Indeed, genes co-expressed in the brain and heart could independently induce epilepsy and/or arrhythmia. Mutant genes in brain could affect cardiac rhythm through central or peripheral regulation, while in the heart it could also affect cerebral electrical activity by changing the hemodynamics or internal environment. Analysis of mutations in comorbidity of epilepsy and arrhythmia could refine and expand the theory of "cardiocerebral channelopathy" and provide new insights for risk stratification of premature death and corresponding precision therapy in PWE.
Collapse
Affiliation(s)
- Cheng Yu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Xue-Jun Deng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Da Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
12
|
Burglen L, Van Hoeymissen E, Qebibo L, Barth M, Belnap N, Boschann F, Depienne C, De Clercq K, Douglas AGL, Fitzgerald MP, Foulds N, Garel C, Helbig I, Held K, Horn D, Janssen A, Kaindl AM, Narayanan V, Prager C, Rupin-Mas M, Afenjar A, Zhao S, Ramaekers VT, Ruggiero SM, Thomas S, Valence S, Van Maldergem L, Rohacs T, Rodriguez D, Dyment D, Voets T, Vriens J. Gain-of-function variants in the ion channel gene TRPM3 underlie a spectrum of neurodevelopmental disorders. eLife 2023; 12:81032. [PMID: 36648066 PMCID: PMC9886277 DOI: 10.7554/elife.81032] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
TRPM3 is a temperature- and neurosteroid-sensitive plasma membrane cation channel expressed in a variety of neuronal and non-neuronal cells. Recently, rare de novo variants in TRPM3 were identified in individuals with developmental and epileptic encephalopathy, but the link between TRPM3 activity and neuronal disease remains poorly understood. We previously reported that two disease-associated variants in TRPM3 lead to a gain of channel function . Here, we report a further 10 patients carrying one of seven additional heterozygous TRPM3 missense variants. These patients present with a broad spectrum of neurodevelopmental symptoms, including global developmental delay, intellectual disability, epilepsy, musculo-skeletal anomalies, and altered pain perception. We describe a cerebellar phenotype with ataxia or severe hypotonia, nystagmus, and cerebellar atrophy in more than half of the patients. All disease-associated variants exhibited a robust gain-of-function phenotype, characterized by increased basal activity leading to cellular calcium overload and by enhanced responses to the neurosteroid ligand pregnenolone sulfate when co-expressed with wild-type TRPM3 in mammalian cells. The antiseizure medication primidone, a known TRPM3 antagonist, reduced the increased basal activity of all mutant channels. These findings establish gain-of-function of TRPM3 as the cause of a spectrum of autosomal dominant neurodevelopmental disorders with frequent cerebellar involvement in humans and provide support for the evaluation of TRPM3 antagonists as a potential therapy.
Collapse
Affiliation(s)
- Lydie Burglen
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Developmental Brain Disorders Laboratory, Imagine InstituteParisFrance
| | - Evelien Van Hoeymissen
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| | - Leila Qebibo
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
| | - Magalie Barth
- Department of Genetics, University Hospital of AngersAngersFrance
| | - Newell Belnap
- Translational Genomics Research Institute (TGen), Neurogenomics Division, Center for Rare Childhood DisordersPhoenixUnited States
| | - Felix Boschann
- Charité – Universitäts medizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Genetics and Human GeneticsBerlinGermany
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University Duisburg-EssenEssenGermany
| | - Katrien De Clercq
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| | - Andrew GL Douglas
- University Hospital Southampton NHS Foundation TrustSouthamptonUnited Kingdom
| | | | - Nicola Foulds
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation TrustSouthamptonUnited Kingdom
| | - Catherine Garel
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Service de Radiologie Pédiatrique, Hôpital Armand-Trousseau, Médecine Sorbonne UniversitéParisFrance
| | - Ingo Helbig
- Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Katharina Held
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| | - Denise Horn
- Charité – Universitäts medizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Genetics and Human GeneticsBerlinGermany
| | - Annelies Janssen
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
| | - Angela M Kaindl
- Institute of Cell Biology and Neurobiology, Charité - Universitäts medizin BerlinBerlinGermany
- Department of Pediatric Neurology, Charité - Universitäts medizin BerlinBerlinGermany
- Charité – Universitäts medizin Berlin, Center for Chronically Sick ChildrenBerlinGermany
| | - Vinodh Narayanan
- Translational Genomics Research Institute (TGen), Neurogenomics Division, Center for Rare Childhood DisordersPhoenixUnited States
| | - Christina Prager
- Department of Pediatric Neurology, Charité - Universitäts medizin BerlinBerlinGermany
- Charité – Universitäts medizin Berlin, Center for Chronically Sick ChildrenBerlinGermany
| | - Mailys Rupin-Mas
- Department of Neuropediatrics, University Hospital of AngersAngersFrance
| | - Alexandra Afenjar
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
| | - Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New JerseyNewarkUnited States
| | | | | | - Simon Thomas
- Wessex Regional Genetics Laboratory, Salisbury District HospitalSalisburyUnited Kingdom
| | - Stéphanie Valence
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Sorbonne Université, Service de Neuropédiatrie, Hôpital Trousseau AP-HPParisFrance
| | - Lionel Van Maldergem
- Centre de Génétique Humaine, Université de Franche-Comté BesançonBesanconFrance
- Center of Clinical Investigation 1431, National Institute of Health and Medical ResearchBesanconFrance
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New JerseyNewarkUnited States
| | - Diana Rodriguez
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Sorbonne Université, Service de Neuropédiatrie, Hôpital Trousseau AP-HPParisFrance
| | - David Dyment
- Children's Hospital of Eastern Ontario Research Institute, University of OttawaOttawaCanada
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| |
Collapse
|
13
|
Zhang D, Zheng C, Zhu T, Yang F, Zhou Y. Identification of key module and hub genes in pulpitis using weighted gene co-expression network analysis. BMC Oral Health 2023; 23:2. [PMID: 36593446 PMCID: PMC9808982 DOI: 10.1186/s12903-022-02638-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/30/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Pulpitis is a common disease mainly caused by bacteria. Conventional approaches of diagnosing the state of dental pulp are mainly based on clinical symptoms, thereby harbor deficiencies. The accurate and rapid diagnosis of pulpitis is important for choosing the suitable therapy. The study aimed to identify pulpits related key genes by integrating micro-array data analysis and systems biology network-based methods such as weighted gene co-expression network analysis (WGCNA). METHODS The micro-array data of 13 inflamed pulp and 11 normal pulp were acquired from Gene Expression Omnibus (GEO). WGCNA was utilized to establish a genetic network and categorize genes into diverse modules. Hub genes in the most associated module to pulpitis were screened out using high module group members (MM) methods. Pulpitis model in rat was constructed and iRoot BP plus was applied to cap pulp. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used for validation of hub genes. RESULTS WGCNA was established and genes were categorized into 22 modules. The darkgrey module had the highest correlation with pulpitis among them. A total of 5 hub genes (HMOX1, LOX, ACTG1, STAT3, GNB5) were identified. RT-qPCR proved the differences in expression levels of HMOX1, LOX, ACTG1, STAT3, GNB5 in inflamed dental pulp. Pulp capping reversed the expression level of HMOX1, LOX, ACTG1. CONCLUSION The study was the first to produce a holistic view of pulpitis, screen out and validate hub genes involved in pulpitis using WGCNA method. Pulp capping using iRoot BP plus could reverse partial hub genes.
Collapse
Affiliation(s)
- Denghui Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Chen Zheng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Tianer Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Fan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Yiqun Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
14
|
Genome-wide DNA methylation profiling and exome sequencing resolved a long-time misdiagnosed case. J Hum Genet 2022; 67:547-551. [PMID: 35581385 DOI: 10.1038/s10038-022-01043-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/01/2022] [Accepted: 05/05/2022] [Indexed: 11/08/2022]
Abstract
The search for aetiology of Mendelian disorders is traditionally based on the observation of clinical phenotypes and molecular screening of associated genes. However, a disease-specific diagnosis can be challenging. In this study we detail how the combinatorial genomic and epigenomic assessment allowed to find the underlying molecular event of a clinical case that remained misdiagnosed for years. The individual was referred as affected by an atypical form of Kabuki syndrome with a variant of uncertain significance in the KMT2D gene. However, significant inconsistencies with this diagnosis emerged upon familial segregation of the variant and after the clinical re-evaluation. Therefore, we applied an epigenomic strategy by studying the DNA methylation profile which resulted not consistent with the Kabuki syndrome episignature or with any other disorder-specific episignature described so far, providing strong evidence that the Kabuki syndrome diagnosis does not apply. This result led us to further investigate for epigenetic machinery diseases by using a multigene panel for chromatinopathies. Since this analysis yielded negative results, we applied a whole exome sequencing and identified a de novo pathogenic variant in the CTNNB1 gene associated to NEDSDV syndrome, a neurodevelopmental disorder characterized by intellectual disability and craniofacial anomalies. Based on molecular results and the updated clinical features, we confirmed the NEDSDV diagnosis. Our findings show that the combination of genomic and epigenomics strategies, along with a deeper analysis of clinical phenotype, may provide a significant improvement in the diagnostic protocols for rare genetic disorders and help resolve long-time misdiagnosed and unsolved case.
Collapse
|
15
|
Sahly AN, Shevell M, Sadleir LG, Myers KA. SUDEP risk and autonomic dysfunction in genetic epilepsies. Auton Neurosci 2021; 237:102907. [PMID: 34773737 DOI: 10.1016/j.autneu.2021.102907] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 10/11/2021] [Accepted: 11/06/2021] [Indexed: 01/02/2023]
Abstract
The underlying pathophysiology of sudden unexpected death in epilepsy (SUDEP) remains unclear. This phenomenon is likely multifactorial, and there is considerable evidence that genetic factors play a role. There are certain genetic causes of epilepsy in which the risk of SUDEP appears to be increased relative to epilepsy overall. For individuals with pathogenic variants in genes including SCN1A, SCN1B, SCN8A, SCN2A, GNB5, KCNA1 and DEPDC5, there are varying degrees of evidence to suggest an increased risk for sudden death. Why the risk for sudden death is higher is not completely clear; however, in many cases pathogenic variants in these genes are also associated with autonomic dysfunction, which is hypothesized as a contributing factor to SUDEP. We review the evidence for increased SUDEP risk for patients with epilepsy due to pathogenic variants in these genes, and also discuss what is known about autonomic dysfunction in these contexts.
Collapse
Affiliation(s)
- Ahmed N Sahly
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurosciences, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Michael Shevell
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Research Institute of the McGill University Medical Centre, Montreal, Quebec, Canada
| | - Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Kenneth A Myers
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Research Institute of the McGill University Medical Centre, Montreal, Quebec, Canada.
| |
Collapse
|
16
|
Extended Phenotyping and Functional Validation Facilitate Diagnosis of a Complex Patient Harboring Genetic Variants in MCCC1 and GNB5 Causing Overlapping Phenotypes. Genes (Basel) 2021; 12:genes12091352. [PMID: 34573334 PMCID: PMC8469011 DOI: 10.3390/genes12091352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Identifying multiple ultra-rare genetic syndromes with overlapping phenotypes is a diagnostic conundrum in clinical genetics. This study investigated the pathogenicity of a homozygous missense variant in GNB5 (GNB5L; NM_016194.4: c.920T > G (p. Leu307Arg); GNB5S; NM_006578.4: c.794T > G (p. Leu265Arg)) identified through exome sequencing in a female child who also had 3-methylcrotonyl-CoA carboxylase (3-MCC) deficiency (newborn screening positive) and hemoglobin E trait. The proband presented with early-onset intellectual disability, the severity of which was more in keeping with GNB5-related disorder than 3-MCC deficiency. She later developed bradycardia and cardiac arrest, and upon re-phenotyping showed cone photo-transduction recovery deficit, all known only to GNB5-related disorders. Patient-derived fibroblast assays showed preserved GNB5S expression, but bioluminescence resonance energy transfer assay showed abolished function of the variant reconstituted Gβ5S containing RGS complexes for deactivation of D2 dopamine receptor activity, confirming variant pathogenicity. This study highlights the need for precise phenotyping and functional assays to facilitate variant classification and clinical diagnosis in patients with complex medical conditions.
Collapse
|
17
|
Thorolfsdottir RB, Sveinbjornsson G, Aegisdottir HM, Benonisdottir S, Stefansdottir L, Ivarsdottir EV, Halldorsson GH, Sigurdsson JK, Torp-Pedersen C, Weeke PE, Brunak S, Westergaard D, Pedersen OB, Sorensen E, Nielsen KR, Burgdorf KS, Banasik K, Brumpton B, Zhou W, Oddsson A, Tragante V, Hjorleifsson KE, Davidsson OB, Rajamani S, Jonsson S, Torfason B, Valgardsson AS, Thorgeirsson G, Frigge ML, Thorleifsson G, Norddahl GL, Helgadottir A, Gretarsdottir S, Sulem P, Jonsdottir I, Willer CJ, Hveem K, Bundgaard H, Ullum H, Arnar DO, Thorsteinsdottir U, Gudbjartsson DF, Holm H, Stefansson K. Genetic insight into sick sinus syndrome. Eur Heart J 2021; 42:1959-1971. [PMID: 36282123 PMCID: PMC8140484 DOI: 10.1093/eurheartj/ehaa1108] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/24/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
Aims The aim of this study was to use human genetics to investigate the pathogenesis of sick sinus syndrome (SSS) and the role of risk factors in its development. Methods and results We performed a genome-wide association study of 6469 SSS cases and 1 000 187 controls from deCODE genetics, the Copenhagen Hospital Biobank, UK Biobank, and the HUNT study. Variants at six loci associated with SSS, a reported missense variant in MYH6, known atrial fibrillation (AF)/electrocardiogram variants at PITX2, ZFHX3, TTN/CCDC141, and SCN10A and a low-frequency (MAF = 1.1–1.8%) missense variant, p.Gly62Cys in KRT8 encoding the intermediate filament protein keratin 8. A full genotypic model best described the p.Gly62Cys association (P = 1.6 × 10−20), with an odds ratio (OR) of 1.44 for heterozygotes and a disproportionally large OR of 13.99 for homozygotes. All the SSS variants increased the risk of pacemaker implantation. Their association with AF varied and p.Gly62Cys was the only variant not associating with any other arrhythmia or cardiovascular disease. We tested 17 exposure phenotypes in polygenic score (PGS) and Mendelian randomization analyses. Only two associated with the risk of SSS in Mendelian randomization, AF, and lower heart rate, suggesting causality. Powerful PGS analyses provided convincing evidence against causal associations for body mass index, cholesterol, triglycerides, and type 2 diabetes (P > 0.05). Conclusion We report the associations of variants at six loci with SSS, including a missense variant in KRT8 that confers high risk in homozygotes and points to a mechanism specific to SSS development. Mendelian randomization supports a causal role for AF in the development of SSS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jon K Sigurdsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Christian Torp-Pedersen
- Department of Clinical Research and Cardiology, Nordsjaelland Hospital, Dyrehavevej 29, Hillerød 3400, Denmark
| | - Peter E Weeke
- Department of Cardiology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - Ole B Pedersen
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 77B, Naestved 4700, Denmark
| | - Erik Sorensen
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Kaspar R Nielsen
- Department of Clinical Immunology, Aalborg University Hospital North, Urbansgade 36, Aalborg 9000, Denmark
| | - Kristoffer S Burgdorf
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, Copenhagen 2200, Denmark
| | - Ben Brumpton
- Department of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, Prinsesse Kristinas gate 3, Trondheim 7030, Norway
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109-2218, USA
| | - Asmundur Oddsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | - Kristjan E Hjorleifsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Department of Computing and Mathematical Sciences, California Institute of Technology, 1200 E California Blvd. MC 305-16, Pasadena, CA 91125, USA
| | | | | | - Stefan Jonsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Bjarni Torfason
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Cardiothoracic Surgery, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Atli S Valgardsson
- Department of Cardiothoracic Surgery, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Michael L Frigge
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | | | - Anna Helgadottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | | | - Patrick Sulem
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Immunology, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Cristen J Willer
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109-2218, USA.,Department of Internal Medicine: Cardiology, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109 -5368, USA.,Department of Human Genetics, University of Michigan, 4909 Buhl Building, 1241 E. Catherine St., Ann Arbor, MI 48109 -5618, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gt. 1, Trondheim 7491, Norway.,Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Postboks 8905, Trondheim 7491, Norway.,HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Forskningsveien 2, Levanger 7600, Norway
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen 2100, Denmark.,Statens Serum Institut, Artillerivej 5, Copenhagen 2300, Denmark
| | - David O Arnar
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland.,Department of Medicine, Landspitali-The National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Hjardarhagi 4, Reykjavik 107, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Sturlugata 8, Reykjavik 101, Iceland.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, Reykjavik 101, Iceland
| | | |
Collapse
|
18
|
Tennakoon M, Senarath K, Kankanamge D, Ratnayake K, Wijayaratna D, Olupothage K, Ubeysinghe S, Martins-Cannavino K, Hébert TE, Karunarathne A. Subtype-dependent regulation of Gβγ signalling. Cell Signal 2021; 82:109947. [PMID: 33582184 PMCID: PMC8026654 DOI: 10.1016/j.cellsig.2021.109947] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 01/04/2023]
Abstract
G protein-coupled receptors (GPCRs) transmit information to the cell interior by transducing external signals to heterotrimeric G protein subunits, Gα and Gβγ subunits, localized on the inner leaflet of the plasma membrane. Though the initial focus was mainly on Gα-mediated events, Gβγ subunits were later identified as major contributors to GPCR-G protein signalling. A broad functional array of Gβγ signalling has recently been attributed to Gβ and Gγ subtype diversity, comprising 5 Gβ and 12 Gγ subtypes, respectively. In addition to displaying selectivity towards each other to form the Gβγ dimer, numerous studies have identified preferences of distinct Gβγ combinations for specific GPCRs, Gα subtypes and effector molecules. Importantly, Gβ and Gγ subtype-dependent regulation of downstream effectors, representing a diverse range of signalling pathways and physiological functions have been found. Here, we review the literature on the repercussions of Gβ and Gγ subtype diversity on direct and indirect regulation of GPCR/G protein signalling events and their physiological outcomes. Our discussion additionally provides perspective in understanding the intricacies underlying molecular regulation of subtype-specific roles of Gβγ signalling and associated diseases.
Collapse
Affiliation(s)
- Mithila Tennakoon
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Kanishka Senarath
- Genetics and Molecular Biology Unit, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Dinesh Kankanamge
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Kasun Ratnayake
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dhanushan Wijayaratna
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Koshala Olupothage
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Sithurandi Ubeysinghe
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | | | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada.
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA.
| |
Collapse
|
19
|
Wallace MJ, El Refaey M, Mesirca P, Hund TJ, Mangoni ME, Mohler PJ. Genetic Complexity of Sinoatrial Node Dysfunction. Front Genet 2021; 12:654925. [PMID: 33868385 PMCID: PMC8047474 DOI: 10.3389/fgene.2021.654925] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The pacemaker cells of the cardiac sinoatrial node (SAN) are essential for normal cardiac automaticity. Dysfunction in cardiac pacemaking results in human sinoatrial node dysfunction (SND). SND more generally occurs in the elderly population and is associated with impaired pacemaker function causing abnormal heart rhythm. Individuals with SND have a variety of symptoms including sinus bradycardia, sinus arrest, SAN block, bradycardia/tachycardia syndrome, and syncope. Importantly, individuals with SND report chronotropic incompetence in response to stress and/or exercise. SND may be genetic or secondary to systemic or cardiovascular conditions. Current management of patients with SND is limited to the relief of arrhythmia symptoms and pacemaker implantation if indicated. Lack of effective therapeutic measures that target the underlying causes of SND renders management of these patients challenging due to its progressive nature and has highlighted a critical need to improve our understanding of its underlying mechanistic basis of SND. This review focuses on current information on the genetics underlying SND, followed by future implications of this knowledge in the management of individuals with SND.
Collapse
Affiliation(s)
- Michael J. Wallace
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mona El Refaey
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Pietro Mesirca
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Thomas J. Hund
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Matteo E. Mangoni
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Peter J. Mohler
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
20
|
Mesirca P, Fedorov VV, Hund TJ, Torrente AG, Bidaud I, Mohler PJ, Mangoni ME. Pharmacologic Approach to Sinoatrial Node Dysfunction. Annu Rev Pharmacol Toxicol 2021; 61:757-778. [PMID: 33017571 PMCID: PMC7790915 DOI: 10.1146/annurev-pharmtox-031120-115815] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The spontaneous activity of the sinoatrial node initiates the heartbeat. Sino-atrial node dysfunction (SND) and sick sinoatrial (sick sinus) syndrome are caused by the heart's inability to generate a normal sinoatrial node action potential. In clinical practice, SND is generally considered an age-related pathology, secondary to degenerative fibrosis of the heart pacemaker tissue. However, other forms of SND exist, including idiopathic primary SND, which is genetic, and forms that are secondary to cardiovascular or systemic disease. The incidence of SND in the general population is expected to increase over the next half century, boosting the need to implant electronic pacemakers. During the last two decades, our knowledge of sino-atrial node physiology and of the pathophysiological mechanisms underlying SND has advanced considerably. This review summarizes the current knowledge about SND mechanisms and discusses the possibility of introducing new pharmacologic therapies for treating SND.
Collapse
Affiliation(s)
- Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Vadim V Fedorov
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
| | - Thomas J Hund
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Peter J Mohler
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio 43210, USA
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| |
Collapse
|
21
|
Saidijam M, Afshar S, Taherkhani A. Identifying Potential Biomarkers in Colorectal Cancer and Developing Non-invasive Diagnostic Models Using Bioinformatics Approaches. AVICENNA JOURNAL OF MEDICAL BIOCHEMISTRY 2020. [DOI: 10.34172/ajmb.2020.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the most frequent causes of gastrointestinal tumors. Due to the invasiveness of the current diagnostic methods, there is an urgent need to develop non-invasive diagnostic approaches for CRC. The exact mechanisms and the most important genes associated with the development of CRC are not fully demonstrated. Objectives: This study aimed to identify differentially expressed miRNAs (DEMs), key genes, and their regulators associated with the pathogenesis of CRC. The signaling pathways and biological processes (BPs) that were significantly affected in CRC were also indicated. Moreover, two non-invasive models were constructed for CRC diagnosis. Methods: The miRNA dataset GSE59856 was downloaded from the Gene Expression Omnibus (GEO) database and analyzed to identify DEMs in CRC patients compared with healthy controls (HCs). A protein-protein interaction (PPI) network was built and analyzed. Significant clusters in the PPI networks were identified, and the BPs and pathways associated with these clusters were studied. The hub genes in the PPI network, as well as their regulators were identified. Results: A total of 569 DEMs were demonstrated with the criteria of P value <0.001. A total of 110 essential genes and 30 modules were identified in the PPI network. Functional analysis revealed that 1005 BPs, 9 molecular functions (MFs), 14 cellular components (CCs), and 887 pathways were significantly affected in CRC. A total of 22 transcription factors (TFs) were demonstrated as the regulators of the hubs. Conclusion: Our results may provide new insight into the pathogenesis of CRC and advance the diagnostic and therapeutic methods of the disease. However, confirmation is required in the future.
Collapse
Affiliation(s)
- Massoud Saidijam
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeid Afshar
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
22
|
Inhibition of G protein-gated K + channels by tertiapin-Q rescues sinus node dysfunction and atrioventricular conduction in mouse models of primary bradycardia. Sci Rep 2020; 10:9835. [PMID: 32555258 PMCID: PMC7300035 DOI: 10.1038/s41598-020-66673-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Sinus node (SAN) dysfunction (SND) manifests as low heart rate (HR) and is often accompanied by atrial tachycardia or atrioventricular (AV) block. The only currently available therapy for chronic SND is the implantation of an electronic pacemaker. Because of the growing burden of SND in the population, new pharmacological therapies of chronic SND and heart block are desirable. We developed a collection of genetically modified mouse strains recapitulating human primary SND associated with different degrees of AV block. These mice were generated with genetic ablation of L-type Cav1.3 (Cav1.3-/-), T-type Cav3.1 (Cav3.1-/-), or both (Cav1.3-/-/Cav3.1-/-). We also studied mice haplo-insufficient for the Na+ channel Nav1.5 (Nav1.5+/) and mice in which the cAMP-dependent regulation of hyperpolarization-activated f-(HCN4) channels has been abolished (HCN4-CNBD). We analysed, by telemetric ECG recording, whether pharmacological inhibition of the G-protein-activated K+ current (IKACh) by the peptide tertiapin-Q could improve HR and AV conduction in these mouse strains. Tertiapin-Q significantly improved the HR of Cav1.3-/- (19%), Cav1.3-/-/Cav3.1-/- (23%) and HCN4-CNBD (14%) mice. Tertiapin-Q also improved cardiac conduction of Nav1.5+/- mice by 24%. Our data suggest that the development of pharmacological IKACh inhibitors for the management of SND and conduction disease is a viable approach.
Collapse
|
23
|
Sciacca FL, Ciaccio C, Fontana F, Strano C, Gilardoni F, Pantaleoni C, D'Arrigo S. Severe Phenotype in a Patient With Homozygous 15q21.2 Microdeletion Involving BCL2L10, GNB5, and MYO5C Genes, Resembling Infantile Developmental Disorder With Cardiac Arrhythmias (IDDCA). Front Genet 2020; 11:399. [PMID: 32477400 PMCID: PMC7237723 DOI: 10.3389/fgene.2020.00399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/30/2020] [Indexed: 11/29/2022] Open
Abstract
Homozygous and compound heterozygous mutations in GNB5 gene have been associated with a wide spectrum of clinical presentations, ranging from neurodevelopmental issues with or without cardiac arrhythmia (LADCI) to severe developmental delay with epileptic encephalopathy, retinal dystrophy, and heart rhythm abnormalities (IDDCA). While missense or missense/non-sense mutations usually lead to milder form, the biallelic loss of function of GNB5 gene causes the severe multisystemic IDDCA phenotype. So far, only 27 patients have been described with GNB5-associated disease. We report the first case of a patient carrying a homozygous 15q21.2 microdeletion, encompassing GNB5 and the two contiguous genes BCL2L10 and MYO5C. The clinical features of the child are consistent with the severe IDDCA phenotype, thus confirming the GNB5 loss-of-function mechanism in determining such presentation of the disease.
Collapse
Affiliation(s)
- Francesca L Sciacca
- Neurological Biochemistry and Neuropharmacology Unit, Laboratory of Cytogenetic, Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Ciaccio
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Federica Fontana
- Neurological Biochemistry and Neuropharmacology Unit, Laboratory of Cytogenetic, Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Camilla Strano
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesca Gilardoni
- Neurological Biochemistry and Neuropharmacology Unit, Laboratory of Cytogenetic, Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Pantaleoni
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefano D'Arrigo
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
24
|
Del Caño-Ochoa F, Ng BG, Abedalthagafi M, Almannai M, Cohn RD, Costain G, Elpeleg O, Houlden H, Karimiani EG, Liu P, Manzini MC, Maroofian R, Muriello M, Al-Otaibi A, Patel H, Shimon E, Sutton VR, Toosi MB, Wolfe LA, Rosenfeld JA, Freeze HH, Ramón-Maiques S. Cell-based analysis of CAD variants identifies individuals likely to benefit from uridine therapy. Genet Med 2020; 22:1598-1605. [PMID: 32461667 PMCID: PMC7521996 DOI: 10.1038/s41436-020-0833-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Pathogenic autosomal recessive variants in CAD, encoding the multienzymatic protein initiating pyrimidine de novo biosynthesis, cause a severe inborn metabolic disorder treatable with a dietary supplement of uridine. This condition is difficult to diagnose given the large size of CAD with over 1000 missense variants and the nonspecific clinical presentation. We aimed to develop a reliable and discerning assay to assess the pathogenicity of CAD variants and to select affected individuals that might benefit from uridine therapy. Methods Using CRISPR/Cas9, we generated a human CAD-knockout cell line that requires uridine supplements for survival. Transient transfection of the knockout cells with recombinant CAD restores growth in absence of uridine. This system determines missense variants that inactivate CAD and do not rescue the growth phenotype. Results We identified 25 individuals with biallelic variants in CAD and a phenotype consistent with a CAD deficit. We used the CAD-knockout complementation assay to test a total of 34 variants, identifying 16 as deleterious for CAD activity. Combination of these pathogenic variants confirmed 11 subjects with a CAD deficit, for whom we describe the clinical phenotype. Conclusions We designed a cell-based assay to test the pathogenicity of CAD variants, identifying 11 CAD-deficient individuals who could benefit from uridine therapy.
Collapse
Affiliation(s)
- Francisco Del Caño-Ochoa
- Genome Dynamics and Function Program, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Group 739, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-Instituto de Salud Carlos III, Valencia, Spain.,Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| | - Bobby G Ng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Malak Abedalthagafi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Mohammed Almannai
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ronald D Cohn
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Paediatric Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Paediatrics, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gregory Costain
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada.,Centre for Genetic Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Orly Elpeleg
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Henry Houlden
- Department of Neuromuscular disorders, UCL Institute of Neurology University College, London, UK
| | - Ehsan Ghayoor Karimiani
- Molecular and Clinical Sciences Institute, St. George's, University of London, Cranmer Terrace, London, UK
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Baylor Genetics Laboratories, Houston, TX, USA
| | - M Chiara Manzini
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Reza Maroofian
- Department of Neuromuscular disorders, UCL Institute of Neurology University College, London, UK
| | - Michael Muriello
- Department of Pediatrics/Division of Genetics, Medical College of Wisconsin, Milwaukee, WI, USA.,Genomic Science and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ali Al-Otaibi
- Department of Pediatric Neurology, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Hema Patel
- Department of Neurology (Section of pediatric neurology) Children's Hospital of Wisconsin, Medical of College of Wisconsin, Milwaukee, WI, USA
| | - Edvardson Shimon
- Pediatric Neurology Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - V Reid Sutton
- Department of Molecular, Human Genetics Baylor College of Medicine & Texas Children's Hospital, Houston, TX, USA
| | - Mehran Beiraghi Toosi
- Department of Pediatric Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Lynne A Wolfe
- Undiagnosed Diseases Program, Common Fund, National Institutes of Health, Bethesda, MD, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Baylor Genetics Laboratories, Houston, TX, USA
| | - Hudson H Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Santiago Ramón-Maiques
- Genome Dynamics and Function Program, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain. .,Group 739, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-Instituto de Salud Carlos III, Valencia, Spain. .,Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain.
| |
Collapse
|
25
|
Yazdani S, Badjatiya A, Dorrani N, Lee H, Grody WW, Nelson SF, Dipple KM. Genetic characterization and long-term management of severely affected siblings with intellectual developmental disorder with cardiac arrhythmia syndrome. Mol Genet Metab Rep 2020; 23:100582. [PMID: 32280589 PMCID: PMC7138921 DOI: 10.1016/j.ymgmr.2020.100582] [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: 02/03/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 11/30/2022] Open
Abstract
We report two brothers with severe global cognitive and motor delay, cortical visual impairment and sick sinus syndrome who were born to consanguineous parents. Standard genetic evaluations did not reveal the cause of their mental retardation. As expected, chromosomal microarray (CMA) revealed extensive regions of homozygosity. Exome sequencing revealed that both affected boys were homozygous for a nonsense mutation in the G-protein β5 (GNB5) gene (NM_016194.3:c.1032C > G; Tyr344Ter), and that the parents were carriers of this mutation. No other DNA variants that were explanatory for the sick sinus or the developmental delay/intellectual disability were identified, and no other clinical parameters are likely to have contributed to this unusual combination of phenotypes. The neurologic features of our patients are more severe than those of most of the other patients previously reported with GNB5 variants, probably because of the homozygous, complete loss-of-function (nonsense/stop-gain) nature of their variant, and their clinical course has been monitored for longer duration.
Collapse
Affiliation(s)
- Shahram Yazdani
- Departments of Pediatrics and Mattel Children's Hospital at UCLA, Los Angeles, CA, United States of America
| | - Anish Badjatiya
- Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Naghmeh Dorrani
- Departments of Pediatrics and Mattel Children's Hospital at UCLA, Los Angeles, CA, United States of America
| | - Hane Lee
- Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America.,Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Wayne W Grody
- Departments of Pediatrics and Mattel Children's Hospital at UCLA, Los Angeles, CA, United States of America.,Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America.,Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Stanley F Nelson
- Departments of Pediatrics and Mattel Children's Hospital at UCLA, Los Angeles, CA, United States of America.,Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America.,Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Katrina M Dipple
- Departments of Pediatrics and Mattel Children's Hospital at UCLA, Los Angeles, CA, United States of America.,Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| |
Collapse
|
26
|
Tang M, Wang Y, Xu Y, Tong W, Jin D, Yang XA. IDDCA syndrome in a Chinese infant due to GNB5 biallelic mutations. J Hum Genet 2020; 65:627-631. [PMID: 32203251 DOI: 10.1038/s10038-020-0742-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 11/10/2022]
Abstract
Herein, we present a Chinese infant with an early-onset intellectual developmental disorder with cardiac arrhythmia syndrome. A 6-month-old boy visited our hospital because of convulsions and paroxysmal cyanosis for 1 day. Mental development analysis showed that the patient had a neurodevelopmental delay. Frequent seizures occurred, and ECG monitoring demonstrated severe cardiac arrhythmia. Whole-exome sequencing showed that the infant had two compound heterozygous variants, NM_016194:c.458G>A/p.Cys153Tyr and NM_016194:c.1032C>A/p.Tyr344*, in GNB5. The first variant was inherited from his mother, while the other one was a de novo variant. Haplotype analysis indicated that the de novo variant was located in the paternal chromosome. Structural modeling indicated that both mutations could influence the interaction of GNB5 with its binding protein. Our study expanded the known genetic and phenotypic spectrum of GNB5-associated diseases, by presenting a Chinese male infant with IDDCA.
Collapse
Affiliation(s)
- Mingxing Tang
- Pediatric Intensive Care Unit, Anhui Provincial Children's Hospital, Hefei, 230029, PR China
| | - Yajian Wang
- Beijing Chigene Translational Medicine Research Center Co., Ltd, Beijing, 100875, PR China
| | - Yuanyuan Xu
- Pediatric Intensive Care Unit, Anhui Provincial Children's Hospital, Hefei, 230029, PR China
| | - Wenjia Tong
- Pediatric Intensive Care Unit, Anhui Provincial Children's Hospital, Hefei, 230029, PR China
| | - Danqun Jin
- Pediatric Intensive Care Unit, Anhui Provincial Children's Hospital, Hefei, 230029, PR China.
| | - Xiu-An Yang
- School of Basic Medical Science, Chengde Medical University, Chengde, 067000, PR China.
| |
Collapse
|
27
|
Malerba N, De Nittis P, Merla G. The Emerging Role of Gβ Subunits in Human Genetic Diseases. Cells 2019; 8:E1567. [PMID: 31817184 PMCID: PMC6952978 DOI: 10.3390/cells8121567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/23/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022] Open
Abstract
Environmental stimuli are perceived and transduced inside the cell through the activation of signaling pathways. One common type of cell signaling transduction network is initiated by G-proteins. G-proteins are activated by G-protein-coupled receptors (GPCRs) and transmit signals from hormones, neurotransmitters, and other signaling factors, thus controlling a number of biological processes that include synaptic transmission, visual photoreception, hormone and growth factors release, regulation of cell contraction and migration, as well as cell growth and differentiation. G-proteins mainly act as heterotrimeric complexes, composed of alpha, beta, and gamma subunits. In the last few years, whole exome sequencing and biochemical studies have shown causality of disease-causing variants in genes encoding G-proteins and human genetic diseases. This review focuses on the G-protein β subunits and their emerging role in the etiology of genetically inherited rare diseases in humans.
Collapse
Affiliation(s)
- Natascia Malerba
- Division of Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo (FG), Italy;
| | - Pasquelena De Nittis
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland;
| | - Giuseppe Merla
- Division of Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo (FG), Italy;
| |
Collapse
|
28
|
Pranckėnienė L, Siavrienė E, Gueneau L, Preikšaitienė E, Mikštienė V, Reymond A, Kučinskas V. De novo splice site variant of ARID1B associated with pathogenesis of Coffin-Siris syndrome. Mol Genet Genomic Med 2019; 7:e1006. [PMID: 31628733 PMCID: PMC6900373 DOI: 10.1002/mgg3.1006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/06/2019] [Accepted: 09/18/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Coffin-Siris syndrome is an extremely rare syndrome associated with developmental and congenital anomalies. It is caused by heterozygous pathogenic variants of ARID1A, ARID1B, SMARCA4, SMARCB1, SMARCE1, and SOX11. METHODS This case study presents the whole exome sequencing of a patient with characteristic clinical features of Coffin-Siris syndrome. Analysis included Sanger sequencing of complementary DNA and bioinformatic analysis of the variant. RESULTS Analysis of cDNA Sanger sequencing data revealed that the donor splice site variant led to skipping of exon 19. Further, bioinformatic analysis predicted abnormal splicing in a translational frameshift of 11 amino acids and the creation of a premature termination codon. Results found a novel de novo splice site variant c.5025+2T>C in the ARID1B and truncated 1 633 amino acid protein NP_065783.3:p. (Thr1633Valfs*11). CONCLUSION Truncated ARID1B resulted in loss of the BAF250 domain, which is part of SWI/SNF-like ATP-dependent chromatin remodeling complex. The severe clinical manifestation presented by the proband was attributed to the disappearance of the BAF250 domain in the ARID1B protein. Our finding provides strong evidence that this pathogenic variant of exon 19 caused a frameshift mutation in the ARID1B at the terminal exon, resulting in the expression of a severe phenotype of CSS.
Collapse
Affiliation(s)
- Laura Pranckėnienė
- Department of Human and Medical GeneticsInstitute of Biomedical SciencesFaculty of MedicineVilnius UniversityVilniusLithuania
| | - Evelina Siavrienė
- Department of Human and Medical GeneticsInstitute of Biomedical SciencesFaculty of MedicineVilnius UniversityVilniusLithuania
| | - Lucie Gueneau
- Centre for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Eglė Preikšaitienė
- Department of Human and Medical GeneticsInstitute of Biomedical SciencesFaculty of MedicineVilnius UniversityVilniusLithuania
| | - Violeta Mikštienė
- Department of Human and Medical GeneticsInstitute of Biomedical SciencesFaculty of MedicineVilnius UniversityVilniusLithuania
| | - Alexandre Reymond
- Centre for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Vaidutis Kučinskas
- Department of Human and Medical GeneticsInstitute of Biomedical SciencesFaculty of MedicineVilnius UniversityVilniusLithuania
| |
Collapse
|
29
|
Unique retinal signaling defect in GNB5-related disease. Doc Ophthalmol 2019; 140:273-277. [PMID: 31720979 DOI: 10.1007/s10633-019-09735-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/05/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To report a unique retinal signaling defect in GNB5-related disease. METHODS A 3-year-old female child underwent detailed systemic and ophthalmological evaluation. The eye examination included fundus photography, spectral domain optical coherence tomography and an extended protocol full-field electroretinography (ERG) including the ISCEV recommended standard steps. The dark-adapted (DA) ERGs were performed to a series of white flashes (range 0.006-30.0 cd s m-2) and two red flashes. The DA ERGs to higher stimulus intensities (3.0, 10.0 and 30.0 cd s m-2) were tested using a range of inter-stimulus intervals (ISI) of up to 60 s. In addition to standard light-adapted (LA) ERGs, a short-duration (0.5 s) LA 3.0 30-Hz flicker ERG and a long-duration LA ON-OFF ERG were also performed. Genetic testing included microarray, mitochondrial genome testing and whole exome sequencing. RESULTS The child was diagnosed to have status epilepticus and bradycardia at 6 months of age. Subsequently, she was diagnosed to have global developmental delay and hypotonia. On ophthalmological evaluation, the child fixes and follows light. Fundus evaluation showed mild optic disk pallor; macular SD-OCT was normal. The dim flash DA ERGs (DA 0.006 and DA 0.01 cd s m-2) were non-detectable. DA red flash ERGs showed the presence of an x-wave (cone component) and no rod component. The DA 3.0, 10.0 and 30.0 ERGs showed electronegative configuration regardless of the ISI; the averaged a-wave amplitude (4 flashes) was smaller at shorter ISI but became normal at a prolonged ISI (60 s). The LA 30-Hz flicker ERG was severely reduced but detectable for the initial 0.5 s; this became non-detectable after 5 s of averaging. The LA 3.0 2-Hz ERG showed markedly reduced a- and b-wave amplitudes and a reduced b:a ratio; the LA ON-OFF ERGs were non-detectable. WES identified a homozygous null mutation in G protein subunit beta 5 (GNB5; c.1032C>A/p.Tyr344*). CONCLUSION This report identifies for the first time a unique retinopathy associated with biallelic mutations in GNB5. The observed phenotype is consistent with a dual retinal signaling defect reminiscent of features of bradyopsia and rod ON-bipolar dysfunction.
Collapse
|
30
|
Fukuda T, Hiraide T, Yamoto K, Nakashima M, Kawai T, Yanagi K, Ogata T, Saitsu H. Exome reports A de novo GNB2 variant associated with global developmental delay, intellectual disability, and dysmorphic features. Eur J Med Genet 2019; 63:103804. [PMID: 31698099 DOI: 10.1016/j.ejmg.2019.103804] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/22/2019] [Accepted: 11/02/2019] [Indexed: 11/30/2022]
Abstract
Heterotrimeric G proteins are composed of α, β, and γ subunits and are involved in integrating signals between receptors and effector proteins. The 5 human Gβ proteins (encoded by GNB1, GNB2, GNB3, GNB4, and GNB5) are highly similar. Variants in GNB1 were identified as a genetic cause of developmental delay. De novo variant in GNB2 has recently been reported as a cause of sinus node dysfunction and atrioventricular block but not as a cause of developmental delay. Trio-based whole-exome sequencing was performed on an individual with global developmental delay, muscle hypotonia, multiple congenital joint contractures and dysmorphism such as brachycephalus, thick eyebrows, thin upper lip, micrognathia, prominent chin, and bilateral tapered fingers. We identified a de novo GNB2 variant c.229G>A, p.(Gly77Arg). Notably, pathogenic substitutions of the homologous Gly77 residue including an identical variant (p.Gly77Arg, p.Gly77Val, p.Gly77Ser, p.Gly77Ala) of GNB1, a paralog of GNB2, was reported in individuals with global developmental delay and hypotonia. Clinical features of our case overlap with those of GNB1 variants. Our study suggests that a GNB2 variant may be associated with syndromic global developmental delay.
Collapse
Affiliation(s)
- Tokiko Fukuda
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Takuya Hiraide
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan; Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kaori Yamoto
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoko Kawai
- Department of Maternal-Fetal Biology and National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kumiko Yanagi
- Department of Genome Medicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
31
|
Poke G, King C, Muir A, de Valles-Ibáñez G, Germano M, Moura de Souza CF, Fung J, Chung B, Fung CW, Mignot C, Ilea A, Keren B, Vermersch AI, Davis S, Stanley T, Moharir M, Kannu P, Shao Z, Malerba N, Merla G, Mefford HC, Scheffer IE, Sadleir LG. The epileptology of GNB5 encephalopathy. Epilepsia 2019; 60:e121-e127. [PMID: 31631344 DOI: 10.1111/epi.16372] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 01/23/2023]
Abstract
Pathogenic variants in GNB5 cause an autosomal recessive neurodevelopmental disorder with neonatal sinus bradycardia. Seizures or epilepsy occurred in 10 of 22 previously reported cases, including 6 children from one family. We delineate the epileptology of GNB5 encephalopathy. Our nine patients, including five new patients, were from seven families. Epileptic spasms were the most frequent seizure type, occurring in eight of nine patients, and began at a median age of 3 months (2 months to 3 years). Focal seizures preceded spasms in three children, with onset at 7 days, 11 days, and 4 months. One child presented with convulsive status epilepticus at 6 months. Three children had burst suppression on electroencephalography (EEG), three had hypsarrhythmia, and one evolved from burst suppression to hypsarrhythmia. Background slowing was present in all after age 3 years. Magnetic resonance imaging (MRI) showed cerebral atrophy in one child and cerebellar atrophy in another. All nine had abnormal development prior to seizure onset and ultimately had profound impairment without regression. Hypotonia was present in all, with contractures developing in two older patients. All individuals had biallelic pathogenic variants in GNB5, predicted by in silico tools to result in protein truncation and loss-of-function. GNB5 developmental and epileptic encephalopathy is characterized by epileptic spasms, focal seizures, and profound impairment.
Collapse
Affiliation(s)
- Gemma Poke
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Chontelle King
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Alison Muir
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Michele Germano
- Maternal and Pediatric Department, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | | | - Jasmine Fung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong City, Hong Kong
| | - Brian Chung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong City, Hong Kong
| | - Cheuk Wing Fung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong City, Hong Kong
| | - Cyril Mignot
- Department of Genetics, Reference Center for Intellectual Disorders of Rare Causes, APHP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Adina Ilea
- Department of Pediatric Neurology and Metabolic Diseases, APHP, Robert Debré Hospital, Paris, France
| | - Boris Keren
- Department of Genetics, APHP, Pitié-Salpêtrière University Hospital, Paris, France
| | | | | | - Thorsten Stanley
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | | | - Peter Kannu
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Zhuo Shao
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Natascia Malerba
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Giuseppe Merla
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Heather C Mefford
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Ingrid E Scheffer
- Florey and Murdoch Children's Research Institute, Austin Health and Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| |
Collapse
|
32
|
Malerba N, Benzoni P, Squeo GM, Milanesi R, Giannetti F, Sadleir LG, Poke G, Augello B, Croce AI, Barbuti A, Merla G. Generation of the induced human pluripotent stem cell lines CSSi009-A from a patient with a GNB5 pathogenic variant, and CSSi010-A from a CRISPR/Cas9 engineered GNB5 knock-out human cell line. Stem Cell Res 2019; 40:101547. [PMID: 31479876 DOI: 10.1016/j.scr.2019.101547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/13/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022] Open
Abstract
GNB5 loss-of-function pathogenic variants cause IDDCA, a rare autosomal recessive human genetic disease characterized by infantile onset of intellectual disability, sinus bradycardia, hypotonia, visual abnormalities, and epilepsy. We generated human induced pluripotent stem cells (hiPSCs) from skin fibroblasts of a patient with the homozygous c.136delG frameshift variant, and a GNB5 knock-out (KO) line by CRISPR/Cas9 editing. hiPSCs express common pluripotency markers and differentiate into the three germ layers. These lines represent a powerful cellular model to study the molecular basis of GNB5-related disorders as well as offer an in vitro model for drug screening.
Collapse
Affiliation(s)
- Natascia Malerba
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Patrizia Benzoni
- The PaceLab, Department of Biosciences, Università degli Studi di Milano, Italy
| | - Gabriella Maria Squeo
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Raffaella Milanesi
- The PaceLab, Department of Biosciences, Università degli Studi di Milano, Italy
| | - Federica Giannetti
- The PaceLab, Department of Biosciences, Università degli Studi di Milano, Italy
| | - Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago Wellington, Wellington, New Zealand
| | - Gemma Poke
- Department of Paediatrics and Child Health, University of Otago Wellington, Wellington, New Zealand
| | - Bartolomeo Augello
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Anna Irma Croce
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Andrea Barbuti
- The PaceLab, Department of Biosciences, Università degli Studi di Milano, Italy
| | - Giuseppe Merla
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
33
|
Veerman CC, Mengarelli I, Koopman CD, Wilders R, van Amersfoorth SC, Bakker D, Wolswinkel R, Hababa M, de Boer TP, Guan K, Milnes J, Lodder EM, Bakkers J, Verkerk AO, Bezzina CR. Genetic variation in GNB5 causes bradycardia by augmenting the cholinergic response via increased acetylcholine-activated potassium current ( I K,ACh). Dis Model Mech 2019; 12:dmm.037994. [PMID: 31208990 PMCID: PMC6679373 DOI: 10.1242/dmm.037994] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/06/2019] [Indexed: 12/19/2022] Open
Abstract
Mutations in GNB5, encoding the G-protein β5 subunit (Gβ5), have recently been linked to a multisystem disorder that includes severe bradycardia. Here, we investigated the mechanism underlying bradycardia caused by the recessive p.S81L Gβ5 variant. Using CRISPR/Cas9-based targeting, we generated an isogenic series of human induced pluripotent stem cell (hiPSC) lines that were either wild type, heterozygous or homozygous for the GNB5 p.S81L variant. These were differentiated into cardiomyocytes (hiPSC-CMs) that robustly expressed the acetylcholine-activated potassium channel [I(KACh); also known as IK,ACh]. Baseline electrophysiological properties of the lines did not differ. Upon application of carbachol (CCh), homozygous p.S81L hiPSC-CMs displayed an increased acetylcholine-activated potassium current (I K,ACh) density and a more pronounced decrease of spontaneous activity as compared to wild-type and heterozygous p.S81L hiPSC-CMs, explaining the bradycardia in homozygous carriers. Application of the specific I(KACh) blocker XEN-R0703 resulted in near-complete reversal of the phenotype. Our results provide mechanistic insights and proof of principle for potential therapy in patients carrying GNB5 mutations.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Christiaan C Veerman
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Isabella Mengarelli
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Charlotte D Koopman
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Ronald Wilders
- Amsterdam UMC, University of Amsterdam, Department of Medical Biology, Heart Failure Research Center, 1105 AZ Amsterdam, The Netherlands
| | - Shirley C van Amersfoorth
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Diane Bakker
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Rianne Wolswinkel
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Mariam Hababa
- Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Kaomei Guan
- Department of Pharmacology and Toxicology, Technische Universität Dresden, 01062 Dresden, Germany
| | | | - Elisabeth M Lodder
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Arie O Verkerk
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands.,Amsterdam UMC, University of Amsterdam, Department of Medical Biology, Heart Failure Research Center, 1105 AZ Amsterdam, The Netherlands
| | - Connie R Bezzina
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
34
|
Miller JE, Veturi Y, Ritchie MD. Innovative strategies for annotating the "relationSNP" between variants and molecular phenotypes. BioData Min 2019; 12:10. [PMID: 31114635 PMCID: PMC6518798 DOI: 10.1186/s13040-019-0197-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/18/2019] [Indexed: 11/10/2022] Open
Abstract
Characterizing how variation at the level of individual nucleotides contributes to traits and diseases has been an area of growing interest since the completion of sequencing the first human genome. Our understanding of how a single nucleotide polymorphism (SNP) leads to a pathogenic phenotype on a genome-wide scale is a fruitful endeavor for anyone interested in developing diagnostic tests, therapeutics, or simply wanting to understand the etiology of a disease or trait. To this end, many datasets and algorithms have been developed as resources/tools to annotate SNPs. One of the most common practices is to annotate coding SNPs that affect the protein sequence. Synonymous variants are often grouped as one type of variant, however there are in fact many tools available to dissect their effects on gene expression. More recently, large consortiums like ENCODE and GTEx have made it possible to annotate non-coding regions. Although annotating variants is a common technique among human geneticists, the constant advances in tools and biology surrounding SNPs requires an updated summary of what is known and the trajectory of the field. This review will discuss the history behind SNP annotation, commonly used tools, and newer strategies for SNP annotation. Additionally, we will comment on the caveats that distinguish approaches from one another, along with gaps in the current state of knowledge, and potential future directions. We do not intend for this to be a comprehensive review for any specific area of SNP annotation, but rather it will be an excellent resource for those unfamiliar with computational tools used to functionally characterize SNPs. In summary, this review will help illustrate how each SNP annotation method impacts the way in which the genetic and molecular etiology of a disease is explored in-silico.
Collapse
Affiliation(s)
- Jason E. Miller
- Department of Genetics, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA 19104 USA
| | - Yogasudha Veturi
- Department of Genetics, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA 19104 USA
| | - Marylyn D. Ritchie
- Department of Genetics, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA 19104 USA
| |
Collapse
|
35
|
Pranckėnienė L, Preikšaitienė E, Gueneau L, Reymond A, Kučinskas V. De Novo Duplication in the CHD7 Gene Associated With Severe CHARGE Syndrome. GENOMICS INSIGHTS 2019; 12:1178631019839010. [PMID: 31043788 PMCID: PMC6446253 DOI: 10.1177/1178631019839010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 02/28/2019] [Indexed: 01/14/2023]
Abstract
CHARGE syndrome is an autosomal dominant developmental disorder associated with a constellation of traits involving almost every organ and sensory system, in particular congenital anomalies, including choanal atresia and malformations of the heart, inner ear, and retina. Variants in CHD7 have been shown to cause CHARGE syndrome. Here, we report the identification of a novel de novo p.Asp2119_Pro2120ins6 duplication variant in a conserved region of CHD7 in a severely affected boy presenting with 3 and 5 of the CHARGE cardinal major and minor signs, respectively, combined with congenital umbilical hernia, congenital hernia at the linea alba, mildly hypoplastic inferior vermis, slight dilatation of the lateral ventricles, prominent metopic ridge, and hypoglycemic episodes.
Collapse
Affiliation(s)
- Laura Pranckėnienė
- Department of Human and Medical Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| | - Eglė Preikšaitienė
- Department of Human and Medical Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| | - Lucie Gueneau
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Vaidutis Kučinskas
- Department of Human and Medical Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
36
|
Malerba N, Towner S, Keating K, Squeo GM, Wilson W, Merla G. A NGS-Targeted Autism/ID Panel Reveals Compound Heterozygous GNB5 Variants in a Novel Patient. Front Genet 2018; 9:626. [PMID: 30631341 PMCID: PMC6315145 DOI: 10.3389/fgene.2018.00626] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/23/2018] [Indexed: 11/13/2022] Open
Abstract
Homozygous and compound heterozygous pathogenic variants in GNB5 have been recently associated with a spectrum of clinical presentations varying from a severe multisystem form of the disorder including intellectual disability, early infantile developmental and epileptic encephalopathy, retinal abnormalities and cardiac arrhythmias (IDDCA) to a milder form with language delay, attention-deficit/hyperactivity disorder, cognitive impairment, with or without cardiac arrhythmia (LADCI). Approximately twenty patients have been described so far; here we report a novel case of a 2.5-year-old female who is a compound heterozygote for a frameshift and a missense variant in the GNB5 gene. Her clinical presentation is consistent with a moderate phenotype, corroborating the direct correlation between the type and pathogenic mechanism of the GNB5 genetic variant and the severity of related phenotype.
Collapse
Affiliation(s)
- Natascia Malerba
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Shelley Towner
- Division of Medical Genetics, University of Virginia, Charlottesville, VA, United States
| | - Katherine Keating
- Division of Medical Genetics, University of Virginia, Charlottesville, VA, United States
| | - Gabriella Maria Squeo
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - William Wilson
- Division of Medical Genetics, University of Virginia, Charlottesville, VA, United States
| | - Giuseppe Merla
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
37
|
Sriroopreddy R, Sajeed R, P R, C S. Differentially expressed gene (DEG) based protein-protein interaction (PPI) network identifies a spectrum of gene interactome, transcriptome and correlated miRNA in nondisjunction Down syndrome. Int J Biol Macromol 2018; 122:1080-1089. [PMID: 30218739 DOI: 10.1016/j.ijbiomac.2018.09.056] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 10/28/2022]
Abstract
Down syndrome, a genetic disorder of known attribution reveals several types of brain abnormalities resulting in mental retardation, inadequacy in speech and memory. In this study, we have presented a consolidative network approach to comprehend the intricacy of the associated genes of Down syndrome. In this analysis, the differentially expressed genes (DEG's) were identified and the central networks were constructed as upregulated and downregulated. Subsequently, GNB5, CDC42, SPTAN1, GNG2, GNAZ, PRKACB, SST, CD44, FGF2, PHLPP1, APP, and FYN were identified as the candidate hub genes by using topological parameters. Later, Fpclass a PPI tool identified WASP gene, a co-expression interacting partner with highest network topology. Moreover, an enhanced enrichment pathway namely Opioid signaling was obtained using ClueGo, depicting the roles of the hub genes in signaling and neuronal mechanisms. The transcriptional regulatory factors and the common miRNA connected to them were identified by using MatInspector and miRTarbase. Later, a regulatory network constructed showed that PLAG, T2FB, CREB, NEUR, and GATA were the most commonly connected transcriptional factors and hsa-miR-122-5p was the most prominent miRNA. In a nutshell, these hub genes and the enriched pathway could help understand at a molecular level and eventually used as therapeutic targets for Down syndrome.
Collapse
Affiliation(s)
- Ramireddy Sriroopreddy
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Rakshanda Sajeed
- Department of Analytics, School of Computer Science and Engineering, Vellore Institute of Technology, Vellore 632014, India
| | - Raghuraman P
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Sudandiradoss C
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
38
|
Kang N, Kang JY, Park S, Shin DM. Increased store-operated Ca 2+ entry mediated by GNB5 and STIM1. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:343-348. [PMID: 29719456 PMCID: PMC5928347 DOI: 10.4196/kjpp.2018.22.3.343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/14/2018] [Accepted: 02/19/2018] [Indexed: 01/27/2023]
Abstract
Recent human genetic studies have shown that Gβ5 is related to various clinical symptoms, such as sinus bradycardia, cognitive disability, and attention deficit hyperactivity disorder. Although the calcium signaling cascade is closely associated with a heterotrimeric G-protein, the function of Gβ5 in calcium signaling and its relevance to clinical symptoms remain unknown. In this study, we investigated the in vitro changes of store-operated calcium entry (SOCE) with exogenous expression of Gβ5. The cells expressing Gβ5 had enhanced SOCE after depletion of calcium ion inside the endoplasmic reticulum. Gβ5 also augmented Stim1- and Orai1-dependent SOCE. An ORAI1 loss-of-function mutant did not show inhibition of Gβ5-induced SOCE, and a STIM1-ERM truncation mutant showed no enhancement of SOCE. These results suggested a novel role of GNB5 and Stim1, and provided insight into the regulatory mechanism of SOCE.
Collapse
Affiliation(s)
- Namju Kang
- Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 03722, Korea
| | - Jung Yun Kang
- Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 03722, Korea
| | - Soonhong Park
- Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 03722, Korea
| | - Dong Min Shin
- Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul 03722, Korea
| |
Collapse
|
39
|
Genome-Wide Identification and Characterization of WD40 Protein Genes in the Silkworm, Bombyx mori. Int J Mol Sci 2018; 19:ijms19020527. [PMID: 29425159 PMCID: PMC5855749 DOI: 10.3390/ijms19020527] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/27/2018] [Accepted: 02/06/2018] [Indexed: 12/18/2022] Open
Abstract
WD40 proteins are scaffolding molecules in protein-protein interactions and play crucial roles in fundamental biological processes. Genome-wide characterization of WD40 proteins in animals has been conducted solely in humans. We retrieved 172 WD40 protein genes in silkworm (BmWD40s) and identified these genes in 7 other insects, 9 vertebrates and 5 nematodes. Comparative analysis revealed that the WD40 protein gene family underwent lineage-specific expansions during animal evolution, but did not undergo significant expansion during insect evolution. The BmWD40s were categorized into five clusters and 12 classes according to the phylogenetic classification and their domain architectures, respectively. Sequence analyses indicated that tandem and segmental duplication played minor roles in producing the current number of BmWD40s, and domain recombination events of multi-domain BmWD40s might have occurred mainly after gene duplication events. Gene Ontology (GO) analysis revealed that a higher proportion of BmWD40s was involved in processes, such as binding, transcription-regulation and cellular component biogenesis, compared to all silkworm genes annotated in GO. Microarray-based analysis demonstrated that many BmWD40s had tissue-specific expression and exhibited high and/or sex-related expression during metamorphosis. These findings contribute to a better understanding of the evolution of the animal WD40 protein family and assist the study of the functions of BmWD40s.
Collapse
|
40
|
Vernon H, Cohen J, De Nittis P, Fatemi A, McClellan R, Goldstein A, Malerba N, Guex N, Reymond A, Merla G. Intellectual developmental disorder with cardiac arrhythmia syndrome in a child with compound heterozygous GNB5 variants. Clin Genet 2018; 93:1254-1256. [PMID: 29368331 DOI: 10.1111/cge.13194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 11/30/2022]
Abstract
Identification of a novel compound heterozygous of GNB5 in a patient with intellectual developmental disorder with cardiac arrhytmia (IDDCA), from non-consaguineous family. Three-dimensional modelling and in silico predictions suggest that GNB5 variants are causative of the phenotype, extending the number of IDDCA patients so far identified.
Collapse
Affiliation(s)
- H Vernon
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, Maryland
| | - J Cohen
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, Maryland
| | - P De Nittis
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - A Fatemi
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, Maryland
| | - R McClellan
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, Maryland
| | - A Goldstein
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - N Malerba
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - N Guex
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - A Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - G Merla
- Division of Medical Genetics, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| |
Collapse
|
41
|
Sala L, van Meer BJ, Tertoolen LGJ, Bakkers J, Bellin M, Davis RP, Denning C, Dieben MAE, Eschenhagen T, Giacomelli E, Grandela C, Hansen A, Holman ER, Jongbloed MRM, Kamel SM, Koopman CD, Lachaud Q, Mannhardt I, Mol MPH, Mosqueira D, Orlova VV, Passier R, Ribeiro MC, Saleem U, Smith GL, Burton FL, Mummery CL. MUSCLEMOTION: A Versatile Open Software Tool to Quantify Cardiomyocyte and Cardiac Muscle Contraction In Vitro and In Vivo. Circ Res 2017; 122:e5-e16. [PMID: 29282212 PMCID: PMC5805275 DOI: 10.1161/circresaha.117.312067] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/19/2017] [Accepted: 12/23/2017] [Indexed: 12/31/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: There are several methods to measure cardiomyocyte and muscle contraction, but these require customized hardware, expensive apparatus, and advanced informatics or can only be used in single experimental models. Consequently, data and techniques have been difficult to reproduce across models and laboratories, analysis is time consuming, and only specialist researchers can quantify data. Objective: Here, we describe and validate an automated, open-source software tool (MUSCLEMOTION) adaptable for use with standard laboratory and clinical imaging equipment that enables quantitative analysis of normal cardiac contraction, disease phenotypes, and pharmacological responses. Methods and Results: MUSCLEMOTION allowed rapid and easy measurement of movement from high-speed movies in (1) 1-dimensional in vitro models, such as isolated adult and human pluripotent stem cell-derived cardiomyocytes; (2) 2-dimensional in vitro models, such as beating cardiomyocyte monolayers or small clusters of human pluripotent stem cell-derived cardiomyocytes; (3) 3-dimensional multicellular in vitro or in vivo contractile tissues, such as cardiac “organoids,” engineered heart tissues, and zebrafish and human hearts. MUSCLEMOTION was effective under different recording conditions (bright-field microscopy with simultaneous patch-clamp recording, phase contrast microscopy, and traction force microscopy). Outcomes were virtually identical to the current gold standards for contraction measurement, such as optical flow, post deflection, edge-detection systems, or manual analyses. Finally, we used the algorithm to quantify contraction in in vitro and in vivo arrhythmia models and to measure pharmacological responses. Conclusions: Using a single open-source method for processing video recordings, we obtained reliable pharmacological data and measures of cardiac disease phenotype in experimental cell, animal, and human models.
Collapse
Affiliation(s)
- Luca Sala
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Berend J van Meer
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Leon G J Tertoolen
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Jeroen Bakkers
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Milena Bellin
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Richard P Davis
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Chris Denning
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Michel A E Dieben
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Thomas Eschenhagen
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Elisa Giacomelli
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Catarina Grandela
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Arne Hansen
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Eduard R Holman
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Monique R M Jongbloed
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Sarah M Kamel
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Charlotte D Koopman
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Quentin Lachaud
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Ingra Mannhardt
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Mervyn P H Mol
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Diogo Mosqueira
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Valeria V Orlova
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Robert Passier
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Marcelo C Ribeiro
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Umber Saleem
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Godfrey L Smith
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Francis L Burton
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.)
| | - Christine L Mummery
- From the Department of Anatomy and Embryology, Leiden University Medical Center, The Netherlands (L.S., B.J.v.M., L.G.J.T., M.B., R.P.D., M.A.E.D., E.G., C.G., M.R.M.J., M.P.H.M., V.V.O., R.P., C.L.M.); Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Science, University of Glasgow, United Kingdom (Q.L., G.L.S., F.L.B.); Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); Department of Stem Cell Biology, University of Nottingham, University Park, Nottingham, United Kingdom (C.D., D.M.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany (T.E., A.H., I.M., U.S.); DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck (T.E., A.H., I.M., U.S.); Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (U.S.); Hart Long Centrum, Leiden University Medical Center, The Netherlands (E.R.H., M.R.M.J.); Department of Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands (R.P., M.C.R., C.L.M.).; Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (J.B., S.M.K., C.D.K.); and Clyde Biosciences, Ltd, BioCity Scotland, United Kingdom (G.L.S., F.L.B.).
| |
Collapse
|
42
|
A Genome-Wide Association Study and Complex Network Identify Four Core Hub Genes in Bipolar Disorder. Int J Mol Sci 2017; 18:ijms18122763. [PMID: 29257106 PMCID: PMC5751362 DOI: 10.3390/ijms18122763] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/29/2017] [Accepted: 12/14/2017] [Indexed: 11/25/2022] Open
Abstract
Bipolar disorder is a common and severe mental illness with unsolved pathophysiology. A genome-wide association study (GWAS) has been used to find a number of risk genes, but it is difficult for a GWAS to find genes indirectly associated with a disease. To find core hub genes, we introduce a network analysis after the GWAS was conducted. Six thousand four hundred fifty eight single nucleotide polymorphisms (SNPs) with p < 0.01 were sifted out from Wellcome Trust Case Control Consortium (WTCCC) dataset and mapped to 2045 genes, which are then compared with the protein–protein network. One hundred twelve genes with a degree >17 were chosen as hub genes from which five significant modules and four core hub genes (FBXL13, WDFY2, bFGF, and MTHFD1L) were found. These core hub genes have not been reported to be directly associated with BD but may function by interacting with genes directly related to BD. Our method engenders new thoughts on finding genes indirectly associated with, but important for, complex diseases.
Collapse
|
43
|
Familial early infantile epileptic encephalopathy and cardiac conduction disorder: A rare cause of SUDEP in infancy. Seizure 2017; 50:171-172. [PMID: 28697420 DOI: 10.1016/j.seizure.2017.06.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 06/20/2017] [Indexed: 11/22/2022] Open
|
44
|
Lodder EM, Verkerk AO, Bezzina CR. Pacing Discovery: G-Protein β Subunit Mutations in Sinus Node Dysfunction. Circ Res 2017; 120:1524-1526. [PMID: 28495981 DOI: 10.1161/circresaha.117.310953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Elisabeth M Lodder
- From the Heart Center, Department of Clinical and Experimental Cardiology (E.M.L., A.O.V., C.R.B.) and Department of Medical Biology (A.O.V.), Academic Medical Center, Amsterdam, the Netherlands
| | - Arie O Verkerk
- From the Heart Center, Department of Clinical and Experimental Cardiology (E.M.L., A.O.V., C.R.B.) and Department of Medical Biology (A.O.V.), Academic Medical Center, Amsterdam, the Netherlands
| | - Connie R Bezzina
- From the Heart Center, Department of Clinical and Experimental Cardiology (E.M.L., A.O.V., C.R.B.) and Department of Medical Biology (A.O.V.), Academic Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
45
|
Burkhard S, van Eif V, Garric L, Christoffels VM, Bakkers J. On the Evolution of the Cardiac Pacemaker. J Cardiovasc Dev Dis 2017; 4:jcdd4020004. [PMID: 29367536 PMCID: PMC5715705 DOI: 10.3390/jcdd4020004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 01/26/2023] Open
Abstract
The rhythmic contraction of the heart is initiated and controlled by an intrinsic pacemaker system. Cardiac contractions commence at very early embryonic stages and coordination remains crucial for survival. The underlying molecular mechanisms of pacemaker cell development and function are still not fully understood. Heart form and function show high evolutionary conservation. Even in simple contractile cardiac tubes in primitive invertebrates, cardiac function is controlled by intrinsic, autonomous pacemaker cells. Understanding the evolutionary origin and development of cardiac pacemaker cells will help us outline the important pathways and factors involved. Key patterning factors, such as the homeodomain transcription factors Nkx2.5 and Shox2, and the LIM-homeodomain transcription factor Islet-1, components of the T-box (Tbx), and bone morphogenic protein (Bmp) families are well conserved. Here we compare the dominant pacemaking systems in various organisms with respect to the underlying molecular regulation. Comparative analysis of the pathways involved in patterning the pacemaker domain in an evolutionary context might help us outline a common fundamental pacemaker cell gene programme. Special focus is given to pacemaker development in zebrafish, an extensively used model for vertebrate development. Finally, we conclude with a summary of highly conserved key factors in pacemaker cell development and function.
Collapse
Affiliation(s)
- Silja Burkhard
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| | - Vincent van Eif
- Department of Medical Biology, Academic Medical Center Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Laurence Garric
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| | - Vincent M Christoffels
- Department of Medical Biology, Academic Medical Center Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|