51
|
Monteiro AN, Bouwman P, Kousholt AN, Eccles DM, Millot GA, Masson JY, Schmidt MK, Sharan SK, Scully R, Wiesmüller L, Couch F, Vreeswijk MPG. Variants of uncertain clinical significance in hereditary breast and ovarian cancer genes: best practices in functional analysis for clinical annotation. J Med Genet 2020; 57:509-518. [PMID: 32152249 DOI: 10.1136/jmedgenet-2019-106368] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/28/2019] [Accepted: 12/01/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Alvaro N Monteiro
- Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Peter Bouwman
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Arne N Kousholt
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Diana M Eccles
- Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Gael A Millot
- Hub-DBC, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Jean-Yves Masson
- CHU de Québec-Université Laval, Oncology Division, Laval University Cancer Research Center, Quebec City, Quebec, Canada
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Shyam K Sharan
- National Cancer Institute at Frederick, Frederick, Maryland, USA
| | - Ralph Scully
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
52
|
Li X, Warner JL. A Review of Precision Oncology Knowledgebases for Determining the Clinical Actionability of Genetic Variants. Front Cell Dev Biol 2020; 8:48. [PMID: 32117976 PMCID: PMC7026022 DOI: 10.3389/fcell.2020.00048] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/20/2020] [Indexed: 01/25/2023] Open
Abstract
The increased availability of tumor genetic testing and targeted cancer therapies contributes to the advancement of precision medicine in the field of oncology. Precision oncology knowledgebases provide a way of organizing clinically relevant genetic information in a way that is easily accessible for both oncologists and patients, facilitating the genetic-based clinical decision making. Many organizations and companies have built precision oncology knowledgebases, intended for multiple users. In general, these knowledgebases offer information on cancer-related genetic variants as well as their associated diagnostic, prognostic, and therapeutic implications, but they often differ in their information curations, designs, and user experiences. It is advisable that oncologists use multiple knowledgebases during their practice to have them complement each other. In the future, convergence toward common standards and formats is needed to ensure that the comprehensive knowledge across all sources can be unified to bring the oncology community closer to the achievement of the goal of precision oncology.
Collapse
Affiliation(s)
- Xuanyi Li
- Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Jeremy L. Warner
- Department of Medicine, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
53
|
Zeng J, Slodkowicz G, James LC. Rare missense variants in the human cytosolic antibody receptor preserve antiviral function. eLife 2019; 8:48339. [PMID: 31613747 PMCID: PMC6794091 DOI: 10.7554/elife.48339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/02/2019] [Indexed: 12/30/2022] Open
Abstract
The genetic basis of most human disease cannot be explained by common variants. One solution to this ‘missing heritability problem’ may be rare missense variants, which are individually scarce but collectively abundant. However, the phenotypic impact of rare variants is under-appreciated as gene function is normally studied in the context of a single ‘wild-type’ sequence. Here, we explore the impact of naturally occurring missense variants in the human population on the cytosolic antibody receptor TRIM21, using volunteer cells with variant haplotypes, CRISPR gene editing and functional reconstitution. In combination with data from a panel of computational predictors, the results suggest that protein robustness and purifying selection ensure that function is remarkably well-maintained despite coding variation.
Collapse
Affiliation(s)
- Jingwei Zeng
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Greg Slodkowicz
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Leo C James
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
54
|
Shaik NA, Bokhari HA, Masoodi TA, Shetty PJ, Ajabnoor GMA, Elango R, Banaganapalli B. Molecular modelling and dynamics of CA2 missense mutations causative to carbonic anhydrase 2 deficiency syndrome. J Biomol Struct Dyn 2019; 38:4067-4080. [PMID: 31542996 DOI: 10.1080/07391102.2019.1671899] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Carbonic anhydrase 2 (CA2) enzyme deficiency caused by CA2 gene mutations is an inherited disorder characterized by symptoms like osteopetrosis, renal tubular acidosis, and cerebral calcification. This study has collected the CA2 deficiency causal missense mutations and assessed their pathogenicity using diverse computational programs. The 3D protein models for all missense mutations were built, and analyzed for structural divergence, protein stability, and molecular dynamics properties. We found M-CAP as the most sensitive prediction method to measure the deleterious potential of CA2 missense mutations. Free energy dynamics of tertiary structure models of CA2 mutants with DUET, mCSM, and SDM based consensus methods predicted only 50% of the variants as destabilizing. Superimposition of native and mutant CA2 models revealed the minor structural fluctuations at the amino acid residue level but not at the whole protein structure level. Near native molecular dynamic simulation analysis indicated that CA2 causative missense variants result in residue level fluctuation pattern in the protein structure. This study expands the understanding of genotype-protein phenotype correlations underlying CA2 variant pathogenicity and presents a potential avenue for modifying the CA2 deficiency by targeting biophysical structural features of CA2 protein. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Noor A Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hifaa A Bokhari
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tariq Ahmed Masoodi
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Preetha J Shetty
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, UAE
| | - Ghada M A Ajabnoor
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
55
|
Martin LJ, Benson DW. Identifying Genetic Modifiers in the Age of Exome: Current Considerations. J Pediatr 2019; 213:8-10. [PMID: 31303336 DOI: 10.1016/j.jpeds.2019.06.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/18/2019] [Indexed: 01/28/2023]
Affiliation(s)
- Lisa J Martin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - D Woodrow Benson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
56
|
Jungwirth G, Warta R, Beynon C, Sahm F, von Deimling A, Unterberg A, Herold-Mende C, Jungk C. Intraventricular meningiomas frequently harbor NF2 mutations but lack common genetic alterations in TRAF7, AKT1, SMO, KLF4, PIK3CA, and TERT. Acta Neuropathol Commun 2019; 7:140. [PMID: 31470906 PMCID: PMC6716845 DOI: 10.1186/s40478-019-0793-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/22/2019] [Indexed: 01/28/2023] Open
Abstract
Intraventricular meningiomas (IVMs) account for less than 5% of all intracranial meningiomas; hence their molecular phenotype remains unknown. In this study, we were interested whether genetic alterations in IVMs differ from meningiomas in other locations and analyzed our institutional series with respect to clinical and molecular characteristics. A total of 25 patients with surgical removal of an IVM at our department between 1986 and 2018 were identified from our institutional database. Median progression-free survival (PFS) was 79 months (range of 2-319 months) and PFS at 5 years was 86%. Corresponding tumor tissue was available for 18 patients including one matching recurrence and was subjected to targeted panel sequencing of 130 selected genes frequently mutated in brain cancers by applying a custom hybrid capture approach on a NextSeq500 instrument. Loss of chromosome 22q and 1p occurred frequently in 89 and 44% of cases. Deleterious NF2 mutations were found in 44% of IVMs (n = 8/18). In non-NF2-mutated IVMs, previously reported genetic alterations including TRAF7, AKT1, SMO, KLF4, PIK3CA, and TERT were lacking, suggesting alternative genes in the pathogenesis of non-NF2 IVMs. In silico analysis revealed possible damaging mutations of APC, GABRA6, GSE1, KDR, and two SMO missense mutations differing from previously reported ones. Interestingly, all WHO°II IVMs (n = 3) harbored SMARCB1 and SMARCA4 mutations, indicating a role of the SWI/SNF chromatin remodeling complex in aggressive IVMs.
Collapse
Affiliation(s)
- Gerhard Jungwirth
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, INF 400, D-69120 Heidelberg, Germany
| | - Rolf Warta
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, INF 400, D-69120 Heidelberg, Germany
| | - Christopher Beynon
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, INF 400, D-69120 Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Institute of Pathology, University of Heidelberg, INF 224, D-69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, University of Heidelberg, INF 224, D-69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Unterberg
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, INF 400, D-69120 Heidelberg, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, INF 400, D-69120 Heidelberg, Germany
| | - Christine Jungk
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, INF 400, D-69120 Heidelberg, Germany
| |
Collapse
|
57
|
BRCA1 and BRCA2 Testing through Next Generation Sequencing in a Small Cohort of Italian Breast/Ovarian Cancer Patients: Novel Pathogenic and Unknown Clinical Significance Variants. Int J Mol Sci 2019; 20:ijms20143442. [PMID: 31336956 PMCID: PMC6678297 DOI: 10.3390/ijms20143442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 01/07/2023] Open
Abstract
The aim of this report is to describe results of BRCA1 and BRCA2 Next Generation Sequencing Analysis (NGS) analysis in 132 selected Italian patients with breast/ovarian cancer. A NGS pipeline with a reliable Copy Number Variation (CNV) prediction algorithm was applied. In addition, VarSome and Priors V2.0 Software were employed for in silico analysis of novel missense variants. A total of 37 BRCA1 and BRCA2 pathogenic variants were found in 34 unrelated subjects with a frequency of positive patients of 25.7% (34/132). Twenty-four deleterious variants were detected in BRCA1 (representing the 64.9% of all identified pathogenic defects) and thirteen (35.1% of all identified pathogenic variants) in BRCA2 gene. The percentage of patients carrying a variant of unknown significance (VUS) was 7.5% (10/132). In addition, seven novel variants (five in BRCA2 and two in BRCA1 gene), never previously reported, were identified. Our approach represents a robust and easy-to-use method for full BRCA1/2 screening. However, a consistent number of our high-risk families still remained without a satisfying answer. Necessarily, further collective efforts must be directed to a definitive classification of VUSs. The future auspice is that the use of multi-gene panel and more advanced screenings, such as whole exome sequencing and/or RNA seq, in routine diagnostics increases the detection rate.
Collapse
|
58
|
Padilla N, Moles-Fernández A, Riera C, Montalban G, Özkan S, Ootes L, Bonache S, Díez O, Gutiérrez-Enríquez S, de la Cruz X. BRCA1- and BRCA2-specific in silico tools for variant interpretation in the CAGI 5 ENIGMA challenge. Hum Mutat 2019; 40:1593-1611. [PMID: 31112341 DOI: 10.1002/humu.23802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 11/09/2022]
Abstract
BRCA1 and BRCA2 (BRCA1/2) germline variants disrupting the DNA protective role of these genes increase the risk of hereditary breast and ovarian cancers. Correct identification of these variants then becomes clinically relevant, because it may increase the survival rates of the carriers. Unfortunately, we are still unable to systematically predict the impact of BRCA1/2 variants. In this article, we present a family of in silico predictors that address this problem, using a gene-specific approach. For each protein, we have developed two tools, aimed at predicting the impact of a variant at two different levels: Functional and clinical. Testing their performance in different datasets shows that specific information compensates the small number of predictive features and the reduced training sets employed to develop our models. When applied to the variants of the BRCA1/2 (ENIGMA) challenge in the fifth Critical Assessment of Genome Interpretation (CAGI 5) we find that these methods, particularly those predicting the functional impact of variants, have a good performance, identifying the large compositional bias towards neutral variants in the CAGI sample. This performance is further improved when incorporating to our prediction protocol estimates of the impact on splicing of the target variant.
Collapse
Affiliation(s)
- Natàlia Padilla
- Research Unit in Clinical and Translational Bioinformatics, Vall d'Hebron Institute of Research (VHIR). Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Casandra Riera
- Research Unit in Clinical and Translational Bioinformatics, Vall d'Hebron Institute of Research (VHIR). Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Montalban
- Oncogenetics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Selen Özkan
- Research Unit in Clinical and Translational Bioinformatics, Vall d'Hebron Institute of Research (VHIR). Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lars Ootes
- Research Unit in Clinical and Translational Bioinformatics, Vall d'Hebron Institute of Research (VHIR). Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sandra Bonache
- Oncogenetics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Orland Díez
- Oncogenetics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Area of Clinical and Molecular Genetics, University Hospital of Vall d'Hebron, Barcelona, Spain
| | | | - Xavier de la Cruz
- Research Unit in Clinical and Translational Bioinformatics, Vall d'Hebron Institute of Research (VHIR). Universitat Autònoma de Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
59
|
Rashid MU, Khan FA, Muhammad N, Loya A, Hamann U. Prevalence of PALB2 Germline Mutations in Early-onset and Familial Breast/Ovarian Cancer Patients from Pakistan. Cancer Res Treat 2019; 51:992-1000. [PMID: 30309218 PMCID: PMC6639217 DOI: 10.4143/crt.2018.356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/10/2018] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Partner and localizer of BRCA2 (PALB2) is a breast cancer susceptibility gene that plays an important role in DNA repair. This is the first study assessing the prevalence of PALB2 mutations in early-onset and familial breast/ovarian cancer patients from Pakistan. MATERIALS AND METHODS PALB2 mutation screening was performed in 370 Pakistani patients with early-onset and familial breast/ovarian cancer, who were negative for BRCA1, BRCA2, TP53, CHEK2, and RAD51C mutations, using denaturing high-performance liquid chromatography analysis. Mutations were confirmed by DNA sequencing. Novel PALB2 alterations were analyzed for their potential effect on protein function or splicing using various in silico prediction tools. Three-hundred and seventy-two healthy controls were screened for the presence of the identified (potentially) functional mutations. RESULTS A novel nonsense mutation, p.Y743*, was identified in one familial breast cancer patient (1/127, 0.8%). Besides, four in silico-predicted potentially functional mutations including three missense mutations and one 5' untranslated region mutation were identified: p.D498Y, novel p.G644R, novel p.E744K, and novel c.-134_-133delTCinsGGGT. The mutations p.Y743* and p.D498Y were identified in two familial patients diagnosed with unilateral or synchronous bilateral breast cancer at the ages of 29 and 39, respectively. The other mutations were identified in an early-onset (≤ 30 years of age) breast cancer patient each. All five mutations were absent in 372 healthy controls suggesting that they are disease associated. CONCLUSION Our findings show that PALB2 mutations account for a small proportion of early-onset and hereditary breast/ovarian cancer cases in Pakistan.
Collapse
Affiliation(s)
- Muhammad Usman Rashid
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Faiz Ali Khan
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Noor Muhammad
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Asif Loya
- Department of Pathology, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
60
|
Liu W, Pajusalu S, Lake NJ, Zhou G, Ioannidis N, Mittal P, Johnson NE, Weihl CC, Williams BA, Albrecht DE, Rufibach LE, Lek M. Estimating prevalence for limb-girdle muscular dystrophy based on public sequencing databases. Genet Med 2019; 21:2512-2520. [PMID: 31105274 DOI: 10.1038/s41436-019-0544-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Limb-girdle muscular dystrophies (LGMD) are a genetically heterogeneous category of autosomal inherited muscle diseases. Many genes causing LGMD have been identified, and clinical trials are beginning for treatment of some genetic subtypes. However, even with the gene-level mechanisms known, it is still difficult to get a robust and generalizable prevalence estimation for each subtype due to the limited amount of epidemiology data and the low incidence of LGMDs. METHODS Taking advantage of recently published exome and genome sequencing data from the general population, we used a Bayesian method to develop a robust disease prevalence estimator. RESULTS This method was applied to nine recessive LGMD subtypes. The estimated disease prevalence calculated by this method was largely comparable with published estimates from epidemiological studies; however, it highlighted instances of possible underdiagnosis for LGMD2B and 2L. CONCLUSION The increasing size of aggregated population variant databases will allow for robust and reproducible prevalence estimates of recessive disease, which is critical for the strategic design and prioritization of clinical trials.
Collapse
Affiliation(s)
- Wei Liu
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Sander Pajusalu
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.,Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia
| | - Nicole J Lake
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.,Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia
| | - Geyu Zhou
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Nilah Ioannidis
- Jain Foundation, Seattle, WA, USA.,Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Plavi Mittal
- Jain Foundation, Seattle, WA, USA.,In-Depth Genomics, Bellevue, WA, USA
| | - Nicholas E Johnson
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA
| | - Conrad C Weihl
- Department of Neurology, Washington University School of Medicine, St. Louis,, MO, USA
| | | | | | | | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
61
|
Zhang J, Yang Y, Li P, Yan Y, Lv T, Zhao T, Zeng X, Li D, Zhou X, Chen H, Su J, Yang T, He J, Zhu B. Analysis of deletional hereditary persistence of fetal hemoglobin/δβ-thalassemia and δ-globin gene mutations in Southerwestern China. Mol Genet Genomic Med 2019; 7:e706. [PMID: 31044540 PMCID: PMC6565566 DOI: 10.1002/mgg3.706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 03/31/2019] [Accepted: 04/01/2019] [Indexed: 01/07/2023] Open
Abstract
Background Deletional hereditary persistence of fetal hemoglobin (HPFH)/δβ‐thalassemia and δ‐thalassemia are rare inherited disorders which may complicate the diagnosis of β‐thalassemia. The aim of this study was to reveal the frequency of these two disorders in Southwestern China. Methods A total of 33,596 subjects were enrolled for deletional HPFH/δβ‐thalassemia, and positive individuals with high fetal hemoglobin (Hb F) level were diagnosed by multiplex ligation‐dependent probe amplification (MLPA). A total of 17,834 subjects were analyzed for mutations in the δ‐globin gene. Positive samples with low Hb A2 levels were confirmed by δ‐globin gene sequencing. Furthermore, the pathogenicity and construction of a selected δ‐globin mutation were analyzed. Results A total of 92 suspected cases with Hb F ≥5.0% were further characterized by MLPA. Eight different deletional HPFH/δβ‐thalassemia were observed at a frequency of 0.024%. In addition, 195 cases suspected to have a δ‐globin gene mutation (Hb A2 ≤2.0%) were characterized by molecular analysis. δ‐Globin gene mutation was found at a frequency of 0.49% in Yunnan. The pathogenicity and construction for a selected δ‐globin mutation was predicted. Conclusion Screening of these two disorders was analyzed in Southwestern China, which could define the molecular basis of these conditions in this population.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Obstetrics and GynecologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
- Department of HematologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Yang Yang
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Peng Li
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Yuanlong Yan
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Tao Lv
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Tingting Zhao
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Xiaohong Zeng
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Dongmei Li
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Xiaoyan Zhou
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Hong Chen
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Jie Su
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Tonghua Yang
- Department of HematologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Jing He
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
| | - Baosheng Zhu
- Department of Obstetrics and GynecologyThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Genetic Diagnosis Center, Yunnan Provincial Key Laboratory for Birth Defects and Genetic DiseasesThe First People’s Hospital of Yunnan ProvinceKunmingChina
- Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| |
Collapse
|
62
|
Comparison of Predictive In Silico Tools on Missense Variants in GJB2, GJB6, and GJB3 Genes Associated with Autosomal Recessive Deafness 1A (DFNB1A). ScientificWorldJournal 2019; 2019:5198931. [PMID: 31015822 PMCID: PMC6446107 DOI: 10.1155/2019/5198931] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/25/2019] [Accepted: 02/03/2019] [Indexed: 01/30/2023] Open
Abstract
In silico predictive software allows assessing the effect of amino acid substitutions on the structure or function of a protein without conducting functional studies. The accuracy of in silico pathogenicity prediction tools has not been previously assessed for variants associated with autosomal recessive deafness 1A (DFNB1A). Here, we identify in silico tools with the most accurate clinical significance predictions for missense variants of the GJB2 (Cx26), GJB6 (Cx30), and GJB3 (Cx31) connexin genes associated with DFNB1A. To evaluate accuracy of selected in silico tools (SIFT, FATHMM, MutationAssessor, PolyPhen-2, CONDEL, MutationTaster, MutPred, Align GVGD, and PROVEAN), we tested nine missense variants with previously confirmed clinical significance in a large cohort of deaf patients and control groups from the Sakha Republic (Eastern Siberia, Russia): Сх26: p.Val27Ile, p.Met34Thr, p.Val37Ile, p.Leu90Pro, p.Glu114Gly, p.Thr123Asn, and p.Val153Ile; Cx30: p.Glu101Lys; Cx31: p.Ala194Thr. We compared the performance of the in silico tools (accuracy, sensitivity, and specificity) by using the missense variants in GJB2 (Cx26), GJB6 (Cx30), and GJB3 (Cx31) genes associated with DFNB1A. The correlation coefficient (r) and coefficient of the area under the Receiver Operating Characteristic (ROC) curve as alternative quality indicators of the tested programs were used. The resulting ROC curves demonstrated that the largest coefficient of the area under the curve was provided by three programs: SIFT (AUC = 0.833, p = 0.046), PROVEAN (AUC = 0.833, p = 0.046), and MutationAssessor (AUC = 0.833, p = 0.002). The most accurate predictions were given by two tested programs: SIFT and PROVEAN (Ac = 89%, Se = 67%, Sp = 100%, r = 0.75, AUC = 0.833). The results of this study may be applicable for analysis of novel missense variants of the GJB2 (Cx26), GJB6 (Cx30), and GJB3 (Cx31) connexin genes.
Collapse
|
63
|
Schoonen M, Smuts I, Louw R, Elson JL, van Dyk E, Jonck LM, Rodenburg RJT, van der Westhuizen FH. Panel-Based Nuclear and Mitochondrial Next-Generation Sequencing Outcomes of an Ethnically Diverse Pediatric Patient Cohort with Mitochondrial Disease. J Mol Diagn 2019; 21:503-513. [PMID: 30872186 DOI: 10.1016/j.jmoldx.2019.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/08/2019] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Mitochondrial disease (MD) is a group of rare inherited disorders with clinical heterogeneous phenotypes. Recent advances in next-generation sequencing (NGS) allow for rapid genetic diagnostics in patients who experience MD, resulting in significant strides in determining its etiology. This, however, has not been the case in many patient populations. We report on a molecular diagnostic study using mitochondrial DNA and targeted nuclear DNA (nDNA) NGS of an extensive cohort of predominantly sub-Saharan African pediatric patients with clinical and biochemically defined MD. Patients in this novel cohort presented mostly with muscle involvement (73%). Of the original 212 patients, a muscle respiratory chain deficiency was identified in 127 cases. Genetic analyses were conducted for these 127 cases based on biochemical deficiencies, for both mitochondrial (n = 123) and nDNA using panel-based NGS (n = 86). As a pilot investigation, whole-exome sequencing was performed in a subset of African patients (n = 8). These analyses resulted in the identification of a previously reported pathogenic mitochondrial DNA variant and seven pathogenic or likely pathogenic nDNA variants (ETFDH, SURF1, COQ6, RYR1, STAC3, ALAS2, and TRIOBP), most of which were identified via whole-exome sequencing. This study contributes to knowledge of MD etiology in an understudied, ethnically diverse population; highlights inconsistencies in genotype-phenotype correlations; and proposes future directions for diagnostic approaches in such patient populations.
Collapse
Affiliation(s)
- Maryke Schoonen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Izelle Smuts
- Department of Paediatrics and Child Health, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Joanna L Elson
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa; Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Etresia van Dyk
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Lindi-Maryn Jonck
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Richard J T Rodenburg
- Department of Pediatrics, Radboudumc Amalia Childrens Hospital, Radboud Center for Mitochondrial Medicine, Nijmegen, the Netherlands
| | - Francois H van der Westhuizen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
64
|
Walpole S, Pritchard AL, Cebulla CM, Pilarski R, Stautberg M, Davidorf FH, de la Fouchardière A, Cabaret O, Golmard L, Stoppa-Lyonnet D, Garfield E, Njauw CN, Cheung M, Turunen JA, Repo P, Järvinen RS, van Doorn R, Jager MJ, Luyten GPM, Marinkovic M, Chau C, Potrony M, Höiom V, Helgadottir H, Pastorino L, Bruno W, Andreotti V, Dalmasso B, Ciccarese G, Queirolo P, Mastracci L, Wadt K, Kiilgaard JF, Speicher MR, van Poppelen N, Kilic E, Al-Jamal RT, Dianzani I, Betti M, Bergmann C, Santagata S, Dahiya S, Taibjee S, Burke J, Poplawski N, O’Shea SJ, Newton-Bishop J, Adlard J, Adams DJ, Lane AM, Kim I, Klebe S, Racher H, Harbour JW, Nickerson ML, Murali R, Palmer JM, Howlie M, Symmons J, Hamilton H, Warrier S, Glasson W, Johansson P, Robles-Espinoza CD, Ossio R, de Klein A, Puig S, Ghiorzo P, Nielsen M, Kivelä TT, Tsao H, Testa JR, Gerami P, Stern MH, Paillerets BBD, Abdel-Rahman MH, Hayward NK. Comprehensive Study of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families Worldwide. J Natl Cancer Inst 2018; 110:1328-1341. [PMID: 30517737 PMCID: PMC6292796 DOI: 10.1093/jnci/djy171] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/17/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
Background The BRCA1-associated protein-1 (BAP1) tumor predisposition syndrome (BAP1-TPDS) is a hereditary tumor syndrome caused by germline pathogenic variants in BAP1 encoding a tumor suppressor associated with uveal melanoma, mesothelioma, cutaneous melanoma, renal cell carcinoma, and cutaneous BAP1-inactivated melanocytic tumors. However, the full spectrum of tumors associated with the syndrome is yet to be determined. Improved understanding of the BAP1-TPDS is crucial for appropriate clinical management of BAP1 germline variant carriers and their families, including genetic counseling and surveillance for new tumors. Methods We collated germline variant status, tumor diagnoses, and information on BAP1 immunohistochemistry or loss of somatic heterozygosity on 106 published and 75 unpublished BAP1 germline variant-positive families worldwide to better characterize the genotypes and phenotypes associated with the BAP1-TPDS. Tumor spectrum and ages of onset were compared between missense and null variants. All statistical tests were two-sided. Results The 181 families carried 140 unique BAP1 germline variants. The collated data confirmed the core tumor spectrum associated with the BAP1-TPDS and showed that some families carrying missense variants can exhibit this phenotype. A variety of noncore BAP1-TPDS -associated tumors were found in families of variant carriers. Median ages of onset of core tumor types were lower in null than missense variant carriers for all tumors combined (P < .001), mesothelioma (P < .001), cutaneous melanoma (P < .001), and nonmelanoma skin cancer (P < .001). Conclusions This analysis substantially increases the number of pathogenic BAP1 germline variants and refines the phenotype. It highlights the need for a curated registry of germline variant carriers for proper assessment of the clinical phenotype of the BAP1-TPDS and pathogenicity of new variants, thus guiding management of patients and informing areas requiring further research.
Collapse
Affiliation(s)
- Sebastian Walpole
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- University of Queensland, Brisbane, QLD, Australia
| | - Antonia L Pritchard
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of the Highlands and Islands, Inverness, UK
| | - Colleen M Cebulla
- Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, OH
| | - Robert Pilarski
- Division of Human Genetics, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Meredith Stautberg
- Division of Human Genetics, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Frederick H Davidorf
- Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, OH
| | | | - Odile Cabaret
- Département de Biopathologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Lisa Golmard
- Département De Biologie Des Tumeurs, Institut Curie, Paris, France
| | - Dominique Stoppa-Lyonnet
- Département De Biologie Des Tumeurs, Institut Curie, Paris, France
- Institut Curie, PSL Research University, INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
- Sorbonne Paris Cité, University Paris-Descartes, Paris, France
| | - Erin Garfield
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ching-Ni Njauw
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA
| | - Mitchell Cheung
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Joni A Turunen
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pauliina Repo
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Reetta-Stiina Järvinen
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | | | | | | - Cindy Chau
- Department of Ophthalmology, LUMC, Leiden, The Netherlands
| | - Miriam Potrony
- Dermatology Department, Melanoma Unit, Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Hildur Helgadottir
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - William Bruno
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Virginia Andreotti
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Bruna Dalmasso
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Giulia Ciccarese
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Queirolo
- Medical Oncology Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Mastracci
- Department of Surgical and Diagnostic Sciences, Pathology Unit, University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Karin Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Jens Folke Kiilgaard
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Michael R Speicher
- Institute of Human Genetics, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Natasha van Poppelen
- Department of Ophthalmology
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Emine Kilic
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rana’a T Al-Jamal
- Department of Ophthalmology, Ocular Oncology Service, Helsinki University Central Hospital, Helsinki, Finland
| | - Irma Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marta Betti
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Carsten Bergmann
- Bioscientia Center for Human Genetics, Ingelheim, Germany
- Department of Medicine IV, Faculty of Medicine, Medical Center—University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Saleem Taibjee
- Department of Dermatology, Dorset County Hospital NHS Foundation Trust, Dorchester, UK
| | - Jo Burke
- Tasmanian Clinical Genetics Service, Royal Hobart Hospital, TAS, Australia
| | - Nicola Poplawski
- Adult Genetics Unit, Medicine Directorate, Royal Adelaide Hospital, Adelaide, SA, Australia
- University Department of Paediatrics, University of Adelaide, Adelaide, SA, Australia
| | - Sally J O’Shea
- Dermatology Department, Mater Private Hospital Cork, Citygate, Mahon, Cork, Ireland
| | - Julia Newton-Bishop
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Julian Adlard
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| | - Anne-Marie Lane
- Department of Ophthalmology, Ocular Melanoma Center and Retina Service, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| | - Ivana Kim
- Department of Ophthalmology, Ocular Melanoma Center and Retina Service, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| | - Sonja Klebe
- Department of Anatomical Pathology, Flinders University and SA Pathology at Flinders Medical Centre, Adelaide, SA, Australia
| | | | - J William Harbour
- Bascom Palmer Eye Institute, Sylvester Comprehensive Cancer Center and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Michael L Nickerson
- Laboratory of Translational Genomics, National Cancer Institute, Bethesda, MD
| | - Rajmohan Murali
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jane M Palmer
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Madeleine Howlie
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Judith Symmons
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Hayley Hamilton
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sunil Warrier
- Queensland Ocular Oncology Service, The Terrace Eye Centre, Brisbane, QLD, Australia
| | - William Glasson
- Queensland Ocular Oncology Service, The Terrace Eye Centre, Brisbane, QLD, Australia
| | - Peter Johansson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Carla Daniela Robles-Espinoza
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Santiago de Querétaro, Mexico
| | - Raul Ossio
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Santiago de Querétaro, Mexico
| | - Annelies de Klein
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Susana Puig
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Paola Ghiorzo
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Maartje Nielsen
- Department of Clinical Genetics, LUMC, Leiden, The Netherlands
| | - Tero T Kivelä
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hensin Tsao
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA
- Massachusetts General Hospital Cancer Center, Boston, MA
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Pedram Gerami
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
- The Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL
| | - Marc-Henri Stern
- Département De Biologie Des Tumeurs, Institut Curie, Paris, France
- Institut Curie, PSL Research University, INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Brigitte Bressac-de Paillerets
- Département de Biopathologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Mohamed H Abdel-Rahman
- Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, OH
- Division of Human Genetics, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Department of Pathology, Menoufiya University, Shebin El-Kom, Egypt
| | | |
Collapse
|
65
|
Subbotina E, Williams N, Sampson BA, Tang Y, Coetzee WA. Functional characterization of TRPM4 variants identified in sudden unexpected natural death. Forensic Sci Int 2018; 293:37-46. [PMID: 30391667 DOI: 10.1016/j.forsciint.2018.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/11/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND The TRPM4 gene encodes the subunit of the Ca2+-activated nonselective cation channel, which is enriched in the specialized cardiac conduction system and Purkinje fibers. To date, several putative disease-causing variants in TRPM4 have been reported to be associated with cardiac arrhythmia and progressive conduction disease. Here, we report the functional effects of previously uncharacterized variants of uncertain significance (VUS) that we have found while performing a "genetic autopsy" in individuals who have suffered sudden unexpected death (SUD) in the New York City area. METHODS AND RESULTS We have identified thirteen uncommon missense VUS in TRPM4 by testing 95 targeted genes implicated in channelopathy and cardiomyopathy in 330 cases of SUD. In several cases there were co-existing VUS in one or more other genes that were tested. We selected four TRPM4 VUS (C20S, A380V, L595V and I1082S) for functional characterization, since these cases lacked detectable variants in other genes of our testing panel. Two of the cases were infants, one was a child and one an adult. RNA-seq data analysis showed that the longer TRPM4b splice variant is predominantly expressed in adult and fetal human heart. We therefore used site-directed mutagenesis to introduce these variants in a TRPM4b cDNA. HEK293 cells were transfected with the cDNAs and patch clamping was performed to assess the functional consequences of the TRPM4 mutants. The TRPM4 current was recorded in excised patches and was significantly reduced by each of the mutants. The total protein level of TRPM4-C20S was markedly decreased, whereas the A380V and L595V mutants exhibited decreased surface expression. The TRPM4-A380V current rapidly desensitized following patch excision. CONCLUSIONS Each of the VUS tested caused a defect in TRPM4 channel function via distinctly different mechanisms, hence, it lays the foundation for further co-segregation family studies and animal studies of the TRPM4 variants.
Collapse
Affiliation(s)
- Ekaterina Subbotina
- Department of Pediatrics, Physiology & Neuroscience and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
| | - Nori Williams
- Molecular Genetics Laboratory, Office of Chief Medical Examiner, New York, NY, USA
| | - Barbara A Sampson
- Molecular Genetics Laboratory, Office of Chief Medical Examiner, New York, NY, USA
| | - Yingying Tang
- Molecular Genetics Laboratory, Office of Chief Medical Examiner, New York, NY, USA
| | - William A Coetzee
- Department of Pediatrics, Physiology & Neuroscience and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA; Department of Physiology & Neuroscience and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
66
|
Wierbowski SD, Fragoza R, Liang S, Yu H. Extracting Complementary Insights from Molecular Phenotypes for Prioritization of Disease-Associated Mutations. CURRENT OPINION IN SYSTEMS BIOLOGY 2018; 11:107-116. [PMID: 31086831 PMCID: PMC6510504 DOI: 10.1016/j.coisb.2018.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rapid advances in next-generation sequencing technology have resulted in an explosion of whole-exome/genome sequencing data, providing an unprecedented opportunity to identify disease- and trait-associated variants in humans on a large scale. To date, the long-standing paradigm has leveraged fitness-based approximations to translate this ever-expanding sequencing data into causal insights in disease. However, while this approach robustly identifies variants under evolutionary constraint, it fails to provide molecular insights. Moreover, complex disease phenomena often violate standard assumptions of a direct organismal phenotype to overall fitness effect relationship. Here we discuss the potential of a molecular phenotype-oriented paradigm to uniquely identify candidate disease-causing mutations from the human genetic background. By providing a direct connection between single nucleotide mutations and observable organismal and cellular phenotypes associated with disease, we suggest that molecular phenotypes can readily incorporate alongside established fitness-based methodologies to provide complementary insights to the functional impact of human mutations. Lastly, we discuss how integrated approaches between molecular phenotypes and fitness-based perspectives facilitate new insights into the molecular mechanisms underlying disease-associated mutations while also providing a platform for improved interpretation of epistasis in human disease.
Collapse
Affiliation(s)
- Shayne D. Wierbowski
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Robert Fragoza
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Siqi Liang
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Haiyuan Yu
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
67
|
Lorenz-Guertin JM, Bambino MJ, Jacob TC. γ2 GABA AR Trafficking and the Consequences of Human Genetic Variation. Front Cell Neurosci 2018; 12:265. [PMID: 30190672 PMCID: PMC6116786 DOI: 10.3389/fncel.2018.00265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/02/2018] [Indexed: 11/13/2022] Open
Abstract
GABA type A receptors (GABAARs) mediate the majority of fast inhibitory neurotransmission in the central nervous system (CNS). Most prevalent as heteropentamers composed of two α, two β, and a γ2 subunit, these ligand-gated ionotropic chloride channels are capable of extensive genetic diversity (α1-6, β1-3, γ1-3, δ, 𝜀, 𝜃, π, ρ1-3). Part of this selective GABAAR assembly arises from the critical role for γ2 in maintaining synaptic receptor localization and function. Accordingly, mutations in this subunit account for over half of the known epilepsy-associated genetic anomalies identified in GABAARs. Fundamental structure-function studies and cellular pathology investigations have revealed dynamic GABAAR trafficking and synaptic scaffolding as critical regulators of GABAergic inhibition. Here, we introduce in vitro and in vivo findings regarding the specific role of the γ2 subunit in receptor trafficking. We then examine γ2 subunit human genetic variation and assess disease related phenotypes and the potential role of altered GABAAR trafficking. Finally, we discuss new-age imaging techniques and their potential to provide novel insight into critical regulatory mechanisms of GABAAR function.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew J Bambino
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|