101
|
Zamani G, Hosseini Bereshneh A, Azizi Malamiri R, Bagheri S, Moradi K, Ashrafi MR, Tavasoli AR, Mohammadi M, Badv RS, Ghahvechi Akbari M, Heidari M. The First Comprehensive Cohort of the Duchenne Muscular Dystrophy in Iranian Population: Mutation Spectrum of 314 Patients and Identifying Two Novel Nonsense Mutations. J Mol Neurosci 2020; 70:1565-1573. [DOI: 10.1007/s12031-020-01594-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/14/2020] [Indexed: 12/17/2022]
|
102
|
Liu F, Wang J, Xing Y, Li T. Mutation screening of 17 candidate genes in a cohort of 67 probands with early-onset high myopia. Ophthalmic Physiol Opt 2020; 40:271-280. [PMID: 32215939 DOI: 10.1111/opo.12683] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE To detect variants in 17 known potentially causative genes for non-syndromic myopia in 67 Tujia Chinese patients with early-onset high myopia (eo-HM). METHODS DNA from 67 unrelated patients with early onset (<7 years old) high myopia (refraction error ≤ -6.00D or axial length > 26 mm) were subjected to whole-exome sequencing (WES). Variants in 17 candidate genes were analysed by multistep bioinformatics analysis. Subsequently, Sanger sequencing was used to verify identified candidate mutations and to assess available family members for co-segregation with myopia. RESULTS A multistep systematic analysis of variants in 17 potentially causative genes for eo-HM revealed four novel pathogenic mutations and three potential pathogenic mutations in 4 of 17 genes in 7 of 67 (10.4%) probands. The pathogenic group included one missense mutation (c.100G > C, p.Asp34His) and one splice donor mutation (c.989 + 1G >A) in ARR3, one missense mutation (c.995C > A, p.Thr332Lys) in NDUFAF7 and one novel frameshift mutation (c.726dupA, p.Arg243fs*140) in SLC39A5. The potential pathogenic group included two missense mutations (c.3266A > G, p.Tyr1089Cys; c.913G > A, p.Glu305Lys) in ZNF644 and one missense mutation (c.960T > A, p.His320Gln) in NDUFAF7. Sequence changes were confirmed by Sanger sequencing; all had an allele frequency <0.01 in the 1000G, EVS, ExAC and gnomAD databases. Additionally, both the pathogenic and potentially pathogenic mutations were predicted to be damaging by SIFT, Polyphen-2, PROVEAN, MutationTaster2, CADD and REVEL except the p.Tyr1089Cys and p.Glu305Lys changes were predicted to be neutral by PROVEAN. CONCLUSION Our research provides more evidence to support the hypothesis that mutations in ARR3, SLC39A5 and NDUFAF7 are disease-causing genes for eo-HM and broadens the eo-HM mutation spectrum among different ethnic groups. It also deepens understanding of the contributions of ARR3, SLC39A5, and NDUFAF7 to eo-HM.
Collapse
Affiliation(s)
- Fang Liu
- Department of Ophthalmology, The Central Hospital of Enshi Tujia And Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, China.,Department of Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junwen Wang
- Department of Ophthalmology, The Central Hospital of Enshi Tujia And Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, China
| | - Yiqiao Xing
- Department of Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tuo Li
- Department of Ophthalmology, The Central Hospital of Enshi Tujia And Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, China
| |
Collapse
|
103
|
Ethnogeographic and inter-individual variability of human ABC transporters. Hum Genet 2020; 139:623-646. [PMID: 32206879 PMCID: PMC7170817 DOI: 10.1007/s00439-020-02150-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/16/2020] [Indexed: 12/19/2022]
Abstract
ATP-binding cassette (ABC) transporters constitute a superfamily of 48 structurally similar membrane transporters that mediate the ATP-dependent cellular export of a plethora of endogenous and xenobiotic substances. Importantly, genetic variants in ABC genes that affect gene function have clinically important effects on drug disposition and can be predictors of the risk of adverse drug reactions and efficacy of chemotherapeutics, calcium channel blockers, and protease inhibitors. Furthermore, loss-of-function of ABC transporters is associated with a variety of congenital disorders. Despite their clinical importance, information about the frequencies and global distribution of functionally relevant ABC variants is limited and little is known about the overall genetic complexity of this important gene family. Here, we systematically mapped the genetic landscape of the entire human ABC superfamily using Next-Generation Sequencing data from 138,632 individuals across seven major populations. Overall, we identified 62,793 exonic variants, 98.5% of which were rare. By integrating five computational prediction algorithms with structural mapping approaches using experimentally determined crystal structures, we found that the functional ABC variability is extensive and highly population-specific. Every individual harbored between 9.3 and 13.9 deleterious ABC variants, 76% of which were found only in a single population. Carrier rates of pathogenic variants in ABC transporter genes associated with autosomal recessive congenital diseases, such as cystic fibrosis or pseudoxanthoma elasticum, closely mirrored the corresponding population-specific disease prevalence, thus providing a novel resource for rare disease epidemiology. Combined, we provide the most comprehensive, systematic, and consolidated overview of ethnogeographic ABC transporter variability with important implications for personalized medicine, clinical genetics, and precision public health.
Collapse
|
104
|
Kato M, Yagami A, Tsukamoto T, Shinkai Y, Kato T, Kurahashi H. Novel mutation in the KITLG gene in familial progressive hyperpigmentation with or without hypopigmentation. J Dermatol 2020; 47:669-672. [PMID: 32189379 DOI: 10.1111/1346-8138.15313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/18/2020] [Indexed: 11/27/2022]
Abstract
We herein report a novel mutation in familial progressive hyper- and hypopigmentation (FPHH). The KITLG gene encoding the KIT ligand protein is a disease-causing gene for FPHH. Various disease-causing gain-of-function mutations, which reside within or adjacent to the conserved VTNN motif of this gene, have been described to date. We have now identified a novel KITLG mutation, c.337G>A (p.Glu113Lys), in FPHH which is located within another ligand-receptor interaction site.
Collapse
Affiliation(s)
- Maki Kato
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Akiko Yagami
- Departments of, Department of, Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tetsuya Tsukamoto
- Department of, Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yasuko Shinkai
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Takema Kato
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Hiroki Kurahashi
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| |
Collapse
|
105
|
Li S, Xiao X, Yi Z, Sun W, Wang P, Zhang Q. RPE65 mutation frequency and phenotypic variation according to exome sequencing in a tertiary centre for genetic eye diseases in China. Acta Ophthalmol 2020; 98:e181-e190. [PMID: 31273949 PMCID: PMC7079156 DOI: 10.1111/aos.14181] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/07/2019] [Indexed: 01/14/2023]
Abstract
PURPOSE Retinoid isomerohydrolase RPE65 has received a tremendous amount of attention due to successful clinical gene therapy for Leber congenital amaurosis (LCA) cases caused by RPE65 mutations. This study aimed to evaluate the frequency of RPE65 mutations and the associated phenotypes based on exome sequencing. METHODS RPE65 variants were collected from exome sequencing data obtained from 2133 probands with different forms of hereditary retinal degeneration (HRD). Clinical data were collected from probands with homozygous or compound heterozygous variants in RPE65. Associated phenotypes were characterized based on clinical data. RESULTS Biallelic RPE65 mutations were detected in 18 families, including eight with LCA, five with early-onset retinal degeneration, four with fundus albipunctatus-like (FA-like) changes and one with high hyperopia. These cases accounted for approximately 3.0% (8/269) of LCA and 0.8% (18/2133) of HRD cases. An almost identical FA-like change was identified in seven patients from four unrelated families with RPE65 mutations. Classification of mutations suggested that FA-like changes may be associated with biallelic missense mutations in RPE65. CONCLUSION Fundus albipunctatus-like (FA-like) change, a common characteristic fundus sign in RPE65 biallelic mutations, was unexpected but was confirmed by the finding that affected siblings from different families exhibited similar phenotypes. These results enrich our understanding of RPE65 mutation frequencies and their associated phenotypic variants.
Collapse
Affiliation(s)
- Shiqiang Li
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Xueshan Xiao
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Zhen Yi
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Wenmin Sun
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Panfeng Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Qingjiong Zhang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
106
|
Vallejos-Vidal E, Reyes-Cerpa S, Rivas-Pardo JA, Maisey K, Yáñez JM, Valenzuela H, Cea PA, Castro-Fernandez V, Tort L, Sandino AM, Imarai M, Reyes-López FE. Single-Nucleotide Polymorphisms (SNP) Mining and Their Effect on the Tridimensional Protein Structure Prediction in a Set of Immunity-Related Expressed Sequence Tags (EST) in Atlantic Salmon ( Salmo salar). Front Genet 2020; 10:1406. [PMID: 32174954 PMCID: PMC7056891 DOI: 10.3389/fgene.2019.01406] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Single-nucleotide polymorphisms (SNPs) are single genetic code variations considered one of the most common forms of nucleotide modifications. Such SNPs can be located in genes associated to immune response and, therefore, they may have direct implications over the phenotype of susceptibility to infections affecting the productive sector. In this study, a set of immune-related genes (cc motif chemokine 19 precursor [ccl19], integrin β2 (itβ2, also named cd18), glutathione transferase omega-1 [gsto-1], heat shock 70 KDa protein [hsp70], major histocompatibility complex class I [mhc-I]) were analyzed to identify SNPs by data mining. These genes were chosen based on their previously reported expression on infectious pancreatic necrosis virus (IPNV)-infected Atlantic salmon phenotype. The available EST sequences for these genes were obtained from the Unigene database. Twenty-eight SNPs were found in the genes evaluated and identified most of them as transition base changes. The effect of the SNPs located on the 5'-untranslated region (UTR) or 3'-UTR upon transcription factor binding sites and alternative splicing regulatory motifs was assessed and ranked with a low-medium predicted FASTSNP score risk. Synonymous SNPs were found on itβ2 (c.2275G > A), gsto-1 (c.558G > A), and hsp70 (c.1950C > T) with low FASTSNP predicted score risk. The difference in the relative synonymous codon usage (RSCU) value between the variant codons and the wild-type codon (ΔRSCU) showed one negative (hsp70 c.1950C > T) and two positive ΔRSCU values (itβ2 c.2275G > A; gsto-1 c.558G > A), suggesting that these synonymous SNPs (sSNPs) may be associated to differences in the local rate of elongation. Nonsynonymous SNPs (nsSNPs) in the gsto-1 translatable gene region were ranked, using SIFT and POLYPHEN web-tools, with the second highest (c.205A > G; c484T > C) and the highest (c.499T > C; c.769A > C) predicted score risk possible. Using homology modeling to predict the effect of these nonsynonymous SNPs, the most relevant nucleotide changes for gsto-1 were observed for the nsSNPs c.205A > G, c484T > C, and c.769A > C. Molecular dynamics was assessed to analyze if these GSTO-1 variants have significant differences in their conformational dynamics, suggesting these SNPs could have allosteric effects modulating its catalysis. Altogether, these results suggest that candidate SNPs identified may play a crucial potential role in the immune response of Atlantic salmon.
Collapse
Affiliation(s)
- Eva Vallejos-Vidal
- Department of Cell Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sebastián Reyes-Cerpa
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Jaime Andrés Rivas-Pardo
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Kevin Maisey
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - José M. Yáñez
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - Hector Valenzuela
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Pablo A. Cea
- Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | | | - Lluis Tort
- Department of Cell Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ana M. Sandino
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Mónica Imarai
- Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe E. Reyes-López
- Department of Cell Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
107
|
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
|
108
|
Clayton EA, Khalid S, Ban D, Wang L, Jordan IK, McDonald JF. Tumor suppressor genes and allele-specific expression: mechanisms and significance. Oncotarget 2020; 11:462-479. [PMID: 32064050 PMCID: PMC6996918 DOI: 10.18632/oncotarget.27468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022] Open
Abstract
Recent findings indicate that allele-specific expression (ASE) at specific cancer driver gene loci may be of importance in onset/progression of the disease. Of particular interest are loss-of-function (LOF) of tumor suppressor gene (TSGs) alleles. While LOF tumor suppressor mutations are typically considered to be recessive, if these mutant alleles can be significantly differentially expressed relative to wild-type alleles in heterozygotes, the clinical consequences could be significant. LOF TSG alleles are shown to be segregating at high frequencies in world-wide populations of normal/healthy individuals. Matched sets of normal and tumor tissues isolated from 233 cancer patients representing four diverse tumor types demonstrate functionally important changes in patterns of ASE in individuals heterozygous for LOF TSG alleles associated with cancer onset/progression. While a variety of molecular mechanisms were identified as potentially contributing to changes in ASE patterns in cancer, changes in DNA copy number and allele-specific alternative splicing possibly mediated by antisense RNA emerged as predominant factors. In conclusion, LOF TSGs are segregating in human populations at significant frequencies indicating that many otherwise healthy individuals are at elevated risk of developing cancer. Changes in ASE between normal and cancer tissues indicates that LOF TSG alleles may contribute to cancer onset/progression even when heterozygous with wild-type functional alleles.
Collapse
Affiliation(s)
- Evan A. Clayton
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shareef Khalid
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Dongjo Ban
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Lu Wang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- PanAmerican Bioinformatics Institute, Cali, Colombia
| | - I. King Jordan
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- PanAmerican Bioinformatics Institute, Cali, Colombia
- Applied Bioinformatics Laboratory, Atlanta, GA, USA
| | - John F. McDonald
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
109
|
Shehzadi N, Hussain K, Bukhari NI, Islam M, Salman M, Khan MT. Speeding up the Development of 5-[(4-Chlorophenoxy)-Methyl]-1,3,4-Oxadiazole-2-Thiol as Successful Oral Drug Candidate Based on Physicochemical Characteristics. Pharm Chem J 2020. [DOI: 10.1007/s11094-020-02101-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
110
|
Tang N, Sandahl TD, Ott P, Kepp KP. Computing the Pathogenicity of Wilson's Disease ATP7B Mutations: Implications for Disease Prevalence. J Chem Inf Model 2019; 59:5230-5243. [PMID: 31751128 DOI: 10.1021/acs.jcim.9b00852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genetic variations in the gene encoding the copper-transport protein ATP7B are the primary cause of Wilson's disease. Controversially, clinical prevalence seems much smaller than the prevalence estimated by genetic screening tools, causing fear that many people are undiagnosed, although early diagnosis and treatment is essential. To address this issue, we benchmarked 16 state-of-the-art computational disease-prediction methods against established data of missense ATP7B mutations. Our results show that the quality of the methods varies widely. We show the importance of optimizing the threshold of the methods used to distinguish pathogenic from nonpathogenic mutations against data of clinically confirmed pathogenic and nonpathogenic mutations. We find that most methods use thresholds that predict too many ATP7B mutations to be pathogenic. Thus, our findings explain the current controversy on Wilson's disease prevalence because meta-analysis and text search methods include many computational estimates that lead to higher disease prevalence than clinically observed. As proteins and diseases differ widely, a one-size-fits-all threshold cannot distinguish pathogenic and nonpathogenic mutations efficiently, as shown here. We also show that amino acid changes with small evolutionary substitution probability, mainly due to amino acid volume, are more associated with the disease, implying a pathological effect on the conformational state of the protein, which could affect copper transport or adenosine triphosphate recognition and hydrolysis. These findings may be a first step toward a more quantitative genotype-phenotype relationship of Wilson's disease.
Collapse
Affiliation(s)
- Ning Tang
- DTU Chemistry , Technical University of Denmark , Kemitorvet 206 , 2800 Kongens Lyngby , Denmark
| | - Thomas D Sandahl
- Department of Hepatology and Gastroenterology , Aarhus University Hospital , 8200 Aarhus , Denmark
| | - Peter Ott
- Department of Hepatology and Gastroenterology , Aarhus University Hospital , 8200 Aarhus , Denmark
| | - Kasper P Kepp
- DTU Chemistry , Technical University of Denmark , Kemitorvet 206 , 2800 Kongens Lyngby , Denmark
| |
Collapse
|
111
|
Hernandez Cordero AI, Gonzales NM, Parker CC, Sokolof G, Vandenbergh DJ, Cheng R, Abney M, Sko A, Douglas A, Palmer AA, Gregory JS, Lionikas A. Genome-wide Associations Reveal Human-Mouse Genetic Convergence and Modifiers of Myogenesis, CPNE1 and STC2. Am J Hum Genet 2019; 105:1222-1236. [PMID: 31761296 PMCID: PMC6904802 DOI: 10.1016/j.ajhg.2019.10.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Muscle bulk in adult healthy humans is highly variable even after height, age, and sex are accounted for. Low muscle mass, due to fewer and/or smaller constituent muscle fibers, would exacerbate the impact of muscle loss occurring in aging or disease. Genetic variability substantially influences muscle mass differences, but causative genes remain largely unknown. In a genome-wide association study (GWAS) on appendicular lean mass (ALM) in a population of 85,750 middle-aged (aged 38-49 years) individuals from the UK Biobank (UKB), we found 182 loci associated with ALM (p < 5 × 10-8). We replicated associations for 78% of these loci (p < 5 × 10-8) with ALM in a population of 181,862 elderly (aged 60-74 years) individuals from UKB. We also conducted a GWAS on hindlimb skeletal muscle mass of 1,867 mice from an advanced intercross between two inbred strains (LG/J and SM/J); this GWAS identified 23 quantitative trait loci. Thirty-eight positional candidates distributed across five loci overlapped between the two species. In vitro studies of positional candidates confirmed CPNE1 and STC2 as modifiers of myogenesis. Collectively, these findings shed light on the genetics of muscle mass variability in humans and identify targets for the development of interventions for treatment of muscle loss. The overlapping results between humans and the mouse model GWAS point to shared genetic mechanisms across species.
Collapse
Affiliation(s)
- Ana I Hernandez Cordero
- School of Medicine, Medical Sciences, and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, UK AB24 3FX, UK
| | - Natalia M Gonzales
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Clarissa C Parker
- Department of Psychology, Middlebury College, Middlebury, VT 05753, USA; Program in Neuroscience, Middlebury College, Middlebury, VT, 05753, USA
| | - Greta Sokolof
- Department of Psychological and Brain Sciences, The University of Iowa, Iowa City, IA 52242, USA
| | - David J Vandenbergh
- Department of Biobehavioral Health, Penn State Institute for the Neurosciences, and Molecular, Cellular, and Integrative Sciences Program, Pennsylvania State University, University Park, PA 16802, USA
| | - Riyan Cheng
- Department of Health Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Mark Abney
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Andrew Sko
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer S Gregory
- School of Medicine, Medical Sciences, and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, UK AB24 3FX, UK
| | - Arimantas Lionikas
- School of Medicine, Medical Sciences, and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, UK AB24 3FX, UK.
| |
Collapse
|
112
|
Huang Y, Zhao J, Mao G, Lee GS, Zhang J, Bi L, Gu L, Chang Z, Valentino J, Li GM. Identification of novel genetic variants predisposing to familial oral squamous cell carcinomas. Cell Discov 2019; 5:57. [PMID: 31798960 PMCID: PMC6877579 DOI: 10.1038/s41421-019-0126-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common subtype of head and neck squamous cell carcinoma (HNSCC), but the pathogenesis underlying familial OSCCs is unknown. Here, we analyzed whole-genome sequences of a family with autosomal dominant expression of oral tongue cancer and identified proto-oncogenes VAV2 and IQGAP1 as the primary factors responsible for oral cancer in the family. These two genes are also frequently mutated in sporadic OSCCs and HNSCCs. Functional analysis revealed that the detrimental variants target tumorigenesis-associated pathways, thus confirming that these novel genetic variants help to establish a predisposition to familial OSCC.
Collapse
Affiliation(s)
- Yaping Huang
- 1Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jizhi Zhao
- 4Department of Stomatology, Peking Union Medical College Hospital, Beijing, 100730 China
| | - Guogen Mao
- 2Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| | - Grace Sanghee Lee
- 2Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| | - Jia Zhang
- 5Insititute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Lijun Bi
- 5Insititute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Liya Gu
- 1Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Zhijie Chang
- 6Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084 China
| | - Joseph Valentino
- 3Department of Otolaryngology, Head & Neck Surgery, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| | - Guo-Min Li
- 1Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA.,2Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| |
Collapse
|
113
|
Giri J, Moyer AM, Bielinski SJ, Caraballo PJ. Concepts Driving Pharmacogenomics Implementation Into Everyday Healthcare. Pharmgenomics Pers Med 2019; 12:305-318. [PMID: 31802928 PMCID: PMC6826176 DOI: 10.2147/pgpm.s193185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022] Open
Abstract
Pharmacogenomics (PGx) is often promoted as the domain of precision medicine with the greatest potential to readily impact everyday healthcare. Rapid advances in PGx knowledge derived from extensive basic and clinical research along with decreasing costs of laboratory testing have led to an increased interest in PGx and expectations of imminent clinical translation with substantial clinical impact. However, the implementation of PGx into clinical workflows is neither simple nor straightforward, and comprehensive processes and multidisciplinary collaboration are required. Several national and international institutions have pioneered models for implementing clinical PGx, and these initial models have led to a better understanding of unresolved challenges. In this review, we have categorized and explored the most relevant of these challenges to highlight potential gaps and present possible solutions. We describe the ongoing need for basic and clinical research to drive further developments in evidence-based medicine. Integration into daily clinical workflows introduces new challenges requiring innovative solutions; specifically those related to the electronic health record and embedded clinical decision support. We describe advances in PGx testing and result reporting and describe the critical need for increased standardization in these areas across laboratories. We also explore the complexity of the PGx knowledge required for clinical practice and the need for educational strategies to ensure adequate understanding among members of current and future healthcare teams. Finally, we evaluate knowledge obtained from previous implementation efforts and discuss how to best apply these learnings to future projects. Despite these challenges, the future of precision medicine appears promising due to the rapidity of recent advances in the field and current multidisciplinary efforts to effectively translate PGx to everyday clinical practice.
Collapse
Affiliation(s)
- Jyothsna Giri
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ann M Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Pedro J Caraballo
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
114
|
Yi Z, Xiao X, Li S, Sun W, Zhang Q. Pathogenicity discrimination and genetic test reference for CRX variants based on genotype-phenotype analysis. Exp Eye Res 2019; 189:107846. [PMID: 31626798 DOI: 10.1016/j.exer.2019.107846] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/27/2019] [Accepted: 10/14/2019] [Indexed: 12/22/2022]
Abstract
The cone-rod homeobox (CRX) gene is specifically expressed in developing and mature photoreceptors and is relatively conserved, with limited polymorphisms in coding regions. Rare variants in CRX are usually considered causative for different forms of retinal degeneration, but this might be problematic based on recent data. This study aimed to classify CRX variants based on a genotype-phenotype analysis of our data and the literature. Twenty-four CRX variants, including 14 novel variants, were detected in 37 Chinese families based on exome sequencing data obtained from 4971 Chinese probands with different forms of eye diseases. After detailed phenotypic analysis and cosegregation analysis in families with CRX variants, the 24 variants could be classified into three groups: benign (six), likely benign (six), and pathogenic (12). Somatic mosaicism was identified in a family with unaffected parents (the father had a mutant allele that was detected in approximately 17% of his leukocyte DNA) and two affected sons. Furthermore, a thorough reassessment was systematically performed for all 113 heterozygous variants as well as for their associated phenotypes from our cohort and patients previously reported. Two critical findings on the pathogenicity of CRX variants were obtained based on the genotype-phenotype correlation, family segregation and ensemble predicting methods: 1) approximately half of heterozygous missense variants are likely benign, and 2) heterozygous truncating variants affecting the homeodomain are likely benign. Truncating mutations after the homeodomain are likely associated with a more severe phenotype. Although most heterozygous pathogenic variants in CRX are associated with autosomal dominant retinal degeneration, a homozygous c.268C> T (p.Arg90Trp) substitution and homozygous complete deletion of CRX have been reported to cause Leber congenital amaurosis. In conclusion, many rare missense variants and some truncating variants in CRX are likely benign, although previously, they might have been predicted to be damaging by some online tools. Evaluation of the pathogenicity of a CRX variant should consider both its nature and location. The information obtained in this study is critical in the era of routine clinical genetic test, not only for CRX but also for many other genes with many more variants. Functional studies and additional genotype-phenotype analyses are expected to confirm these associations.
Collapse
Affiliation(s)
- Zhen Yi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xueshan Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Shiqiang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Wenmin Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Qingjiong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 Xianlie Road, Guangzhou, 510060, China.
| |
Collapse
|
115
|
Maffini F, Lorenzini D, Lepanto D, De Fiori E, Fumagalli C, Rappa A, Tagliabue M, Barberis M. A case of Warthin-like papillary thyroid carcinoma with diffuse sclerosing stroma and a novel RET mutation: a new entity or a combined tumour? Ecancermedicalscience 2019; 13:965. [PMID: 31921336 PMCID: PMC6834387 DOI: 10.3332/ecancer.2019.965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Indexed: 01/11/2023] Open
Abstract
Activating mutations of the RET gene have been described for both papillary (chromosomal rearrangement) and medullary (missense mutations) thyroid carcinomas. Here, we describe a case of a Warthin-like variant of papillary thyroid carcinoma displaying some morphological aspects that mimic the diffuse sclerosing variant. The tumour harboured BRAF V600E mutation and a novel germline point mutation in the RET gene, with unknown clinical and pathological meaning.
Collapse
Affiliation(s)
- Fausto Maffini
- Department of Pathology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy.,These authors contributed equally to writing this article
| | - Daniele Lorenzini
- University of Milan School of Medicine, Via Ripamonti 435, 20141 Milan, Italy.,These authors contributed equally to writing this article
| | - Daniela Lepanto
- Department of Pathology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | - Elvio De Fiori
- Department of Radiology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | - Caterina Fumagalli
- Department of Pathology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | - Alessandra Rappa
- Department of Pathology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | - Marta Tagliabue
- Division of Otolaryngology and Head and Neck Surgery, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | - Massimo Barberis
- Department of Pathology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
116
|
Shaul E, Kogan-Liberman D, Schuckalo S, Jan D, Ewart M, Nguyen T, Martinez M, Ovchinsky N. Novel mutations in NOTCH2 gene in infants with neonatal cholestasis. Pediatr Rep 2019; 11:8206. [PMID: 31595186 PMCID: PMC6778839 DOI: 10.4081/pr.2019.8206] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023] Open
Abstract
One cause of neonatal cholestasis (NC) is paucity of intrahepatic bile ducts which can be associated with Alagille syndrome or non- syndromic. Alagille syndrome is caused by autosomal dominant mutations in the Notch signaling pathway ligand Jagged1 in 94% of patients and mutations in the NOTCH2 receptor in <1% of patients. This is a retrospective case series studying infants with neonatal cholestasis found to have variants of unknown significance (VOUS) in NOTCH2. Sorting intolerant from tolerant (SIFT) and polymorphism phenotyping (PolyPhen) were utilized to predict a damaging effect. Five infants with NC without other features of Alagille syndrome were found to have one copy of a VOUS in NOTCH2, predicted to be damaging by SIFT and PolyPhen. Our cases support the notion that NOTCH2 mutations may result in hypoplastic biliary system. Further characterization of these variants is important to assist with our clinical approach to NC.
Collapse
Affiliation(s)
- Eliana Shaul
- Department of Pediatrics, Children's Hospital at Montefiore, Bronx, NY
| | - Debora Kogan-Liberman
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital at Montefiore, Bronx, NY
| | - Stephanie Schuckalo
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Goryeb Children's Hospital - Atlantic Health System, Morristown, NJ
| | - Dominique Jan
- Department of Pediatric Surgery, Children's Hospital at Montefiore, Bronx, NY
| | - Michelle Ewart
- Division of Surgical Pathology, Montefiore Medical Center, Bronx, NY
| | - Trang Nguyen
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital at Montefiore, Bronx, NY
| | - Mercedes Martinez
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Morgan Stanley Children's Hospital of New York, NY, USA
| | - Nadia Ovchinsky
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital at Montefiore, Bronx, NY.,Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Goryeb Children's Hospital - Atlantic Health System, Morristown, NJ
| |
Collapse
|
117
|
Formankova R, Kanderova V, Rackova M, Svaton M, Brdicka T, Riha P, Keslova P, Mejstrikova E, Zaliova M, Freiberger T, Grombirikova H, Zemanova Z, Vlkova M, Fencl F, Copova I, Bronsky J, Jabandziev P, Sedlacek P, Soukalova J, Zapletal O, Stary J, Trka J, Kalina T, Skvarova Kramarzova K, Hlavackova E, Litzman J, Fronkova E. Novel SAMD9 Mutation in a Patient With Immunodeficiency, Neutropenia, Impaired Anti-CMV Response, and Severe Gastrointestinal Involvement. Front Immunol 2019; 10:2194. [PMID: 31620126 PMCID: PMC6759462 DOI: 10.3389/fimmu.2019.02194] [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: 01/23/2019] [Accepted: 08/30/2019] [Indexed: 12/27/2022] Open
Abstract
Mutations in the Sterile alpha motif domain containing 9 (SAMD9) gene have been described in patients with severe multisystem disorder, MIRAGE syndrome, but also in patients with bone marrow (BM) failure in the absence of other systemic symptoms. The role of hematopoietic stem cell transplantation (HSCT) in the management of the disease is still unclear. Here, we present a patient with a novel mutation in SAMD9 (c.2471 G>A, p.R824Q), manifesting with prominent gastrointestinal tract involvement and immunodeficiency, but without any sign of adrenal insufficiency typical for MIRAGE syndrome. He suffered from severe CMV (cytomegalovirus) infection at 3 months of age, with a delayed development of T lymphocyte functional response against CMV, profound T cell activation, significantly reduced B lymphocyte counts and impaired lymphocyte proliferative response. Cultured T cells displayed slightly lower calcium flux and decreased survival. At the age of 6 months, he developed severe neutropenia requiring G-CSF administration, and despite only mild morphological and immunophenotypical disturbances in the BM, 78% of the BM cells showed monosomy 7 at the age of 18 months. Surprisingly, T cell proliferation after CD3 stimulation and apoptosis of the cells normalized during the follow-up, possibly reflecting the gradual development of monosomy 7. Among other prominent symptoms, he had difficulty swallowing, requiring percutaneous endoscopic gastrostomy (PEG), frequent gastrointestinal infections, and perianal erosions. He suffered from repeated infections and periodic recurring fevers with the elevation of inflammatory markers. At 26 months of age, he underwent HSCT that significantly improved hematological and immunological laboratory parameters. Nevertheless, he continued to suffer from other conditions, and subsequently, he died at day 440 post-transplant due to sepsis. Pathogenicity of this novel SAMD9 mutation was confirmed experimentally. Expression of mutant SAMD9 caused a significant decrease in proliferation and increase in cell death of the transfected cells. Conclusion: We describe a novel SAMD9 mutation in a patient with prominent gastrointestinal and immunological symptoms but without adrenal hypoplasia. Thus, SAMD9 mutations should be considered as cause of enteropathy in pediatric patients. The insufficient therapeutic outcome of transplantation further questions the role of HSCT in the management of patients with SAMD9 mutations and multisystem involvement.
Collapse
Affiliation(s)
- Renata Formankova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Veronika Kanderova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Marketa Rackova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Michael Svaton
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomas Brdicka
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Petr Riha
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Petra Keslova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Ester Mejstrikova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Marketa Zaliova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomas Freiberger
- Molecular Genetics Laboratory, Center of Cardiovascular Surgery and Transplantation, Brno, Czechia.,CEITEC, Masaryk University, Brno, Czechia.,Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Hana Grombirikova
- Molecular Genetics Laboratory, Center of Cardiovascular Surgery and Transplantation, Brno, Czechia.,Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Zuzana Zemanova
- Center of Oncocytogenetics, Institute of Clinical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Marcela Vlkova
- Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Clinical Immunology and Allergology, St. Anne's University Hospital Brno, Brno, Czechia
| | - Filip Fencl
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Ivana Copova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jiri Bronsky
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Petr Jabandziev
- CEITEC, Masaryk University, Brno, Czechia.,Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Paediatrics, University Hospital Brno, Brno, Czechia
| | - Petr Sedlacek
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jana Soukalova
- Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Medical Genetics, University Hospital Brno, Brno, Czechia
| | - Ondrej Zapletal
- Department of Pediatric Hematology, University Hospital Brno, Brno, Czechia
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jan Trka
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomas Kalina
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Karolina Skvarova Kramarzova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Eva Hlavackova
- Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Clinical Immunology and Allergology, St. Anne's University Hospital Brno, Brno, Czechia
| | - Jiri Litzman
- Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Clinical Immunology and Allergology, St. Anne's University Hospital Brno, Brno, Czechia
| | - Eva Fronkova
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| |
Collapse
|
118
|
Guttula PK, Chandrasekaran G, Gupta MK. Screening and insilico analysis of deleterious nsSNPs (missense) in human CSF3 for their effects on protein structure, stability and function. Comput Biol Chem 2019; 82:57-64. [PMID: 31272062 DOI: 10.1016/j.compbiolchem.2019.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/25/2019] [Accepted: 06/02/2019] [Indexed: 10/26/2022]
Abstract
Human granulocyte colony stimulating factor (hG-CSF), known as CSF3, plays an important role in the growth, differentiation, proliferation, survival, and activation of the granulocyte cell lineage such as neutrophils and their precursors. Functional reduction in native CSF3 protein results in reduced proliferation and activation of neutrophils and leads to neutropenia. Single nucleotide polymorphisms (SNPs) in the CSF3 gene may have deleterious effects on the CSF3 protein function. This study was undertaken to find the functional SNPs in the human CSF3 gene. Results suggest that 18.9% of all the SNPs in the dbSNP database for CSF3 gene were present in the coding region. Out of 59 non-synonymous SNPs (nsSNPs), 26 nsSNPs were predicted to be non-tolerable by SIFT whereas 18 and 7 nsSNPs were predicted as probably damaging and possibly damaging, respectively by PolyPhen. Among this 31 nsSNPs, 16 nsSNPs were identified to be potentially deleterious by PANTHER server, and 4 nsSNPs were found to be neutral by PROVEAN. SNPAnalyzer predicted 7 nsSNPs to be neutral phenotype and the remaining 24 nsSNPs to be associated with diseases. Among the predicted nsSNPs, rs144408123, rs144408123, rs145136406, rs145311241, rs373191696, rs762945096, rs763688260, rs767572172, rs775326370, rs777777864, rs777983866, rs781596455, rs139072004, rs757612684, rs772911210, rs139072004, rs746634544, rs749993200, rs763426127, rs772466210 were identified as deleterious and potentially damaging. I-Mutant analysis revealed that th 20 nsSNPs were important for protein stability of CSF3. Therefore, th 20 nsSNPs may be used for the wider population-based genetic studies and also should be taken into account while engineering the recombinant CSF3 protein for clinical use.
Collapse
Affiliation(s)
- Praveen Kumar Guttula
- Gene Manipulation Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Odisha, 769008, India
| | - Gopalakrishnan Chandrasekaran
- Gene Manipulation Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Odisha, 769008, India
| | - Mukesh Kumar Gupta
- Gene Manipulation Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Odisha, 769008, India.
| |
Collapse
|
119
|
Chiu YH, Liu YN, Chen HJ, Chang YC, Kao SM, Liu MY, Weng YY, Hsiao KJ, Liu TT. Prediction of functional consequences of the five newly discovered G6PD variations in Taiwan. Data Brief 2019; 25:104129. [PMID: 31294066 PMCID: PMC6595892 DOI: 10.1016/j.dib.2019.104129] [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: 04/25/2019] [Revised: 05/28/2019] [Accepted: 06/03/2019] [Indexed: 01/04/2023] Open
Abstract
Glucose-6-phosphate dehydrogenase deficiency (G6PD deficiency; OMIM #300908) is the most common inborn error disorders worldwide. While the G6PD is the key enzyme of removing oxidative stress in erythrocytes, the early diagnosis is utmost vital to prevent chronic and drug-, food- or infection-induced hemolytic anemia. The characterization of the mutations is also important for the subsequent genetic counseling, especially for female carrier with ambiguous enzyme activities and males with mild mutations. While multiplex SNaPshot assay and Sanger sequencing were performed on 500 G6PD deficient males, five newly discovered variations, namely c.187G > A (p.E63K), c.585G > C (p.Q195H), c.586A > T (p.I196F), c.743G > A (p.G248D), and c.1330G > A (p.V444I) were detected in the other six patients. These variants were previously named as the Pingtung, Tainan, Changhua, Chiayi, and Tainan-2 variants, respectively. The in silico analysis, as well as the prediction of the structure of the resultant mutant G6PD protein indicated that these five newly discovered variants might be disease causing mutations.
Collapse
Affiliation(s)
- Yen-Hui Chiu
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan.,Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Yu-Ning Liu
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Hsiao-Jan Chen
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Ying-Chen Chang
- Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Shu-Min Kao
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Mei-Ying Liu
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Ying-Yen Weng
- Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Kwang-Jen Hsiao
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan.,Preventive Medicine Foundation, Taipei, Taiwan
| | - Tze-Tze Liu
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
120
|
Gulilat M, Lamb T, Teft WA, Wang J, Dron JS, Robinson JF, Tirona RG, Hegele RA, Kim RB, Schwarz UI. Targeted next generation sequencing as a tool for precision medicine. BMC Med Genomics 2019; 12:81. [PMID: 31159795 PMCID: PMC6547602 DOI: 10.1186/s12920-019-0527-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/13/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Targeted next-generation sequencing (NGS) enables rapid identification of common and rare genetic variation. The detection of variants contributing to therapeutic drug response or adverse effects is essential for implementation of individualized pharmacotherapy. Successful application of short-read based NGS to pharmacogenes with high sequence homology, nearby pseudogenes and complex structure has been previously shown despite anticipated technical challenges. However, little is known regarding the utility of such panels to detect copy number variation (CNV) in the highly polymorphic cytochrome P450 (CYP) 2D6 gene, or to identify the promoter (TA)7 TAA repeat polymorphism UDP glucuronosyltransferase (UGT) 1A1*28. Here we developed and validated PGxSeq, a targeted exome panel for pharmacogenes pertinent to drug disposition and/or response. METHODS A panel of capture probes was generated to assess 422 kb of total coding region in 100 pharmacogenes. NGS was carried out in 235 subjects, and sequencing performance and accuracy of variant discovery validated in clinically relevant pharmacogenes. CYP2D6 CNV was determined using the bioinformatics tool CNV caller (VarSeq). Identified SNVs were assessed in terms of population allele frequency and predicted functional effects through in silico algorithms. RESULTS Adequate performance of the PGxSeq panel was demonstrated with a depth-of-coverage (DOC) ≥ 20× for at least 94% of the target sequence. We showed accurate detection of 39 clinically relevant gene variants compared to standard genotyping techniques (99.9% concordance), including CYP2D6 CNV and UGT1A1*28. Allele frequency of rare or novel variants and predicted function in 235 subjects mirrored findings from large genomic datasets. A large proportion of patients (78%, 183 out of 235) were identified as homozygous carriers of at least one variant necessitating altered pharmacotherapy. CONCLUSIONS PGxSeq can serve as a comprehensive, rapid, and reliable approach for the detection of common and novel SNVs in pharmacogenes benefiting the emerging field of precision medicine.
Collapse
Affiliation(s)
- Markus Gulilat
- Division of Clinical Pharmacology, Department of Medicine, Western University, London Health Sciences Centre - University Hospital, 339 Windermere Road, London, ON, N6A 5A5, Canada.,Department of Physiology and Pharmacology, Western University, Medical Sciences Building, Room 216, London, ON, N6A 5C1, Canada
| | - Tyler Lamb
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building, Room 216, London, ON, N6A 5C1, Canada
| | - Wendy A Teft
- Division of Clinical Pharmacology, Department of Medicine, Western University, London Health Sciences Centre - University Hospital, 339 Windermere Road, London, ON, N6A 5A5, Canada
| | - Jian Wang
- Robarts Research Institute, Western University, 1151 Richmond St. N, London, ON, N6A 5B7, Canada
| | - Jacqueline S Dron
- Robarts Research Institute, Western University, 1151 Richmond St. N, London, ON, N6A 5B7, Canada
| | - John F Robinson
- Robarts Research Institute, Western University, 1151 Richmond St. N, London, ON, N6A 5B7, Canada
| | - Rommel G Tirona
- Division of Clinical Pharmacology, Department of Medicine, Western University, London Health Sciences Centre - University Hospital, 339 Windermere Road, London, ON, N6A 5A5, Canada.,Department of Physiology and Pharmacology, Western University, Medical Sciences Building, Room 216, London, ON, N6A 5C1, Canada
| | - Robert A Hegele
- Robarts Research Institute, Western University, 1151 Richmond St. N, London, ON, N6A 5B7, Canada
| | - Richard B Kim
- Division of Clinical Pharmacology, Department of Medicine, Western University, London Health Sciences Centre - University Hospital, 339 Windermere Road, London, ON, N6A 5A5, Canada.,Department of Physiology and Pharmacology, Western University, Medical Sciences Building, Room 216, London, ON, N6A 5C1, Canada
| | - Ute I Schwarz
- Division of Clinical Pharmacology, Department of Medicine, Western University, London Health Sciences Centre - University Hospital, 339 Windermere Road, London, ON, N6A 5A5, Canada.
| |
Collapse
|
121
|
de Carvalho-Siqueira GQ, Ananina G, de Souza BB, Borges MG, Ito MT, da Silva-Costa SM, de Farias Domingos I, Falcão DA, Lopes-Cendes I, Bezerra MAC, da Silva Araújo A, Lucena-Araújo AR, de Souza Gonçalves M, Saad STO, Costa FF, de Melo MB. Whole-exome sequencing indicates FLG2 variant associated with leg ulcers in Brazilian sickle cell anemia patients. Exp Biol Med (Maywood) 2019; 244:932-939. [PMID: 31079484 DOI: 10.1177/1535370219849592] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although sickle cell anemia results from homozygosity for a single mutation at position 7 of the β-globin chain, the clinical aspects of this condition are very heterogeneous. Complications include leg ulcers, which have a negative impact on patients’ quality of life and are related to the severity of the disease. Nevertheless, the complex pathogenesis of this complication has yet to be elucidated. To identify novel genes associated with leg ulcers in sickle cell anemia, we performed whole-exome sequencing of extreme phenotypes in a sample of Brazilian sickle cell anemia patients and validated our findings in another sample. Our discovery cohort consisted of 40 unrelated sickle cell anemia patients selected based on extreme phenotypes: 20 patients without leg ulcers, aged from 40 to 61 years, and 20 with chronic leg ulcers. DNA was extracted from peripheral blood leukocytes and used for whole-exome sequencing. After the bioinformatics analysis, eight variants were selected for validation by Sanger sequencing and TaqMan® genotyping in 293 sickle cell anemia patients (153 without leg ulcers) from two different locations in Brazil. After the validation, Fisher’s exact test revealed a statistically significant difference in a stop codon variant (rs12568784 G/T) in the FLG2 gene between the GT and GG genotypes ( P = 0.035). We highlight the importance of rs12568784 in leg ulcer development as this variant of the FLG2 gene results in impairment of the skin barrier, predisposing the individual to inflammation and infection. Additionally, we suggest that the remaining seven variants and the genes in which they occur could be strong candidates for leg ulcers in sickle cell anemia. Impact statement To our knowledge, the present study is the first to use whole-exome sequencing based on extreme phenotypes to identify new candidate genes associated with leg ulcers in sickle cell anemia patients. There are few studies about this complication; the pathogenesis remains complex and has yet to be fully elucidated. We identified interesting associations in genes never related with this complication to our knowledge, especially the variant in the FLG2 gene. The knowledge of variants related with leg ulcer in sickle cell anemia may lead to a better comprehension of the disease’s etiology, allowing prevention and early treatment options in risk genotypes while improving quality of life for these patients.
Collapse
Affiliation(s)
| | - Galina Ananina
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Bruno Batista de Souza
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Murilo Guimarães Borges
- 2 Department of Medical Genetics and Genome Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| | - Mirta Tomie Ito
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Sueli Matilde da Silva-Costa
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Igor de Farias Domingos
- 3 Genetics Postgraduate Program, Federal University of Pernambuco, Recife, PE 50670-901, Brazil
| | - Diego Arruda Falcão
- 3 Genetics Postgraduate Program, Federal University of Pernambuco, Recife, PE 50670-901, Brazil
| | - Iscia Lopes-Cendes
- 2 Department of Medical Genetics and Genome Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| | | | | | | | | | | | | | - Mônica Barbosa de Melo
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| |
Collapse
|
122
|
Chiu YH, Chen HJ, Chang YC, Liu YN, Kao SM, Liu MY, Weng YY, Hsiao KJ, Liu TT. Applying a multiplexed primer extension method on dried blood spots increased the detection of carriers at risk of glucose-6-phosphate dehydrogenase deficiency in newborn screening program. Clin Chim Acta 2019; 495:271-277. [PMID: 31022393 DOI: 10.1016/j.cca.2019.04.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/15/2019] [Accepted: 04/19/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Patients with glucose-6-phosphate dehydrogenase deficiency might develop acute hemolytic anemia, chronic hemolytic anemia, and neonatal hyperbilirubinemia when exposed to high levels of oxidative stress. Severe hemolysis may occur in not only patients but also female carriers under certain conditions. However, 80%-85% of female carriers were undetected in an existing newborn screening program because of their wide-ranging levels of enzyme activity. METHODS We developed a cost- and time-efficient multiplex SNaPshot assay using dried blood spots. RESULTS By detecting 21 common mutations in Taiwan and Southeast Asia, the assay could determine 98.2% of the mutant alleles in our cohort of Taiwanese newborns. The 9 undetermined mutant alleles were consequently detected by Sanger sequencing, of which 5 unpublished variations-c.187G > A (Pingtung), c.585G > C (Tainan), c.586A > T (Changhua), c.743G > A (Chiayi), and c.1330G > A (Tainan-2)-were detected. Furthermore, 13% of mild mutations were missed in male infants whose enzyme levels at 6.1-7.0 U/gHb in the newborn screening program when set the cutoff value at 6.0 U/gHb. We therefore suggest increasing the cutoff value and applying the multiplex SNaPshot assay as the second tier for neonatal screening. CONCLUSIONS Our approach could significantly increase the detection rate of male patients and female carriers with a reasonable cost and a reasonable number of clinic referrals.
Collapse
Affiliation(s)
- Yen-Hui Chiu
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan; Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Hsiao-Jan Chen
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Ying-Chen Chang
- Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ning Liu
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Shu-Min Kao
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Mei-Ying Liu
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Ying-Yen Weng
- Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Kwang-Jen Hsiao
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan; Preventive Medicine Foundation, Taipei, Taiwan.
| | - Tze-Tze Liu
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan; Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
123
|
Myoglobinopathy is an adult-onset autosomal dominant myopathy with characteristic sarcoplasmic inclusions. Nat Commun 2019; 10:1396. [PMID: 30918256 PMCID: PMC6437160 DOI: 10.1038/s41467-019-09111-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/14/2019] [Indexed: 11/08/2022] Open
Abstract
Myoglobin, encoded by MB, is a small cytoplasmic globular hemoprotein highly expressed in cardiac myocytes and oxidative skeletal myofibers. Myoglobin binds O2, facilitates its intracellular transport and serves as a controller of nitric oxide and reactive oxygen species. Here, we identify a recurrent c.292C>T (p.His98Tyr) substitution in MB in fourteen members of six European families suffering from an autosomal dominant progressive myopathy with highly characteristic sarcoplasmic inclusions in skeletal and cardiac muscle. Myoglobinopathy manifests in adulthood with proximal and axial weakness that progresses to involve distal muscles and causes respiratory and cardiac failure. Biochemical characterization reveals that the mutant myoglobin has altered O2 binding, exhibits a faster heme dissociation rate and has a lower reduction potential compared to wild-type myoglobin. Preliminary studies show that mutant myoglobin may result in elevated superoxide levels at the cellular level. These data define a recognizable muscle disease associated with MB mutation. Myoglobin is a hemeprotein that reversibly binds oxygen and gives muscle its red color. Here, the authors report a genetic variant in the MB gene that associates with myoglobinopathy, an autosomal dominant progressive myopathy, and altered oxygen binding properties of the mutant protein.
Collapse
|
124
|
Lin PC, Yeh YM, Wu PY, Hsu KF, Chang JY, Shen MR. Germline susceptibility variants impact clinical outcome and therapeutic strategies for stage III colorectal cancer. Sci Rep 2019; 9:3931. [PMID: 30850667 PMCID: PMC6408483 DOI: 10.1038/s41598-019-40571-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 02/19/2019] [Indexed: 12/30/2022] Open
Abstract
Although somatic mutations are the main cause of cancer, underlying germline alterations may affect cancer outcome. There is little information on comprehensive analysis of germline genome sequencing for cancer healthcare strategy. Here we studied the implication of germline cancer-associated variants on cancer counselling and therapeutic strategies by germline whole genome and tumor targeted sequencing. Fifty-five gynecological and 104 colorectal cancer (CRC) patients were enrolled. We identified 22 germline pathogenic variants in 16 cancer-associated genes. Most of them are involved in DNA repair signaling, including MLH1, BRCA1/2, MUTYH, ATM, PMS2, MSH6, BAP1, and FANCA. About 6% of cancer patients presented the secondary findings of germline variants with non-oncogenic impact, mainly on the cardiovascular system which should be carefully monitored during chemotherapy. CRC patients carrying germline susceptibility variants had better disease-free survival than those without variants. Importantly, in the CRC model, the underlying germline alterations mold the tumor somatic alteration landscape. NOTCH1 mutation was the most common somatic mutation in recurrent CRC, implying a potential therapeutic target in adjuvant setting. In conclusion, both tumor genome and germline sequence data have to be analyzed to have a more complete picture of the overall genetic foundation of cancer.
Collapse
Affiliation(s)
- Peng-Chan Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Computer Science and Information Engineering, College of Electrical Engineering and Computer Science, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Yeh
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Graduate Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Ying Wu
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan
| | - Keng-Fu Hsu
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan
| | - Jang-Yang Chang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Meng-Ru Shen
- Department of Obstetrics and Gynecology, National Cheng Kung University, Tainan, Taiwan. .,Department of Pharmacology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
125
|
Devarajan S, Moon I, Ho MF, Larson NB, Neavin DR, Moyer AM, Black JL, Bielinski SJ, Scherer SE, Wang L, Weinshilboum RM, Reid JM. Pharmacogenomic Next-Generation DNA Sequencing: Lessons from the Identification and Functional Characterization of Variants of Unknown Significance in CYP2C9 and CYP2C19. Drug Metab Dispos 2019; 47:425-435. [PMID: 30745309 DOI: 10.1124/dmd.118.084269] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/15/2019] [Indexed: 02/06/2023] Open
Abstract
CYP2C9 and CYP2C19 are highly polymorphic pharmacogenes; however, clinically actionable genetic variability in drug metabolism due to these genes has been limited to a few common alleles. The identification and functional characterization of less-common open reading frame sequence variation might help to individualize therapy with drugs that are substrates for the enzymes encoded by these genes. The present study identified seven uncharacterized variants each in CYP2C9 and CYP2C19 using next-generation sequence data for 1013 subjects, and functionally characterized the encoded proteins. Constructs were created and transiently expressed in COS-1 cells for the assay of protein concentration and enzyme activities using fluorometric substrates and liquid chromatography- tandem mass spectrometry with tolbutamide (CYP2C9) and (S)-mephenytoin (CYP2C19) as prototypic substrates. The results were compared with the SIFT, Polyphen, and Provean functional prediction software programs. Cytochrome P450 oxidoreductase (CPR) activities were also determined. Positive correlations were observed between protein content and fluorometric enzyme activity for variants of CYP2C9 (P < 0.05) and CYP2C19 (P < 0.0005). However, CYP2C9 709G>C and CYP2C19 65A>G activities were much lower than predicted based on protein content. Substrate intrinsic clearance values for CYP2C9 218C>T, 343A>C, and CYP2C19 337G>A, 518C>T, 556C>T, and 557G>A were less than 25% of wild-type allozymes. CPR activity levels were similar for all variants. In summary, sequencing of CYP2C9 and CYP2C19 in 1013 subjects identified low-frequency variants that had not previously been functionally characterized. In silico predictions were not always consistent with functional assay results. These observations emphasize the need for high-throughput methods for pharmacogene variant mutagenesis and functional characterization.
Collapse
Affiliation(s)
- Sandhya Devarajan
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Irene Moon
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Ming-Fen Ho
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Nicholas B Larson
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Drew R Neavin
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Ann M Moyer
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - John L Black
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Suzette J Bielinski
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Steven E Scherer
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Liewei Wang
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Richard M Weinshilboum
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| | - Joel M Reid
- Departments of Molecular Pharmacology and Experimental Therapeutics (S.D., I.M., M.-F.H., L.W., R.M.W., J.M.R.) and Health Sciences Research (N.B.L., S.J.B.), Personalized Genomics Laboratory, Department of Laboratory Medicine and Pathology (A.M.M., J.L.B.), and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences (D.R.N.), Mayo Clinic, Rochester, Minnesota; and Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (S.E.S.)
| |
Collapse
|
126
|
Martins R, Fraga S, Esteves I, Calhau P. Child with unusual combination of sickle cell disease and autosomal recessive agammaglobulinemia associated with a novel CD79a gene mutation. BMJ Case Rep 2019. [DOI: 10.1136/bcr-2018-227346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
This article describes a novel mutation in CD79a gene identified in a child with sickle cell disease (SCD), who was diagnosed with autosomal recessive agammaglobulinaemia in the context of prolonged febrile syndrome. The association of a primary immunodeficiency with SCD in the same child was unexpected.
Collapse
|
127
|
Jones EG, Landstrom AP. Determining the Likelihood of Variant Pathogenicity Using Amino Acid-level Signal-to-Noise Analysis of Genetic Variation. J Vis Exp 2019. [PMID: 30735170 DOI: 10.3791/58907] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Advancements in the cost and speed of next generation genetic sequencing have generated an explosion of clinical whole exome and whole genome testing. While this has led to increased identification of likely pathogenic mutations associated with genetic syndromes, it has also dramatically increased the number of incidentally found genetic variants of unknown significance (VUS). Determining the clinical significance of these variants is a major challenge for both scientists and clinicians. An approach to assist in determining the likelihood of pathogenicity is signal-to-noise analysis at the protein sequence level. This protocol describes a method for amino acid-level signal-to-noise analysis that leverages variant frequency at each amino acid position of the protein with known protein topology to identify areas of the primary sequence with elevated likelihood of pathologic variation (relative to population "background" variation). This method can identify amino acid residue location "hotspots" of high pathologic signal, which can be used to refine the diagnostic weight of VUSs such as those identified by next generation genetic testing.
Collapse
Affiliation(s)
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine;
| |
Collapse
|
128
|
Gorakshakar AC, Breganza PV, Colaco SP, Shaikh RF, Bohra MY, Sawant PM, Nadkarni AH, Colah RB, Ghosh KK. Rare β- and δ-Globin Gene Mutations in the Pathare Prabhus: Original Inhabitants of Mumbai, India. Hemoglobin 2019; 42:297-301. [PMID: 30626279 DOI: 10.1080/03630269.2018.1544909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Genetic structure of the Indian population is influenced by waves of several immigrants from West Eurasia. Therefore, genetic information of various ethnic groups is valuable to understand their origins, the pattern of migration as well as the genetic relationship between them. No genetic data is available on Pathare Prabhu, which is a small indigenous Hindu community from Mumbai, Maharashtra State, India. The aim of this study was to screen the Pathare Prabhus for hemoglobinopathies, which is a major public health problem in India. Two hundred and fifty-seven unrelated Pathare Prabhus subjects were screened for various hemoglobinopathies. Complete blood counts (CBC) were done on an automated hematology counter. High performance liquid chromatography (HPLC) was used to identify β-thalassemia (β-thal) carriers. Molecular characterization of the β gene defects was done by reverse dot-blot hybridization, amplification refractory mutation system (ARMS) and DNA sequencing. Deletional α-thalassemia (α-thal) was detected by multiplex polymerase chain reaction (PCR). Hb A2-Saurashtra (HBD: c.301C>T) was identified by DNA sequencing; its modeling was also done. The prevalence of β-thal was 3.89%, while deletional α-thal was 5.4%. The initiation codon (ATG>ACG) (HBB: c.2T>C) was seen in eight individuals (80.0%), Hb D-Punjab (HBB: c.364G>C) and Hb A2-Saurashtra, was found in two and one individual, respectively. A community-specific β-thal mutation was found in Pathare Prabhus in significant proportions. This information is useful in developing an algorithm for a prenatal diagnosis (PND) program.
Collapse
Affiliation(s)
- Ajit C Gorakshakar
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Pearl V Breganza
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Stacy P Colaco
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Roshan F Shaikh
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Meenu Y Bohra
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Pratibha M Sawant
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Anita H Nadkarni
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Roshan B Colah
- a Department of Transfusion Medicine , National Institute of Immunohaematology, King Edward Memorial Hospital Campus , Parel , Mumbai , Maharashtra State , India
| | - Kanjaksha K Ghosh
- b Surat Raktadan Kendra, Udhna Khatodara Urban Health Centre , Near Chosath Joganio Mata Mandir , Surat , Gujarat State , India
| |
Collapse
|
129
|
Sun S, Thorson JA, Murray SS. Annotation of Variant Data from High-Throughput DNA Sequencing from Tumor Specimens: Filtering Strategies to Identify Driver Mutations. Methods Mol Biol 2019; 1908:49-60. [PMID: 30649720 DOI: 10.1007/978-1-4939-9004-7_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The use of next-generation sequencing technologies has enabled the analysis of a wide spectrum of somatic mutations in tumors. This analysis can be carried out using various strategies including the use of small panels of focused, clinically actionable genes, large panels of cancer-related genes, whole exomes, and the entire genome. One of the main goals in these analyses is to identify key mutations in these tumors that drive the oncogenic process. Depending on the gene, mutations can have altering effects, such as loss of function mutations in tumor suppressor genes, to mutations that activate genes such as kinases involved with cell cycle progression or proliferation. Once the sequencing process is complete, and the alignment of the large collection of reads to the reference genome and variant calling has been carried out, one is left with a large collection of variants. The challenge then becomes assigning where the variant resides in the genome with respect to coding regions, splice site regions, regulatory regions, and what potential functional effect these variants may have on the resulting protein. Other helpful information includes determining if the variant has been identified before, and if so, the tumor type associated with the variant. In addition, if the tumor profiling experiment is not conducted with a matched specimen representing the inherited genome, various tools are helpful to determine if the variant is likely to be an inherited polymorphism or a somatic event. In this chapter, we review the various tools available for annotating variants to assist in filtering down and prioritizing the hundreds to thousands of variants down to the key variants likely to be driver mutations and relevant to the tumor being profiled.
Collapse
Affiliation(s)
- Shulei Sun
- Center for Advanced Laboratory Medicine, University of California San Diego Health, La Jolla, CA, USA
| | - John A Thorson
- Center for Advanced Laboratory Medicine, University of California San Diego Health, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Sarah S Murray
- Center for Advanced Laboratory Medicine, University of California San Diego Health, La Jolla, CA, USA.
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
130
|
Lovato DV, Herai RR, Pignatari GC, Beltrão-Braga PCB. The Relevance of Variants With Unknown Significance for Autism Spectrum Disorder Considering the Genotype-Phenotype Interrelationship. Front Psychiatry 2019; 10:409. [PMID: 31231258 PMCID: PMC6567929 DOI: 10.3389/fpsyt.2019.00409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/23/2019] [Indexed: 11/13/2022] Open
Abstract
Several efforts in basic and clinical research have been contributing to unveiling the genetics behind autism spectrum disorders (ASD). However, despite these advancements, many individuals diagnosed with ASD and related neuropsychiatric conditions have been genetically investigated without elucidative results. The enormous genetic complexity of ASD-related conditions makes it a significant challenge to achieve, with a growing number of genes (close to a thousand) involved, belonging to different molecular pathways and presenting distinct genetic variations. Next-generation sequencing (NGS) is the approach most used in genetic research related to ASD, identifying de novo mutation, which is closely related to more severe clinical phenotypes, especially when they affect constrained and loss-of-function intolerant genes. On the other hand, de novo mutation findings contribute to a small percentage of the ASD population, since most of the cases and genetic variants associated with neuropsychiatric conditions are inherited and phenotypes are results of additive polygenic models, which makes statistical efforts more difficult. As a result, NGS investigation can sound vainly or unsuccessful, and new mutations on genes already related with ASD are classified as variants of unknown significance (VUS), hampering their endorsement to a clinical phenotype. This review is focused on currently available strategies to clarify the impact of VUS and to describe the efforts to identify more pieces of evidence throughout clinical interpretation and genetic curation process.
Collapse
Affiliation(s)
- Diogo V Lovato
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Roberto R Herai
- Experimental Multiuser Laboratory, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil.,Lico Kaesemodel Institute (ILK), Curitiba, Brazil
| | - Graciela C Pignatari
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patricia C B Beltrão-Braga
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Laboratory of Disease Modeling, Scientific Platform Pasteur-USP, São Paulo, Brazil
| |
Collapse
|
131
|
Trinh J, Lohmann K, Baumann H, Balck A, Borsche M, Brüggemann N, Dure L, Dean M, Volkmann J, Tunc S, Prasuhn J, Pawlack H, Imhoff S, Lill CM, Kasten M, Bauer P, Rolfs A, Klein C. Utility and implications of exome sequencing in early‐onset Parkinson's disease. Mov Disord 2018; 34:133-137. [DOI: 10.1002/mds.27559] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 01/12/2023] Open
Affiliation(s)
- Joanne Trinh
- Institute of Neurogenetics University of Lübeck Lübeck Germany
| | - Katja Lohmann
- Institute of Neurogenetics University of Lübeck Lübeck Germany
| | - Hauke Baumann
- Institute of Neurogenetics University of Lübeck Lübeck Germany
| | - Alexander Balck
- Institute of Neurogenetics University of Lübeck Lübeck Germany
- Department of Neurology University of Lübeck Lübeck Germany
| | - Max Borsche
- Institute of Neurogenetics University of Lübeck Lübeck Germany
- Department of Neurology University of Lübeck Lübeck Germany
| | - Norbert Brüggemann
- Institute of Neurogenetics University of Lübeck Lübeck Germany
- Department of Neurology University of Lübeck Lübeck Germany
| | - Leon Dure
- Department of Neurology University of Alabama at Birmingham Birmingham Alabama USA
| | - Marissa Dean
- Department of Neurology University of Alabama at Birmingham Birmingham Alabama USA
| | - Jens Volkmann
- Departement of Neurology Universitatsklinikum Würzburg Würzburg Germany
| | - Sinem Tunc
- Institute of Neurogenetics University of Lübeck Lübeck Germany
- Department of Neurology University of Lübeck Lübeck Germany
| | - Jannik Prasuhn
- Institute of Neurogenetics University of Lübeck Lübeck Germany
- Department of Neurology University of Lübeck Lübeck Germany
| | - Heike Pawlack
- Institute of Neurogenetics University of Lübeck Lübeck Germany
| | - Sophie Imhoff
- Institute of Neurogenetics University of Lübeck Lübeck Germany
| | | | - Meike Kasten
- Institute of Neurogenetics University of Lübeck Lübeck Germany
- Psychiatry University of Lübeck Lübeck Germany
| | - Peter Bauer
- Centogene AG, Institute for Rare Diseases Rostock Germany
| | - Arndt Rolfs
- Centogene AG, Institute for Rare Diseases Rostock Germany
- Albrecht Kossel Institute for Neuroregeneration University Hospital Rostock Rostock Germany
| | - Christine Klein
- Institute of Neurogenetics University of Lübeck Lübeck Germany
| | | |
Collapse
|
132
|
Zhou Y, Fujikura K, Mkrtchian S, Lauschke VM. Computational Methods for the Pharmacogenetic Interpretation of Next Generation Sequencing Data. Front Pharmacol 2018; 9:1437. [PMID: 30564131 PMCID: PMC6288784 DOI: 10.3389/fphar.2018.01437] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022] Open
Abstract
Up to half of all patients do not respond to pharmacological treatment as intended. A substantial fraction of these inter-individual differences is due to heritable factors and a growing number of associations between genetic variations and drug response phenotypes have been identified. Importantly, the rapid progress in Next Generation Sequencing technologies in recent years unveiled the true complexity of the genetic landscape in pharmacogenes with tens of thousands of rare genetic variants. As each individual was found to harbor numerous such rare variants they are anticipated to be important contributors to the genetically encoded inter-individual variability in drug effects. The fundamental challenge however is their functional interpretation due to the sheer scale of the problem that renders systematic experimental characterization of these variants currently unfeasible. Here, we review concepts and important progress in the development of computational prediction methods that allow to evaluate the effect of amino acid sequence alterations in drug metabolizing enzymes and transporters. In addition, we discuss recent advances in the interpretation of functional effects of non-coding variants, such as variations in splice sites, regulatory regions and miRNA binding sites. We anticipate that these methodologies will provide a useful toolkit to facilitate the integration of the vast extent of rare genetic variability into drug response predictions in a precision medicine framework.
Collapse
Affiliation(s)
- Yitian Zhou
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kohei Fujikura
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Souren Mkrtchian
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Volker M. Lauschke
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
133
|
McKenna B, Koomar T, Vervier K, Kremsreiter J, Michaelson JJ. Whole-genome sequencing in a family with twin boys with autism and intellectual disability suggests multimodal polygenic risk. Cold Spring Harb Mol Case Stud 2018; 4:a003285. [PMID: 30559312 PMCID: PMC6318775 DOI: 10.1101/mcs.a003285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/10/2018] [Indexed: 01/02/2023] Open
Abstract
Over the past decade, a focus on de novo mutations has rapidly accelerated gene discovery in autism spectrum disorder (ASD), intellectual disability (ID), and other neurodevelopmental disorders (NDDs). However, recent studies suggest that only a minority of cases are attributable to de novo mutations, and instead these disorders often result from an accumulation of various forms of genetic risk. Consequently, we adopted an inclusive approach to investigate the genetic risk contributing to a case of male monozygotic twins with ASD and ID. At the time of the study, the probands were 7 yr old and largely nonverbal. Medical records indicated a history of motor delays, sleep difficulties, and significant cognitive deficits. Through whole-genome sequencing of the probands and their parents, we uncovered elevated common polygenic risk, a coding de novo point mutation in CENPE, an ultra-rare homozygous regulatory variant in ANK3, inherited rare variants in NRXN3, and a maternally inherited X-linked deletion situated in a noncoding regulatory region between ZNF81 and ZNF182 Although each of these genes has been directly or indirectly associated with NDDs, evidence suggests that no single variant adequately explains the probands' phenotype. Instead, we propose that the probands' condition is due to the confluence of multiple rare variants in the context of a high-risk genetic background. This case emphasizes the multifactorial nature of genetic risk underlying most instances of NDDs and aligns with the "female protective model" of ASD.
Collapse
Affiliation(s)
- Brooke McKenna
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Department of Psychology, Emory University, Atlanta, Georgia 30322, USA
| | - Tanner Koomar
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | - Kevin Vervier
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Host-Microbiota Interactions Laboratory, Wellcome Trust Sanger Institute, Cambridge CB10 1SA, United Kingdom
| | - Jamie Kremsreiter
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | - Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| |
Collapse
|
134
|
Zhang X, Zhang L, Wu Y, Li G, Chen S, Xia Y, Li H. Identification of novel compound heterozygous SPG7 mutations-related hereditary spastic paraplegia in a Chinese family: a case report. BMC Neurol 2018; 18:196. [PMID: 30497413 PMCID: PMC6263041 DOI: 10.1186/s12883-018-1199-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 11/16/2018] [Indexed: 11/10/2022] Open
Abstract
Background Autosomal recessive hereditary spastic paraplegias (ARHSPs) are a group of clinically and genetically heterogeneous neurodegenerative diseases with progressive spasticity and weakness in the lower limbs. Mutations in the Spastic Paraplegia gene 7 (SPG7) account for about 5–21% of ARHSP cases. However, in Asians, few reports about the mutations exist. In this study, we firstly report a novel finding from a Chinese family with compound heterozygous SPG7 mutations, in which three siblings were affected with a complicated form of ARHSP. Case presentation A 56-year-old man presented with progressive stiffness, weakness and ataxia in the lower limbs. Two sisters of him had similar symptoms and dysarthria. Brain magnetic resonance imaging (MRI) revealed cerebellar atrophy in each of the patients. Genetic analysis, which exerted a targeted next generation sequencing (NGS) panel covering 917 comprehensive ataxia genes to the proband, followed by Sanger sequencing of candidate genes in other eight family members, was used to find the etiology of the disease. Ultimately, we identified compound heterozygous SPG7 mutations with two mutations: (c.1150_1150-1insCTAC and c.2062C > T, p.Arg688Trp) and one single nucleotide polymorphism (c.2063G > A, p.Arg688Gln). Conclusions The four bases insertion mutation (4bIM) was predicted to cause frameshift mutation or affect the splicing, and the last two variants were led to a stop codon mutation (p.Arg688Ter). As located in highly conserved positions and encoded paraplegin, the mutations were speculated to result in a truncated or defective protein and would be pathogenic factors of the disease. This paper proves to be the first case report of SPG7 mutation in ARHSP reported in Chinese population. Our findings widen the spectrum of SPG7 mutations of ARHSP and indicate that the SPG7 mutation is an important cause of adult-onset undiagnosed ataxia. Electronic supplementary material The online version of this article (10.1186/s12883-018-1199-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoqian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang avenue, Wuhan, 430022, Hubei, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang avenue, Wuhan, 430022, Hubei, China
| | - Yanqing Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang avenue, Wuhan, 430022, Hubei, China
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang avenue, Wuhan, 430022, Hubei, China
| | - Shengcai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang avenue, Wuhan, 430022, Hubei, China
| | - Yuanpeng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang avenue, Wuhan, 430022, Hubei, China.
| | - Hongge Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang avenue, Wuhan, 430022, Hubei, China.
| |
Collapse
|
135
|
Identification of genetic variants associated with tacrolimus metabolism in kidney transplant recipients by extreme phenotype sampling and next generation sequencing. THE PHARMACOGENOMICS JOURNAL 2018; 19:375-389. [PMID: 30442921 PMCID: PMC6522337 DOI: 10.1038/s41397-018-0063-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/11/2018] [Accepted: 09/27/2018] [Indexed: 12/26/2022]
Abstract
An extreme phenotype sampling (EPS) model with targeted next-generation sequencing (NGS) identified genetic variants associated with tacrolimus (Tac) metabolism in subjects from the Deterioration of Kidney Allograft Function (DeKAF) Genomics cohort which included 1,442 European Americans (EA) and 345 African Americans (AA). This study included 48 subjects separated into 4 groups of 12 (AA high, AA low, EA high, EA low). Groups were selected by the extreme phenotype of dose-normalized Tac trough concentrations after adjusting for common genetic variants and clinical factors. NGS spanned >3 Mb of 28 genes and identified 18,661 genetic variants (3,961 previously unknown). A group of 125 deleterious variants, by SIFT analysis, were associated with Tac troughs in EAs (burden test, p=0.008), CYB5R2 was associated with Tac troughs in AAs (SKAT, p=0.00079). In CYB5R2, rs61733057 (increased allele frequency in AAs) was predicted to disrupt protein function by SIFT and PolyPhen2 analysis. The variants merit further validation.
Collapse
|
136
|
McWilliams RR, Wieben ED, Chaffee KG, Antwi SO, Raskin L, Olopade OI, Li D, Highsmith WE, Colon-Otero G, Khanna LG, Permuth JB, Olson JE, Frucht H, Genkinger J, Zheng W, Blot WJ, Wu L, Almada LL, Fernandez-Zapico ME, Sicotte H, Pedersen KS, Petersen GM. CDKN2A Germline Rare Coding Variants and Risk of Pancreatic Cancer in Minority Populations. Cancer Epidemiol Biomarkers Prev 2018; 27:1364-1370. [PMID: 30038052 PMCID: PMC6214745 DOI: 10.1158/1055-9965.epi-17-1065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/13/2018] [Accepted: 07/11/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Pathogenic germline mutations in the CDKN2A tumor suppressor gene are rare and associated with highly penetrant familial melanoma and pancreatic cancer in non-Hispanic whites (NHW). To date, the prevalence and impact of CDKN2A rare coding variants (RCV) in racial minority groups remain poorly characterized. We examined the role of CDKN2A RCVs on the risk of pancreatic cancer among minority subjects.Methods: We sequenced CDKN2A in 220 African American (AA) pancreatic cancer cases, 900 noncancer AA controls, and 183 Nigerian controls. RCV frequencies were determined for each group and compared with that of 1,537 NHW patients with pancreatic cancer. Odds ratios (OR) and 95% confidence intervals (CI) were calculated for both a case-case comparison of RCV frequencies in AAs versus NHWs, and case-control comparison between AA cases versus noncancer AA controls plus Nigerian controls. Smaller sets of Hispanic and Native American cases and controls also were sequenced.Results: One novel missense RCV and one novel frameshift RCV were found among AA patients: 400G>A and 258_278del. RCV carrier status was associated with increased risk of pancreatic cancer among AA cases (11/220; OR, 3.3; 95% CI, 1.5-7.1; P = 0.004) compared with AA and Nigerian controls (17/1,083). Further, AA cases had higher frequency of RCVs: 5.0% (OR, 13.4; 95% CI, 4.9-36.7; P < 0.001) compared with NHW cases (0.4%).Conclusions: CDKN2A RCVs are more common in AA than in NHW patients with pancreatic cancer and associated with moderately increased pancreatic cancer risk among AAs.Impact: RCVs in CDKN2A are frequent in AAs and are associated with risk for pancreatic cancer. Cancer Epidemiol Biomarkers Prev; 27(11); 1364-70. ©2018 AACR.
Collapse
Affiliation(s)
| | - Eric D Wieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kari G Chaffee
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Samuel O Antwi
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - Leon Raskin
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Olufunmilayo I Olopade
- Departments of Medicine and Human Genetics, University of Chicago Medical Center, Chicago, Illinois
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - W Edward Highsmith
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Gerardo Colon-Otero
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Jacksonville, Florida
| | - Lauren G Khanna
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Jennifer B Permuth
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Janet E Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Harold Frucht
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Jeanine Genkinger
- Department of Epidemiology, Columbia University Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Wei Zheng
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - William J Blot
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Lang Wu
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota
| | - Hugues Sicotte
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
137
|
Wang X, Liu X, Huang L, Fang S, Jia X, Xiao X, Li S, Guo X. Mutation Survey of Candidate Genes and Genotype-Phenotype Analysis in 20 Southeastern Chinese Patients with Axenfeld-Rieger Syndrome. Curr Eye Res 2018; 43:1334-1341. [PMID: 29939776 DOI: 10.1080/02713683.2018.1493129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 10/28/2022]
Abstract
Purpose/aim of the study: To conduct a survey spectrum of the PITX2, FOXC1, and PRDM5 genes to reveal genotype-phenotype correlations in a cohort of Southeastern Chinese patients with Axenfeld-Rieger syndrome (ARS). MATERIALS AND METHODS A total of 20 probands with ARS were recruited in Southeast China. All patients underwent full ocular and systemic examinations. Sanger sequencing was used to analyze all coding regions of and regions adjacent to PITX2, FOXC1, and PRDM5 and 13 upstream regulatory elements of PITX2. Multiplex ligation-dependent probe amplification was performed to detect gross insertions and deletions in PITX2 and FOXC1. Quantitative polymerase chain reaction was used to detect copy number variations in regulatory elements of PITX2. A bioinformatics analysis was conducted to evaluate the pathogenicity of variants. RESULTS Eleven mutations, including eight novel mutations, were identified in PITX2. Seven of the mutations were truncations. A genotype-phenotype correlation analysis showed that 81.8% (9/11) of patients with mutations in PITX2 developed glaucoma before reaching 10 years old. The proportion of patients without detected mutations was only 33.3% (3/9, P = 0.0399). In patient G1399, ultrasound biomicroscopy revealed that the left eye exhibited a phenotype similar to aniridia with complete angle closure and a remaining stub of iris tissue. CONCLUSION This is the first genetic study of a cohort of Southeastern Chinese patients with ARS. Eight novel mutations were detected, expanding the mutation spectrum of PITX2. PITX2 may be a major candidate gene for ARS in Southeastern Chinese patients. Truncations may be the primary mutation type in PITX2. Glaucoma onset may be earlier in patients with mutations in PITX2 than in those without mutations in PITX2 and FOXC1. A block of the anterior chamber angle by the end of the iris might represent the main factor influencing the development of glaucoma in ARS patients with an asymmetric aniridia phenotype.
Collapse
Affiliation(s)
- Xun Wang
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| | - Xing Liu
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| | - Liqin Huang
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| | - Shaohua Fang
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| | - Xiaoyun Jia
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| | - Xueshan Xiao
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| | - Shiqiang Li
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| | - Xiangming Guo
- a State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center , Sun Yat-Sen University , Guangzhou , China
| |
Collapse
|
138
|
Tessier L, Côté O, Bienzle D. Sequence variant analysis of RNA sequences in severe equine asthma. PeerJ 2018; 6:e5759. [PMID: 30324028 PMCID: PMC6186407 DOI: 10.7717/peerj.5759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 09/15/2018] [Indexed: 12/13/2022] Open
Abstract
Background Severe equine asthma is a chronic inflammatory disease of the lung in horses similar to low-Th2 late-onset asthma in humans. This study aimed to determine the utility of RNA-Seq to call gene sequence variants, and to identify sequence variants of potential relevance to the pathogenesis of asthma. Methods RNA-Seq data were generated from endobronchial biopsies collected from six asthmatic and seven non-asthmatic horses before and after challenge (26 samples total). Sequences were aligned to the equine genome with Spliced Transcripts Alignment to Reference software. Read preparation for sequence variant calling was performed with Picard tools and Genome Analysis Toolkit (GATK). Sequence variants were called and filtered using GATK and Ensembl Variant Effect Predictor (VEP) tools, and two RNA-Seq predicted sequence variants were investigated with both PCR and Sanger sequencing. Supplementary analysis of novel sequence variant selection with VEP was based on a score of <0.01 predicted with Sorting Intolerant from Tolerant software, missense nature, location within the protein coding sequence and presence in all asthmatic individuals. For select variants, effect on protein function was assessed with Polymorphism Phenotyping 2 and screening for non-acceptable polymorphism 2 software. Sequences were aligned and 3D protein structures predicted with Geneious software. Difference in allele frequency between the groups was assessed using a Pearson’s Chi-squared test with Yates’ continuity correction, and difference in genotype frequency was calculated using the Fisher’s exact test for count data. Results RNA-Seq variant calling and filtering correctly identified substitution variants in PACRG and RTTN. Sanger sequencing confirmed that the PACRG substitution was appropriately identified in all 26 samples while the RTTN substitution was identified correctly in 24 of 26 samples. These variants of uncertain significance had substitutions that were predicted to result in loss of function and to be non-neutral. Amino acid substitutions projected no change of hydrophobicity and isoelectric point in PACRG, and a change in both for RTTN. For PACRG, no difference in allele frequency between the two groups was detected but a higher proportion of asthmatic horses had the altered RTTN allele compared to non-asthmatic animals. Discussion RNA-Seq was sensitive and specific for calling gene sequence variants in this disease model. Even moderate coverage (<10–20 counts per million) yielded correct identification in 92% of samples, suggesting RNA-Seq may be suitable to detect sequence variants in low coverage samples. The impact of amino acid alterations in PACRG and RTTN proteins, and possible association of the sequence variants with asthma, is of uncertain significance, but their role in ciliary function may be of future interest.
Collapse
Affiliation(s)
- Laurence Tessier
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada.,BenchSci, Toronto, ON, Canada
| | - Olivier Côté
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada.,BioAssay Works, Ijamsville, MD, USA
| | - Dorothee Bienzle
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
139
|
Challenges of Identifying Clinically Actionable Genetic Variants for Precision Medicine. JOURNAL OF HEALTHCARE ENGINEERING 2018; 2016:3617572. [PMID: 27195526 PMCID: PMC4955563 DOI: 10.1155/2016/3617572] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 03/17/2016] [Indexed: 12/30/2022]
Abstract
Advances in genomic medicine have the potential to change the way we treat human disease, but translating these advances into reality for improving healthcare outcomes depends essentially on our ability to discover disease- and/or drug-associated clinically actionable genetic mutations. Integration and manipulation of diverse genomic data and comprehensive electronic health records (EHRs) on a big data infrastructure can provide an efficient and effective way to identify clinically actionable genetic variants for personalized treatments and reduce healthcare costs. We review bioinformatics processing of next-generation sequencing (NGS) data, bioinformatics infrastructures for implementing precision medicine, and bioinformatics approaches for identifying clinically actionable genetic variants using high-throughput NGS data and EHRs.
Collapse
|
140
|
Taeubner J, Wieczorek D, Yasin L, Brozou T, Borkhardt A, Kuhlen M. Penetrance and Expressivity in Inherited Cancer Predisposing Syndromes. Trends Cancer 2018; 4:718-728. [PMID: 30352675 DOI: 10.1016/j.trecan.2018.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/01/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023]
Abstract
Inherited diseases are not always expressed in the same way in every individual that carries the same variant in a disease-causing gene. This phenomenon is known as reduced or incomplete penetrance. Variable and incomplete penetrance may explain why inherited diseases are occasionally transmitted through unaffected parents, but also why clinically healthy individuals can carry potentially pathogenic variants without expressing features of the disease. Here, we will provide an overview of factors that play a fundamental role in the concept of penetrance and expressivity of cancer predisposing genes in children with malignancies. These findings are important to understand the complexity of inherited diseases and cancer development and to improve genetic counselling for the affected families.
Collapse
Affiliation(s)
- Julia Taeubner
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Dagmar Wieczorek
- Institute of Human Genetics, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Layal Yasin
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Triantafyllia Brozou
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michaela Kuhlen
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
141
|
Mutational Profiles of F8 and F9 in a Cohort of Haemophilia A and Haemophilia B Patients in the Multi-ethnic Malaysian Population. Mediterr J Hematol Infect Dis 2018; 10:e2018056. [PMID: 30210749 PMCID: PMC6131101 DOI: 10.4084/mjhid.2018.056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/10/2018] [Indexed: 12/30/2022] Open
Abstract
Background Haemophilia A (HA) and Haemophilia B (HB) are X-linked blood disorders that are caused by various mutations in the factor VIII (F8) and factor IX (F9) genes respectively. Identification of mutations is essential as some of the mutations are associated with the development of inhibitors. This study is the first comprehensive study of the F8 mutational profile in Malaysia. Materials and methods We analysed 100 unrelated HA and 15 unrelated HB patients for genetic alterations in the F8 and F9 genes by using the long-range PCR, DNA sequencing, and the multiplex-ligation-dependent probe amplification assays. The prediction software was used to confirm the effects of these mutations on factor VIII and IX proteins. Results 44 (53%) of the severe HA patients were positive for F8 intron 22 inversion, and three (3.6%) were positive for intron one inversion. There were 22 novel mutations in F8, including missense (8), frameshift (9), splice site (3), large deletion (1) and nonsense (1) mutations. In HB patients, four novel mutations were identified including the splice site (1), small deletion (1), large deletion (1) and missense (1) mutation. Discussion The mutational spectrum of F8 in Malaysian patients is heterogeneous, with a slightly higher frequency of intron 22 inversion in these severe HA patients when compared to other Asian populations. Identification of these mutational profiles in F8 and F9 genes among Malaysian patients will provide a useful reference for the early detection and diagnosis of HA and HB in the Malaysian population.
Collapse
|
142
|
Abstract
"Conservation genomics" encompasses the idea that genome-scale data will improve the capacity of resource managers to protect species. Although genetic approaches have long been used in conservation research, it has only recently become tractable to generate genome-wide data at a scale that is useful for conservation. In this Review, we discuss how genome-scale data can inform species delineation in the face of admixture, facilitate evolution through the identification of adaptive alleles, and enhance evolutionary rescue based on genomic patterns of inbreeding. As genomic approaches become more widely adopted in conservation, we expect that they will have a positive impact on management and policy decisions.
Collapse
Affiliation(s)
- Megan A Supple
- Department of Ecology and Evolutionary Biology, University of California Santa Cruz, Santa Cruz, CA, 95060, USA.
| | - Beth Shapiro
- Department of Ecology and Evolutionary Biology, University of California Santa Cruz, Santa Cruz, CA, 95060, USA.
- UCSC Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, 95060, USA.
| |
Collapse
|
143
|
Suvatha A, Sibin MK, Bhat DI, Narasingarao KVL, Vazhayil V, Chetan GK. Factor XIII polymorphism and risk of aneurysmal subarachnoid haemorrhage in a south Indian population. BMC MEDICAL GENETICS 2018; 19:159. [PMID: 30185149 PMCID: PMC6126001 DOI: 10.1186/s12881-018-0674-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/29/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND The rupture of a brain aneurysm causes bleeding in the subarachnoid space and is known as aneurysmal subarachnoid haemorrhage (aSAH). In our study, we evaluated the association of factor XIII polymorphism and the risk of Aneurysmal subarachnoid haemorrhage (aSAH) in South Indian population. METHODS The study was performed in 200 subjects with aSAH and 205 healthy control subjects. Genotyping of rs5985(c.103G > T (p.Val35Leu)) and rs5982(c.1694C > T (p.Pro564Leu)) polymorphism was performed by Taqman® allelic discrimination assay. RESULTS In our study, Val/Leu genotype frequency was higher in control subjects (18%) compared to aSAH patients (9%).The Val/Leu genotype was associated with lower risk of aSAH (OR = 0.48, 95%CI = 0.26-0.88, p = 0.02). When compared with Val allele, Leu allele was significantly associated with lower risk of aSAH (OR = 0.55, 95%CI = 0.32-0.95, p = 0.03). In subtyping, we found a significant association of Leu/Leu genotype with the Basilar top aneurysm (OR = 3.59, 95%CI = 1.11-11.64, p = 0.03). In c.1694C > T (p.Pro565Leu) variant, Pro/Pro Vs Pro/Leu genotype (OR = 2.06, 95%CI = 1.10-3.85, p = 0.02) was significantly associated with higher risk of aSAH. The 564Leu allelic frequency in aSAH patients (36%) was higher when compared with that in healthy controls (30%) in our study. When allele frequency (Pro Vs Leu) was compared, 564Leu allele was found to be significantly associated with higher aSAH risk (OR = 1.36, 95%CI = 1.01-1.83, p = 0.04). (OR = 1.36, 95%CI = 1.01-1.83, p = 0.04). Regarding rs5985 and rs5982, significant association was found in the log-additive model (OR = 0.57, 95%CI = 0.33-0.97, p = 0.034; OR = 1.32, 95%CI = 1.00-1.72, p = 0.043). CONCLUSION These results suggest that 34Leu allele was a protective factor for lower risk of aSAH whereas 564Leu allele was associated with higher risk of aSAH in South Indian population.
Collapse
Affiliation(s)
- Arati Suvatha
- Department of Human Genetics, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, 560029, India
| | - M K Sibin
- Department of Biochemistry, Armed Forces Medical College, Pune, 411040, India
| | - Dhananjaya I Bhat
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore, 560029, India
| | - K V L Narasingarao
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore, 560029, India
| | - Vikas Vazhayil
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore, 560029, India
| | - G K Chetan
- Department of Human Genetics, National Institute of Mental Health and Neuro Sciences, Bangalore, Karnataka, 560029, India.
| |
Collapse
|
144
|
Blood lipid-related low-frequency variants in LDLR and PCSK9 are associated with onset age and risk of myocardial infarction in Japanese. Sci Rep 2018; 8:8107. [PMID: 29802317 PMCID: PMC5970143 DOI: 10.1038/s41598-018-26453-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 05/14/2018] [Indexed: 12/24/2022] Open
Abstract
Recent studies have revealed the importance of rare variants in myocardial infarction (MI) susceptibility in European populations. Because genetic architectures vary in different populations, we investigated how they contribute to MI susceptibility in Japanese subjects. We performed targeted sequencing of 36 coronary artery disease risk genes, identified by genome-wide association studies, in 9,956 cases and 8,373 controls. Gene-based association tests identified significant enrichment of rare variants in LDLR and PCSK9 in MI cases. We identified 52 (novel 22) LDLR variants predicted to be damaging. Carriers of these variants showed a higher risk of MI (carriers/non-carriers 89/9867 in cases, 17/8356 controls, OR = 4.4, P = 7.2 × 10−10), higher LDL-cholesterol levels and younger age of onset for MI. With respect to PCSK9, E32K carriers showed higher LDL-cholesterol levels and younger age of onset for MI, whereas R93C carriers had lower LDL-cholesterol levels. A significant correlation between LDL-cholesterol levels and onset age of MI was observed in these variant carriers. In good agreement with previous studies in patients with familial hypercholesterolaemia, our study in the Japanese general population showed that rare variants in LDLR and PCSK9 were associated with the onset age of MI by altering LDL-cholesterol levels.
Collapse
|
145
|
Lin Y, Liu L, Sheng Y, Shen C, Zheng X, Zhou F, Yang S, Yin X, Zhang X. A catalog of potential putative functional variants in psoriasis genome-wide association regions. PLoS One 2018; 13:e0196635. [PMID: 29715312 PMCID: PMC5929547 DOI: 10.1371/journal.pone.0196635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/15/2018] [Indexed: 01/20/2023] Open
Abstract
Psoriasis is a common inflammatory skin disease, with considerable genetic contribution. Genome-wide association studies have successfully identified a number of genomic regions for the risk of psoriasis. However, it is challenging to pinpoint the functional causal variants and then further decipher the genetic mechanisms underlying each region. In order to prioritize potential functional causal variants within psoriasis susceptibility regions, we integrated the genetic association findings and functional genomic data publicly available, i.e. histone modifications in relevant immune cells. We characterized a pervasive enrichment pattern of psoriasis variants in five core histone marks across immune cells/tissues. We discovered that genetic alleles within psoriasis association regions might influence gene expression levels through significantly affecting the binding affinities of 17 transcription factors. We established a catalog of 654 potential functional causal variants for psoriasis and suggested that they significantly overlapped with causal variants for autoimmune diseases. We identified potential causal variant rs79824801 overlay with the peaks of five histone marks in primary CD4+ T cells. Its alternative allele affected the binding affinity of transcription factor IKZF1. This study highlights the complex genetic architecture and complicated mechanisms for psoriasis. The findings will inform the functional experiment design for psoriasis.
Collapse
Affiliation(s)
- Yan Lin
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Department of Dermatology, The Fourth Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
| | - Lu Liu
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
| | - Yujun Sheng
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
| | - Changbing Shen
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Xiaodong Zheng
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
| | - Fusheng Zhou
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
| | - Sen Yang
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
| | - Xianyong Yin
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Xuejun Zhang
- Institute of Dermatology, Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Key lab of Dermatology, Ministry of Education, Anhui Medical University and State Key lab of Dermatology Incubation, Hefei, Anhui, China
| |
Collapse
|
146
|
Al-Tahan S, Al-Obeidi E, Yoshioka H, Lakatos A, Weiss L, Grafe M, Palmio J, Wicklund M, Harati Y, Omizo M, Udd B, Kimonis V. Novel valosin-containing protein mutations associated with multisystem proteinopathy. Neuromuscul Disord 2018; 28:491-501. [PMID: 29754758 DOI: 10.1016/j.nmd.2018.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/28/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022]
Abstract
Over fifty missense mutations in the gene coding for valosin-containing protein (VCP) are associated with a unique autosomal dominant adult-onset progressive disease associated with combinations of proximo-distal inclusion body myopathy (IBM), Paget's disease of bone (PDB), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS). We report the clinical, histological, and molecular findings in four new patients/families carrying novel VCP mutations: c.474 G > A (p.M158I); c.478 G > C (p.A160P); c.383G > C (p.G128A); and c.382G > T (p.G128C). Clinical features included myopathy, PDB, ALS and Parkinson's disease though frontotemporal dementia was not an associated feature in these families. One of the patients was noted to have severe manifestations of PDB and was suspected of having neoplasia. There were wide inter- and intra-familial variations making genotype-phenotype correlations difficult between the novel mutations and frequency or age of onset of IBM, PDB, FTD, ALS and Parkinson's disease. Increasing awareness of the full spectrum of clinical presentations will improve diagnosis of VCP-related diseases and thus proactively manage or prevent associated clinical features such as PDB.
Collapse
Affiliation(s)
- Sejad Al-Tahan
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA
| | - Ebaa Al-Obeidi
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA
| | - Hiroshi Yoshioka
- Department of Radiological Sciences, University of California, Irvine, CA
| | - Anita Lakatos
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA
| | - Lan Weiss
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA
| | - Marjorie Grafe
- Department of Pathology, Oregon Health and Science University, Portland, OR
| | - Johanna Palmio
- Neuromuscular Research Center, Tampere University and University Hospital, Neurology, Tampere, Finland
| | - Matt Wicklund
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO
| | - Yadollah Harati
- Department of Neurology, Baylor College of Medicine, Houston, TX
| | | | - Bjarne Udd
- Neuromuscular Research Center, Tampere University and University Hospital, Neurology, Tampere, Finland; Folkhälsan Institute of Genetics and the Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland; Neurology Department, Vasa Central Hospital, Vasa, Finland
| | - Virginia Kimonis
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA.
| |
Collapse
|
147
|
Seifi M, Walter MA. Accurate prediction of functional, structural, and stability changes in PITX2 mutations using in silico bioinformatics algorithms. PLoS One 2018; 13:e0195971. [PMID: 29664915 PMCID: PMC5903617 DOI: 10.1371/journal.pone.0195971] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/03/2018] [Indexed: 11/24/2022] Open
Abstract
Mutations in PITX2 have been implicated in several genetic disorders, particularly Axenfeld-Rieger syndrome. In order to determine the most reliable bioinformatics tools to assess the likely pathogenicity of PITX2 variants, the results of bioinformatics predictions were compared to the impact of variants on PITX2 structure and function. The MutPred, Provean, and PMUT bioinformatic tools were found to have the highest performance in predicting the pathogenicity effects of all 18 characterized missense variants in PITX2, all with sensitivity and specificity >93%. Applying these three programs to assess the likely pathogenicity of 13 previously uncharacterized PITX2 missense variants predicted 12/13 variants as deleterious, except A30V which was predicted as benign variant for all programs. Molecular modeling of the PITX2 homoedomain predicts that of the 31 known PITX2 variants, L54Q, F58L, V83F, V83L, W86C, W86S, and R91P alter PITX2's structure. In contrast, the remaining 24 variants are not predicted to change PITX2's structure. The results of molecular modeling, performed on all the PITX2 missense mutations located in the homeodomain, were compared with the findings of eight protein stability programs. CUPSAT was found to be the most reliable in predicting the effect of missense mutations on PITX2 stability. Our results showed that for PITX2, and likely other members of this homeodomain transcription factor family, MutPred, Provean, PMUT, molecular modeling, and CUPSAT can reliably be used to predict PITX2 missense variants pathogenicity.
Collapse
Affiliation(s)
- Morteza Seifi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Michael A. Walter
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
148
|
Broncy L, Njima BB, Méjean A, Béroud C, Romdhane KB, Ilie M, Hofman V, Muret J, Hofman P, Bouhamed HC, Paterlini-Bréchot AP. Single-cell genetic analysis validates cytopathological identification of circulating cancer cells in patients with clear cell renal cell carcinoma. Oncotarget 2018; 9:20058-20074. [PMID: 29732003 PMCID: PMC5929446 DOI: 10.18632/oncotarget.25102] [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: 10/07/2017] [Accepted: 03/24/2018] [Indexed: 12/14/2022] Open
Abstract
CONTEXT Circulating Rare Cells (CRC) are non-haematological cells circulating in blood. They include Circulating Cancer Cells (CCC) and cells with uncertain malignant features (CRC-UMF) according to cytomorphology. Clear cell renal cell carcinomas frequently bear a mutated Von Hippel-Lindau (VHL) gene. AIM To match blind genetic analysis of CRC and tumor samples with CRC cytopathological diagnosis. RESULTS 29/30 patients harboured CRC (20 harboured CCC, 29 CRC-UMF) and 25/29 patients carried VHL mutations in their tumour. 205 single CRC (64 CCC, 141 CRC-UMF) provided genetic data. 57/57 CCC and 104/125 CRC-UMF from the 25 patients with VHL-mutated tumor carried the same VHL mutation detected in the tumor. Seven CCC and 16 CRC-UMF did not carry VHL mutations but were found in patients with wild-type VHL tumor tissue. CONCLUSIONS All the CCC and 83,2% (104/125) of the CRC-UMF were found to carry the same VHL mutation identified in the corresponding tumorous tissue, validating cytopathological identification of CCC in patients with clear cell renal cell carcinoma. METHODS The blood of 30 patients with clear cell renal cell carcinoma was treated by ISET® for CRC isolation, cytopathology and single-cell VHL mutations analysis, performed blindly and compared to VHL mutations of corresponding tumor tissues and leukocytes.
Collapse
Affiliation(s)
- Lucile Broncy
- INSERM Unit 1151, Faculté de Médecine Paris Descartes, Paris, France
| | - Basma Ben Njima
- Genetics and Pathology Departments, University of Tunis, Tunis, Tunisia
| | - Arnaud Méjean
- Service d'Urologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Christophe Béroud
- Aix Marseille University, INSERM, MMG, Marseille, France
- APHM, Hôpital TIMONE Enfants, Laboratoire de Génétique Moléculaire, Marseille, France
| | | | - Marius Ilie
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | - Veronique Hofman
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | - Jane Muret
- Institut Curie, PSL Research University, Département d'Anesthésie Réanimation Douleur, Paris, France
| | - Paul Hofman
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | | | - And Patrizia Paterlini-Bréchot
- INSERM Unit 1151, Faculté de Médecine Paris Descartes, Paris, France
- Laboratoire de Biochimie A, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
149
|
Hess K, Oliverio R, Nguyen P, Le D, Ellis J, Kdeiss B, Ord S, Chalkia D, Nikolaidis N. Concurrent action of purifying selection and gene conversion results in extreme conservation of the major stress-inducible Hsp70 genes in mammals. Sci Rep 2018; 8:5082. [PMID: 29572464 PMCID: PMC5865164 DOI: 10.1038/s41598-018-23508-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/14/2018] [Indexed: 12/28/2022] Open
Abstract
Several evolutionary mechanisms alter the fate of mutations and genes within populations based on their exhibited functional effects. To understand the underlying mechanisms involved in the evolution of the cellular stress response, a very conserved mechanism in the course of organismal evolution, we studied the patterns of natural genetic variation and functional consequences of polymorphisms of two stress-inducible Hsp70 genes. These genes, HSPA1A and HSPA1B, are major orchestrators of the cellular stress response and are associated with several human diseases. Our phylogenetic analyses revealed that the duplication of HSPA1A and HSPA1B originated in a lineage proceeding to placental mammals, and henceforth they remained in conserved synteny. Additionally, analyses of synonymous and non-synonymous changes suggest that purifying selection shaped the HSPA1 gene diversification, while gene conversion resulted in high sequence conservation within species. In the human HSPA1-cluster, the vast majority of mutations are synonymous and specific genic regions are devoid of mutations. Furthermore, functional characterization of several human polymorphisms revealed subtle differences in HSPA1A stability and intracellular localization. Collectively, the observable patterns of HSPA1A-1B variation describe an evolutionary pattern, in which purifying selection and gene conversion act simultaneously and conserve a major orchestrator of the cellular stress response.
Collapse
Affiliation(s)
- Kyle Hess
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA.,Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Ryan Oliverio
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Peter Nguyen
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Dat Le
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Jacqueline Ellis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Brianna Kdeiss
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Sara Ord
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Dimitra Chalkia
- UCLA Center for Systems Biomedicine, Division of Digestive Diseases, School of Medicine, Los Angeles, CA, USA
| | - Nikolas Nikolaidis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA.
| |
Collapse
|
150
|
Abstract
BACKGROUND Given the etiologic heterogeneity of disease classification using clinical phenomenology, we employed contemporary criteria to classify variants associated with myoclonic epilepsy with ragged-red fibers (MERRF) syndrome and to assess the strength of evidence of gene-disease associations. Standardized approaches are used to clarify the definition of MERRF, which is essential for patient diagnosis, patient classification, and clinical trial design. METHODS Systematic literature and database search with application of standardized assessment of gene-disease relationships using modified Smith criteria and of variants reported to be associated with MERRF using modified Yarham criteria. RESULTS Review of available evidence supports a gene-disease association for two MT-tRNAs and for POLG. Using modified Smith criteria, definitive evidence of a MERRF gene-disease association is identified for MT-TK. Strong gene-disease evidence is present for MT-TL1 and POLG. Functional assays that directly associate variants with oxidative phosphorylation impairment were critical to mtDNA variant classification. In silico analysis was of limited utility to the assessment of individual MT-tRNA variants. With the use of contemporary classification criteria, several mtDNA variants previously reported as pathogenic or possibly pathogenic are reclassified as neutral variants. CONCLUSIONS MERRF is primarily an MT-TK disease, with pathogenic variants in this gene accounting for ~90% of MERRF patients. Although MERRF is phenotypically and genotypically heterogeneous, myoclonic epilepsy is the clinical feature that distinguishes MERRF from other categories of mitochondrial disorders. Given its low frequency in mitochondrial disorders, myoclonic epilepsy is not explained simply by an impairment of cellular energetics. Although MERRF phenocopies can occur in other genes, additional data are needed to establish a MERRF disease-gene association. This approach to MERRF emphasizes standardized classification rather than clinical phenomenology, thus improving patient diagnosis and clinical trial design.
Collapse
|