651
|
Ileana Dumbrava E, Brusco L, Daniels MS, Wathoo C, Shaw KR, Lu KH, Zheng X, Strong LC, Litton J, Arun BK, Eterovic AK, Routbort MJ, Patel KP, Qi Y, Piha-Paul SA, Subbiah V, Hong DS, Rodon J, Kopetz S, Mendelsohn J, Mills GB, Chen K, Meric-Bernstam F. Expanded analysis of secondary germline findings from matched tumor/normal sequencing identifies additional clinically significant mutations. JCO Precis Oncol 2019; 3:PO.18.00143. [PMID: 31517177 PMCID: PMC6741435 DOI: 10.1200/po.18.00143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) for tumor molecular profiling can reveal secondary germline pathogenic and likely pathogenic variants (LPV/PV). The American College of Medical Genetics (ACMG) recommends return of secondary results for a subset of 59 genes, but other genes with evidence of clinical utility are emerging. We previously reported that 4.3% of patients who underwent NGS of a targeted panel of 201 genes had LPV/PV based on the ACMG list. Here we report the frequency of additional germline cancer-related gene variants and discuss their clinical utility. PATIENTS AND METHODS Matched tumor and germline DNA NGS of a targeted panel of 201 genes was performed in a research laboratory on samples from 1000 patients with advanced or metastatic solid tumors enrolled in a molecular testing protocol (NCT01772771). The frequency of germline LPV/PV in 54 cancer-related genes, beyond the genes in ACMG list, were analyzed. RESULTS Among 1000 patients who underwent tumor/normal DNA sequencing, 46 (4.6%) were found to have a germline LPV/PV in the following genes: AR-(5), ATM-(4), BAP1-(1), CDH1-(1), CDKN2A-(1), CHEK1-(2), CHEK2-(10), EGFR-(1), ERCC3-(4), ERCC5-(1), HNF1B-(1), HRAS-(1), MITF-(4), MLL3-(1), NF1-(3), PKHD1-(4), PTCH1-(1), and SMARCA4-(1). Thus, a total 8.7% of patients had an LPV/PV with 2 patients having 2 concomitant germline LPV/PV. Five mutations in high-penetrance hereditary cancer predisposition genes were selected to be returned to patients or their representatives: BAP1, CDH1, CDKN2A, EGFR, and SMARCA4. CONCLUSIONS Broader genomic testing is likely to identify additional secondary pathogenic germline alterations, some with potential clinical utility for return to patients and their relatives. The recommended genes for which germline results should be returned are continually changing, warranting continued study.
Collapse
Affiliation(s)
| | - Lauren Brusco
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Chetna Wathoo
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kenna R. Shaw
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Karen H. Lu
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiaofeng Zheng
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Jennifer Litton
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Banu K. Arun
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Keyur P. Patel
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yuan Qi
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David S. Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jordi Rodon
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - John Mendelsohn
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gordon B. Mills
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ken Chen
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
652
|
Estimating carrier frequencies of newborn screening disorders using a whole-genome reference panel of 3552 Japanese individuals. Hum Genet 2019; 138:389-409. [PMID: 30887117 DOI: 10.1007/s00439-019-01998-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/06/2019] [Indexed: 12/19/2022]
Abstract
Incidence rates of Mendelian diseases vary among ethnic groups, and frequencies of variant types of causative genes also vary among human populations. In this study, we examined to what extent we can predict population frequencies of recessive disorders from genomic data, and explored better strategies for variant interpretation and classification. We used a whole-genome reference panel from 3552 general Japanese individuals constructed by the Tohoku Medical Megabank Organization (ToMMo). Focusing on 32 genes for 17 congenital metabolic disorders included in newborn screening (NBS) in Japan, we identified reported and predicted pathogenic variants through variant annotation, interpretation, and multiple ways of classifications. The estimated carrier frequencies were compared with those from the Japanese NBS data based on 1,949,987 newborns from a previous study. The estimated carrier frequency based on genomic data with a recent guideline of variant interpretation for the PAH gene, in which defects cause hyperphenylalaninemia (HPA) and phenylketonuria (PKU), provided a closer estimate to that by the observed incidence than the other methods. In contrast, the estimated carrier frequencies for SLC25A13, which causes citrin deficiency, were much higher compared with the incidence rate. The results varied greatly among the 11 NBS diseases with single responsible genes; the possible reasons for departures from the carrier frequencies by reported incidence rates were discussed. Of note, (1) the number of pathogenic variants increases by including additional lines of evidence, (2) common variants with mild effects also contribute to the actual frequency of patients, and (3) penetrance of each variant remains unclear.
Collapse
|
653
|
Carli D, Giorgio E, Pantaleoni F, Bruselles A, Barresi S, Riberi E, Licciardi F, Gazzin A, Baldassarre G, Pizzi S, Niceta M, Radio FC, Molinatto C, Montin D, Calvo PL, Ciolfi A, Fleischer N, Ferrero GB, Brusco A, Tartaglia M. NBAS
pathogenic variants: Defining the associated clinical and facial phenotype and genotype–phenotype correlations. Hum Mutat 2019; 40:721-728. [DOI: 10.1002/humu.23734] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/05/2019] [Accepted: 02/28/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Diana Carli
- Department of Public Health and PediatricsUniversity of TorinoTorino Italy
| | - Elisa Giorgio
- Department of Medical SciencesUniversity of TorinoTorino Italy
| | - Francesca Pantaleoni
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù IRCSSRome Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular MedicineIstituto Superiore di SanitàRome Italy
| | - Sabina Barresi
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù IRCSSRome Italy
| | - Evelise Riberi
- Department of Public Health and PediatricsUniversity of TorinoTorino Italy
| | | | - Andrea Gazzin
- Department of Public Health and PediatricsUniversity of TorinoTorino Italy
| | | | - Simone Pizzi
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù IRCSSRome Italy
| | - Marcello Niceta
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù IRCSSRome Italy
| | - Francesca C. Radio
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù IRCSSRome Italy
| | - Cristina Molinatto
- Department of Public Health and PediatricsUniversity of TorinoTorino Italy
| | - Davide Montin
- Department of Public Health and PediatricsUniversity of TorinoTorino Italy
| | - Pier L. Calvo
- Pediatric Gastroenterology UnitCittà della Salute e della Scienza University HospitalTorino Italy
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù IRCSSRome Italy
| | | | | | - Alfredo Brusco
- Department of Medical SciencesUniversity of TorinoTorino Italy
- Medical Genetics UnitCittà della Salute e della Scienza University HospitalTorino Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù IRCSSRome Italy
| |
Collapse
|
654
|
Systematic evaluation of gene variants linked to hearing loss based on allele frequency threshold and filtering allele frequency. Sci Rep 2019; 9:4583. [PMID: 30872718 PMCID: PMC6418148 DOI: 10.1038/s41598-019-41068-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/25/2019] [Indexed: 02/02/2023] Open
Abstract
As the number of genes identified for linkage to hearing loss has been increasing and more public databases have become available, we aimed to systematically evaluate all variants reported for nonsyndromic hearing loss (NSHL) based on their allele frequencies (AFs) in the general population. Among the 3,549 variants in 97 NSHL genes reported as pathogenic/likely pathogenic in ClinVar and HGMD, 1,618 were found in public databases (gnomAD, ExAC, EVS, and 1000G). To evaluate the pathogenicity of these variants, we employed AF thresholds and NSHL-optimized ACMG guidelines. AF thresholds were determined using a high-resolution variant frequency framework and Hardy-Weinberg equilibrium calculation: 0.6% and 0.1% for recessive and dominant genes, respectively. Filtering AFs of variants linked to NSHL were obtained based on AFs reported in gnomAD and ExAC. We found that 48 variants in 23 genes had filtering AFs above the suggested thresholds and assumed that these variants might be benign based on their filtering AFs. 47 variants, except for one notorious high-frequency GJB2 mutation (c.109G > A; p.Val37Ile), were confirmed to be benign/likely benign by the NSHL-optimized ACMG guidelines. The proposed systematic approach will aid in precise evaluation of NSHL variant pathogenicity in the context of filtering AFs, AF thresholds, and NSHL-specific ACMG guidelines, thus improving NSHL diagnostics.
Collapse
|
655
|
Kudryavtseva AV, Lukyanova EN, Kalinin DV, Zaretsky AR, Pokrovsky AV, Golovyuk AL, Fedorova MS, Pudova EA, Kharitonov SL, Pavlov VS, Kobelyatskaya AA, Melnikova NV, Dmitriev AA, Polyakov AP, Alekseev BY, Kiseleva MV, Kaprin AD, Krasnov GS, Snezhkina AV. Mutational load in carotid body tumor. BMC Med Genomics 2019; 12:39. [PMID: 30871634 PMCID: PMC6416835 DOI: 10.1186/s12920-019-0483-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Carotid body tumor (CBT) is a rare neoplasm arising from paraganglion located near the bifurcation of the carotid artery. There is great intra-tumor heterogeneity, and CBT development could be associated with both germline and somatic allelic variants. Studies on the molecular genetics of CBT are limited, and the molecular mechanisms of its pathogenesis are not fully understood. This work is focused on the estimation of mutational load (ML) in CBT. Methods Using the NextSeq 500 platform, we performed exome sequencing of tumors with matched lymph node tissues and peripheral blood obtained from six patients with CBT. To obtain reliable results in tumors with low ML, we developed and successfully applied a complex approach for the analysis of sequencing data. ML was evaluated as the number of somatic variants per megabase (Mb) of the target regions covered by the Illumina TruSeq Exome Library Prep Kit. Results The ML in CBT varied in the range of 0.09–0.28/Mb. Additionally, we identified several pathogenic/likely pathogenic somatic and germline allelic variants across six patients studied (including TP53 variants). Conclusions Using the developed approach, we estimated the ML in CBT, which is much lower than in common malignant tumors. Identified variants in known paraganglioma/pheochromocytoma-causative genes and novel genes could be associated with the pathogenesis of CBT. The obtained results expand our knowledge of the mutation process in CBT as well as the biology of tumor development.
Collapse
Affiliation(s)
- Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Elena N Lukyanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry V Kalinin
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrew R Zaretsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anatoly V Pokrovsky
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander L Golovyuk
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergey L Kharitonov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladislav S Pavlov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey P Polyakov
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Marina V Kiseleva
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|
656
|
Ravichandran V, Shameer Z, Kemel Y, Walsh M, Cadoo K, Lipkin S, Mandelker D, Zhang L, Stadler Z, Robson M, Offit K, Vijai J. Toward automation of germline variant curation in clinical cancer genetics. Genet Med 2019; 21:2116-2125. [PMID: 30787465 PMCID: PMC6703969 DOI: 10.1038/s41436-019-0463-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 02/06/2019] [Indexed: 01/01/2023] Open
Abstract
Purpose: Cancer care professionals are confronted with interpreting results from multiplexed gene sequencing of patients at hereditary risk for cancer. Assessments for variant classification now require orthogonal data searches and aggregation of multiple lines of evidence from diverse resources. The clinical genetics community needs a fast algorithm that automates ACMG based variant classification and provides uniform results. Methods: Pathogenicity of Mutation Analyzer (PathoMAN) automates germline genomic variant curation from clinical sequencing based on ACMG guidelines. PathoMAN aggregates multiple tracks of genomic, protein and disease specific information from public sources. We compared expertly curated variant data from clinical laboratories to assess performance. Results: PathoMAN achieved a high overall concordance of 94.4% for pathogenic and 81.1% for benign variants. We observed negligible discordance (0.3% pathogenic, 0% benign) when contrasted against expert curated variants. Some loss of resolution (5.3% pathogenic, 18.9% benign) and gain of resolution (1.6% pathogenic, 3.8% benign) was also observed. Conclusion: Automation of variant curation enables unbiased, fast, efficient delivery of results in both clinical and laboratory research. We highlight the advantages and weaknesses related to the programmable automation of variant classification. PathoMAN will aid in rapid variant classification by generating robust models using a knowledge-base of diverse genetic data. https://pathoman.mskcc.org
Collapse
Affiliation(s)
- Vignesh Ravichandran
- Niehaus Center For Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zarina Shameer
- Niehaus Center For Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yelena Kemel
- Niehaus Center For Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Walsh
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karen Cadoo
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven Lipkin
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Diana Mandelker
- Diagnostic Molecular Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Liying Zhang
- Diagnostic Molecular Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zsofia Stadler
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Robson
- Niehaus Center For Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, USA.,Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Offit
- Niehaus Center For Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, USA.,Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph Vijai
- Niehaus Center For Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
657
|
Oh SH, Sung YH, Kim I, Sim CK, Lee JH, Baek M, Pack CG, Seok C, Seo EJ, Lee MS, Kim KM. Novel Compound Heterozygote Mutation in IL10RA in a Patient With Very Early-Onset Inflammatory Bowel Disease. Inflamm Bowel Dis 2019; 25:498-509. [PMID: 30462267 DOI: 10.1093/ibd/izy353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Very early-onset inflammatory bowel disease (VEO-IBD) is often associated with monogenetic disorders. IL-10RA deficiency is one of the major causal mutations in VEO-IBD. Here, we aimed to identify the causal mutation associated with severe IBD in a 1-year-old patient, validate the pathogenicity of the mutation, and characterize the mutant protein. METHODS To identify the causal mutation, targeted exome sequencing (ES) was performed using the genomic DNA from the patient. To validate the pathogenicity, IL-10RA functional tests were performed using the patient's peripheral blood mononuclear cells (PBMCs). Additionally, flow cytometry analysis, confocal microscopy on overexpressed green fluorescent protein-fused mutants, and computational analysis on the structures of IL-10RA proteins were performed. RESULTS We identified a novel compound heterozygote mutation p.[Tyr91Cys];[Pro146Alafs*40] in the IL10RA gene of the patient. The missense variant p.Tyr91Cys was previously identified but not functionally tested, and a frameshift variant, p.Pro146Alafs*40, is novel and nonfunctional. PBMCs from the patient showed defective signal transducer and activator of transcription 3 activation. The p.Tyr91Cys mutant protein failed to properly localize on the plasma membrane. The p.Tyr91Cys mutation seems to disrupt the hydrophobic core structure surrounding the tyrosine 91 residue, causing structural instability. CONCLUSIONS Targeted ES and linkage analysis identified novel compound heterozygous mutations p.[Tyr91Cys];[Pro146Alafs*40] in the IL10RA gene of a child with severe VEO-IBD. p.Tyr91Cys proteins were functionally defective in IL-10RA signaling and failed to properly localize on the plasma membrane, probably due to its structural instability.
Collapse
Affiliation(s)
- Seak Hee Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Hoon Sung
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inki Kim
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chan Kyu Sim
- Lab of Molecular Immunology and Medicine, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung Hoon Lee
- Lab of Molecular Immunology and Medicine, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Minkyung Baek
- Department of Chemistry, Seoul National University, Seoul, Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine, Asan Institutes for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chaok Seok
- Department of Chemistry, Seoul National University, Seoul, Korea
| | - Eul Ju Seo
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Myeong Sup Lee
- Lab of Molecular Immunology and Medicine, Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Mo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
658
|
Németh E, Krzystanek M, Reiniger L, Ribli D, Pipek O, Sztupinszki Z, Glasz T, Csabai I, Moldvay J, Szallasi Z, Szüts D. The genomic imprint of cancer therapies helps timing the formation of metastases. Int J Cancer 2019; 145:694-704. [PMID: 30694556 DOI: 10.1002/ijc.32159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/14/2018] [Accepted: 01/16/2019] [Indexed: 11/07/2022]
Abstract
A retrospective determination of the time of metastasis formation is essential for a better understanding of the evolution of oligometastatic cancer. This study was based on the hypothesis that genomic alterations induced by cancer therapies could be used to determine the temporal order of the treatment and the formation of metastases. We analysed the whole genome sequence of a primary tumour sample and three metastatic sites derived from autopsy samples from a young never-smoker lung adenocarcinoma patient with an activating EGFR mutation. Mutation detection methods were refined to accurately detect and distinguish clonal and subclonal mutations. In comparison to a panel of samples from untreated smoker or never-smoker patients, we showed that the mutagenic effect of cisplatin treatment could be specifically detected from the base substitution mutations. Metastases that arose before or after chemotherapeutic treatment could be distinguished based on the allele frequency of cisplatin-induced dinucleotide mutations. In addition, genomic rearrangements and late amplification of the EGFR gene likely induced by afatinib treatment following the acquisition of a T790M gefitinib resistance mutation provided further evidence to tie the time of metastasis formation to treatment history. The established analysis pipeline for the detection of treatment-derived mutations allows the drawing of tumour evolutionary paths based on genomic data, showing that metastases may be seeded well before they become detectable by clinical imaging.
Collapse
Affiliation(s)
- Eszter Németh
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Marcin Krzystanek
- Translational Cancer Genomics, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Lilla Reiniger
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,SE-NAP Brain Metastasis Research group, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Dezső Ribli
- Department of Physics of Complex Systems, Eötvös University, Budapest, Hungary
| | - Orsolya Pipek
- Department of Physics of Complex Systems, Eötvös University, Budapest, Hungary
| | - Zsófia Sztupinszki
- Translational Cancer Genomics, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Tibor Glasz
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - István Csabai
- Department of Physics of Complex Systems, Eötvös University, Budapest, Hungary
| | - Judit Moldvay
- SE-NAP Brain Metastasis Research group, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Department of Tumor Biology, National Korányi Institute of Pulmonology-Semmelweis University, Budapest, Hungary
| | - Zoltan Szallasi
- Translational Cancer Genomics, Danish Cancer Society Research Center, Copenhagen, Denmark.,SE-NAP Brain Metastasis Research group, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Dávid Szüts
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
659
|
Xie Z, Hou Y, Yu M, Liu Y, Fan Y, Zhang W, Wang Z, Xiong H, Yuan Y. Clinical and genetic spectrum of sarcoglycanopathies in a large cohort of Chinese patients. Orphanet J Rare Dis 2019; 14:43. [PMID: 30764848 PMCID: PMC6376703 DOI: 10.1186/s13023-019-1021-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/03/2019] [Indexed: 11/20/2022] Open
Abstract
Background Sarcoglycanopathies comprise four subtypes of autosomal recessive limb-girdle muscular dystrophy (LGMD2C, LGMD2D, LGMD2E, and LGMD2F) that are caused, respectively, by mutations in the SGCG, SGCA, SGCB, and SGCD genes. Knowledge about the clinical and genetic features of sarcoglycanopathies in Chinese patients is limited. The aims of this study were to investigate in detail the clinical manifestations, sarcoglycan expression, and gene mutations in Chinese patients with sarcoglycanopathies and to identify possible correlations between them. Results Of 3638 patients for suspected neuromuscular diseases (1733 with inherited myopathies, 1557 with acquired myopathies, and 348 unknown), 756 patients had next-generation sequencing (NGS) diagnostic panel. Twenty-five patients with sarcoglycanopathies (11.5%) were identified from 218 confirmed LGMDs, comprising 18 with LGMD2D, 6 with LGMD2E, and one with LGMD2C. One patient with LGMD2D also had Charcot-Marie-Tooth 1A. The clinical phenotypes of the patients with LGMD2D or LGMD2E were markedly heterogeneous. Muscle biopsy showed a dystrophic pattern in 19 patients and mild myopathic changes in 6. The percentage of correct prediction of genotype based on expression of sarcoglycan was 36.0% (4 LGMD2D, 4 LGMD2E, and one LGMD2C). There was a statistically significant positive correlation between reduction of α-sarcoglycan level and disease severity in LGMD2D. Thirty-five mutations were identified in SGCA, SGCB, SGCG, and PMP22, 16 of which were novel. Exon 3 of SGCA was a hotspot region for mutations in LGMD2D. The missense mutation c.662G > A (p.R221H) was the most common mutation in SGCA. Missense mutations in both alleles of SGCA were associated with a relative benign disease course. No obvious clinical, sarcoglycan expression, and genetic correlation was found in LGMD2E. Conclusions This study expands the clinical and genetic spectrum of sarcoglycanopathies in Chinese patients and provides evidence that disease severity of LGMD2D may be predicted by α-sarcoglycan expression and SGCA mutation. Electronic supplementary material The online version of this article (10.1186/s13023-019-1021-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhiying Xie
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yue Hou
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yilin Liu
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yanbin Fan
- Department of Pediatrics, Peking University First Hospital, Xishiku St, Xicheng District, Beijing, 100034, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Hui Xiong
- Department of Pediatrics, Peking University First Hospital, Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
660
|
Bhaskaran SP, Chandratre K, Gupta H, Zhang L, Wang X, Cui J, Kim YC, Sinha S, Jiang L, Lu B, Wu X, Qin Z, Huang T, Wang SM. Germline variation in BRCA1/2 is highly ethnic-specific: Evidence from over 30,000 Chinese hereditary breast and ovarian cancer patients. Int J Cancer 2019; 145:962-973. [PMID: 30702160 PMCID: PMC6617753 DOI: 10.1002/ijc.32176] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/14/2019] [Accepted: 01/25/2019] [Indexed: 01/04/2023]
Abstract
BRCA1 and BRCA2 play essential roles in maintaining the genome stability. Pathogenic germline mutations in these two genes disrupt their function, lead to genome instability and increase the risk of developing breast and ovarian cancers. BRCA mutations have been extensively screened in Caucasian populations, and the resulting information are used globally as the standard reference in clinical diagnosis, treatment and prevention of BRCA-related cancers. Recent studies suggest that BRCA mutations can be ethnic-specific, raising the question whether a Caucasian-based BRCA mutation information can be used as a universal standard worldwide, or whether an ethnicity-based BRCA mutation information system need to be developed for the corresponding ethnic populations. In this study, we used Chinese population as a model to test ethnicity-specific BRCA mutations considering that China has one of the latest numbers of breast cancer patients therefore BRCA mutation carriers. Through comprehensive data mining, standardization and annotation, we collected 1,088 distinct BRCA variants derived from over 30,000 Chinese individuals, one of the largest BRCA data set from a non-Caucasian population covering nearly all known BRCA variants in the Chinese population (https://dbBRCA-Chinese.fhs.umac.mo). Using this data, we performed multi-layered analyses to determine the similarities and differences of BRCA variation between Chinese and non-Chinese ethnic populations. The results show the substantial differences of BRCA data between Chinese and non-Chinese ethnicities. Our study indicates that the current Caucasian population-based BRCA data is not adequate to represent the BRCA status in non-Caucasian populations. Therefore, ethnic-based BRCA standards need to be established to serve for the non-Caucasian populations.
Collapse
Affiliation(s)
- Shanmuga Priya Bhaskaran
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Khyati Chandratre
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Hemant Gupta
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Li Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Xiaoyu Wang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Jian Cui
- Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE
| | - Yeong C Kim
- Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE
| | - Siddharth Sinha
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Luhan Jiang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Boya Lu
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Xiaobing Wu
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Zixin Qin
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Teng Huang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - San Ming Wang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
661
|
Hebebrand M, Hüffmeier U, Trollmann R, Hehr U, Uebe S, Ekici AB, Kraus C, Krumbiegel M, Reis A, Thiel CT, Popp B. The mutational and phenotypic spectrum of TUBA1A-associated tubulinopathy. Orphanet J Rare Dis 2019; 14:38. [PMID: 30744660 PMCID: PMC6371496 DOI: 10.1186/s13023-019-1020-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/03/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The TUBA1A-associated tubulinopathy is clinically heterogeneous with brain malformations, microcephaly, developmental delay and epilepsy being the main clinical features. It is an autosomal dominant disorder mostly caused by de novo variants in TUBA1A. RESULTS In three individuals with developmental delay we identified heterozygous de novo missense variants in TUBA1A using exome sequencing. While the c.1307G > A, p.(Gly436Asp) variant was novel, the two variants c.518C > T, p.(Pro173Leu) and c.641G > A, p.(Arg214His) were previously described. We compared the variable phenotype observed in these individuals with a carefully conducted review of the current literature and identified 166 individuals, 146 born and 20 fetuses with a TUBA1A variant. In 107 cases with available clinical information we standardized the reported phenotypes according to the Human Phenotype Ontology. The most commonly reported features were developmental delay (98%), anomalies of the corpus callosum (96%), microcephaly (76%) and lissencephaly (agyria-pachygyria) (70%), although reporting was incomplete in the different studies. We identified a total of 121 specific variants, including 15 recurrent ones. Missense variants cluster in the C-terminal region around the most commonly affected amino acid position Arg402 (13.3%). In a three-dimensional protein model, 38.6% of all disease-causing variants including those in the C-terminal region are predicted to affect the binding of microtubule-associated proteins or motor proteins. Genotype-phenotype analysis for recurrent variants showed an overrepresentation of certain clinical features. However, individuals with these variants are often reported in the same publication. CONCLUSIONS With 166 individuals, we present the most comprehensive phenotypic and genotypic standardized synopsis for clinical interpretation of TUBA1A variants. Despite this considerable number, a detailed genotype-phenotype characterization is limited by large inter-study variability in reporting.
Collapse
Affiliation(s)
- Moritz Hebebrand
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Ulrike Hüffmeier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Regina Trollmann
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ute Hehr
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Steffen Uebe
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Mandy Krumbiegel
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Christian T Thiel
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany.
| | - Bernt Popp
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| |
Collapse
|
662
|
Wang X, Liu A, Lu Y, Hu Q. Novel compound heterozygous mutations in the SPTA1 gene, causing hereditary spherocytosis in a neonate with Coombs‑negative hemolytic jaundice. Mol Med Rep 2019; 19:2801-2807. [PMID: 30816434 PMCID: PMC6423610 DOI: 10.3892/mmr.2019.9947] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Hereditary spherocytosis (HS) is a common heterogeneous type of inherited hemolytic anemia characterized by jaundice and splenomegaly. Diagnosis of HS in neonates is considered unreliable, and is generally based on positive family history, spherocytes in peripheral smears, increased osmotic fragility, and jaundice. In the present study, routine laboratory tests, next‑generation sequencing, and Sanger sequencing were applied to diagnose a neonatal patient with Coombs‑negative hemolytic jaundice. The neonate had no family history of HS; however, spherocytes were observed in peripheral smears, and the patient exhibited Coombs‑negative and severe hemolytic jaundice, normal mean corpuscular hemoglobin concentration (MCHC) and mean corpuscular volume (MCV), normal glucose‑6‑phosphate dehydrogenase activity, negative thalassemia genetic mutation screening results, and negative autoimmune antibody tests. Novel compound heterozygous mutations in the spectrin‑α, erythrocytic 1 (SPTA1) gene (c.3897‑1G>C and c.5029G>A) were identified. The SPTA1 c.3897‑1G>C mutation in intron 27‑1, which disrupted the consensus splice site, was inherited from his asymptomatic mother, and the SPTA1 c.5029G>A (p.Gly1677Arg) mutation in trans with the SPTA1 c.3897‑1G>C mutation was inherited from his asymptomatic father. Sanger sequencing of mRNA reverse transcribed into cDNA identified a deletion of the first 10 nucleotides of exon 28, confirming the splicing mutation. In conclusion, the present study reports a rare case of autosomal‑recessive HS with a severe clinical phenotype, but normal MCHC and MCV.
Collapse
Affiliation(s)
- Xiong Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Aiguo Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yanjun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qun Hu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
663
|
Kotan LD, Isik E, Turan I, Mengen E, Akkus G, Tastan M, Gurbuz F, Yuksel B, Topaloglu AK. Prevalence and associated phenotypes of PLXNA1 variants in normosmic and anosmic idiopathic hypogonadotropic hypogonadism. Clin Genet 2019; 95:320-324. [PMID: 30467832 DOI: 10.1111/cge.13482] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 01/08/2023]
Abstract
Idiopathic hypogonadotropic hypogonadism (IHH) can be divided into two major forms, normosmic IHH and Kallmann syndrome (KS). Genetic mutations are responsible for the majority of IHH. PLXNA1 has recently been implicated in the GnRH neuron migration and the etiology of KS. We aimed to investigate the prevalence and associated phenotypes of PLXNA1 variants in a large cohort of IHH patients. We screened the whole exome data of 215 IHH patients in a single center for causative PLXNA1 variants. Our studies showed eight novel (p.Arg836His, p.Lys1451Arg, p.Val287Met, p.Val536Ile, p.Ser1850Arg, p.Ile1701Val, p.Arg319Trp, and p.Pro485Leu) and two previously described (p.Arg528Trp and p.Gly720Glu) heterozygous PLXNA1 variants in nine affected individuals from seven unrelated families. Only three of nine patients were anosmic (KS) while the remaining patients showed normal olfactory function (nIHH). Seven of nine patients (77.7%) harbored additional one or two variants in other nIHH/KS-associated genes, including PROKR2, IGSF10, HS6ST1, SEMA3E, CCDC141, FGFR1, NRP1, POLR3A, and SRA1. Our findings indicate that PLXNA1 variants cause not only anosmic but also normosmic IHH with a relatively high prevalence (3.9%). Heterozygous missense PLXNA1 variants appear to be involved together with other IHH gene variants in bringing about the IHH disease phenotype.
Collapse
Affiliation(s)
- Leman D Kotan
- Division of Pediatric Endocrinology, Cukurova University, Faculty of Medicine, Adana, Turkey
| | - Emregul Isik
- Gaziantep Children's Hospital, Department of Pediatrics, Clinics of Pediatric Endocrinology, Gaziantep, Turkey
| | - Ihsan Turan
- Division of Pediatric Endocrinology, Cukurova University, Faculty of Medicine, Adana, Turkey
| | - Eda Mengen
- Department of Pediatrics, Division of Pediatric Endocrinology, Ankara Children's Hematology and Oncology Training Hospital, Ankara, Turkey
| | - Gamze Akkus
- Division of Endocrinology, Cukurova University, Faculty of Medicine, Adana, Turkey
| | - Mehmet Tastan
- Division of Pediatric Endocrinology, Cukurova University, Faculty of Medicine, Adana, Turkey
| | - Fatih Gurbuz
- Division of Pediatric Endocrinology, Cukurova University, Faculty of Medicine, Adana, Turkey
| | - Bilgin Yuksel
- Division of Pediatric Endocrinology, Cukurova University, Faculty of Medicine, Adana, Turkey
| | - A Kemal Topaloglu
- Division of Pediatric Endocrinology, Cukurova University, Faculty of Medicine, Adana, Turkey
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
664
|
Nicolas E, Demidova EV, Iqbal W, Serebriiskii IG, Vlasenkova R, Ghatalia P, Zhou Y, Rainey K, Forman AF, Dunbrack RL, Golemis EA, Hall MJ, Daly MB, Arora S. Interaction of germline variants in a family with a history of early-onset clear cell renal cell carcinoma. Mol Genet Genomic Med 2019; 7:e556. [PMID: 30680959 PMCID: PMC6418363 DOI: 10.1002/mgg3.556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/31/2022] Open
Abstract
Background Identification of genetic factors causing predisposition to renal cell carcinoma has helped improve screening, early detection, and patient survival. Methods We report the characterization of a proband with renal and thyroid cancers and a family history of renal and other cancers by whole‐exome sequencing (WES), coupled with WES analysis of germline DNA from additional affected and unaffected family members. Results This work identified multiple predicted protein‐damaging variants relevant to the pattern of inherited cancer risk. Among these, the proband and an affected brother each had a heterozygous Ala45Thr variant in SDHA, a component of the succinate dehydrogenase (SDH) complex. SDH defects are associated with mitochondrial disorders and risk for various cancers; immunochemical analysis indicated loss of SDHB protein expression in the patient’s tumor, compatible with SDH deficiency. Integrated analysis of public databases and structural predictions indicated that the two affected individuals also had additional variants in genes including TGFB2, TRAP1, PARP1, and EGF, each potentially relevant to cancer risk alone or in conjunction with the SDHA variant. In addition, allelic imbalances of PARP1 and TGFB2 were detected in the tumor of the proband. Conclusion Together, these data suggest the possibility of risk associated with interaction of two or more variants.
Collapse
Affiliation(s)
- Emmanuelle Nicolas
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Elena V Demidova
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Kazan Federal University, Kazan, Russia
| | - Waleed Iqbal
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ilya G Serebriiskii
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Kazan Federal University, Kazan, Russia
| | | | - Pooja Ghatalia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yan Zhou
- Biostatistics and Bioinformatics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Kim Rainey
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Andrea F Forman
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Roland L Dunbrack
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Michael J Hall
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mary B Daly
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Sanjeevani Arora
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
665
|
Ling C, Sui R, Yao F, Wu Z, Zhang X, Zhang S. Whole exome sequencing identified a novel truncation mutation in the NHS gene associated with Nance-Horan syndrome. BMC MEDICAL GENETICS 2019; 20:14. [PMID: 30642278 PMCID: PMC6332535 DOI: 10.1186/s12881-018-0725-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 11/21/2018] [Indexed: 11/10/2022]
Abstract
Background Nance-Horan syndrome (NHS) is an X-linked inheritance disorder characterized by bilateral congenital cataracts, and facial and dental dysmorphism. This disorder is caused by mutations in the NHS gene. However, NHS may be difficult to detect in individuals with subtle facial dysmorphism and dental abnormalities in whom congenital cataracts are the primary clinical manifestations. Methods In this study, we present a three-generation family with NHS. Whole exome sequencing was performed to determine the potential pathogenic variant in the proband. Further validation was explored with Sanger sequencing in 9 of the available individuals of the family and additional 200 controls. Results A novel truncation mutation in gene NHS (c.C4449G, p.Tyr1483Ter) was found in the proband, who presented with a long-narrow face, prominent nose and large anteverted pinnae ear, screw-driver like incisors, mild mulberry like molars, one missing maxillary second molar and malocclusion. We found this mutation was detected in 2 male patients and 4 female carriers in the family. However, the mutation was never detected in the control subjects. Conclusions In conclusion, we identified a novel truncation mutation in the NHS gene, which might associate with NHS. Our review on the NHS studies illustrated that NHS has significantly clinical heterogeneity. And NHS mutations in the NHS-affected individuals typically result in premature truncation of the protein. And the new mutation revealed in this study would highlight the understanding of the causative mutations of NHS. Electronic supplementary material The online version of this article (10.1186/s12881-018-0725-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chao Ling
- Laboratory of Clinical Genetics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruifang Sui
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Fengxia Yao
- Laboratory of Clinical Genetics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhihong Wu
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xue Zhang
- Laboratory of Clinical Genetics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China. .,McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
666
|
Garibaldi M, Rendu J, Brocard J, Lacene E, Fauré J, Brochier G, Beuvin M, Labasse C, Madelaine A, Malfatti E, Bevilacqua JA, Lubieniecki F, Monges S, Taratuto AL, Laporte J, Marty I, Antonini G, Romero NB. 'Dusty core disease' (DuCD): expanding morphological spectrum of RYR1 recessive myopathies. Acta Neuropathol Commun 2019; 7:3. [PMID: 30611313 PMCID: PMC6320585 DOI: 10.1186/s40478-018-0655-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
Several morphological phenotypes have been associated to RYR1-recessive myopathies. We recharacterized the RYR1-recessive morphological spectrum by a large monocentric study performed on 54 muscle biopsies from a large cohort of 48 genetically confirmed patients, using histoenzymology, immunohistochemistry, and ultrastructural studies. We also analysed the level of RyR1 expression in patients’ muscle biopsies. We defined “dusty cores” the irregular areas of myofibrillar disorganisation characterised by a reddish-purple granular material deposition with uneven oxidative stain and devoid of ATPase activity, which represent the characteristic lesion in muscle biopsy in 54% of patients. We named Dusty Core Disease (DuCD) the corresponding entity of congenital myopathy. Dusty cores had peculiar histological and ultrastructural characteristics compared to the other core diseases. DuCD muscle biopsies also showed nuclear centralization and type1 fibre predominance. Dusty cores were not observed in other core myopathies and centronuclear myopathies. The other morphological groups in our cohort of patients were: Central Core (CCD: 21%), Core-Rod (C&R:15%) and Type1 predominance “plus” (T1P+:10%). DuCD group was associated to an earlier disease onset, a more severe clinical phenotype and a lowest level of RyR1 expression in muscle, compared to the other groups. Variants located in the bridge solenoid and the pore domains were more frequent in DuCD patients. In conclusion, DuCD is the most frequent histopathological presentation of RYR1-recessive myopathies. Dusty cores represent the unifying morphological lesion among the DuCD pathology spectrum and are the morphological hallmark for the recessive form of disease.
Collapse
|
667
|
Choi MH, Mejlænder-Andersen E, Manueldas S, El Jellas K, Steine SJ, Tjensvoll K, Sætran HA, Knappskog S, Hoem D, Nordgård O, Hovland R, Molven A. Mutation analysis by deep sequencing of pancreatic juice from patients with pancreatic ductal adenocarcinoma. BMC Cancer 2019; 19:11. [PMID: 30611220 PMCID: PMC6321709 DOI: 10.1186/s12885-018-5195-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
Background Reliable methods are needed to identify patients with early-stage cancer or high-grade precancerous lesions in the pancreas. Analysis of pancreatic juice to detect somatic mutations could represent one such approach. Here we investigated the concordance between mutations found in the primary tumor and pancreatic juice from the same patient. Methods Amplicon-based targeted deep sequencing was performed on samples from 21 patients with pancreatic ductal adenocarcinoma (PDAC) who had undergone Whipple’s operation. Mutation profiles were determined in formalin-fixed sections of the primary tumor and in pancreatic juice sampled from the main pancreatic duct during surgery. Results Using a cut-off of 3% for variant allele frequency, KRAS mutations were detected in 20/21 primary tumors (95%) and in 15/21 (71%) juice samples. When also considering low-frequency variants, KRAS mutations were found in 20/21 juice samples. Most juice samples exhibited multiple KRAS variants not seen in the primary tumor, and only in 11 cases (52%) did the most abundant variant of the juice correspond to the KRAS mutation detected in the tumor. TP53 mutations were found in 16 tumors (76%) and six juice samples (29%). Among the positive juice samples, only one exhibited more than a single TP53 mutation. Detection of both KRAS and TP53 mutations was fully concordant in the primary tumor and juice sample in 7/21 cases (33%). Conclusions Pancreatic juice from PDAC patients is rich in KRAS mutations often not seen in the primary tumor and possibly reflecting precancerous lesions in other regions of the pancreas. The inclusion of TP53 mutation detection and additional markers must therefore be considered for fully exploiting the clinical potential of pancreatic juice samples in early cancer detection. Electronic supplementary material The online version of this article (10.1186/s12885-018-5195-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Man Hung Choi
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Eline Mejlænder-Andersen
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Sophia Manueldas
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Khadija El Jellas
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Solrun J Steine
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kjersti Tjensvoll
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Hege Aase Sætran
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Dag Hoem
- Department of Gastrointestinal Surgery, Haukeland University Hospital, Bergen, Norway
| | - Oddmund Nordgård
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Randi Hovland
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway. .,Department of Pathology, Haukeland University Hospital, Bergen, Norway. .,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
668
|
Abstract
Genetic testing has an increasingly important role in the diagnosis and management of cardiac disorders, where it confirms the diagnosis, aids prognostication and risk stratification and guides treatment. A genetic diagnosis in the proband also enables clarification of the risk for family members by cascade testing. Genetics in cardiac disorders is complex where epigenetic and environmental factors might come into interplay. Incomplete penetrance and variable expressivity is also common. Genetic results in cardiac conditions are mostly probabilistic and should be interpreted with all available clinical information. With this complexity in cardiac genetics, testing is only indicated in patients with a strong suspicion of an inheritable cardiac disorder after a full clinical evaluation. In this review we discuss the genetics underlying the major cardiomyopathies and channelopathies, and the practical aspects of diagnosing these conditions in the laboratory.
Collapse
|
669
|
Kim J, Luo W, Wang M, Wegman-Ostrosky T, Frone MN, Johnston JJ, Nickerson ML, Rotunno M, Li SA, Achatz MI, Brodie SA, Dean M, de Andrade KC, Fortes FP, Gianferante M, Khincha P, McMaster ML, McReynolds LJ, Pemov A, Pinheiro M, Santiago KM, Alter BP, Caporaso NE, Gadalla SM, Goldin LR, Greene MH, Loud J, Yang XR, Freedman ND, Gapstur SM, Gaudet MM, Calista D, Ghiorzo P, Fargnoli MC, Nagore E, Peris K, Puig S, Landi MT, Hicks B, Zhu B, Liu J, Sampson JN, Chanock SJ, Mirabello LJ, Morton LM, Biesecker LG, Tucker MA, Savage SA, Goldstein AM, Stewart DR. Prevalence of pathogenic/likely pathogenic variants in the 24 cancer genes of the ACMG Secondary Findings v2.0 list in a large cancer cohort and ethnicity-matched controls. Genome Med 2018; 10:99. [PMID: 30583724 PMCID: PMC6305568 DOI: 10.1186/s13073-018-0607-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Prior research has established that the prevalence of pathogenic/likely pathogenic (P/LP) variants across all of the American College of Medical Genetics (ACMG) Secondary Findings (SF) genes is approximately 0.8-5%. We investigated the prevalence of P/LP variants in the 24 ACMG SF v2.0 cancer genes in a family-based cancer research cohort (n = 1173) and in cancer-free ethnicity-matched controls (n = 982). METHODS We used InterVar to classify variants and subsequently conducted a manual review to further examine variants of unknown significance (VUS). RESULTS In the 24 genes on the ACMG SF v2.0 list associated with a cancer phenotype, we observed 8 P/LP unique variants (8 individuals; 0.8%) in controls and 11 P/LP unique variants (14 individuals; 1.2%) in cases, a non-significant difference. We reviewed 115 VUS. The median estimated per-variant review time required was 30 min; the first variant within a gene took significantly (p = 0.0009) longer to review (median = 60 min) compared with subsequent variants (median = 30 min). The concordance rate was 83.3% for the variants examined by two reviewers. CONCLUSION The 115 VUS required database and literature review, a time- and labor-intensive process hampered by the difficulty in interpreting conflicting P/LP determinations. By rigorously investigating the 24 ACMG SF v2.0 cancer genes, our work establishes a benchmark P/LP variant prevalence rate in a familial cancer cohort and controls.
Collapse
Affiliation(s)
- Jung Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Wen Luo
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA
| | - Mingyi Wang
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA
| | - Talia Wegman-Ostrosky
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
- División de Investigación, Instituto Nacional de Cancerología, 14080, Mexico City, Mexico
| | - Megan N Frone
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Jennifer J Johnston
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, 20892, USA
| | - Michael L Nickerson
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Gaithersburg, MD, 20877, USA
| | - Melissa Rotunno
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Shengchao A Li
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA
| | - Maria I Achatz
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
- Centro de Oncologia, Hospital Sirio-Libanes, Sao Paulo, SP, 013050-050, Brazil
| | - Seth A Brodie
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA
| | - Michael Dean
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Gaithersburg, MD, 20877, USA
| | - Kelvin C de Andrade
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
- International Research Center, A.C. Camargo Cancer Center, São Paulo, 01508-010, Brazil
| | - Fernanda P Fortes
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
- International Research Center, A.C. Camargo Cancer Center, São Paulo, 01508-010, Brazil
| | - Matthew Gianferante
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Payal Khincha
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Mary L McMaster
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Lisa J McReynolds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Alexander Pemov
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Maisa Pinheiro
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Karina M Santiago
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
- International Research Center, A.C. Camargo Cancer Center, São Paulo, 01508-010, Brazil
| | - Blanche P Alter
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Neil E Caporaso
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Shahinaz M Gadalla
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Lynn R Goldin
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Mark H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Jennifer Loud
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Xiaohong R Yang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Neal D Freedman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Susan M Gapstur
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, GA, USA
| | - Mia M Gaudet
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, GA, USA
| | - Donato Calista
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa and Genetics of Rare Cancers, IRCCS Ospedale Policinico San Martino, Genoa, Italy
| | | | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncologia, Valencia, Spain
| | - Ketty Peris
- Institute of Dermatology, Catholic University - Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Susana Puig
- Dermatology Department, Melanoma Unit, Hospital Clinic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain & Centro de Investigacion Biomedica en Red en Enfermedades Raras (CIBERER), Valencia, Spain
| | - Maria Teresa Landi
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Belynda Hicks
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA
| | - Bin Zhu
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA
| | - Jia Liu
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA
| | - Joshua N Sampson
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Stephen J Chanock
- Office of the Director, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Lisa J Mirabello
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Lindsay M Morton
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Leslie G Biesecker
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, 20892, USA
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, Human Genetics Program National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Alisa M Goldstein
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA.
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, 20850, USA.
| |
Collapse
|
670
|
Suzuki H, Kurosawa K, Fukuda K, Ijima K, Sumazaki R, Saito S, Kosaki R, Hirasawa A, Okazaki Y, Imai K, Matsunaga T, Iwata T, Kosaki K. Japanese pathogenic variant database: DPV. ACTA ACUST UNITED AC 2018. [DOI: 10.3233/trd-180027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hisato Suzuki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Kurosawa
- Division of Medical Genetics, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Kazumoto Ijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryo Sumazaki
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Shinji Saito
- Department of Pediatrics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Rika Kosaki
- Division of Medical Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Akira Hirasawa
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yasushi Okazaki
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kohsuke Imai
- Department of Pediatrics Perinatal and Maternal Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsuo Matsunaga
- National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Takeshi Iwata
- National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
671
|
Tsipi M, Poulou M, Fylaktou I, Kosma K, Tsoutsou E, Pons MR, Kokkinou E, Kitsiou-Tzeli S, Fryssira H, Tzetis M. Phenotypic expression of a spectrum of Neurofibromatosis Type 1 (NF1) mutations identified through NGS and MLPA. J Neurol Sci 2018; 395:95-105. [PMID: 30308447 DOI: 10.1016/j.jns.2018.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 09/06/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Neurofibromatosis Type 1 (NF1) is caused by mutations of the NF1 gene. The aim of this study was to identify the genetic causes underlying the disease, attempt possible phenotype/genotype correlations and add to the NF1 mutation spectrum. A screening protocol based on genomic DNA was established in 168 patients, encompassing sequencing of all coding exons and adjoining introns using a custom targeted next generation sequencing protocol and subsequent confirmation of findings with Sanger sequencing. MLPA was used to detect deletions/duplications and positive findings were confirmed by RNA analysis. All novel findings were evaluated according to ACMG Standards and guidelines for the interpretation of sequence variants with the aid of in-silico bioinformatic tools and family segregation analysis. A germline variant was identified in 145 patients (86%). In total 49 known and 70 novel variants in coding and non-coding regions were identified. Seven patients carried whole or partial gene deletions. NF1 patients, present with high phenotypic variability even in cases where the same germline disease causing variant has been identified. Our findings will contribute to a better knowledge of the genetic causes and the phenotypic expression related to the disease.
Collapse
Affiliation(s)
- Maria Tsipi
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Myrto Poulou
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| | - Irene Fylaktou
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| | - Konstantina Kosma
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eirini Tsoutsou
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| | - Maria-Roser Pons
- 1(st) Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Childrens Hospital, Athens, Greece
| | - Eleftheria Kokkinou
- 1(st) Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Childrens Hospital, Athens, Greece
| | - Sofia Kitsiou-Tzeli
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece; Research Institute for the Study of Genetic and Malignant Diseases in Childhood, "Aghia Sophia" Childrens Hospital, Athens, Greece
| | - Helen Fryssira
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| | - Maria Tzetis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
672
|
Lippert J, Appenzeller S, Liang R, Sbiera S, Kircher S, Altieri B, Nanda I, Weigand I, Gehrig A, Steinhauer S, Riemens RJM, Rosenwald A, Müller CR, Kroiss M, Rost S, Fassnacht M, Ronchi CL. Targeted Molecular Analysis in Adrenocortical Carcinomas: A Strategy Toward Improved Personalized Prognostication. J Clin Endocrinol Metab 2018; 103:4511-4523. [PMID: 30113656 DOI: 10.1210/jc.2018-01348] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/30/2018] [Indexed: 12/20/2022]
Abstract
CONTEXT Adrenocortical carcinoma (ACC) has a heterogeneous prognosis, and current medical therapies have limited efficacy in its advanced stages. Genome-wide multiomics studies identified molecular patterns associated with clinical outcome. OBJECTIVE Here, we aimed at identifying a molecular signature useful for both personalized prognostic stratification and druggable targets, using methods applicable in clinical routine. DESIGN In total, 117 tumor samples from 107 patients with ACC were analyzed. Targeted next-generation sequencing of 160 genes and pyrosequencing of 4 genes were applied to formalin-fixed, paraffin-embedded (FFPE) specimens to detect point mutations, copy number alterations, and promoter region methylation. Molecular results were combined with clinical/histopathological parameters (tumor stage, age, symptoms, resection status, and Ki-67) to predict progression-free survival (PFS). RESULTS In addition to known driver mutations, we detected recurrent alterations in genes not previously associated with ACC (e.g., NOTCH1, CIC, KDM6A, BRCA1, BRCA2). Best prediction of PFS was obtained integrating molecular results (more than one somatic mutation, alterations in Wnt/β-catenin and p53 pathways, high methylation pattern) and clinical/histopathological parameters into a combined score (P < 0.0001, χ2 = 68.6). Accuracy of prediction for early disease progress was 83.3% (area under the receiver operating characteristic curve: 0.872, 95% confidence interval 0.80 to 0.94). Furthermore, 17 potentially targetable alterations were found in 64 patients (e.g., in CDK4, NOTCH1, NF1, MDM2, and EGFR and in DNA repair system). CONCLUSIONS This study demonstrates that molecular profiling of FFPE tumor samples improves prognostication of ACC beyond clinical/histopathological parameters and identifies new potential drug targets. These findings pave the way to precision medicine in this rare disease.
Collapse
Affiliation(s)
- Juliane Lippert
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Silke Appenzeller
- Core Unit Bioinformatics, Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany
| | - Raimunde Liang
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Silviu Sbiera
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Stefan Kircher
- Institute for Pathology, University of Würzburg, Würzburg, Germany
| | - Barbara Altieri
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
- Division of Endocrinology and Metabolic Diseases, Catholic University of the Sacred Heart, Rome, Italy
| | - Indrajit Nanda
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Isabel Weigand
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Andrea Gehrig
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Sonja Steinhauer
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Renzo J M Riemens
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, LK Maastricht, Netherlands
| | - Andreas Rosenwald
- Institute for Pathology, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Clemens R Müller
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Matthias Kroiss
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Simone Rost
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Martin Fassnacht
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
- Central Labor, University Hospital of Würzburg, Würzburg, Germany
| | - Cristina L Ronchi
- Department of Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, England
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, England
| |
Collapse
|
673
|
Malkowska M, Zubek J, Plewczynski D, Wyrwicz LS. ShapeGTB: the role of local DNA shape in prioritization of functional variants in human promoters with machine learning. PeerJ 2018; 6:e5742. [PMID: 30519505 PMCID: PMC6275119 DOI: 10.7717/peerj.5742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 09/13/2018] [Indexed: 02/01/2023] Open
Abstract
Motivation The identification of functional sequence variations in regulatory DNA regions is one of the major challenges of modern genetics. Here, we report results of a combined multifactor analysis of properties characterizing functional sequence variants located in promoter regions of genes. Results We demonstrate that GC-content of the local sequence fragments and local DNA shape features play significant role in prioritization of functional variants and outscore features related to histone modifications, transcription factors binding sites, or evolutionary conservation descriptors. Those observations allowed us to build specialized machine learning classifier identifying functional single nucleotide polymorphisms within promoter regions—ShapeGTB. We compared our method with more general tools predicting pathogenicity of all non-coding variants. ShapeGTB outperformed them by a wide margin (average precision 0.93 vs. 0.47–0.55). On the external validation set based on ClinVar database it displayed worse performance but was still competitive with other methods (average precision 0.47 vs. 0.23–0.42). Such results suggest unique characteristics of mutations located within promoter regions and are a promising signal for the development of more accurate variant prioritization tools in the future.
Collapse
Affiliation(s)
- Maja Malkowska
- Laboratory of Bioinformatics and Biostatistics, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Julian Zubek
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Dariusz Plewczynski
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland.,Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Lucjan S Wyrwicz
- Laboratory of Bioinformatics and Biostatistics, Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| |
Collapse
|
674
|
Kullo IJ, Olson J, Fan X, Jose M, Safarova M, Radecki Breitkopf C, Winkler E, Kochan DC, Snipes S, Pacyna JE, Carney M, Chute CG, Gupta J, Jose S, Venner E, Murugan M, Jiang Y, Zordok M, Farwati M, Philogene M, Smith E, Shaibi GQ, Caraballo P, Freimuth R, Lindor NM, Sharp R, Thibodeau SN. The Return of Actionable Variants Empirical (RAVE) Study, a Mayo Clinic Genomic Medicine Implementation Study: Design and Initial Results. Mayo Clin Proc 2018; 93:1600-1610. [PMID: 30392543 PMCID: PMC6652203 DOI: 10.1016/j.mayocp.2018.06.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/04/2018] [Accepted: 06/08/2018] [Indexed: 12/02/2022]
Abstract
OBJECTIVES To identify clinically actionable genetic variants from targeted sequencing of 68 disease-related genes, estimate their penetrance, and assess the impact of disclosing results to participants and providers. PATIENTS AND METHODS The Return of Actionable Variants Empirical (RAVE) Study investigates outcomes following the return of pathogenic/likely pathogenic (P/LP) variants in 68 disease-related genes. The study was initiated in December 2016 and is ongoing. Targeted sequencing was performed in 2533 individuals with hyperlipidemia or colon polyps. The electronic health records (EHRs) of participants carrying P/LP variants in 36 cardiovascular disease (CVD) genes were manually reviewed to ascertain the presence of relevant traits. Clinical outcomes, health care utilization, family communication, and ethical and psychosocial implications of disclosure of genomic results are being assessed by surveys, telephone interviews, and EHR review. RESULTS Of 29,208 variants in the 68 genes, 1915 were rare (frequency <1%) and putatively functional, and 102 of these (60 in 36 CVD genes) were labeled P/LP based on the American College of Medical Genetics and Genomics framework. Manual review of the EHRs of participants (n=73 with P/LP variants in CVD genes) revealed that 33 had the expected trait(s); however, only 6 of 45 participants with non-familial hypercholesterolemia (FH) P/LP variants had the expected traits. CONCLUSION Expected traits were present in 13% of participants with P/LP variants in non-FH CVD genes, suggesting low penetrance; this estimate may change with additional testing performed as part of the clinical evaluation. Ongoing analyses of the RAVE Study will inform best practices for genomic medicine.
Collapse
Affiliation(s)
- Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN.
| | - Janet Olson
- Department of Health Sciences Research, Biomedical Ethics Program, Mayo Clinic, Rochester, MN
| | - Xiao Fan
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Merin Jose
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Maya Safarova
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Erin Winkler
- Center for Individualized Medicine-Genomics, Mayo Clinic, Rochester, MN
| | - David C Kochan
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Sara Snipes
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Joel E Pacyna
- Department of Health Sciences Research, Biomedical Ethics Program, Mayo Clinic, Rochester, MN
| | - Meaghan Carney
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Christopher G Chute
- Johns Hopkins University, Schools of Medicine, Public Health and Nursing, Baltimore, MD
| | - Jyoti Gupta
- National Human Genome Research Institute, Bethesda, MD
| | - Sheethal Jose
- National Human Genome Research Institute, Bethesda, MD
| | - Eric Venner
- Baylor College of Medicine Human Genome Sequencing Center, Houston, TX
| | - Mullai Murugan
- Baylor College of Medicine Human Genome Sequencing Center, Houston, TX
| | - Yunyun Jiang
- Baylor College of Medicine Human Genome Sequencing Center, Houston, TX
| | - Magdi Zordok
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Medhat Farwati
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Erica Smith
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Gabriel Q Shaibi
- Center for Health Promotion and Disease Prevention, Arizona State University, Phoenix, AZ
| | | | - Robert Freimuth
- Department of Health Sciences Research, Biomedical Ethics Program, Mayo Clinic, Rochester, MN
| | | | - Richard Sharp
- Department of Health Sciences Research, Biomedical Ethics Program, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
675
|
Nicora G, Limongelli I, Gambelli P, Memmi M, Malovini A, Mazzanti A, Napolitano C, Priori S, Bellazzi R. CardioVAI: An automatic implementation of ACMG-AMP variant interpretation guidelines in the diagnosis of cardiovascular diseases. Hum Mutat 2018; 39:1835-1846. [PMID: 30298955 DOI: 10.1002/humu.23665] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/24/2018] [Accepted: 10/04/2018] [Indexed: 11/09/2022]
Abstract
Variant interpretation for the diagnosis of genetic diseases is a complex process. The American College of Medical Genetics and Genomics, with the Association for Molecular Pathology, have proposed a set of evidence-based guidelines to support variant pathogenicity assessment and reporting in Mendelian diseases. Cardiovascular disorders are a field of application of these guidelines, but practical implementation is challenging due to the genetic disease heterogeneity and the complexity of information sources that need to be integrated. Decision support systems able to automate variant interpretation in the light of specific disease domains are demanded. We implemented CardioVAI (Cardio Variant Interpreter), an automated system for guidelines based variant classification in cardiovascular-related genes. Different omics-resources were integrated to assess pathogenicity of every genomic variant in 72 cardiovascular diseases related genes. We validated our method on benchmark datasets of high-confident assessed variants, reaching pathogenicity and benignity concordance up to 83 and 97.08%, respectively. We compared CardioVAI to similar methods and analyzed the main differences in terms of guidelines implementation. We finally made available CardioVAI as a web resource (http://cardiovai.engenome.com/) that allows users to further specialize guidelines recommendations.
Collapse
Affiliation(s)
- Giovanna Nicora
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | | | - Patrick Gambelli
- Laboratory of Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Mirella Memmi
- Laboratory of Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Alberto Malovini
- Laboratory of Informatics and Systems Engineering for Clinical Research, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Andrea Mazzanti
- Laboratory of Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Carlo Napolitano
- Laboratory of Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Silvia Priori
- Laboratory of Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Riccardo Bellazzi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy.,Laboratory of Informatics and Systems Engineering for Clinical Research, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| |
Collapse
|
676
|
Abolhassani H, Kiaee F, Tavakol M, Chavoshzadeh Z, Mahdaviani SA, Momen T, Yazdani R, Azizi G, Habibi S, Gharagozlou M, Movahedi M, Hamidieh AA, Behniafard N, Nabavi M, Bemanian MH, Arshi S, Molatefi R, Sherkat R, Shirkani A, Amin R, Aleyasin S, Faridhosseini R, Jabbari-Azad F, Mohammadzadeh I, Ghaffari J, Shafiei A, Kalantari A, Mansouri M, Mesdaghi M, Babaie D, Ahanchian H, Khoshkhui M, Soheili H, Eslamian MH, Cheraghi T, Dabbaghzadeh A, Tavassoli M, Kalmarzi RN, Mortazavi SH, Kashef S, Esmaeilzadeh H, Tafaroji J, Khalili A, Zandieh F, Sadeghi-Shabestari M, Darougar S, Behmanesh F, Akbari H, Zandkarimi M, Abolnezhadian F, Fayezi A, Moghtaderi M, Ahmadiafshar A, Shakerian B, Sajedi V, Taghvaei B, Safari M, Heidarzadeh M, Ghalebaghi B, Fathi SM, Darabi B, Bazregari S, Bazargan N, Fallahpour M, Khayatzadeh A, Javahertrash N, Bashardoust B, Zamani M, Mohsenzadeh A, Ebrahimi S, Sharafian S, Vosughimotlagh A, Tafakoridelbari M, Rahimi M, Ashournia P, Razaghian A, Rezaei A, Mamishi S, Parvaneh N, Rezaei N, Hammarström L, Aghamohammadi A. Fourth Update on the Iranian National Registry of Primary Immunodeficiencies: Integration of Molecular Diagnosis. J Clin Immunol 2018; 38:816-832. [PMID: 30302726 DOI: 10.1007/s10875-018-0556-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/26/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND The number of inherited diseases and the spectrum of clinical manifestations of primary immunodeficiency disorders (PIDs) are ever-expanding. Molecular diagnosis using genomic approaches should be performed for all PID patients since it provides a resource to improve the management and to estimate the prognosis of patients with these rare immune disorders. METHOD The current update of Iranian PID registry (IPIDR) contains the clinical phenotype of newly registered patients during last 5 years (2013-2018) and the result of molecular diagnosis in patients enrolled for targeted and next-generation sequencing. RESULTS Considering the newly diagnosed patients (n = 1395), the total number of registered PID patients reached 3056 (1852 male and 1204 female) from 31 medical centers. The predominantly antibody deficiency was the most common subcategory of PID (29.5%). The putative causative genetic defect was identified in 1014 patients (33.1%) and an autosomal recessive pattern was found in 79.3% of these patients. Among the genetically different categories of PID patients, the diagnostic rate was highest in defects in immune dysregulation and lowest in predominantly antibody deficiencies and mutations in the MEFV gene were the most frequent genetic disorder in our cohort. CONCLUSIONS During a 20-year registration of Iranian PID patients, significant changes have been observed by increasing the awareness of the medical community, national PID network establishment, improving therapeutic facilities, and recently by inclusion of the molecular diagnosis. The current collective study of PID phenotypes and genotypes provides a major source for ethnic surveillance, newborn screening, and genetic consultation for prenatal and preimplantation genetic diagnosis.
Collapse
Affiliation(s)
- Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran
| | - Fatemeh Kiaee
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran
| | - Marzieh Tavakol
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Zahra Chavoshzadeh
- Pediatric Infections Research Center, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Alireza Mahdaviani
- Pediatric Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tooba Momen
- Department of Allergy and Clinical Immunology, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran
| | - Gholamreza Azizi
- Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Sima Habibi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran
| | - Mohammad Gharagozlou
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Masoud Movahedi
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Hematology, Oncology and Stem Cell Transplantation Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Behniafard
- Department of Allergy and Clinical Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammamd Nabavi
- Department of Allergy and Clinical Immunology, Rasool e Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Bemanian
- Department of Allergy and Clinical Immunology, Rasool e Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Saba Arshi
- Department of Allergy and Clinical Immunology, Rasool e Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Rasol Molatefi
- Department of Pediatrics, Bo-Ali children's Hospital of Ardabil University of Medical Sciences, Ardabil, Iran
| | - Roya Sherkat
- Acquired Immunodeficiency Research Center, lsfahan University of Medical Sciences, Isfahan, Iran
| | - Afshin Shirkani
- Allergy and Clinical Immunology Department, Bushehr University of Medical Science, School of Medicine, Bushehr, Iran
| | - Reza Amin
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Aleyasin
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Faridhosseini
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Iraj Mohammadzadeh
- Noncommunicable Pediatric Diseases Research Center, Amirkola Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Javad Ghaffari
- Department of Pediatrics, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Shafiei
- Department of Immunology, Bahrami Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Kalantari
- Department of Immunology and Allergy, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Mansouri
- Immunology and Allergy Department, Mofid Children's Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mehrnaz Mesdaghi
- Immunology and Allergy Department, Mofid Children's Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Delara Babaie
- Pediatric Infections Research Center, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ahanchian
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Khoshkhui
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Habib Soheili
- Department of Pediatrics, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | | | - Taher Cheraghi
- Department of Pediatrics, 17th Shahrivar Children's Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Abbas Dabbaghzadeh
- Noncommunicable Pediatric Diseases Research Center, Amirkola Hospital, Babol University of Medical Sciences, Babol, Iran.,Department of Allergy and Clinical Immunology, Pediatric Infectious Diseases Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahmoud Tavassoli
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Nasiri Kalmarzi
- Cellular & Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - Sara Kashef
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Javad Tafaroji
- Department of Pediatrics, Qom University of Medical Sciences, Qom, Iran
| | - Abbas Khalili
- Department of Pediatrics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fariborz Zandieh
- Department of Immunology, Bahrami Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sepideh Darougar
- Pediatric Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Behmanesh
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hedayat Akbari
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Farhad Abolnezhadian
- Department of Immunology and Allergy, Ahvaz University of Medical Sciences, Ahvaz, Iran
| | - Abbas Fayezi
- Department of Immunology and Allergy, Ahvaz University of Medical Sciences, Ahvaz, Iran
| | - Mojgan Moghtaderi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Behzad Shakerian
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Sajedi
- Department of Immunology and Allergy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Behrang Taghvaei
- Department of Immunology and Allergy, Semnan University of Medical Sciences, Semnan, Iran
| | - Mojgan Safari
- Department of Pediatrics, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Heidarzadeh
- Department of Immunology and Allergy, Kashan University of Medical Sciences, Kashan, Iran
| | - Babak Ghalebaghi
- Department of Pediatrics, 17th Shahrivar Children's Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Mohammad Fathi
- Department of Immunology and Allergy, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Behzad Darabi
- Department of Immunology and Allergy, Ilam University of Medical Sciences, Ilam, Iran
| | - Saeed Bazregari
- Allergy and Clinical Immunology Department, Bushehr University of Medical Science, School of Medicine, Bushehr, Iran
| | - Nasrin Bazargan
- Department of Immunology and Allergy, Kerman University of Medical Sciences, Kerman, Iran
| | - Morteza Fallahpour
- Department of Allergy and Clinical Immunology, Rasool e Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Khayatzadeh
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Naser Javahertrash
- Department of Allergy and Clinical Immunology, Rasool e Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Bahram Bashardoust
- Pediatric Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadali Zamani
- Department of Immunology and Allergy, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Azam Mohsenzadeh
- Department of Pediatrics, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Sarehsadat Ebrahimi
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Samin Sharafian
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Ahmad Vosughimotlagh
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Mitra Tafakoridelbari
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Maziar Rahimi
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Parisa Ashournia
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Anahita Razaghian
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Pediatrics Center of Excellence, Children's Medical Center, Tehran, University of Medical Sciences, Tehran, Iran
| | - Arezou Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran
| | - Setareh Mamishi
- Pediatric Infectious Diseases Research Center, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Nima Parvaneh
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran
| | - Lennart Hammarström
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran. .,Iranian Primary Immunodeficiencies Network (IPIN), Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
677
|
Azaiez H, Booth KT, Ephraim SS, Crone B, Black-Ziegelbein EA, Marini RJ, Shearer AE, Sloan-Heggen CM, Kolbe D, Casavant T, Schnieders MJ, Nishimura C, Braun T, Smith RJ. Genomic Landscape and Mutational Signatures of Deafness-Associated Genes. Am J Hum Genet 2018; 103:484-497. [PMID: 30245029 PMCID: PMC6174355 DOI: 10.1016/j.ajhg.2018.08.006] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022] Open
Abstract
The classification of genetic variants represents a major challenge in the post-genome era by virtue of their extraordinary number and the complexities associated with ascribing a clinical impact, especially for disorders exhibiting exceptional phenotypic, genetic, and allelic heterogeneity. To address this challenge for hearing loss, we have developed the Deafness Variation Database (DVD), a comprehensive, open-access resource that integrates all available genetic, genomic, and clinical data together with expert curation to generate a single classification for each variant in 152 genes implicated in syndromic and non-syndromic deafness. We evaluate 876,139 variants and classify them as pathogenic or likely pathogenic (more than 8,100 variants), benign or likely benign (more than 172,000 variants), or of uncertain significance (more than 695,000 variants); 1,270 variants are re-categorized based on expert curation and in 300 instances, the change is of medical significance and impacts clinical care. We show that more than 96% of coding variants are rare and novel and that pathogenicity is driven by minor allele frequency thresholds, variant effect, and protein domain. The mutational landscape we define shows complex gene-specific variability, making an understanding of these nuances foundational for improved accuracy in variant interpretation in order to enhance clinical decision making and improve our understanding of deafness biology.
Collapse
|
678
|
Mutations in KCNK4 that Affect Gating Cause a Recognizable Neurodevelopmental Syndrome. Am J Hum Genet 2018; 103:621-630. [PMID: 30290154 DOI: 10.1016/j.ajhg.2018.09.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/31/2018] [Indexed: 12/15/2022] Open
Abstract
Aberrant activation or inhibition of potassium (K+) currents across the plasma membrane of cells has been causally linked to altered neurotransmission, cardiac arrhythmias, endocrine dysfunction, and (more rarely) perturbed developmental processes. The K+ channel subfamily K member 4 (KCNK4), also known as TRAAK (TWIK-related arachidonic acid-stimulated K+ channel), belongs to the mechano-gated ion channels of the TRAAK/TREK subfamily of two-pore-domain (K2P) K+ channels. While K2P channels are well known to contribute to the resting membrane potential and cellular excitability, their involvement in pathophysiological processes remains largely uncharacterized. We report that de novo missense mutations in KCNK4 cause a recognizable syndrome with a distinctive facial gestalt, for which we propose the acronym FHEIG (facial dysmorphism, hypertrichosis, epilepsy, intellectual disability/developmental delay, and gingival overgrowth). Patch-clamp analyses documented a significant gain of function of the identified KCNK4 channel mutants basally and impaired sensitivity to mechanical stimulation and arachidonic acid. Co-expression experiments indicated a dominant behavior of the disease-causing mutations. Molecular dynamics simulations consistently indicated that mutations favor sealing of the lateral intramembrane fenestration that has been proposed to negatively control K+ flow by allowing lipid access to the central cavity of the channel. Overall, our findings illustrate the pleiotropic effect of dysregulated KCNK4 function and provide support to the hypothesis of a gating mechanism based on the lateral fenestrations of K2P channels.
Collapse
|
679
|
Elrharchi S, Riahi Z, Salime S, Nahili H, Rouba H, Kabine M, Bonnet C, Petit C, Barakat A. Two novel homozygous missense mutations identified in the BSND gene in Moroccan patients with Bartter's syndrome. Int J Pediatr Otorhinolaryngol 2018; 113:46-50. [PMID: 30174009 DOI: 10.1016/j.ijporl.2018.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/05/2018] [Accepted: 07/07/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Hearing loss (HL) is one of the most common sensorineural disorders. In the present study, we identified two novel missense mutations in BSND gene causing Bartter syndrome type IV which is a genetic disease with an autosomal recessive transmission, characterized by hypokalaemia, metabolic alkalosis, an elevation in plasma renin activity and hyperaldosteronism as well as sensorineural deafness. METHODS Whole-exome sequencing was performed to study the genetic causes of Hearing loss in two unrelated patients from two Moroccan families. RESULTS The two novel homozygous mutations p.Arg8Gly (c.22C > G), p.Thr36Asn (c.107C > A) in exon 1 of BSND gene which encodes barttin were identified in 7 patients belonging to two unrelated families originated from central region of Morocco. CONCLUSION We identified two novel missense mutations p.Arg8Gly and p.Thr36Asn in exon 1 of BSND gene; both mutations were described for the first time in Moroccan patients with Bartter syndrome type IV.
Collapse
Affiliation(s)
- Soukaina Elrharchi
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco; Laboratoire de santé et environnement, Faculté des Sciences Ain Chock, Université Hassan II, Casablanca, Morocco
| | - Zied Riahi
- INSERM UMRS1120, Institut de la Vision, Paris, France; UPMC-Sorbonnes Universités Paris VI, Paris, France
| | - Sara Salime
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Halima Nahili
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hassan Rouba
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Mostafa Kabine
- Laboratoire de santé et environnement, Faculté des Sciences Ain Chock, Université Hassan II, Casablanca, Morocco
| | - Crystel Bonnet
- INSERM UMRS1120, Institut de la Vision, Paris, France; UPMC-Sorbonnes Universités Paris VI, Paris, France
| | - Christine Petit
- INSERM UMRS1120, Institut de la Vision, Paris, France; UPMC-Sorbonnes Universités Paris VI, Paris, France; Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France; Collège de France, Paris, France
| | - Abdelhamid Barakat
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco.
| |
Collapse
|
680
|
Miao H, Zhou J, Yang Q, Liang F, Wang D, Ma N, Gao B, Du J, Lin G, Wang K, Zhang Q. Long-read sequencing identified a causal structural variant in an exome-negative case and enabled preimplantation genetic diagnosis. Hereditas 2018; 155:32. [PMID: 30279644 PMCID: PMC6162922 DOI: 10.1186/s41065-018-0069-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND For a proportion of individuals judged clinically to have a recessive Mendelian disease, only one heterozygous pathogenic variant can be found from clinical whole exome sequencing (WES), posing a challenge to genetic diagnosis and genetic counseling. One possible reason is the limited ability to detect disease causal structural variants (SVs) from short reads sequencing technologies. Long reads sequencing can produce longer reads (typically 1000 bp or longer), therefore offering greatly improved ability to detect SVs that may be missed by short-read sequencing. RESULTS Here we describe a case study, where WES identified only one heterozygous pathogenic variant for an individual suspected to have glycogen storage disease type Ia (GSD-Ia), which is an autosomal recessive disease caused by bi-allelic mutations in the G6PC gene. Through Nanopore long-read whole-genome sequencing, we identified a 7.1 kb deletion covering two exons on the other allele, suggesting that complex structural variants (SVs) may explain a fraction of cases when the second pathogenic allele is missing from WES on recessive diseases. Both breakpoints of the deletion are within Alu elements, and we designed Sanger sequencing and quantitative PCR assays based on the breakpoints for preimplantation genetic diagnosis (PGD) for the family planning on another child. Four embryos were obtained after in vitro fertilization (IVF), and an embryo without deletion in G6PC was transplanted after PGD and was confirmed by prenatal diagnosis, postnatal diagnosis, and subsequent lack of disease symptoms after birth. CONCLUSIONS In summary, we present one of the first examples of using long-read sequencing to identify causal yet complex SVs in exome-negative patients, which subsequently enabled successful personalized PGD.
Collapse
Affiliation(s)
- Hefan Miao
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410078 Hunan China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078 Hunan China
| | | | - Qi Yang
- GrandOmics Biosciences, Beijing, 102206 China
| | - Fan Liang
- GrandOmics Biosciences, Beijing, 102206 China
| | - Depeng Wang
- GrandOmics Biosciences, Beijing, 102206 China
| | - Na Ma
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410078 Hunan China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078 Hunan China
| | - Bodi Gao
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410078 Hunan China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078 Hunan China
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410078 Hunan China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078 Hunan China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410078 Hunan China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078 Hunan China
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Qianjun Zhang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410078 Hunan China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078 Hunan China
| |
Collapse
|
681
|
Quezada Urban R, Díaz Velásquez CE, Gitler R, Rojo Castillo MP, Sirota Toporek M, Figueroa Morales A, Moreno García O, García Esquivel L, Torres Mejía G, Dean M, Delgado Enciso I, Ochoa Díaz López H, Rodríguez León F, Jan V, Garzón Barrientos VH, Ruiz Flores P, Espino Silva PK, Haro Santa Cruz J, Martínez Gregorio H, Rojas Jiménez EA, Romero Cruz LE, Méndez Catalá CF, Álvarez Gómez RM, Fragoso Ontiveros V, Herrera LA, Romieu I, Terrazas LI, Chirino YI, Frecha C, Oliver J, Perdomo S, Vaca Paniagua F. Comprehensive Analysis of Germline Variants in Mexican Patients with Hereditary Breast and Ovarian Cancer Susceptibility. Cancers (Basel) 2018; 10:E361. [PMID: 30262796 PMCID: PMC6211045 DOI: 10.3390/cancers10100361] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/05/2018] [Accepted: 09/15/2018] [Indexed: 12/11/2022] Open
Abstract
Hereditary breast and ovarian cancer syndrome (HBOC) represents 5⁻10% of all patients with breast cancer and is associated with high-risk pathogenic alleles in BRCA1/2 genes, but only for 25% of cases. We aimed to find new pathogenic alleles in a panel of 143 cancer-predisposing genes in 300 Mexican cancer patients with suspicion of HBOC and 27 high-risk patients with a severe family history of cancer, using massive parallel sequencing. We found pathogenic variants in 23 genes, including BRCA1/2. In the group of cancer patients 15% (46/300) had a pathogenic variant; 11% (33/300) harbored variants with unknown clinical significance (VUS) and 74% (221/300) were negative. The high-risk group had 22% (6/27) of patients with pathogenic variants, 4% (1/27) had VUS and 74% (20/27) were negative. The most recurrent mutations were the Mexican founder deletion of exons 9-12 and the variant p.G228fs in BRCA1, each found in 5 of 17 patients with alterations in this gene. Rare VUS with potential impact at the protein level were found in 21 genes. Our results show for the first time in the Mexican population a higher contribution of pathogenic alleles in other susceptibility cancer genes (54%) than in BRCA1/2 (46%), highlighting the high locus heterogeneity of HBOC and the necessity of expanding genetic tests for this disease to include broader gene panels.
Collapse
Affiliation(s)
- Rosalía Quezada Urban
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
| | - Clara Estela Díaz Velásquez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
| | | | | | | | | | | | | | | | - Michael Dean
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | - Héctor Ochoa Díaz López
- Department of Health, El Colegio de la Frontera Sur (ECOSUR), San Cristóbal de Las Casas 29290, Chiapas, Mexico.
| | - Fernando Rodríguez León
- Department of Health, El Colegio de la Frontera Sur (ECOSUR), San Cristóbal de Las Casas 29290, Chiapas, Mexico.
| | - Virginia Jan
- Internal Medicine, Hospital de Especialidades Vida Mejor, ISSTECH, Tuxtla Gutiérrez 29040, Chiapas, Mexico.
| | | | - Pablo Ruiz Flores
- Centro de Investigación Biomédica, Universidad Autónoma de Coahuila, Torreón 27000, Coahuila, Mexico.
| | - Perla Karina Espino Silva
- Centro de Investigación Biomédica, Universidad Autónoma de Coahuila, Torreón 27000, Coahuila, Mexico.
| | - Jorge Haro Santa Cruz
- Centro de Investigación Biomédica, Universidad Autónoma de Coahuila, Torreón 27000, Coahuila, Mexico.
| | - Héctor Martínez Gregorio
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
| | - Ernesto Arturo Rojas Jiménez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
| | - Luis Enrique Romero Cruz
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
| | - Claudia Fabiola Méndez Catalá
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
| | | | | | - Luis Alonso Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas-Instituto Nacional de Cancerología, CDMX 14080, Mexico.
| | - Isabelle Romieu
- Center for Center for Research on Population Health, National Institute of Public Health, Cuernavaca 62100, Morelos, Mexico.
- Hubert Department of Global Health, Emory University, Atlanta, GA 30322, USA.
| | - Luis Ignacio Terrazas
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, 54090 Tlalnepantla, Estado de México, Mexico.
| | - Yolanda Irasema Chirino
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, 54090 Tlalnepantla, Estado de México, Mexico.
| | | | - Javier Oliver
- Hospital Italiano, Buenos Aires C1199ABB, Argentina.
| | - Sandra Perdomo
- Investigación en Nutrición, Genética y Metabolismo, Facultad de Medicina, Universidad El Bosque, Bogotá 110121, Colombia.
- Department of Pathology and Laboratories, Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá 110100, Colombia.
| | - Felipe Vaca Paniagua
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, Estado de México 54090, Mexico.
- Instituto Nacional de Cancerología, CDMX 14080, Mexico.
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, 54090 Tlalnepantla, Estado de México, Mexico.
| |
Collapse
|
682
|
Shen T, Pu J, Lai HY, Xu L, Si X, Yan Y, Jiang Y, Zhang B. Genetic analysis of ATP13A2, PLA2G6 and FBXO7 in a cohort of Chinese patients with early-onset Parkinson's disease. Sci Rep 2018; 8:14028. [PMID: 30232368 PMCID: PMC6145881 DOI: 10.1038/s41598-018-32217-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/24/2018] [Indexed: 12/15/2022] Open
Abstract
Several genes have already been certified as causative genes in patients with autosomal recessive early-onset Parkinson's syndrome with pyramidal tract signs, including ATP13A2, PLA2G6 and FBXO7. Variants in these three genes may also play roles in early-onset Parkinson's disease (EOPD). In order to investigate the contribution of genetic variants in these three genes to Chinese sporadic EOPD patients, we screened 101 Chinese sporadic EOPD patients and 83 age- and sex-matched healthy controls using direct sequencing. Interpretation of those detected variants was performed based on the guidelines developed by the American College of Medical Genetics and Genomics (ACMG). Two missense variants, p.G360E and p.T733M, with "uncertain significance" classification were identified in the ATP13A2 gene and five synonymous variants were significantly over-represented in EOPD patients. Two missense variants, p.R53C and p.T319M, were absent in both our control group and online databases, classified as "likely pathogenic" in the PLA2G6 gene. Only benign variants were identified in the FBXO7 gene. These results indicate that rare variants of PLA2G6 may contribute to PD susceptibility in Chinese population, the ATP13A2 might be associated with higher risk for sporadic EOPD, while the FBXO7 gene doesn't seem to be a risk factor to develop sporadic PD in Chinese population. Further biochemical and molecular biological studies needs to be conducted to support our main results in our future researches.
Collapse
Affiliation(s)
- Ting Shen
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.,Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, 310029, China
| | - Jiali Pu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hsin-Yi Lai
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.,Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, 310029, China
| | - Lingjia Xu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Xiaoli Si
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yaping Yan
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yasi Jiang
- Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, 310029, China
| | - Baorong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
683
|
A Bayesian framework for efficient and accurate variant prediction. PLoS One 2018; 13:e0203553. [PMID: 30212499 PMCID: PMC6136750 DOI: 10.1371/journal.pone.0203553] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 08/22/2018] [Indexed: 12/04/2022] Open
Abstract
There is a growing need to develop variant prediction tools capable of assessing a wide spectrum of evidence. We present a Bayesian framework that involves aggregating pathogenicity data across multiple in silico scores on a gene-by-gene basis and multiple evidence statistics in both quantitative and qualitative forms, and performs 5-tiered variant classification based on the resulting probability credible interval. When evaluated in 1,161 missense variants, our gene-specific in silico model-based meta-predictor yielded an area under the curve (AUC) of 96.0% and outperformed all other in silico predictors. Multifactorial model analysis incorporating all available evidence yielded 99.7% AUC, with 22.8% predicted as variants of uncertain significance (VUS). Use of only 3 auto-computed evidence statistics yielded 98.6% AUC with 56.0% predicted as VUS, which represented sufficient accuracy to rapidly assign a significant portion of VUS to clinically meaningful classifications. Collectively, our findings support the use of this framework to conduct large-scale variant prioritization using in silico predictors followed by variant prediction and classification with a high degree of predictive accuracy.
Collapse
|
684
|
Mignot C, McMahon AC, Bar C, Campeau PM, Davidson C, Buratti J, Nava C, Jacquemont ML, Tallot M, Milh M, Edery P, Marzin P, Barcia G, Barnerias C, Besmond C, Bienvenu T, Bruel AL, Brunga L, Ceulemans B, Coubes C, Cristancho AG, Cunningham F, Dehouck MB, Donner EJ, Duban-Bedu B, Dubourg C, Gardella E, Gauthier J, Geneviève D, Gobin-Limballe S, Goldberg EM, Hagebeuk E, Hamdan FF, Hančárová M, Hubert L, Ioos C, Ichikawa S, Janssens S, Journel H, Kaminska A, Keren B, Koopmans M, Lacoste C, Laššuthová P, Lederer D, Lehalle D, Marjanovic D, Métreau J, Michaud JL, Miller K, Minassian BA, Morales J, Moutard ML, Munnich A, Ortiz-Gonzalez XR, Pinard JM, Prchalová D, Putoux A, Quelin C, Rosen AR, Roume J, Rossignol E, Simon MEH, Smol T, Shur N, Shelihan I, Štěrbová K, Vyhnálková E, Vilain C, Soblet J, Smits G, Yang SP, van der Smagt JJ, van Hasselt PM, van Kempen M, Weckhuysen S, Helbig I, Villard L, Héron D, Koeleman B, Møller RS, Lesca G, Helbig KL, Nabbout R, Verbeek NE, Depienne C. IQSEC2-related encephalopathy in males and females: a comparative study including 37 novel patients. Genet Med 2018; 21:837-849. [PMID: 30206421 PMCID: PMC6752297 DOI: 10.1038/s41436-018-0268-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/31/2018] [Indexed: 12/19/2022] Open
Abstract
Purpose Variants in IQSEC2, escaping X inactivation, cause X-linked intellectual disability with frequent epilepsy in males and females. We aimed to investigate sex-specific differences. Methods We collected the data of 37 unpublished patients (18 males and 19 females) with IQSEC2 pathogenic variants and 5 individuals with variants of unknown significance and reviewed published variants. We compared variant types and phenotypes in males and females and performed an analysis of IQSEC2 isoforms. Results IQSEC2 pathogenic variants mainly led to premature truncation and were scattered throughout the longest brain-specific isoform, encoding the synaptic IQSEC2/BRAG1 protein. Variants occurred de novo in females but were either de novo (2/3) or inherited (1/3) in males, with missense variants being predominantly inherited. Developmental delay and intellectual disability were overall more severe in males than in females. Likewise, seizures were more frequently observed and intractable, and started earlier in males than in females. No correlation was observed between the age at seizure onset and severity of intellectual disability or resistance to antiepileptic treatments. Conclusion This study provides a comprehensive overview of IQSEC2-related encephalopathy in males and females, and suggests that an accurate dosage of IQSEC2 at the synapse is crucial during normal brain development.
Collapse
Affiliation(s)
- Cyril Mignot
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universites, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle epiniere, ICM, Paris, France. .,APHP, Hôpital Pitie-Salpetriere, Departement de Genetique et de Cytogenetique; Centre de Reference Deficience Intellectuelle de Causes Rares; GRC UPMC «Deficience Intellectuelle et Autisme», Paris, France.
| | - Aoife C McMahon
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Claire Bar
- APHP, Reference Centre for Rare Epilepsies, Necker-Enfants Malades Hospital, Imagine Institute, Paris Descartes University, Paris, France.,INSERM U1163, Imagine Institute, Paris, France.,Paris Descartes University, Paris, France
| | - Philippe M Campeau
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and University of Montreal, Montreal, QC, Canada
| | - Claire Davidson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Julien Buratti
- APHP, Hôpital Pitie-Salpetriere, Departement de Genetique et de Cytogenetique; Centre de Reference Deficience Intellectuelle de Causes Rares; GRC UPMC «Deficience Intellectuelle et Autisme», Paris, France
| | - Caroline Nava
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universites, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle epiniere, ICM, Paris, France.,APHP, Hôpital Pitie-Salpetriere, Departement de Genetique et de Cytogenetique; Centre de Reference Deficience Intellectuelle de Causes Rares; GRC UPMC «Deficience Intellectuelle et Autisme», Paris, France
| | | | - Marilyn Tallot
- CHU La Reunion-Groupe Hospitalier Sud Reunion, La Reunion, France
| | - Mathieu Milh
- APHM, Hôpital d'Enfants de La Timone, Service de Neurologie Pediatrique, centre de reference deficiences intellectuelles de cause rare, Marseille, France.,Aix Marseille University, INSERM, MMG, UMR-S 1251, Faculte de medecine, Marseille, France
| | - Patrick Edery
- Service de Genetique, Centre de Reference Anomalies du Developpement, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Universite Claude Bernard Lyon 1, Bron, France.,Claude Bernard Lyon I University, Lyon, France
| | - Pauline Marzin
- APHP, Hôpital Pitie-Salpetriere, Departement de Genetique et de Cytogenetique; Centre de Reference Deficience Intellectuelle de Causes Rares; GRC UPMC «Deficience Intellectuelle et Autisme», Paris, France
| | - Giulia Barcia
- INSERM U1163, Imagine Institute, Paris, France.,Paris Descartes University, Paris, France.,APHP, Service de genetique medicale, Necker-Enfants Malades Hospital, Imagine Institute, Paris Descartes University, Paris, France
| | - Christine Barnerias
- APHP, Unite fonctionnelle de Neurologie, Necker-Enfants Malades Hospital, Imagine Institute, Paris Descartes University, Paris, France
| | - Claude Besmond
- INSERM U1163, Imagine Institute, Paris, France.,Paris Descartes University, Paris, France
| | - Thierry Bienvenu
- APHP, Laboratoire de Genetique et Biologie Moleculaires, Hôpital Cochin, HUPC, Paris, France.,Universite Paris Descartes Paris, Institut de Psychiatrie et de Neurosciences de Paris, Inserm U894, Paris, France
| | - Ange-Line Bruel
- FHU-TRANSLAD, Universite de Bourgogne/CHU Dijon, Dijon, France.,INSERM UMR 1231 GAD team, Genetics of Developmental disorders, Universite de Bourgogne-Franche Comte, Dijon, France
| | - Ledia Brunga
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Berten Ceulemans
- Department of Pediatric Neurology, University Hospital and University of Antwerp, Antwerp, Belgium
| | - Christine Coubes
- Departement de Genetique Medicale, Maladies rares et Medecine Personnalisee, CHU de Montpellier, Montpellier, France
| | - Ana G Cristancho
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fiona Cunningham
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Elizabeth J Donner
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Bénédicte Duban-Bedu
- Centre de Genetique Chromosomique, Hôpital St-Vincent-de-Paul, GHICL, Lille, France
| | - Christèle Dubourg
- CHU Rennes, Service de Genetique Moleculaire et Genomique, Rennes, France
| | - Elena Gardella
- Danish Epilepsy Centre Filadelfia, Dianalund, Denmark.,Institute for Regional Health Services, University of Southern Denmark, Odense, Denmark
| | - Julie Gauthier
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and University of Montreal, Montreal, QC, Canada
| | - David Geneviève
- Departement de Genetique Medicale, Maladies rares et Medecine Personnalisee, CHU de Montpellier, Montpellier, France.,INSERM U1183, Montpellier, France
| | - Stéphanie Gobin-Limballe
- APHP, Service de genetique medicale, Necker-Enfants Malades Hospital, Imagine Institute, Paris Descartes University, Paris, France
| | - Ethan M Goldberg
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Eveline Hagebeuk
- Stichting Epilepsie Instellingen Nederland, SEIN, Zwolle, The Netherlands
| | - Fadi F Hamdan
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and University of Montreal, Montreal, QC, Canada
| | - Miroslava Hančárová
- Department of Biology and Medical Genetics, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Laurence Hubert
- INSERM U1163, Imagine Institute, Paris, France.,Paris Descartes University, Paris, France
| | - Christine Ioos
- APHP, University Hospital of Paris ïle-de-France ouest, Raymond Poincare Hospital, Garches, France
| | - Shoji Ichikawa
- Department of Clinical Diagnostics, Ambry Genetics, Aliso Viejo, CA, USA
| | - Sandra Janssens
- Centre for Medical Genetics Ghent, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium
| | - Hubert Journel
- Service de Genetique Medicale, Hôpital Chubert, Vannes, France
| | - Anna Kaminska
- APHP, Department of Clinical Neurophysiology, Necker-Enfants Malades Hospital, Paris, France
| | - Boris Keren
- APHP, Hôpital Pitie-Salpetriere, Departement de Genetique et de Cytogenetique; Centre de Reference Deficience Intellectuelle de Causes Rares; GRC UPMC «Deficience Intellectuelle et Autisme», Paris, France
| | - Marije Koopmans
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Lacoste
- Departement de Genetique Medicale, APHM, Hopital d'Enfants de La Timone, Marseille, France
| | - Petra Laššuthová
- Child Neurology Department, 2nd Faculty of Medicine, Charles University and Motol Hospital, Prague, Czech Republic
| | - Damien Lederer
- Centre de Genetique Humaine, Institut de Pathologie et de Genetique, Gosselies, Belgium
| | - Daphné Lehalle
- FHU-TRANSLAD, Universite de Bourgogne/CHU Dijon, Dijon, France.,Unite fonctionnelle de genetique clinique, Centre Hospitalier Intercommunal de Creteil, Creteil, France
| | | | - Julia Métreau
- APHP, Service de neurologie pediatrique, Hôpital Universitaire Bicetre, Le Kremlin-Bicetre, France
| | - Jacques L Michaud
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and University of Montreal, Montreal, QC, Canada
| | - Kathryn Miller
- Department of Pediatrics, Albany Medical Center, Albany, NY, USA
| | - Berge A Minassian
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Joannella Morales
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Marie-Laure Moutard
- APHP, Hôpital Trousseau, service de neuropediatrie, Paris, France.,Sorbonne Universite, GRC n°19, pathologies Congenitales du Cervelet-LeucoDystrophies, APHP, Hôpital Armand Trousseau, Paris, France
| | - Arnold Munnich
- INSERM U1163, Imagine Institute, Paris, France.,Paris Descartes University, Paris, France.,APHP, Service de genetique medicale, Necker-Enfants Malades Hospital, Imagine Institute, Paris Descartes University, Paris, France
| | | | - Jean-Marc Pinard
- Division of Neuropediatrics, CHU Raymond Poincare (APHP), Garches, France
| | - Darina Prchalová
- Department of Biology and Medical Genetics, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Audrey Putoux
- Service de Genetique, Centre de Reference Anomalies du Developpement, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Universite Claude Bernard Lyon 1, Bron, France.,Claude Bernard Lyon I University, Lyon, France
| | - Chloé Quelin
- Service de Genetique Medicale, CLAD Ouest CHU Hôpital Sud, Rennes, France
| | - Alyssa R Rosen
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joelle Roume
- Unite de Genetique Medicale, Centre de Reference des Maladies rares du Developpement (AnD DI Rares), CHI Poissy-St Germain en Laye, Poissy, France
| | - Elsa Rossignol
- Departments of Pediatrics and Neurosciences, CHU Sainte-Justine and University of Montreal, Montreal, Canada
| | - Marleen E H Simon
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Thomas Smol
- Institut de Genetique Medicale, CHRU Lille, Universite de Lille, Lille, France
| | - Natasha Shur
- Department of Pediatrics, Albany Medical Center, Albany, NY, USA
| | - Ivan Shelihan
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and University of Montreal, Montreal, QC, Canada
| | - Katalin Štěrbová
- Child Neurology Department, 2nd Faculty of Medicine, Charles University and Motol Hospital, Prague, Czech Republic
| | - Emílie Vyhnálková
- Department of Biology and Medical Genetics, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Catheline Vilain
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Universite Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Universite Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Universite Libre de Bruxelles, Brussels, Belgium
| | - Julie Soblet
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Universite Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Universite Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Universite Libre de Bruxelles, Brussels, Belgium
| | - Guillaume Smits
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Universite Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Universite Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Universite Libre de Bruxelles, Brussels, Belgium
| | - Samuel P Yang
- Clinical Genomics & Predictive Medicine, Providence Medical Group, Dayton, WA, USA
| | | | - Peter M van Hasselt
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center, Utrecht, The Netherlands
| | - Marjan van Kempen
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sarah Weckhuysen
- Neurogenetics Group, Center of Molecular Neurology, VIB, Antwerp, Belgium.,Neurology Department, University Hospital Antwerp, Antwerp, Belgium
| | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurent Villard
- Aix Marseille University, INSERM, MMG, UMR-S 1251, Faculte de medecine, Marseille, France.,Departement de Genetique Medicale, APHM, Hopital d'Enfants de La Timone, Marseille, France
| | - Delphine Héron
- APHP, Hôpital Pitie-Salpetriere, Departement de Genetique et de Cytogenetique; Centre de Reference Deficience Intellectuelle de Causes Rares; GRC UPMC «Deficience Intellectuelle et Autisme», Paris, France
| | - Bobby Koeleman
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rikke S Møller
- CHU Rennes, Service de Genetique Moleculaire et Genomique, Rennes, France.,Danish Epilepsy Centre Filadelfia, Dianalund, Denmark
| | - Gaetan Lesca
- Service de Genetique, Centre de Reference Anomalies du Developpement, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Universite Claude Bernard Lyon 1, Bron, France.,Claude Bernard Lyon I University, Lyon, France
| | - Katherine L Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rima Nabbout
- APHP, Reference Centre for Rare Epilepsies, Necker-Enfants Malades Hospital, Imagine Institute, Paris Descartes University, Paris, France.,INSERM U1163, Imagine Institute, Paris, France.,Paris Descartes University, Paris, France
| | - Nienke E Verbeek
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christel Depienne
- INSERM, U 1127, CNRS UMR 7225, Sorbonne Universites, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle epiniere, ICM, Paris, France. .,IGBMC, CNRS UMR 7104/INSERM U964/Universite de Strasbourg, Illkirch, France. .,Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
685
|
Ladino LY, Galvis J, Yasnó D, Ramírez A, Beltrán OI. A pathogenic homozygous variant of the BBS10 gene in a patient with Bardet Biedl syndrome. BIOMEDICA : REVISTA DEL INSTITUTO NACIONAL DE SALUD 2018; 38:308-320. [PMID: 30335236 DOI: 10.7705/biomedica.v38i4.4199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/23/2018] [Accepted: 04/02/2018] [Indexed: 06/08/2023]
Abstract
The Bardet-Biedl syndrome is an autosomal recessive hereditary disorder with vast locus heterogeneity that belongs to the so-called ciliopathies, whose proteins are localized in the primary cilia and present functional deficiency. The multisystemic features of the disease include ocular, renal, cognitive, skeletal, as well as gonadal involvement and obesity, among others, with high inter- and intrafamilial variability. We describe the clinical case of an adolescent male patient with Bardet-Biedl syndrome, including the approach, the results from a 22-gene sequencing panel, and the analysis of updated scientific literature. We collected the clinical data of the patient and, after obtaining the informed consent, we conducted a multigenic sequencing panel oriented to known implicated genes. The patient was born to consanguineous parents and was the first affected member of the family. He presented with postaxial polydactyly, obesity, micropenis, retinitis pigmentosa, and learning disability. The multigenic panel allowed the identification of the homozygous pathogenic variant c.39_46del in the BBS10 gene and in other BBS genes variants associated with obesity. As the Bardet-Biedl syndrome is a rare disease, it is challenging to interpret its pleiotropism and gene/allelic heterogeneity. Its confirmation by molecular tests allows an adequate approach, follow-up, and genetic counseling of the patient and the family.
Collapse
Affiliation(s)
- Luz Yaqueline Ladino
- Departamento de Genética, Grupo de Investigación GenHOMI, Fundación Hospital Pediátrico La Misericordia-HOMI, Bogotá, D.C., Colombia Maestría en Genética Humana, Universidad Nacional de Colombia, Bogotá, D.C., Colombia.
| | | | | | | | | |
Collapse
|
686
|
Cao Z, Zhu Y, Liu L, Wu S, Liu B, Zhuang J, Tong Y, Chen X, Xie Y, Nie K, Lu C, Ma X, Yang J. Novel mutations in HSF4 cause congenital cataracts in Chinese families. BMC MEDICAL GENETICS 2018; 19:150. [PMID: 30143024 PMCID: PMC6109319 DOI: 10.1186/s12881-018-0636-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/28/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Congenital cataract, a kind of cataract presenting at birth or during early childhood, is a leading cause of childhood blindness. To date, more than 30 genes on different chromosomes are known to cause this disorder. This study aimed to identify the HSF4 mutations in a cohort from Chinese families affected with congenital cataracts. METHODS Forty-two unrelated non-syndromic congenital cataract families and 112 ethnically matched controls from southeast China were recruited from the southeast of China. We employed Sanger sequencing method to discover the variants. To confirm the novel mutations, STR haplotypes were constructed to check the co-segregation with congenital cataract. The pathogenic potential of the novel mutations were assessed using bioinformatics tools including SIFT, Polyphen2, and Human Splicing Finder. The pathogenicity of all the mutations was evaluated by the guidelines of American College of Medical Genetics and InterVar software. RESULTS No previously reported HSF4 mutations were found in all the congenital cataract families. Five novel HSF4 mutations including c.187 T > C (p.Phe63Leu), c.218G > T (p.Arg73Leu), c.233A > G (p.Tyr78Cys), IVS5 c.233-1G > A and c.314G > C (p.Ser105Thr) were identified in five unrelated families with congenital cataracts, respectively. These mutations co-segregated with all affected individuals in each family were not observed in the unaffected family members or in 112 unrelated controls. All five mutations were categorized to be the disease "pathogenic" according to ACMG guidelines and using InterVar software. Mutations in the HSF4 were responsible for 11.90% Chinese families with congenital cataracts in our cohort. CONCLUSIONS In the study, we identified five novel HSF4 mutations in Chinese families with congenital cataracts. Our results expand the spectrum of HSF4 mutations causing congenital cataracts, which may be helpful for the molecular diagnosis of congenital cataracts in the era of precision medicine.
Collapse
Affiliation(s)
- Zongfu Cao
- Graduate School of Peking Union Medical College, Beijing, China.,National Center for Human Genetics, Beijing, China.,National Human Genetic Resources Center, National Research Institute for Family Planning, Peking Union Medical College, 12 Da-hui-si, Hai Dian, Beijing, 100081, China
| | - Yihua Zhu
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lijuan Liu
- Fuzhou Southeast Eye Hospital, Fuzhou, Fujian, China
| | - Shuangqing Wu
- Department of Ophthalmology, Hangzhou Red-cross hospital, Zhejiang, Hangzhou, China
| | - Bing Liu
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jianfu Zhuang
- Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Yi Tong
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaole Chen
- Biomedical Engineering Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Yongqing Xie
- Biomedical Engineering Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Kaimei Nie
- Biomedical Engineering Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Cailing Lu
- Graduate School of Peking Union Medical College, Beijing, China.,National Human Genetic Resources Center, National Research Institute for Family Planning, Peking Union Medical College, 12 Da-hui-si, Hai Dian, Beijing, 100081, China
| | - Xu Ma
- Graduate School of Peking Union Medical College, Beijing, China. .,National Center for Human Genetics, Beijing, China. .,National Human Genetic Resources Center, National Research Institute for Family Planning, Peking Union Medical College, 12 Da-hui-si, Hai Dian, Beijing, 100081, China.
| | - Juhua Yang
- Biomedical Engineering Center, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
687
|
Noncardiac genetic predisposition in sudden infant death syndrome. Genet Med 2018; 21:641-649. [DOI: 10.1038/s41436-018-0131-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/28/2018] [Indexed: 01/18/2023] Open
|
688
|
Paldino A, De Angelis G, Merlo M, Gigli M, Dal Ferro M, Severini GM, Mestroni L, Sinagra G. Genetics of Dilated Cardiomyopathy: Clinical Implications. Curr Cardiol Rep 2018; 20:83. [DOI: 10.1007/s11886-018-1030-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
689
|
Yang Y, Guo J, Dai L, Zhu Y, Hu H, Tan L, Chen W, Liang D, He J, Tu M, Wang K, Wu L. XRCC2 mutation causes meiotic arrest, azoospermia and infertility. J Med Genet 2018; 55:628-636. [PMID: 30042186 PMCID: PMC6119352 DOI: 10.1136/jmedgenet-2017-105145] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 05/31/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Background Meiotic homologous recombination (HR) plays an essential role in gametogenesis. In most eukaryotes, meiotic HR is mediated by two recombinase systems: ubiquitous RAD51 and meiosis-specific DMC1. In the RAD51-mediated HR system, RAD51 and five RAD51 paralogues are essential for normal RAD51 function, but the role of RAD51 in human meiosis is unclear. The knockout of Rad51 or any Rad51 paralogue in mice exhibits embryonic lethality. We investigated a family with meiotic arrest, azoospermia and infertility but without other abnormalities. Methods Homozygosity mapping and whole-exome sequencing were performed in a consanguineous family. An animal model carrying a related mutation was created by using a CRISPR/Cas9 system. Results We identified a 1 bp homozygous substitution (c.41T>C/p.Leu14Pro) on a RAD51 paralogue, namely, XRCC2, in the consanguineous family. We did not detect any XRCC2 recessive mutation in a cohort of 127 males with non-obstructive-azoospermia. Knockin mice with Xrcc2-c.T41C/p.Leu14Pro mutation were generated successfully by the CRISPR/Cas9 method. The homozygotes survived and exhibited meiotic arrest, azoospermia, premature ovarian failure and infertility. Conclusion A XRCC2 recessive mutation causing meiotic arrest and infertility in humans was duplicated with knockin mice. Our results revealed a new Mendelian hereditary entity and provided an experimental model of RAD51-HR gene defect in mammalian meiosis.
Collapse
Affiliation(s)
- Yongjia Yang
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,State Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Jihong Guo
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,State Key Laboratory of Medical Genetics, Central South University, Changsha, China.,Xiangya Hospital, Central South University, Changsha, China
| | - Lei Dai
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,State Key Laboratory of Medical Genetics, Central South University, Changsha, China.,Xiangya Hospital, Central South University, Changsha, China
| | - Yimin Zhu
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,Hunan People's Hospital, Hunan Normal University, Changsha, China
| | - Hao Hu
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,Department of Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, China
| | - Lihong Tan
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China
| | - Weijian Chen
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China
| | - Desheng Liang
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,State Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Jingliang He
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,State Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Ming Tu
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China
| | - Kewei Wang
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China
| | - Lingqian Wu
- The Laboratory of Genetics and Metabolism, Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, University of South China, Changsha, China.,State Key Laboratory of Medical Genetics, Central South University, Changsha, China
| |
Collapse
|
690
|
Sztupinszki Z, Diossy M, Krzystanek M, Reiniger L, Csabai I, Favero F, Birkbak NJ, Eklund AC, Syed A, Szallasi Z. Migrating the SNP array-based homologous recombination deficiency measures to next generation sequencing data of breast cancer. NPJ Breast Cancer 2018; 4:16. [PMID: 29978035 PMCID: PMC6028448 DOI: 10.1038/s41523-018-0066-6] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 02/01/2023] Open
Abstract
The first genomic scar-based homologous recombination deficiency (HRD) measures were produced using SNP arrays. As array-based technology has been largely replaced by next generation sequencing approaches, it has become important to develop algorithms that derive the same type of genomic scar scores from next generation sequencing (whole exome “WXS”, whole genome “WGS”) data. In order to perform this analysis, we introduce here the scarHRD R package and show that using this method the SNP array-based and next generation sequencing-based derivation of HRD scores show good correlation (Pearson correlation between 0.73 and 0.87 depending on the actual HRD measure) and that the NGS-based HRD scores distinguish similarly well between BRCA mutant and BRCA wild-type cases in a cohort of triple-negative breast cancer patients of the TCGA data set.
Collapse
Affiliation(s)
- Zsofia Sztupinszki
- 1Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, Lyngby, 2800 Denmark
| | - Miklos Diossy
- 1Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, Lyngby, 2800 Denmark
| | - Marcin Krzystanek
- 1Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, Lyngby, 2800 Denmark
| | - Lilla Reiniger
- 21st Department of Pathology and Experimental Research, Semmelweis University, Budapest, Hungary.,32nd Department of Pathology, MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - István Csabai
- 4Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | - Francesco Favero
- 5Biotech Research & Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, DK-2200 Denmark
| | - Nicolai J Birkbak
- 6The Francis Crick Institute, London, UK.,7University College London Cancer Institute, London, UK
| | - Aron C Eklund
- 1Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, Lyngby, 2800 Denmark
| | - Ali Syed
- Danish National Life Science Supercomputing Center, Copenhagen, Denmark
| | - Zoltan Szallasi
- 1Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, Lyngby, 2800 Denmark.,32nd Department of Pathology, MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary.,9Computational Health Informatics Program, Boston Children's Hospital, Boston, MA USA.,10Harvard Medical School, Boston, MA USA
| |
Collapse
|
691
|
Abid A, Shahid S, Shakoor M, Lanewala AA, Hashmi S, Khaliq S. Screening of the LAMB2, WT1, NPHS1, and NPHS2 Genes in Pediatric Nephrotic Syndrome. Front Genet 2018; 9:214. [PMID: 30013592 PMCID: PMC6036290 DOI: 10.3389/fgene.2018.00214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 05/25/2018] [Indexed: 02/04/2023] Open
Abstract
Mutations in the NPHS1, NPHS2, LAMB2, and the WT1 genes are responsible for causing nephrotic syndrome (NS) in two third of the early onset cases. This study was carried out to assess the frequencies of mutations in these genes in a cohort of pediatric NS patients. A total of 64 pediatric familial or sporadic SRNS cases were recruited. Among these, 74% had a disease onset of up to 3 years of age. We found one homozygous frameshift mutation in the NPHS1 gene in one CNS case and two homozygous mutations in the NPHS2 gene. Six mutations in four cases in the LAMB2 gene were also identified. No mutation was detected in the WT1 gene in isolated SRNS cases. LAMB2 gene missense mutations were segregating in NS cases with no extra-renal abnormalities. Analysis of the population genomic data (1000 genome and gnomAD databases) for the prevalence estimation revealed that NS is more prevalent than previously determined from clinical cohorts especially in Asian population compared with overall world populations (prevalence worldwide was 1in 189036 and in South-Asian was 1in 56689). Our results reiterated a low prevalence of mutations in the NPHS1, NPHS2, LAMB2, and WT1 genes in the studied population from Pakistan as compared to some European population that showed a high prevalence of mutations in these genes. This is a comprehensive screening of the genes causing early onset NS in sporadic and familial NS cases suggesting a more systematic and robust approach for mutation identification in all the 45 disease-causing genes in NS in our population is required.
Collapse
Affiliation(s)
- Aiysha Abid
- Centre for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Saba Shahid
- Department of Genomic, National Institute of Blood Diseases, Karachi, Pakistan
| | - Madiha Shakoor
- Department of Human Genetics, University of Health Sciences, Lahore, Pakistan
| | - Ali A Lanewala
- Department of Pediatric Nephrology, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Seema Hashmi
- Department of Pediatric Nephrology, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Shagufta Khaliq
- Department of Human Genetics, University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
692
|
The genotypic and phenotypic spectrum of PARS2-related infantile-onset encephalopathy. J Hum Genet 2018; 63:971-980. [PMID: 29915213 DOI: 10.1038/s10038-018-0478-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022]
Abstract
Mitochondrial aminoacyl-tRNA synthetases (mt-aaRSs) are a family of enzymes that play critical roles in protein biosynthesis. Mutations in mt-aaRSs are associated with various diseases. As a member of the mt-aaRS family, PARS2 encoding prolyl-tRNA synthetase 2 was recently shown to be associated with Alpers syndrome and certain infantile-onset neurodegenerative disorders in four patients. Here, we present two patients in a pedigree with early developmental delay, epileptic spasms, delayed myelination combined with cerebellar white matter abnormalities, and progressive cortical atrophy. Whole-exome sequencing revealed pathogenic compound heterozygous variants [c.283 G > A (p.95 V > I)] and [c.604 G > C (p.202 R > G)] in PARS2. Nearly all patients had epileptic spasms with early response to treatment, early developmental delay and/or regression followed by generalized hypotonia, postnatal microcephaly, elevated lactate levels, and progressive cerebral atrophy. Our study provides further evidence for validating the role of PARS2 in the pathology of related infantile-onset encephalopathy, contributing to the phenotypic features of this condition, and providing clinical and molecular insight for the diagnosis of this disease entity.
Collapse
|
693
|
Turan I, Kotan LD, Tastan M, Gurbuz F, Topaloglu AK, Yuksel B. Molecular genetic studies in a case series of isolated hypoaldosteronism due to biosynthesis defects or aldosterone resistance. Clin Endocrinol (Oxf) 2018; 88:799-805. [PMID: 29582446 DOI: 10.1111/cen.13603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIM Hypoaldosteronism is associated with either insufficient aldosterone production or aldosterone resistance (pseudohypoaldosteronism). Patients with aldosterone defects typically present with similar symptoms and findings, which include failure to thrive, vomiting, hyponatremia, hyperkalemia and metabolic acidosis. Accurate diagnosis of these clinical conditions therefore can be challenging. Molecular genetic analyses can help to greatly clarify this complexity. The aim of this study was to obtain an overview of the clinical and genetic characteristics of patients with aldosterone defects due to biosynthesis defects or aldosterone resistance. DESIGN AND PATIENTS We investigated the clinical and molecular genetic features of 8 consecutive patients with a clinical picture of aldosterone defects seen in our clinics during the period of May 2015 through October 2017. We screened CYP11B2 for aldosterone synthesis defects and NR3C2 and the three EnaC subunits (SCNN1A, SCNN1B and SCNN1G) for aldosterone resistance. RESULTS We found 4 novel and 2 previously reported mutations in the genes CYP11B2, NR3C2, SCNN1A and SCNN1G in 9 affected individuals from 7 unrelated families. CONCLUSION Molecular genetic investigations can help confidently diagnose these conditions and clarify the pathogenicity of aldosterone defects. This study may expand the clinical and genetic correlations of defects in aldosterone synthesis or resistance.
Collapse
Affiliation(s)
- Ihsan Turan
- Faculty of Medicine, Division of Pediatric Endocrinology, Cukurova University, Adana, Turkey
| | - Leman Damla Kotan
- Faculty of Medicine, Division of Pediatric Endocrinology, Cukurova University, Adana, Turkey
| | - Mehmet Tastan
- Faculty of Medicine, Division of Pediatric Endocrinology, Cukurova University, Adana, Turkey
| | - Fatih Gurbuz
- Faculty of Medicine, Division of Pediatric Endocrinology, Cukurova University, Adana, Turkey
| | - Ali Kemal Topaloglu
- Faculty of Medicine, Division of Pediatric Endocrinology, Cukurova University, Adana, Turkey
| | - Bilgin Yuksel
- Faculty of Medicine, Division of Pediatric Endocrinology, Cukurova University, Adana, Turkey
| |
Collapse
|
694
|
Xu C, Messina A, Somm E, Miraoui H, Kinnunen T, Acierno J, Niederländer NJ, Bouilly J, Dwyer AA, Sidis Y, Cassatella D, Sykiotis GP, Quinton R, De Geyter C, Dirlewanger M, Schwitzgebel V, Cole TR, Toogood AA, Kirk JM, Plummer L, Albrecht U, Crowley WF, Mohammadi M, Tena-Sempere M, Prevot V, Pitteloud N. KLB, encoding β-Klotho, is mutated in patients with congenital hypogonadotropic hypogonadism. EMBO Mol Med 2018; 9:1379-1397. [PMID: 28754744 PMCID: PMC5623842 DOI: 10.15252/emmm.201607376] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic form of isolated gonadotropin‐releasing hormone (GnRH) deficiency caused by mutations in > 30 genes. Fibroblast growth factor receptor 1 (FGFR1) is the most frequently mutated gene in CHH and is implicated in GnRH neuron development and maintenance. We note that a CHH FGFR1 mutation (p.L342S) decreases signaling of the metabolic regulator FGF21 by impairing the association of FGFR1 with β‐Klotho (KLB), the obligate co‐receptor for FGF21. We thus hypothesized that the metabolic FGF21/KLB/FGFR1 pathway is involved in CHH. Genetic screening of 334 CHH patients identified seven heterozygous loss‐of‐function KLB mutations in 13 patients (4%). Most patients with KLB mutations (9/13) exhibited metabolic defects. In mice, lack of Klb led to delayed puberty, altered estrous cyclicity, and subfertility due to a hypothalamic defect associated with inability of GnRH neurons to release GnRH in response to FGF21. Peripheral FGF21 administration could indeed reach GnRH neurons through circumventricular organs in the hypothalamus. We conclude that FGF21/KLB/FGFR1 signaling plays an essential role in GnRH biology, potentially linking metabolism with reproduction.
Collapse
Affiliation(s)
- Cheng Xu
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Andrea Messina
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Emmanuel Somm
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Hichem Miraoui
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Tarja Kinnunen
- Department of Biology, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - James Acierno
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Nicolas J Niederländer
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Justine Bouilly
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Andrew A Dwyer
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland.,University of Lausanne Institute of Higher Education and Research in Healthcare, Lausanne, Switzerland
| | - Yisrael Sidis
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Daniele Cassatella
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Gerasimos P Sykiotis
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Richard Quinton
- Institute for Genetic Medicine, University of Newcastle-on-Tyne, Newcastle-on Tyne, UK
| | - Christian De Geyter
- Clinic of Gynecological Endocrinology and Reproductive Medicine, University Hospital, University of Basel, Basel, Switzerland
| | - Mirjam Dirlewanger
- Pediatric Endocrine and Diabetes Unit, Children's Hospital, University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Valérie Schwitzgebel
- Pediatric Endocrine and Diabetes Unit, Children's Hospital, University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Trevor R Cole
- Department of Clinical Genetics, Birmingham Women's Hospital, Birmingham, UK
| | - Andrew A Toogood
- Department of Endocrinology, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK
| | - Jeremy Mw Kirk
- Department of Endocrinology, Birmingham Children's Hospital, Birmingham, UK
| | - Lacey Plummer
- National Center for Translational Research in Reproduction and Infertility, Harvard Reproductive Endocrine Sciences Center of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Urs Albrecht
- Department of Biology, Biochemistry, Faculty of Science, University of Fribourg, Fribourg, Switzerland
| | - William F Crowley
- National Center for Translational Research in Reproduction and Infertility, Harvard Reproductive Endocrine Sciences Center of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Moosa Mohammadi
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Instituto Maimonides de Investigación Biomédica de Cordoba (IMIBIC/HURS), Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, JPARC, Lille, France.,FHU 1000 Days for Health, School of Medicine, University of Lille, Lille, France
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology & Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
695
|
Maeda A, Yoshida A, Kawai K, Arai Y, Akiba R, Inaba A, Takagi S, Fujiki R, Hirami Y, Kurimoto Y, Ohara O, Takahashi M. Development of a molecular diagnostic test for Retinitis Pigmentosa in the Japanese population. Jpn J Ophthalmol 2018; 62:451-457. [PMID: 29785639 DOI: 10.1007/s10384-018-0601-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 04/30/2018] [Indexed: 11/29/2022]
Abstract
PURPOSE Retinitis Pigmentosa (RP) is the most common form of inherited retinal dystrophy caused by different genetic variants. More than 60 causative genes have been identified to date. The establishment of cost-effective molecular diagnostic tests with high sensitivity and specificity can be beneficial for patients and clinicians. Here, we developed a clinical diagnostic test for RP in the Japanese population. STUDY DESIGN Evaluation of diagnostic technology, Prospective, Clinical and experimental study. METHODS A panel of 39 genes reported to cause RP in Japanese patients was established. Next generation sequence (NGS) technology was applied for the analyses of 94 probands with RP and RP-related diseases. After interpretation of detected genetic variants, molecular diagnosis based on a study of the genetic variants and a clinical phenotype was made by a multidisciplinary team including clinicians, researchers and genetic counselors. RESULTS NGS analyses found 14,343 variants from 94 probands. Among them, 189 variants in 83 probands (88.3% of all cases) were selected as pathogenic variants and 64 probands (68.1%) have variants which can cause diseases. After the deliberation of these 64 cases, molecular diagnosis was made in 43 probands (45.7%). The final molecular diagnostic rate with the current system combining supplemental Sanger sequencing was 47.9% (45 of 94 cases). CONCLUSIONS The RP panel provides the significant advantage of detecting genetic variants with a high molecular diagnostic rate. This type of race-specific high-throughput genotyping allows us to conduct a cost-effective and clinically useful genetic diagnostic test.
Collapse
Affiliation(s)
- Akiko Maeda
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan. .,Division of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe, Japan. .,Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, USA.
| | - Akiko Yoshida
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.,Division of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe, Japan
| | - Kanako Kawai
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Yuki Arai
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Ryutaro Akiba
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Akira Inaba
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Seiji Takagi
- Division of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe, Japan.,Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Ryoji Fujiki
- Department of Technology Development, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Yasuhiko Hirami
- Division of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe, Japan.,Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yasuo Kurimoto
- Division of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe, Japan.,Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Osamu Ohara
- Department of Technology Development, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.,Division of Ophthalmology, Institute of Biomedical Research and Innovation Hospital, Kobe, Japan.,Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe, Japan
| |
Collapse
|
696
|
Kosaki R, Ono H, Terashima H, Kosaki K. Timothy syndrome-like condition with syndactyly but without prolongation of the QT interval. Am J Med Genet A 2018; 176:1657-1661. [PMID: 29736926 DOI: 10.1002/ajmg.a.38833] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/18/2018] [Accepted: 04/17/2018] [Indexed: 01/03/2023]
Abstract
Timothy syndrome is characterized by a unique combination of a prolongation of the corrected QT interval of the electrocardiogram and bilateral cutaneous syndactyly of the fingers and the toes and is caused by heterozygous mutations in CACNA1C, a gene encoding a calcium channel. After the discovery of the CACNA1C gene as the causative gene for Timothy syndrome, patients with CACNA1C mutations with QT prolongation but without syndactyly were described. Here, we report a 5-year-old female patient with cutaneous syndactyly, developmental delay, and pulmonary hypertension. Exome analysis showed a previously undescribed de novo heterozygous mutation in the CACNA1C gene, p.Arg1024Gly. To our knowledge, this patient is the first to exhibit syndactyly and to carry a CACNA1C mutation but to not have QT prolongation, which has long been considered an obligatory feature of Timothy syndrome.
Collapse
Affiliation(s)
- Rika Kosaki
- Division of Medical Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Hiroshi Ono
- Division of Cardiology, National Center for Child Health and Development, Tokyo, Japan
| | - Hiroshi Terashima
- Division of Neulorogy, National Center for Child Health and Development, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
697
|
Traboulsi EI. Words of Uncertain Significance. Ophthalmol Retina 2018; 2:387-388. [PMID: 31047319 DOI: 10.1016/j.oret.2018.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/16/2018] [Accepted: 01/22/2018] [Indexed: 06/09/2023]
Affiliation(s)
- Elias I Traboulsi
- Center for Genetic Eye Diseases, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
698
|
Di Resta C, Spiga I, Presi S, Merella S, Pipitone GB, Manitto MP, Querques G, Parodi MB, Ferrari M, Carrera P. Integration of multigene panels for the diagnosis of hereditary retinal disorders using Next Generation Sequencing and bioinformatics approaches. EJIFCC 2018; 29:15-25. [PMID: 29765283 PMCID: PMC5949615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
In recent years, Next-Generation Sequencing (NGS) opened a new way for the study of pathogenic mechanisms and for molecular diagnosis of inherited disorders. In the present work, we focused our attention on the inherited retinal dystrophies (IRDs), a group of specific disorders of the retina, displaying a very high clinical and genetic heterogeneity, whose genetic diagnosis is not easily feasible. It represents a paradigmatic example for the integration of clinical and molecular examination toward precision medicine. In this paper, we discuss the use of targeted NGS resequencing of selected gene panels in a cohort of patients affected by IRDs. We tested the hypothesis to apply a selective approach based on a careful clinical examination. By this approach we reached a 66% overall detection rate for pathogenic variants, with a 52% diagnostic yield. Reduction of the efforts for validation and classification of variants is a clear advantage for the management of genetic testing in a clinical setting.
Collapse
Affiliation(s)
- Chiara Di Resta
- Vita-Salute San Raffaele University, Milan, Italy, Unit of Genomics for Human Disease Diagnosis, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ivana Spiga
- Clinical Molecular Biology Laboratory, IRCCS San Raffaele Hospital, Milan, Italy
| | - Silvia Presi
- Clinical Molecular Biology Laboratory, IRCCS San Raffaele Hospital, Milan, Italy
| | - Stefania Merella
- Clinical Molecular Biology Laboratory, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Maria Pia Manitto
- Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Querques
- Medical Retina & Imaging Unit, Department of Ophthalmology IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Maurizio Ferrari
- Vita-Salute San Raffaele University, Milan, Italy, Unit of Genomics for Human Disease Diagnosis, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy, Clinical Molecular Biology Laboratory, IRCCS San Raffaele Hospital, Milan, Italy,Corresponding author: Maurizio Ferrari; Genomic Unit for the Diagnosis of Human Pathologies; Division of Genetics and Cellular Biology; IRCCS San Raffaele Hospital; Via Olgettina 60; 20132 Milan, Italy; Phone: 02-26432303; Fax: 02-26434351; E-mail:
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy, Clinical Molecular Biology Laboratory, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
699
|
Mutation of IPO13 causes recessive ocular coloboma, microphthalmia, and cataract. Exp Mol Med 2018; 50:1-11. [PMID: 29700284 PMCID: PMC5938035 DOI: 10.1038/s12276-018-0079-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/22/2018] [Accepted: 02/14/2018] [Indexed: 11/12/2022] Open
Abstract
Ocular coloboma is a developmental structural defect of the eye that often occurs as complex ocular anomalies. However, its genetic etiology remains largely unexplored. Here we report the identification of mutation (c.331C>T, p.R111C) in the IPO13 gene in a consanguineous family with ocular coloboma, microphthalmia, and cataract by a combination of whole-exome sequencing and homozygosity mapping. IPO13 encodes an importin-B family protein and has been proven to be associated with the pathogenesis of coloboma and microphthalmia. We found that Ipo13 was expressed in the cornea, sclera, lens, and retina in mice. Additionally, the mRNA expression level of Ipo13 decreased significantly in the patient compared with its expression in a healthy individual. Morpholino-oligonucleotide-induced knockdown of ipo13 in zebrafish caused dose-dependent microphthalmia and coloboma, which is highly similar to the ocular phenotypes in the patient. Moreover, both visual motor response and optokinetic response were impaired severely. Notably, these ocular phenotypes in ipo13-deficient zebrafish could be rescued remarkably by full-length ipo13 mRNA, suggesting that the phenotypes observed in zebrafish were due to insufficient ipo13 function. Altogether, our findings demonstrate, for the first time, a new role of IPO13 in eye morphogenesis and that loss of function of IPO13 could lead to ocular coloboma, microphthalmia, and cataract in humans and zebrafish. In-depth genomic analysis of the family of a young man with severe visual impairment reveals a new gene involved in eye development. Ocular coloboma encompasses various hereditary disorders in which the eyes form improperly. Many of the underlying genetic factors remain unidentified. Researchers led by Zi-Bing Jin at Wenzhou Medical University in China sequenced the genes of 28-year-old man with a recessive form of ocular coloboma. By comparing these genetic data against equivalent genome sequences from his healthy parents, Jin’s team identified a gene called IPO13 as the culprit. IPO13 has not been linked to human disease before, but the researchers demonstrated that switching off IPO13 expression in zebrafish embryos gave rise to underdeveloped eyes with defects in the iris and cornea. These findings give clinicians another potential indicator for early diagnosis of ocular coloboma.
Collapse
|
700
|
Chakravorty S, Hegde M. Inferring the effect of genomic variation in the new era of genomics. Hum Mutat 2018; 39:756-773. [PMID: 29633501 DOI: 10.1002/humu.23427] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/20/2018] [Accepted: 03/28/2018] [Indexed: 12/11/2022]
Abstract
Accurate and detailed understanding of the effects of variants in the coding and noncoding regions of the genome is the next big challenge in the new genomic era of personalized medicine, especially to tackle newer findings of genetic and phenotypic heterogeneity of diseases. This is necessary to resolve the gene-variant-disease relationship, the pathogenic variant spectrum of genes, pathogenic variants with variable clinical consequences, and multiloci diseases. In turn, this will facilitate patient recruitment for relevant clinical trials. In this review, we describe the trends in research at the intersection of basic and clinical genomics aiming to (a) overcome molecular diagnostic challenges and increase the clinical utility of next-generation sequencing (NGS) platforms, (b) elucidate variants associated with disease, (c) determine overall genomic complexity including epistasis, complex inheritance patterns such as "synergistic heterozygosity," digenic/multigenic inheritance, modifier effect, and rare variant load. We describe the newly emerging field of integrated functional genomics, in vivo or in vitro large-scale functional approaches, statistical bioinformatics algorithms that support NGS genomics data to interpret variants for timely clinical diagnostics and disease management. Thus, facilitating the discovery of new therapeutic or biomarker options, and their roles in the future of personalized medicine.
Collapse
Affiliation(s)
- Samya Chakravorty
- Department of Human Genetics, Emory University School of Medicine, Whitehead Biomedical Research Building Suite 301, Atlanta, Georgia
| | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine, Whitehead Biomedical Research Building Suite 301, Atlanta, Georgia
| |
Collapse
|