201
|
Zou S, Wang X, Chen H, Lin J, Wen C, Zhan Q, Chen H, Lu X, Deng X, Shen B. Postoperative hyperprogression disease of pancreatic ductal adenocarcinoma after curative resection: a retrospective cohort study. BMC Cancer 2022; 22:649. [PMID: 35698045 PMCID: PMC9190100 DOI: 10.1186/s12885-022-09719-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prognosis for patients recurred rapidly after resection of pancreatic ductal adenocarcinoma (PDAC) was extremely poor. We proposed the concept of postoperative hyper-progression disease (PO-HPD) to define recurrence within 2 months after surgery, explored the role of surgery for postoperative HPD patients and determined the predictive preoperative risk factors and genomic features of PO-HPD. METHODS 976 patients undergoing curative resection of PDAC were enrolled. Survival data of 1733 stage IV patients from the US Surveillance, Epidemiology and End Results database was also collected. Patients relapsed were grouped into 3 groups regarding of the recurrence time (within 2 months were PO-HPD, within 2 to 12 months were early recurrence (ER) and within > 12 months were late recurrence (LR)). Risk factors for PO-HPD were explored with logistic regression models. Genomic features of 113 patients were investigated using next-generation sequencing-based gene panel testing. RESULTS 718 of 976 cases relapsed, 101were PO-HPD, 418 were ER and 199 were LR. Total survival of PO-HPD was 12.5 months, shorter than that of ER (16.7 months) and LR (35.1 months), and verged on that of stage IV patients (10.6 months). Preoperative risk factors for PO-HPD included red blood cell count < 3.94*10^12/L, CA19-9 ≥ 288.6 U/mL, CA125 ≥ 22.3 U/mL and tumor size≥3.45 cm. Mutations of CEBPA, ATR and JAK1 were only identified in PO-HPD and they owned lower level of CN gain compared to others. CONCLUSIONS Prognosis of PO-HPD was extremely poor and the role of surgery for PO-HPD should be prudently assessed.
Collapse
Affiliation(s)
- Siyi Zou
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
| | - Xinjing Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Haoda Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
| | - Jiewei Lin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
| | - Chenlei Wen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Qian Zhan
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Xiongxiong Lu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China.
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China.
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China.
- Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, 200025, China.
| |
Collapse
|
202
|
Gao Y, Wu D, Chen B, Chen Y, Zhang Q, Zhao P. Rare Variants in Novel Candidate Genes Associated With Nonsyndromic Patent Ductus Arteriosus Identified With Whole-Exome Sequencing. Front Genet 2022; 13:921925. [PMID: 35734438 PMCID: PMC9207465 DOI: 10.3389/fgene.2022.921925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Patent ductus arteriosus (PDA) is one of the most common congenital heart defects causing pulmonary hypertension, infective endocarditis, and even death. The important role of genetics in determining spontaneous ductal closure has been well-established. However, as many of the identified variants are rare, thorough identification of the associated genetic factors is necessary to further explore the genetic etiology of PDA. Methods: We performed whole-exome sequencing (WES) on 39 isolated nonsyndromic PDA patients and 100 healthy controls. Rare variants and novel genes were identified through bioinformatic filtering strategies. The expression patterns of candidate genes were explored in human embryo heart samples. Results: Eighteen rare damaging variants of six novel PDA-associated genes (SOX8, NES, CDH2, ANK3, EIF4G1, and HIPK1) were newly identified, which were highly expressed in human embryo hearts. Conclusions: WES is an efficient diagnostic tool for exploring the genetic pathogenesis of PDA. These findings contribute new insights into the molecular basis of PDA and may inform further studies on genetic risk factors for congenital heart defects.
Collapse
Affiliation(s)
- Ying Gao
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Dan Wu
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Bo Chen
- Department of Cardiothoracic Surgery, School of Medicine, Heart Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinghui Chen
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Zhang
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pengjun Zhao
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Pengjun Zhao,
| |
Collapse
|
203
|
A novel missense variant in the LMNB2 gene causes progressive myoclonus epilepsy. Acta Neurol Belg 2022; 122:659-667. [PMID: 33783721 DOI: 10.1007/s13760-021-01650-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022]
Abstract
Progressive myoclonus epilepsies (PMEs) are a group of disorders embracing myoclonus, seizures, and neurological dysfunctions. Because of the genetic and clinical heterogeneity, a large proportion of PMEs cases have remained molecularly undiagnosed. The present study aimed to determine the underlying genetic factors that contribute to the PME phenotype in an Iranian female patient. We describe a consanguineous Iranian family with autosomal recessive PME that had remained undiagnosed despite extensive genetic and pathological tests. After performing neuroimaging and clinical examinations, due to heterogeneity of PMEs, the proband was subjected to paired-end whole-exome sequencing and the candidate variant was confirmed by Sanger sequencing. Various in-silico tools were also used to predict the pathogenicity of the variant. In this study, we identified a novel homozygous missense variant (NM_032737.4:c.472C > T; p.(Arg158Trp)) in the LMNB2 gene (OMIM: 150341) as the most likely disease-causing variant. Neuroimaging revealed a progressive significant generalized atrophy in the cerebral and cerebellum without significant white matter signal changes. Video-electroencephalography monitoring showed a generalized pattern of high-voltage sharp waves in addition to multifocal spikes and waves compatible with mixed type seizures and epileptic encephalopathic pattern. Herein, we introduce the second case of PME caused by a novel variant in the LMNB2 gene. This study also underscores the potentiality of next-generation sequencing in the genetic diagnosis of patients with neurologic diseases with an unknown cause.
Collapse
|
204
|
Livesey BJ, Marsh JA. Interpreting protein variant effects with computational predictors and deep mutational scanning. Dis Model Mech 2022; 15:275742. [PMID: 35736673 PMCID: PMC9235876 DOI: 10.1242/dmm.049510] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Computational predictors of genetic variant effect have advanced rapidly in recent years. These programs provide clinical and research laboratories with a rapid and scalable method to assess the likely impacts of novel variants. However, it can be difficult to know to what extent we can trust their results. To benchmark their performance, predictors are often tested against large datasets of known pathogenic and benign variants. These benchmarking data may overlap with the data used to train some supervised predictors, which leads to data re-use or circularity, resulting in inflated performance estimates for those predictors. Furthermore, new predictors are usually found by their authors to be superior to all previous predictors, which suggests some degree of computational bias in their benchmarking. Large-scale functional assays known as deep mutational scans provide one possible solution to this problem, providing independent datasets of variant effect measurements. In this Review, we discuss some of the key advances in predictor methodology, current benchmarking strategies and how data derived from deep mutational scans can be used to overcome the issue of data circularity. We also discuss the ability of such functional assays to directly predict clinical impacts of mutations and how this might affect the future need for variant effect predictors.
Collapse
Affiliation(s)
- Benjamin J Livesey
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
205
|
Liu Y, Qu HQ, Chang X, Qu J, Mentch FD, Nguyen K, Tian L, Glessner J, Sleiman PMA, Hakonarson H. Mutation Burden Analysis of Six Common Mental Disorders in African Americans by Whole Genome Sequencing. Hum Mol Genet 2022; 31:3769-3776. [PMID: 35642741 DOI: 10.1093/hmg/ddac129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/12/2022] [Accepted: 05/27/2022] [Indexed: 11/14/2022] Open
Abstract
Mental disorders present a global health concern, with limited treatment options. In today's medical practice, medications such as antidepressants are prescribed not only for depression, but also for conditions such as anxiety and attention deficit hyperactivity disorder (ADHD). Therefore, identifying gene targets for specific disorders is important and offers improved precision. In this study, we performed a genetic analysis of six common mental disorders, ADHD, anxiety, depression, delays in mental developments, intellectual disabilities (ID), and speech/language disorder in the ethnic minority of African Americans (AA) using whole genome sequencing (WGS). WGS data was generated from blood-derived DNA from 4178 AA individuals, including 1384 patients with the diagnosis of at least one mental disorder. Mutation burden analysis was applied based on rare and deleterious mutations in the AA population between cases and controls, and further analyzed in the context of patients with single mental disorder diagnosis. Certain genes uncovered demonstrated significant p values in mutation burden analysis. In addition, exclusive recurrences in specific type of disorder were scanned through gene-drug interaction databases to assess for availability of potential medications. We uncovered 15 genes harboring deleterious mutations, including HMGCR and UST for ADHD; FNTB for anxiety, XIRP2, NPPC, , STK33, PANX1 and NTS for depression; RUNX3, TACR1, and NDUFS7 for delays in mental developments; HPN for ID; COL6A3, DDB1, and NDUFA11 for speech/language disorder. Taken together, we have established critical insight into the development of new precision medicine approaches for mental disorders in African Americans.
Collapse
Affiliation(s)
- Yichuan Liu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Hui-Qi Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Xiao Chang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Jingchun Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Frank D Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Kenny Nguyen
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Lifeng Tian
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Joseph Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Patrick M A Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| |
Collapse
|
206
|
Next generation sequencing for diagnosis of hereditary anemia: Experience in a Spanish reference center. Clin Chim Acta 2022; 531:112-119. [DOI: 10.1016/j.cca.2022.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/19/2022]
|
207
|
Steyaert W, Varney MJ, Benovic JL, Creemers J, Speeckaert MM, Coucke PJ, Delanghe JR. Hypergastrinemia, a clue leading to the identification of an atypical form of diabetes mellitus type 2. Clin Chim Acta 2022; 532:79-83. [PMID: 35623402 DOI: 10.1016/j.cca.2022.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND A hitherto undescribed form of diabetes mellitus type 2 is reported in a Flemish family. In these patients, markedly elevated gastrin levels were observed, which could not be linked to gastrointestinal symptoms. MATERIALS AND METHODS Gel permeation chromatography was performed for gastrin, insulin, and proinsulin. Proprotein convertase subtilisin/kexin type (PCSK1 and PCSK2)] were sequenced. Whole-exome sequencing was performed on the genomic DNA extracted from leukocytes of the proband of the family. RESULTS Gel permeation chromatography revealed that the apparent hypergastrinemia was caused by the accumulation of biologically inactive progastrin. Besides, high serum concentrations of proinsulin and intact fibroblast growth factor 23 (FGF23) were also detected. Sequencing of PCSK1 and PCSK2 genes did not reveal any mutations in these genes. Whole exome sequencing revealed a c.1150C>T (p.Pro384Ser) mutation in G protein-coupled receptor kinase 6 (GRK6), which cosegregated with the disease. Expression of the mutant enzyme in mammalian cells revealed that it was mislocalized compared to the wild-type GRK6. CONCLUSIONS In the affected patients, prohormone processing is impaired likely due to the altered function of mutant GRK6. Delayed pro-insulin processing causes hypoglycaemia episodes a couple of hours following meals. In addition, increased plasma concentrations of progastrin and intact FGF23 in the affected individuals can be explained by incomplete processing of the precursor hormones.
Collapse
Affiliation(s)
- Wouter Steyaert
- Department of Genetics, Ghent University, Ghent, Belgium; Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Matthew J Varney
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - John Creemers
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Marijn M Speeckaert
- Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium
| | - Paul J Coucke
- Department of Genetics, Ghent University, Ghent, Belgium
| | - Joris R Delanghe
- Department of Clinical Chemistry, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
208
|
Novel role of the synaptic scaffold protein Dlgap4 in ventricular surface integrity and neuronal migration during cortical development. Nat Commun 2022; 13:2746. [PMID: 35585091 PMCID: PMC9117333 DOI: 10.1038/s41467-022-30443-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/29/2022] [Indexed: 11/08/2022] Open
Abstract
Subcortical heterotopias are malformations associated with epilepsy and intellectual disability, characterized by the presence of ectopic neurons in the white matter. Mouse and human heterotopia mutations were identified in the microtubule-binding protein Echinoderm microtubule-associated protein-like 1, EML1. Further exploring pathological mechanisms, we identified a patient with an EML1-like phenotype and a novel genetic variation in DLGAP4. The protein belongs to a membrane-associated guanylate kinase family known to function in glutamate synapses. We showed that DLGAP4 is strongly expressed in the mouse ventricular zone (VZ) from early corticogenesis, and interacts with key VZ proteins including EML1. In utero electroporation of Dlgap4 knockdown (KD) and overexpression constructs revealed a ventricular surface phenotype including changes in progenitor cell dynamics, morphology, proliferation and neuronal migration defects. The Dlgap4 KD phenotype was rescued by wild-type but not mutant DLGAP4. Dlgap4 is required for the organization of radial glial cell adherens junction components and actin cytoskeleton dynamics at the apical domain, as well as during neuronal migration. Finally, Dlgap4 heterozygous knockout (KO) mice also show developmental defects in the dorsal telencephalon. We hence identify a synapse-related scaffold protein with pleiotropic functions, influencing the integrity of the developing cerebral cortex.
Collapse
|
209
|
Yaung SJ, Woestmann C, Ju C, Ma XM, Gattam S, Zhou Y, Xi L, Pal S, Balasubramanyam A, Tikoo N, Heussel CP, Thomas M, Kriegsmann M, Meister M, Schneider MA, Herth FJ, Wehnl B, Diehn M, Alizadeh AA, Palma JF, Muley T. Early Assessment of Chemotherapy Response in Advanced Non-Small Cell Lung Cancer with Circulating Tumor DNA. Cancers (Basel) 2022; 14:cancers14102479. [PMID: 35626082 PMCID: PMC9139958 DOI: 10.3390/cancers14102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 11/16/2022] Open
Abstract
Monitoring treatment efficacy early during therapy could enable a change in treatment to improve patient outcomes. We report an early assessment of response to treatment in advanced NSCLC using a plasma-only strategy to measure changes in ctDNA levels after one cycle of chemotherapy. Plasma samples were collected from 92 patients with Stage IIIB-IV NSCLC treated with first-line chemo- or chemoradiation therapies in an observational, prospective study. Retrospective ctDNA analysis was performed using next-generation sequencing with a targeted 198-kb panel designed for lung cancer surveillance and monitoring. We assessed whether changes in ctDNA levels after one or two cycles of treatment were associated with clinical outcomes. Subjects with ≤50% decrease in ctDNA level after one cycle of chemotherapy had a lower 6-month progression-free survival rate (33% vs. 58%, HR 2.3, 95% CI 1.2 to 4.2, log-rank p = 0.009) and a lower 12-month overall survival rate (25% vs. 70%, HR 4.3, 95% CI 2.2 to 9.7, log-rank p < 0.001). Subjects with ≤50% decrease in ctDNA level after two cycles of chemotherapy also had shorter survival. Using non-invasive liquid biopsies to measure early changes in ctDNA levels in response to chemotherapy may help identify non-responders before standard-of-care imaging in advanced NSCLC.
Collapse
Affiliation(s)
- Stephanie J. Yaung
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
- Correspondence: ; Tel.: +1-925-523-8824
| | | | - Christine Ju
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Xiaoju Max Ma
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
| | - Sandeep Gattam
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Yiyong Zhou
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Liu Xi
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
| | - Subrata Pal
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Aarthi Balasubramanyam
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Nalin Tikoo
- Alector, Inc., South San Francisco, CA 94080, USA;
| | - Claus Peter Heussel
- Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik, University Hospital, 69126 Heidelberg, Germany;
- Diagnostic and Interventional Radiology, University Hospital, 69120 Heidelberg, Germany
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
| | - Michael Thomas
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Michael Meister
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Marc A. Schneider
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Felix J. Herth
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Birgit Wehnl
- Roche Diagnostics GmbH, 82377 Penzberg, Germany;
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.A.A.)
| | - Ash A. Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.A.A.)
| | - John F. Palma
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
| | - Thomas Muley
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| |
Collapse
|
210
|
Gan M, Zhang C, Qiu L, Wang Y, Bao H, Yu R, Liu R, Wu X, Shao Y, Hou P, Fei Z. Molecular landscape and therapeutic alterations in Asian soft-tissue sarcoma patients. Cancer Med 2022; 11:4070-4078. [PMID: 35586877 PMCID: PMC9636498 DOI: 10.1002/cam4.4725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/27/2022] [Accepted: 03/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background Soft‐tissue sarcoma (STS) is a rare solid malignant tumor with numerous histologic subtypes. Current studies on targeted therapy for STS are in preclinical and early‐phase trials. Genomic differences largely influence the prognosis of patients even with the same subtype. To investigate the genomic alterations (GAs) and the potential of targeted therapy in STS, we analyzed the genomic landscape, the therapeutic GAs, and biomarkers of immunotherapy in Chinese STS patients. Methods Targeted sequencing covering 425 genes was performed, from which we obtained the results of tissue samples from 351 Chinese STS patients of all ages covering different histologic subtypes. Bioinformatics analysis of altered genes with nonsynonymous mutations, copy‐number variations, and gene fusions were performed. OncoKB therapeutic GAs and relevant biomarkers including TMB, MSI, and HRD were further examined for potential targeted therapy. Results In total, 2743 GAs were identified in 330 genes with a median of 6 (1–38) per case. The top 11 frequently altered genes were: TP53, MCL1, MDM2, CDK4, MYC, CDKN2A, GNAS, RB1, ATRX, CDKN2B, and FGFR1. OncoKB defined therapeutic GAs were found in 23 genes in 43% of the patients. In general, 9.4% of the patients had high‐TMB, 2.8% had MSI, and 13.7% had HRD. A significant difference in the percentage of patients with OncoKB therapeutic GAs were observed between the most frequent two subtypes, leiomyosarcoma and liposarcoma. Altogether, 54% of the patients had the potential to respond to a targeted therapy. Conclusion This study indicated the potential efficacy of targeted therapy on many STS patients, and also provided insight for novel precision therapy. The clinical efficacy of combining targeted therapy and immunotherapy can be further investigated.
Collapse
Affiliation(s)
- Meifu Gan
- Department of PathologyTaizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical UniversityLinhaiChina
| | - Chen Zhang
- Radiotherapy & Chemotherapy Department 2, HwaMei HospitalUniversity of Chinese Academy of ScienceNingboChina
| | - Liqing Qiu
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer HospitalZhejiang University School of MedicineHangzhouChina
| | - Yue Wang
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Hua Bao
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Ruoying Yu
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Rui Liu
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Xue Wu
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Yang Shao
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
- School of Public HealthNanjing Medical UniversityNanjingChina
| | - Peifeng Hou
- Department of Medical OncologyFujian Medical University Union HospitalFuzhouChina
| | - Zhenglei Fei
- Anorectal SurgeryNingbo Medical Center Lihuili HospitalNingBoChina
| |
Collapse
|
211
|
Dai H, Wei Y, Liu Y, Liu J, Yu R, Zhang J, Pang J, Shao Y, Li Q, Yang Z. Pathway-Based Analysis Revealed the Role of Keap1-Nrf2 Pathway and PI3K-Akt Pathway in Chinese Esophageal Squamous Cell Carcinoma Patients With Definitive Chemoradiotherapy. Front Genet 2022; 12:799663. [PMID: 35548450 PMCID: PMC9081370 DOI: 10.3389/fgene.2021.799663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/13/2021] [Indexed: 12/30/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the major type of EC in China. Chemoradiotherapy is a standard definitive treatment for early-stage EC and significantly improves local control and overall survival for late-stage patients. However, chemoradiotherapy resistance, which limits therapeutic efficacy and treatment-induced toxicity, is still a leading problem for treatment break. To optimize the selection of ESCC patients for chemoradiotherapy, we retrospectively analyzed the clinical features and genome landscape of a Chinese ESCC cohort of 58 patients. TP53 was the most frequent mutation gene, followed by NOTCH1. Frequently, copy number variants were found in MCL1 (24/58, 41.4%), FGF19 (23/58, 39.7%), CCND1 (22/58, 37.9%), and MYC (20/58, 34.5%). YAP1 and SOX2 amplifications were mutually exclusive in this cohort. Using univariate and multivariate analyses, the YAP1 variant and BRIP1 mutant were identified as adverse factors for OS. Patients with PI3K-Akt pathway alterations displayed longer PFS and OS than patients with an intact PI3K-Akt pathway. On the contrary, two patients with Keap1-Nrf2 pathway alterations displayed significantly shortened PFS and OS, which may be associated with dCRT resistance. Our data highlighted the prognostic value of aberrant cancer pathways in ESCC patients, which may provide guidance for better chemoradiotherapy management.
Collapse
Affiliation(s)
- Honghai Dai
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yanjun Wei
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yunxia Liu
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingwen Liu
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Ruoying Yu
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Junli Zhang
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | | | - Yang Shao
- Nanjing Geneseeq Technology Inc, Nanjing, China.,School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiang Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhe Yang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
212
|
He J, Hu X, Chen L, Liu Q, Jiang Y. Characteristics of Genomic Alterations in Pericardial Effusion of Advanced Non-small Cell Lung Cancer. Front Genet 2022; 13:850290. [PMID: 35646096 PMCID: PMC9133843 DOI: 10.3389/fgene.2022.850290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The feasibility and value of pericardial effusion as a liquid biopsy sample for actionable alteration detection in patients with non-small cell lung cancer (NSCLC) has not been adequately investigated. Here, we aim to reveal genomic alterations between pericardial effusion and paired tumor tissue, plasma (plasma cfDNA), and pleural effusion supernatant (PE-cfDNA) based on second-generation sequencing technology. Material and methods: A total of 26 advanced NSCLC patients were retrospectively studied. The following samples were collected and sequenced using two targeted next-generation sequencing panels: pericardial effusion (n = 26), matched tumor tissue (n = 6), plasma (n = 16), and pleural effusion supernatant (n = 5). Results: A total of 10 actionable alterations were identified in pericardial effusion of the NSCLC patients, including MET amplification, EGFR L858R, EGFR T790M, EGFR exon 19 deletion, EGFR L861Q, KRAS G12C, EML4-ALK (exon 18: exon 20) fusion, EML4-ALK (exon 20: exon 20) fusion, EML4-ALK (exon 6: exon 20) fusion, and ERBB2 exon 20 insertion. All these actionable alterations harbored multiple drug-sensitive targets as well as several drug-resistant targets, such as EGFR T790M. Compared to plasma cfDNA of 16 patients, paired pericardial effusion had higher number of actionable alterations (p = 0.08) as well as higher percentage of the population with actionable alterations (p = 0.16). Moreover, 8 out of 10 actionable alterations with single nucleotide variations (SNVs) or insertions/deletions (indels) had a higher variant allele frequency (VAF) in pericardial effusion than plasma cfDNA. In addition, we identified two actionable alterations in paired pericardial effusion, which were absence in PE-cfDNA. Clearly, 2 out of 3 actionable alterations with SNVs/indels in pericardial effusion had a higher VAF than those in PE-cfDNA. Our finding suggested the importance of pericardial effusion in the optimal selection of patients for targeted therapy. Conclusion: Among liquid biopsy specimens from the advanced NSCLC patients, pericardial effusion may be a better candidate for genomic profiling than plasma cfDNA, while it could serve as a supplement to PE-cfDNA in detecting actionable alterations. Therefore, pericardial effusion might provide a new alternative for selection of patients for better treatment management.
Collapse
Affiliation(s)
- Jiaxue He
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Xintong Hu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Liguo Chen
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Qiaoliang Liu
- College of Communication Engineering, Jilin University, Changchun, China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Yanfang Jiang,
| |
Collapse
|
213
|
Scrutinizing Deleterious Nonsynonymous SNPs and Their Effect on Human POLD1 Gene. Genet Res (Camb) 2022; 2022:1740768. [PMID: 35620275 PMCID: PMC9117041 DOI: 10.1155/2022/1740768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/17/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
POLD1 (DNA polymerase delta 1, catalytic subunit) is a protein-coding gene that encodes the large catalytic subunit (POLD1/p125) of the DNA polymerase delta (Polδ) complex. The consequence of missense or nonsynonymous SNPs (nsSNPs), which occur in the coding region of a specific gene, is the replacement of single amino acid. It may also change the structure, stability, and/or functions of the protein. Mutation in the POLD1 gene is associated with autosomal dominant predisposition to colonic adenomatous polyps, colon cancer, endometrial cancer (EDMC), breast cancer, and brain tumors. These de novo mutations in the POLD1 gene also result in autosomal dominant MDPL syndrome (mandibular hypoplasia, deafness, progeroid features, and lipodystrophy). In this study, genetic variations of POLD1 which may affect the structure and/or function were analyzed using different types of bioinformatics tools. A total of 17038 nsSNPs for POLD1 were collected from the NCBI database, among which 1317 were missense variants. Out of all missense nsSNPs, 28 were found to be deleterious functionally and structurally. Among these deleterious nsSNPs, 23 showed a conservation scale of >5, 2 were predicted to be associated with binding site formation, and one acted as a posttranslational modification site. All of them were involved in coil, extracellular structures, or helix formation, and some cause the change in size, charge, and hydrophobicity.
Collapse
|
214
|
Liu Y, Qu HQ, Qu J, Chang X, Mentch FD, Nguyen K, Tian L, Glessner J, Sleiman PMA, Hakonarson H. Burden of rare coding variants reveals genetic heterogeneity between obese and non-obese asthma patients in the African American population. Respir Res 2022; 23:116. [PMID: 35524249 PMCID: PMC9078008 DOI: 10.1186/s12931-022-02039-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
Background Asthma is a complex condition largely attributed to the interactions among genes and environments as a heterogeneous phenotype. Obesity is significantly associated with asthma development, and genetic studies on obese vs. non-obese asthma are warranted. Methods To investigate asthma in the minority African American (AA) population with or without obesity, we performed a whole genome sequencing (WGS) study on blood-derived DNA of 4289 AA individuals, included 2226 asthma patients (1364 with obesity and 862 without obesity) and 2006 controls without asthma. The burden analysis of functional rare coding variants was performed by comparing asthma vs. controls and by stratified analysis of obese vs. non-obese asthma, respectively. Results Among the top 66 genes with P < 0.01 in the asthma vs. control analysis, stratified analysis by obesity showed inverse correlation of natural logarithm (LN) of P value between obese and non-obese asthma (r = − 0.757, P = 1.90E−13). Five genes previously reported in a genome-wide association study (GWAS) on asthma, including TSLP, SLC9A4, PSMB8, IGSF5, and IKZF4 were demonstrated association in the asthma vs. control analysis. The associations of IKZF4 and IGSF5 are only associated with obese asthma; and the association of SLC9A4 is only observed in non-obese asthma. In addition, the association of RSPH3 (the gene is related to primary ciliary dyskinesia) is observed in non-obese asthma. Conclusions These findings highlight genetic heterogeneity between obese and non-obese asthma in patients of AA ancestry. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02039-0.
Collapse
Affiliation(s)
- Yichuan Liu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Hui-Qi Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jingchun Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Xiao Chang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Frank D Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Kenny Nguyen
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Lifeng Tian
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Joseph Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Patrick M A Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA. .,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA. .,Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA. .,Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| |
Collapse
|
215
|
Dang LT, Terrell JR, Konia T, Fung MA, McPherson JD, Kiuru M. Characteristics of amelanotic acantholytic-like melanoma resembling squamous cell carcinoma. J Cutan Pathol 2022; 49:500-503. [PMID: 35118708 PMCID: PMC9473664 DOI: 10.1111/cup.14210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 01/26/2023]
Affiliation(s)
- Luke T. Dang
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA
| | - Jessica R. Terrell
- Department of Dermatology, University of California, Davis, Sacramento, CA
| | - Thomas Konia
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA,Department of Dermatology, University of California, Davis, Sacramento, CA
| | - Maxwell A. Fung
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA,Department of Dermatology, University of California, Davis, Sacramento, CA
| | - John D. McPherson
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA
| | - Maija Kiuru
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA,Department of Dermatology, University of California, Davis, Sacramento, CA
| |
Collapse
|
216
|
Narang A, Lacaze P, Ronaldson KJ, McNeil JJ, Jayaram M, Thomas N, Sellmer R, Crockford DN, Stowe R, Greenway SC, Pantelis C, Bousman CA. Whole-genome sequencing analysis of clozapine-induced myocarditis. THE PHARMACOGENOMICS JOURNAL 2022; 22:173-179. [PMID: 35461379 DOI: 10.1038/s41397-022-00271-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 06/14/2023]
Abstract
One of the concerns limiting the use of clozapine in schizophrenia treatment is the risk of rare but potentially fatal myocarditis. Our previous genome-wide association study and human leucocyte antigen analyses identified putative loci associated with clozapine-induced myocarditis. However, the contribution of DNA variation in cytochrome P450 genes, copy number variants and rare deleterious variants have not been investigated. We explored these unexplored classes of DNA variation using whole-genome sequencing data from 25 cases with clozapine-induced myocarditis and 25 demographically-matched clozapine-tolerant control subjects. We identified 15 genes based on rare variant gene-burden analysis (MLLT6, CADPS, TACC2, L3MBTL4, NPY, SLC25A21, PARVB, GPR179, ACAD9, NOL8, C5orf33, FAM127A, AFDN, SLC6A11, PXDN) nominally associated (p < 0.05) with clozapine-induced myocarditis. Of these genes, 13 were expressed in human myocardial tissue. Although independent replication of these findings is required, our study provides preliminary insights into the potential role of rare genetic variants in susceptibility to clozapine-induced myocarditis.
Collapse
Affiliation(s)
- Ankita Narang
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada
| | - Paul Lacaze
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Kathlyn J Ronaldson
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - John J McNeil
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Mahesh Jayaram
- Adult Mental Health Rehabilitation Unit, North Western Mental Health, Melbourne Health, Western Health, Sunshine Hospital, St Albans, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Western Centre for Health & Education, St Albans, VIC, Australia
| | - Naveen Thomas
- Adult Mental Health Rehabilitation Unit, North Western Mental Health, Melbourne Health, Western Health, Sunshine Hospital, St Albans, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Western Centre for Health & Education, St Albans, VIC, Australia
| | - Rory Sellmer
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - David N Crockford
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Robert Stowe
- Departments of Psychiatry and Neurology (Medicine), Neuropsychiatry Program, and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Steven C Greenway
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christos Pantelis
- Adult Mental Health Rehabilitation Unit, North Western Mental Health, Melbourne Health, Western Health, Sunshine Hospital, St Albans, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Western Centre for Health & Education, St Albans, VIC, Australia
| | - Chad A Bousman
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada.
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
217
|
Dai H, Wei Y, Liu Y, Liu J, Yu R, Zhang J, Pang J, Shao Y, Li Q, Yang Z. Pathway-Based Analysis Revealed the Role of Keap1-Nrf2 Pathway and PI3K-Akt Pathway in Chinese Esophageal Squamous Cell Carcinoma Patients With Definitive Chemoradiotherapy. Front Genet 2022; 12:799663. [PMID: 35548450 DOI: 10.3389/fgene.2021.625867/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/13/2021] [Indexed: 05/26/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the major type of EC in China. Chemoradiotherapy is a standard definitive treatment for early-stage EC and significantly improves local control and overall survival for late-stage patients. However, chemoradiotherapy resistance, which limits therapeutic efficacy and treatment-induced toxicity, is still a leading problem for treatment break. To optimize the selection of ESCC patients for chemoradiotherapy, we retrospectively analyzed the clinical features and genome landscape of a Chinese ESCC cohort of 58 patients. TP53 was the most frequent mutation gene, followed by NOTCH1. Frequently, copy number variants were found in MCL1 (24/58, 41.4%), FGF19 (23/58, 39.7%), CCND1 (22/58, 37.9%), and MYC (20/58, 34.5%). YAP1 and SOX2 amplifications were mutually exclusive in this cohort. Using univariate and multivariate analyses, the YAP1 variant and BRIP1 mutant were identified as adverse factors for OS. Patients with PI3K-Akt pathway alterations displayed longer PFS and OS than patients with an intact PI3K-Akt pathway. On the contrary, two patients with Keap1-Nrf2 pathway alterations displayed significantly shortened PFS and OS, which may be associated with dCRT resistance. Our data highlighted the prognostic value of aberrant cancer pathways in ESCC patients, which may provide guidance for better chemoradiotherapy management.
Collapse
Affiliation(s)
- Honghai Dai
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yanjun Wei
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yunxia Liu
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingwen Liu
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Ruoying Yu
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Junli Zhang
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | | | - Yang Shao
- Nanjing Geneseeq Technology Inc, Nanjing, China
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiang Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhe Yang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
218
|
Dap M, Gicquel F, Lambert L, Perdriolle‐Galet E, Bonnet C, Morel O. Utility of Chromosomal Microarray Analysis for the exploration of isolated and severe fetal growth restriction diagnosed before 24 weeks’ gestation. Prenat Diagn 2022; 42:1281-1287. [DOI: 10.1002/pd.6149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Matthieu Dap
- Obstetrics and Fetal medicine Unit CHRU of Nancy F‐54000 Nancy France
- Department of fetopathology and placental pathology CHRU of Nancy F‐54000 Nancy France
- Université de Lorraine Inserm IADI F‐54000 Nancy France
| | - Fanny Gicquel
- Obstetrics and Fetal medicine Unit CHRU of Nancy F‐54000 Nancy France
| | - Laetitia Lambert
- Department of Medical Genetics CHRU of Nancy Nancy FranceF‐54000
- INSERM UMRS 1256 NGERE Nutrition, Genetics, and Environmental Risk Exposure Université de Lorraine F‐54000 Nancy France
| | | | - Céline Bonnet
- INSERM UMRS 1256 NGERE Nutrition, Genetics, and Environmental Risk Exposure Université de Lorraine F‐54000 Nancy France
- Genetics Laboratory CHRU of Nancy F‐54000 Nancy France
| | - Olivier Morel
- Obstetrics and Fetal medicine Unit CHRU of Nancy F‐54000 Nancy France
- Université de Lorraine Inserm IADI F‐54000 Nancy France
| |
Collapse
|
219
|
Nguyen K, Alsaati N, Le Coz C, Romberg N. Genetic obstacles to developing and tolerizing human B cells. WIREs Mech Dis 2022; 14:e1554. [DOI: 10.1002/wsbm.1554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Kim Nguyen
- Division of Immunology and Allergy Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Nouf Alsaati
- Division of Immunology and Allergy Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Carole Le Coz
- Division of Immunology and Allergy Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Neil Romberg
- Division of Immunology and Allergy Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
- Department of Pediatrics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
- Institute for Immunology University of Pennsylvania Philadelphia Pennsylvania USA
| |
Collapse
|
220
|
Disease Modeling of Rare Neurological Disorders in Zebrafish. Int J Mol Sci 2022; 23:ijms23073946. [PMID: 35409306 PMCID: PMC9000079 DOI: 10.3390/ijms23073946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023] Open
Abstract
Rare diseases are those which affect a small number of people compared to the general population. However, many patients with a rare disease remain undiagnosed, and a large majority of rare diseases still have no form of viable treatment. Approximately 40% of rare diseases include neurologic and neurodevelopmental disorders. In order to understand the characteristics of rare neurological disorders and identify causative genes, various model organisms have been utilized extensively. In this review, the characteristics of model organisms, such as roundworms, fruit flies, and zebrafish, are examined, with an emphasis on zebrafish disease modeling in rare neurological disorders.
Collapse
|
221
|
Takai E, Nakamura H, Chiku S, Kubo E, Ohmoto A, Totoki Y, Shibata T, Higuchi R, Yamamoto M, Furuse J, Shimizu K, Takahashi H, Morizane C, Furukawa T, Yachida S. Whole-exome Sequencing Reveals New Potential Susceptibility Genes for Japanese Familial Pancreatic Cancer. Ann Surg 2022; 275:e652-e658. [PMID: 32826389 DOI: 10.1097/sla.0000000000004213] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The primary objective of this study was to identify novel genes that predispose people in the Japanese population to FPC. SUMMARY OF BACKGROUND DATA Familial history of pancreatic cancer is an important risk factor but, to date, few genes predisposing individuals to increased risk of developing FPC have been identified. METHODS We performed whole-exome sequencing of germline DNA from 81 Japanese FPC patients. We also investigated somatic gene alterations in 21 matched tumor tissues through whole-exome sequencing and copy number analysis. RESULTS Our germline variants identified previously known FPC susceptibility genes such as ATM and BRCA2, and several novel tumor suppressor genes with potentially deleterious variants for FPC. Interestingly, somatic whole-exome analysis demonstrated that most tumor samples with suspicious loss of heterozygosity of candidate genes were KRAS wild-types, implying that these cases may not have required KRAS activation as a driver event for carcinogenesis. CONCLUSIONS Our findings indicate that FPC patients harbor potentially deleterious causative germline variants in tumor suppressor genes, which are known to acquire somatic mutations in pancreatic cancer, and that somatic loss of heterozygosity of some FPC susceptibility genes may contribute to the development of FPC in the absence of somatic KRAS-activating mutation. Genetic testing for a wider variety of FPC-predisposition genes could provide better screening approach for high-risk groups of pancreatic cancer.
Collapse
Affiliation(s)
- Erina Takai
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiromi Nakamura
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Suenori Chiku
- Information and Communication Research Division, Mizuho Information and Research Institute, Tokyo, Japan
| | - Emi Kubo
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akihiro Ohmoto
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Totoki
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Ryota Higuchi
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masakazu Yamamoto
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Kyoko Shimizu
- Department of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hideaki Takahashi
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
222
|
Zhao J, Benton S, Zhang B, Olivares S, Asadbeigi S, Busam K, Gerami P. Benign and Intermediate-grade Melanocytic Tumors With BRAF Mutations and Spitzoid Morphology: A Subset of Melanocytic Neoplasms Distinct From Melanoma. Am J Surg Pathol 2022; 46:476-485. [PMID: 34753863 DOI: 10.1097/pas.0000000000001831] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The current classification of Spitz neoplasms in the World Health Organization (WHO), Fourth Edition defines Spitz neoplasms as melanocytic proliferations with characteristic Spitz morphology and a Spitz-associated genomic fusion or HRAS mutation. In contrast, melanocytic neoplasms with BRAF mutations are considered typical of common acquired nevi, dysplastic nevi, and melanomas from intermittent sun-damaged skin. However, increased utilization of ancillary testing methods such as BRAFV600E immunohistochemistry and sequencing studies have made apparent a subgroup of benign-grade and intermediate-grade melanocytic neoplasms with Spitzoid morphology that harbor BRAFV600E mutations. We refer to these cases as BRAF mutated and morphologically Spitzoid (BAMS) nevi and tumors. Two experienced dermatopathologists reviewed a series of 36 BAMS nevi/tumors. Cases in which a diagnosis of melanoma was favored were excluded. The histomorphologic, clinical, and molecular findings were assessed by immunohistochemistry, fluorescence in situ hybridization, and next-generation sequencing using validated gene panels. Characteristics of BAMS nevi/tumors were compared with a control set of Spitz tumors with previously reported fusion proteins. BAMS nevi/tumors had a decreased proportion of Kamino bodies (P=0.03) and a higher proportion of cytoplasmic pigmentation (P<0.00001). There were no differences in other morphologic features such as the silhouette, epidermal hyperplasia, pagetosis, and cytologic atypia compared with fusion-induced Spitz tumors. In 6/17 cases where next-generation sequencing studies were available, recurrent mutations in the KMT gene family were seen. This was higher than the proportion of such mutations seen in fusion Spitz tumors and lower than the frequency in cutaneous melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | - Klaus Busam
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Pedram Gerami
- Department of Dermatology
- Robert H. Lurie Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
223
|
Genome-wide compound heterozygote analysis highlights DPY19L2 alleles in a non-consanguineous Spanish family with a complete form of globozoospermia. Reprod Biomed Online 2022; 45:332-340. [DOI: 10.1016/j.rbmo.2022.03.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/18/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022]
|
224
|
Vanlallawma A, Lallawmzuali D, Pautu JL, Scaria V, Sivasubbu S, Kumar NS. Whole exome sequencing of pediatric leukemia reveals a novel InDel within FLT-3 gene in AML patient from Mizo tribal population, Northeast India. BMC Genom Data 2022; 23:23. [PMID: 35350997 PMCID: PMC8961913 DOI: 10.1186/s12863-022-01037-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background Leukemia is the most common type of cancer in pediatrics. Genomic mutations contribute towards the molecular mechanism of disease progression and also helps in diagnosis and prognosis. This is the first scientific mutational exploration in whole exome of pediatric leukemia patients from a cancer prone endogamous Mizo tribal population, Northeast India. Result Three non-synonymous exonic variants in NOTCH1 (p.V1699E), MUTYH (p.G143E) and PTPN11 (p.S502P) were found to be pathogenic. A novel in-frame insertion-deletion within the juxtamembrane domain of FLT3 (p.Tyr589_Tyr591delinsTrpAlaGlyAsp) was also observed. Conclusion These unique variants could have a potential mutational significance and these could be candidate genes in elucidating the possibility of predisposition to cancers within the population. This study merits further investigation for its role in diagnosis and prognosis and also suggests the need for population wide screening to identify unique mutations that might play a key role towards precision medicine. Supplementary Information The online version contains supplementary material available at 10.1186/s12863-022-01037-x.
Collapse
Affiliation(s)
- Andrew Vanlallawma
- Department of Biotechnology, Mizoram University, Aizawl, Mizoram, 796004, India
| | - Doris Lallawmzuali
- Department of Pathology, Mizoram State Cancer Institute, Zemabawk, Aizawl, Mizoram, 796017, India
| | - Jeremy L Pautu
- Department of Medical Oncology, Mizoram State Cancer Institute, Zemabawk, Aizawl, Mizoram, 796017, India
| | - Vinod Scaria
- CSIR - Institute of Genomics and Integrative Biology, South Campus, Mathura Road, New Delhi, 110025, India
| | - Sridhar Sivasubbu
- CSIR - Institute of Genomics and Integrative Biology, South Campus, Mathura Road, New Delhi, 110025, India
| | | |
Collapse
|
225
|
Inherited and De Novo Variation in Lithuanian Genomes: Introduction to the Analysis of the Generational Shift. Genes (Basel) 2022; 13:genes13040569. [PMID: 35456375 PMCID: PMC9028680 DOI: 10.3390/genes13040569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
Most genetic variants are rare and specific to the population, highlighting the importance of characterizing local population genetic diversity. Many countries have initiated population-based whole-genome sequencing (WGS) studies. Genomic variation within Lithuanian families are not available in the public databases. Here, we describe initial findings of a high-coverage (an average of 36.27×) whole genome sequencing for 25 trios of the Lithuanian population. Each genome on average carried approximately 4,701,473 (±28,255) variants, where 80.6% (3,787,626) were single nucleotide polymorphisms (SNPs), and the rest 19.4% were indels. An average of 12.45% was novel according to dbSNP (build 150). The WGS structural variation (SV) analysis identified on average 9133 (±85.10) SVs, of which 95.85% were novel. De novo single nucleotide variation (SNV) analysis identified 4417 variants, where 1.1% de novo SNVs were exonic, 43.9% intronic, 51.9% intergenic, and the rest 3.13% in UTR or downstream sequence. Three potential pathogenic de novo variants in the ZSWIM8, CDC42EP1, and RELA genes were identified. Our findings provide useful information on local human population genomic variation, especially for de novo variants, and will be a valuable resource for further genetic studies, and medical implications.
Collapse
|
226
|
A Type 3 Gaucher-Like Disease Due To Saposin C Deficiency in Two Emirati Families Caused by a Novel Splice Site Variant in the PSAP Gene. J Mol Neurosci 2022; 72:1322-1333. [DOI: 10.1007/s12031-022-01987-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/20/2022] [Indexed: 10/18/2022]
|
227
|
Lee W, de Prisco N, Gennarino VA. Identifying patients and assessing variant pathogenicity for an autosomal dominant disease-driving gene. STAR Protoc 2022; 3:101150. [PMID: 35146449 PMCID: PMC8819039 DOI: 10.1016/j.xpro.2022.101150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Identifying a disease gene and determining its causality in patients can be challenging. Here, we present an approach to predicting the pathogenicity of deletions and missense variants for an autosomal dominant gene. We provide online resources for identifying patients and determining constraint metrics to isolate the causal gene among several candidates encompassed in a shared region of deletion. We also provide instructions for optimizing functional annotation programs that may be otherwise inaccessible to a nonexpert or novice in computational approaches. For complete details on the use and execution of this protocol, please refer to Gennarino et al. (2018).
Collapse
Affiliation(s)
- Winston Lee
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nicola de Prisco
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Vincenzo A. Gennarino
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
- Departments of Pediatrics and Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
- Initiative for Columbia Ataxia and Tremor, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
228
|
Kohaar I, Zhang X, Tan SH, Nousome D, Babcock K, Ravindranath L, Sukumar G, Mcgrath-Martinez E, Rosenberger J, Alba C, Ali A, Young D, Chen Y, Cullen J, Rosner IL, Sesterhenn IA, Dobi A, Chesnut G, Turner C, Dalgard C, Wilkerson MD, Pollard HB, Srivastava S, Petrovics G. Germline mutation landscape of DNA damage repair genes in African Americans with prostate cancer highlights potentially targetable RAD genes. Nat Commun 2022; 13:1361. [PMID: 35292633 PMCID: PMC8924169 DOI: 10.1038/s41467-022-28945-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 02/18/2022] [Indexed: 11/09/2022] Open
Abstract
In prostate cancer, emerging data highlight the role of DNA damage repair genes (DDRGs) in aggressive forms of the disease. However, DDRG mutations in African American men are not yet fully defined. Here, we profile germline mutations in all known DDRGs (N = 276) using whole genome sequences from blood DNA of a matched cohort of patients with primary prostate cancer comprising of 300 African American and 300 European Ancestry prostate cancer patients, to determine whether the mutation status can enhance patient stratification for specific targeted therapies. Here, we show that only 13 of the 46 DDRGs identified with pathogenic/likely pathogenic mutations are present in both African American and European ancestry patients. Importantly, RAD family genes (RAD51, RAD54L, RAD54B), which are potentially targetable, as well as PMS2 and BRCA1, are among the most frequently mutated DDRGs in African American, but not in European Ancestry patients.
Collapse
Affiliation(s)
- Indu Kohaar
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA. .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA.
| | - Xijun Zhang
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Shyh-Han Tan
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Darryl Nousome
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Kevin Babcock
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA
| | - Lakshmi Ravindranath
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Gauthaman Sukumar
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Elisa Mcgrath-Martinez
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - John Rosenberger
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Camille Alba
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Amina Ali
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA.,Urology Service, Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
| | - Denise Young
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Yongmei Chen
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA
| | - Jennifer Cullen
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA
| | - Inger L Rosner
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA
| | | | - Albert Dobi
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Gregory Chesnut
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Urology Service, Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA
| | - Clesson Turner
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Clifton Dalgard
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Matthew D Wilkerson
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Harvey B Pollard
- The American Genome Center, Precision Medicine Initiative for Military Medical Education and Research (PRIMER), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Shiv Srivastava
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA.,Department of Biochemistry and Molecular & Cell biology, Georgetown University School of Medicine, Washington, DC, 20057, USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, 20817, USA. .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA.
| |
Collapse
|
229
|
Qu HQ, Qu J, Vaccaro C, Chang X, Mentch F, Li J, Mafra F, Nguyen K, Gonzalez M, March M, Pellegrino R, Glessner J, Sleiman P, Kao C, Hakonarson H. Genetic Analysis for Type 1 Diabetes Genes in Juvenile Dermatomyositis Unveils Genetic Disease Overlap. Rheumatology (Oxford) 2022; 61:3497-3501. [PMID: 35171267 DOI: 10.1093/rheumatology/keac100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/10/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Juvenile dermatomyositis (JDM) is a serious autoimmune and complex genetic disease. Another autoimmune genetic disease, type 1 diabetes (T1D), has been observed for significantly increased prevalence in families with JDM, while increased JDM risk has also been observed in T1D cases. This study aimed to study whether these two autoimmune diseases, JDM and T1D, share common genetic susceptibility. METHODS From 169 JDM families, 121 unrelated cases with European ancestry (EA) were identified by genome-wide genotyping, principal component analysis (PCA), and identical-by-descent (IBD) analysis. T1D genetic risk score (GRS) were calculated in these cases, and were compared with 361 EA T1D cases and 1943 non-diabetes EA controls. 113 cases of the 121 unrelated European cases were sequenced by whole exome sequencing (WES). RESULTS We observed increased T1D GRS in JDM cases (P=9.42E-05). Using whole exome sequencing (WES), we uncovered the T1D genes, phospholipase B1 (PLB1), cystic fibrosis transmembrane conductance regulator (CFTR), tyrosine hydroxylase (TH), CD6 molecule (CD6), perforin 1 (PRF1), and dynein axonemal heavy chain 2 (DNAH2), potentially associated with JDM by the burden test of rare functional coding variants. CONCLUSION Novel mechanisms of JDM related to these T1D genes are suggested by this study, which may imply novel therapeutic targets for JDM and warrant further study.
Collapse
Affiliation(s)
- Hui-Qi Qu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Jingchun Qu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Courtney Vaccaro
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Xiao Chang
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Frank Mentch
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Jin Li
- Department of Cell Biology, Tianjin Medical University, Tianjin, 300070, China
| | - Fernanda Mafra
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Kenny Nguyen
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Michael Gonzalez
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Michael March
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Renata Pellegrino
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Joseph Glessner
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Patrick Sleiman
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Charlly Kao
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA.,Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, USA
| |
Collapse
|
230
|
Ware SM, Bhatnagar S, Dexheimer PJ, Wilkinson JD, Sridhar A, Fan X, Shen Y, Tariq M, Schubert JA, Colan SD, Shi L, Canter CE, Hsu DT, Bansal N, Webber SA, Everitt MD, Kantor PF, Rossano JW, Pahl E, Rusconi P, Lee TM, Towbin JA, Lal AK, Chung WK, Miller EM, Aronow B, Martin LJ, Lipshultz SE. The genetic architecture of pediatric cardiomyopathy. Am J Hum Genet 2022; 109:282-298. [PMID: 35026164 DOI: 10.1016/j.ajhg.2021.12.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/10/2021] [Indexed: 01/27/2023] Open
Abstract
To understand the genetic contribution to primary pediatric cardiomyopathy, we performed exome sequencing in a large cohort of 528 children with cardiomyopathy. Using clinical interpretation guidelines and targeting genes implicated in cardiomyopathy, we identified a genetic cause in 32% of affected individuals. Cardiomyopathy sub-phenotypes differed by ancestry, age at diagnosis, and family history. Infants < 1 year were less likely to have a molecular diagnosis (p < 0.001). Using a discovery set of 1,703 candidate genes and informatic tools, we identified rare and damaging variants in 56% of affected individuals. We see an excess burden of damaging variants in affected individuals as compared to two independent control sets, 1000 Genomes Project (p < 0.001) and SPARK parental controls (p < 1 × 10-16). Cardiomyopathy variant burden remained enriched when stratified by ancestry, variant type, and sub-phenotype, emphasizing the importance of understanding the contribution of these factors to genetic architecture. Enrichment in this discovery candidate gene set suggests multigenic mechanisms underlie sub-phenotype-specific causes and presentations of cardiomyopathy. These results identify important information about the genetic architecture of pediatric cardiomyopathy and support recommendations for clinical genetic testing in children while illustrating differences in genetic architecture by age, ancestry, and sub-phenotype and providing rationale for larger studies to investigate multigenic contributions.
Collapse
|
231
|
Barbirou M, Miller A, Manjunath Y, Ramirez AB, Ericson NG, Staveley-O’Carroll KF, Mitchem JB, Warren WC, Chaudhuri AA, Huang Y, Li G, Tonellato PJ, Kaifi JT. Single Circulating-Tumor-Cell-Targeted Sequencing to Identify Somatic Variants in Liquid Biopsies in Non-Small-Cell Lung Cancer Patients. Curr Issues Mol Biol 2022; 44:750-763. [PMID: 35723337 PMCID: PMC8928994 DOI: 10.3390/cimb44020052] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) accounts for most cancer-related deaths worldwide. Liquid biopsy by a blood draw to detect circulating tumor cells (CTCs) is a tool for molecular profiling of cancer using single-cell and next-generation sequencing (NGS) technologies. The aim of the study was to identify somatic variants in single CTCs isolated from NSCLC patients by targeted NGS. Thirty-one subjects (20 NSCLC patients, 11 smokers without cancer) were enrolled for blood draws (7.5 mL). CTCs were identified by immunofluorescence, individually retrieved, and DNA-extracted. Targeted NGS was performed to detect somatic variants (single-nucleotide variants (SNVs) and insertions/deletions (Indels)) across 65 oncogenes and tumor suppressor genes. Cancer-associated variants were classified using OncoKB database. NSCLC patients had significantly higher CTC counts than control smokers (p = 0.0132; Mann–Whitney test). Analyzing 23 CTCs and 13 white blood cells across seven patients revealed a total of 644 somatic variants that occurred in all CTCs within the same subject, ranging from 1 to 137 per patient. The highest number of variants detected in ≥1 CTC within a patient was 441. A total of 18/65 (27.7%) genes were highly mutated. Mutations with oncogenic impact were identified in functional domains of seven oncogenes/tumor suppressor genes (NF1, PTCH1, TP53, SMARCB1, SMAD4, KRAS, and ERBB2). Single CTC-targeted NGS detects heterogeneous and shared mutational signatures within and between NSCLC patients. CTC single-cell genomics have potential for integration in NSCLC precision oncology.
Collapse
Affiliation(s)
- Mouadh Barbirou
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Amanda Miller
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Yariswamy Manjunath
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | | | | | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Jonathan B. Mitchem
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Wesley C. Warren
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Animal Sciences and Surgery, Informatics and Data Sciences Institute, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Aadel A. Chaudhuri
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yi Huang
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Peter J. Tonellato
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Jussuf T. Kaifi
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Correspondence:
| |
Collapse
|
232
|
Hamasaki M, Hosaka N, Freeman LA, Sato M, Hara K, Remaley AT, Kotani K. A novel loop-mediated isothermal amplification-based genotyping method and its application for identifying proprotein convertase subtilisin/kexin type 9 variants in familial hypercholesterolemia. Biochim Biophys Acta Gen Subj 2022; 1866:130063. [PMID: 34848321 DOI: 10.1016/j.bbagen.2021.130063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/14/2021] [Accepted: 11/24/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a key role in regulating low-density lipoprotein levels in plasma. While PCSK9 variants are causatively associated with familial hypercholesterolemia (FH), additional genotyping methods for FH targeting PCSK9 variants are required in a clinical setting. Loop-mediated isothermal amplification (LAMP) is a unique amplification method that amplifies a target gene under isothermal conditions (60-65 °C). However, a robust standardized method has not yet been established for LAMP-based genetic screening tests for genetic diseases, including FH. The present study aimed to develop a novel modification of the LAMP method designed to genotype single nucleotide variants (SNVs) and to apply it for the detection of PCSK9 variants. METHODS Using short quenching probes (≤ 10 nucleotides) for the loop structures of LAMP amplicons, accurate detection of SNVs was verified separately for each allele, without any additional procedures, within 3 h. The diagnostic performance of this method in detecting PCSK9 variants was validated in FH patients. RESULTS All PCSK9 variants tested via conventional sequencing in FH patients were successfully detected using this novel LAMP method. CONCLUSIONS We developed a LAMP-based genotyping method to detect PCSK9 variants in FH. Compared to conventional sequencing, our genotyping method is relatively convenient and time-efficient and is suitable for the screening of FH in clinical settings. Future studies on various genes are also warranted.
Collapse
Affiliation(s)
- Masato Hamasaki
- Division of Community and Family Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi 329-0498, Japan; Eiken Chemical Co., Ltd., 143 Nogi, Nogi-Town, Shimotsuga, Tochigi 329-0114, Japan.
| | - Norimitsu Hosaka
- Eiken Chemical Co., Ltd., 143 Nogi, Nogi-Town, Shimotsuga, Tochigi 329-0114, Japan.
| | - Lita A Freeman
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, 10 Center Dr, Bethesda, MD 20892, USA.
| | - Masaki Sato
- Eiken Chemical Co., Ltd., 143 Nogi, Nogi-Town, Shimotsuga, Tochigi 329-0114, Japan; Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, 10 Center Dr, Bethesda, MD 20892, USA
| | - Kazuo Hara
- Division of Endocrinology and Metabolism, Jichi Medical University Saitama Medical Center, 1-847 Amanuma, Omiya-City, Saitama 330-8503, Japan
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, 10 Center Dr, Bethesda, MD 20892, USA.
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi 329-0498, Japan.
| |
Collapse
|
233
|
Application of Whole Exome Sequencing and Functional Annotations to Identify Genetic Variants Associated with Marfan Syndrome. J Pers Med 2022; 12:jpm12020198. [PMID: 35207686 PMCID: PMC8878617 DOI: 10.3390/jpm12020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 02/04/2023] Open
Abstract
Marfan syndrome (MFS) is a rare disease that affects connective tissue, which causes abnormalities in several organ systems including the heart, eyes, bones, and joints. The autosomal dominant disorder was found to be strongly associated with FBN1, TGFBR1, and TGFBR2 mutations. Although multiple genetic mutations have been reported, data from Asian populations are still limited. As a result, we utilized the whole exome sequencing (WES) technique to identify potential pathogenic variants of MFS in a Taiwan cohort. In addition, a variety of annotation databases were applied to identify the biological functions as well as the potential mechanisms of candidate genes. In this study, we confirmed the pathogenicity of FBN1 to MFS. Our results indicated that TTN and POMT1 may be likely related to MFS phenotypes. Furthermore, we found nine unique variants highly shared in a MFS family cohort, of which eight are novel variants worthy of further investigation.
Collapse
|
234
|
Wang C, Shi M, Zhang L, Ji J, Xie R, Wu C, Guo X, Yang Y, Zhou W, Peng C, Zhang H, Yuan F, Zhang J. Identification of KRAS G12V associated clonal neoantigens and immune microenvironment in long-term survival of pancreatic adenocarcinoma. Cancer Immunol Immunother 2022; 71:491-504. [PMID: 34255132 PMCID: PMC8783870 DOI: 10.1007/s00262-021-03012-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/06/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To investigate the molecular characteristics in tumor immune microenvironment that affect long-term survival of patients with pancreatic adenocarcinoma (PAAD). METHODS The tumor related genetic features of a female PAAD patient (over 13-year survival) who suffered from multiple recurrences and metastases, and six operations over one decade were investigated deeply. Genomic features and immune microenvironment signatures of her primary lesion as well as six metastatic tumors at different time-points were characterized. RESULTS High-frequency clonal neoantigenic mutations identified in these specimens revealed the significant associations between clonal neoantigens with her prognosis after each surgery. Meanwhile, the TCGA and ICGC databases were employed to analyse the function of KRAS G12V in pancreatic cancer. CONCLUSIONS The genomic analysis of clonal neoantigens combined with tumor immune microenvironment could promote the understandings of personalized prognostic evaluation and the stratification of resected PAAD individuals with better outcome.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Lei Zhang
- Genecast Biotechnology Co., Ltd, Wuxi City, 214104, Jiangsu, China
| | - Jun Ji
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Ruyan Xie
- VIP Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Chao Wu
- VIP Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Xianchao Guo
- Genecast Biotechnology Co., Ltd, Wuxi City, 214104, Jiangsu, China
| | - Ying Yang
- Genecast Biotechnology Co., Ltd, Wuxi City, 214104, Jiangsu, China
| | - Wei Zhou
- Genecast Biotechnology Co., Ltd, Wuxi City, 214104, Jiangsu, China
| | - Chenhong Peng
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Henghui Zhang
- Genecast Biotechnology Co., Ltd, Wuxi City, 214104, Jiangsu, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
235
|
Li Y, Liu X, Wang Y, Zhou Y, Hu S, Yang H, Zhong W, Zhao J, Wang X, Chu H, Zheng Y, Zhang J, Zhou L, Wang B. Novel HLA-DRB1 alleles contribute risk for disease susceptibility in primary biliary cholangitis. Dig Liver Dis 2022; 54:228-236. [PMID: 34016546 DOI: 10.1016/j.dld.2021.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Primary biliary cholangitis (PBC) is a complex disease with high heritability. We investigated the association between human leukocyte antigen (HLA)-DRB1 alleles and PBC in families and sporadic cases to evaluate the genetic components of the disease. METHODS We performed whole exome sequencing in three PBC families. We genotyped HLA-DRB1 and calculated the association between HLA-DRB1 alleles and the encoding amino acid sequences with the clinical features. RESULTS Ten variants harboured the HLA-DRB1 gene associated with PBC. DRB1×07:01, 14:01 and 14:05 were highly increased in PBC. Ten coding region polymorphisms were associated with PBC that encode the amino acid variants of HLA-DR β54, β59 and β66 located in the peptide-binding site of the MHC molecule. Glutamine at position 54 was confirmed as a risk amino acid, verifying the results of familial aggregation analysis of PBC families. DISCUSSION Familial aggregation analysis indicated that HLA-DRB1 is a candidate gene for the risk of disease course. Considering that amino acid variations are critical to peptide-binding properties, they underlie the major component of MHC association with PBC.
Collapse
Affiliation(s)
- Yanni Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China; Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Xin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China; Department of General Surgery, YouAn Hospital, Capital Medical University, Beijing, China
| | - Yi Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shixian Hu
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Hui Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoyi Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Chu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanping Zheng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Zhang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China; Department of Gastroenterology and Hepatology, People's Hospital of Hetian District, Xinjiang Uygur Autonomous Region, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
236
|
Grigorenko AP, Protasova MS, Lisenkova AA, Reshetov DA, Andreeva TV, Garcias GDL, Martino Roth MDG, Papassotiropoulos A, Rogaev EI. Neurodevelopmental Syndrome with Intellectual Disability, Speech Impairment, and Quadrupedia Is Associated with Glutamate Receptor Delta 2 Gene Defect. Cells 2022; 11:400. [PMID: 35159210 PMCID: PMC8834146 DOI: 10.3390/cells11030400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/16/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
Bipedalism, speech, and intellect are the most prominent traits that emerged in the evolution of Homo sapiens. Here, we describe a novel genetic cause of an "involution" phenotype in four patients, who are characterized by quadrupedal locomotion, intellectual impairment, the absence of speech, small stature, and hirsutism, observed in a consanguineous Brazilian family. Using whole-genome sequencing analysis and homozygous genetic mapping, we identified genes bearing homozygous genetic variants and found a homozygous 36.2 kb deletion in the gene of glutamate receptor delta 2 (GRID2) in the patients, resulting in the lack of a coding region from the fifth to the seventh exons. The GRID2 gene is highly expressed in the cerebellum cortex from prenatal development to adulthood, specifically in Purkinje neurons. Deletion in this gene leads to the loss of the alpha chain in the extracellular amino-terminal protein domain (ATD), essential in protein folding and transport from the endoplasmic reticulum (ER) to the cell surface. Then, we studied the evolutionary trajectories of the GRID2 gene. There was no sign of strong selection of the highly conservative GRID2 gene in ancient hominids (Neanderthals and Denisovans) or modern humans; however, according to in silico tests using the Mfold tool, the GRID2 gene possibly gained human-specific mutations that increased the stability of GRID2 mRNA.
Collapse
Affiliation(s)
- Anastasia P. Grigorenko
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.G.); (T.V.A.)
- Laboratory of Evolutionary Genomics, Department of Genomics and Human Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (M.S.P.); (A.A.L.); (D.A.R.)
- Department of Psychiatry, UMass Chan Medical School, Shrewsbury, MA 01545, USA
| | - Maria S. Protasova
- Laboratory of Evolutionary Genomics, Department of Genomics and Human Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (M.S.P.); (A.A.L.); (D.A.R.)
| | - Alexandra A. Lisenkova
- Laboratory of Evolutionary Genomics, Department of Genomics and Human Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (M.S.P.); (A.A.L.); (D.A.R.)
| | - Denis A. Reshetov
- Laboratory of Evolutionary Genomics, Department of Genomics and Human Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (M.S.P.); (A.A.L.); (D.A.R.)
| | - Tatiana V. Andreeva
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.G.); (T.V.A.)
- Laboratory of Evolutionary Genomics, Department of Genomics and Human Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (M.S.P.); (A.A.L.); (D.A.R.)
- Center for Genetics and Genetic Technologies, Department of Genetics, Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Gilberto De Lima Garcias
- Catholic University of Pelotas, Pelotas 96015-560, RS, Brazil; (G.D.L.G.); (M.D.G.M.R.)
- Federal University of Pelotas, Pelotas 96010-610, RS, Brazil
| | | | - Andreas Papassotiropoulos
- Transfaculty Research Platform, University of Basel, CH-4055 Basel, Switzerland;
- Psychiatric University Clinics, University of Basel, CH-4055 Basel, Switzerland
| | - Evgeny I. Rogaev
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.G.); (T.V.A.)
- Laboratory of Evolutionary Genomics, Department of Genomics and Human Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (M.S.P.); (A.A.L.); (D.A.R.)
- Department of Psychiatry, UMass Chan Medical School, Shrewsbury, MA 01545, USA
- Center for Genetics and Genetic Technologies, Department of Genetics, Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| |
Collapse
|
237
|
Case series of congenital pseudarthrosis of the tibia unfulfilling neurofibromatosis type 1 diagnosis: 21% with somatic NF1 haploinsufficiency in the periosteum. Hum Genet 2022; 141:1371-1383. [DOI: 10.1007/s00439-021-02429-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/29/2021] [Indexed: 12/17/2022]
|
238
|
Long JM, Powers JM, Katona BW. Evaluation of Classic, Attenuated, and Oligopolyposis of the Colon. Gastrointest Endosc Clin N Am 2022; 32:95-112. [PMID: 34798989 PMCID: PMC8607742 DOI: 10.1016/j.giec.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The goal of this review is to provide an overview of evaluating patients with adenomatous polyposis of the colon, including elements such as generating a differential diagnosis, referral considerations for genetic testing, genetic testing options, and expected outcomes from genetic testing in these individuals. In more recent years, adenomatous colonic polyposis has evolved beyond the more robustly characterized familial adenomatous polyposis (FAP) and MUTYH-associated polyposis (MAP) now encompassing more newly described genes and associated syndromes. Technological innovation, from whole-exome sequencing to multigene panel testing, has dramatically increased the amount of genotypic and phenotypic data amassed in adenomatous polyposis cohorts, which has contributed greatly to informing diagnosis and clinical management of affected individuals and their families.
Collapse
Affiliation(s)
- Jessica M. Long
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacquelyn M. Powers
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bryson W. Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
239
|
Annear DJ, Vandeweyer G, Sanchis-Juan A, Raymond FL, Kooy RF. Non-Mendelian inheritance patterns and extreme deviation rates of CGG repeats in autism. Genome Res 2022; 32:1967-1980. [PMID: 36351771 PMCID: PMC9808627 DOI: 10.1101/gr.277011.122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/14/2022] [Indexed: 11/10/2022]
Abstract
As expansions of CGG short tandem repeats (STRs) are established as the genetic etiology of many neurodevelopmental disorders, we aimed to elucidate the inheritance patterns and role of CGG STRs in autism-spectrum disorder (ASD). By genotyping 6063 CGG STR loci in a large cohort of trios and quads with an ASD-affected proband, we determined an unprecedented rate of CGG repeat length deviation across a single generation. Although the concept of repeat length being linked to deviation rate was solidified, we show how shorter STRs display greater degrees of size variation. We observed that CGG STRs did not segregate by Mendelian principles but with a bias against longer repeats, which appeared to magnify as repeat length increased. Through logistic regression, we identified 19 genes that displayed significantly higher rates and degrees of CGG STR expansion within the ASD-affected probands (P < 1 × 10-5). This study not only highlights novel repeat expansions that may play a role in ASD but also reinforces the hypothesis that CGG STRs are specifically linked to human cognition.
Collapse
Affiliation(s)
- Dale J. Annear
- Department of Medical Genetics, University of Antwerp, 2600 Antwerp, Belgium
| | - Geert Vandeweyer
- Department of Medical Genetics, University of Antwerp, 2600 Antwerp, Belgium
| | - Alba Sanchis-Juan
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, United Kingdom;,Department of Haematology, University of Cambridge, NHS Blood and Transplant Centre, Cambridge, CB2 0PT, United Kingdom
| | - F. Lucy Raymond
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, United Kingdom;,Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, United Kingdom
| | - R. Frank Kooy
- Department of Medical Genetics, University of Antwerp, 2600 Antwerp, Belgium
| |
Collapse
|
240
|
OUP accepted manuscript. Hum Mol Genet 2022; 31:2236-2261. [DOI: 10.1093/hmg/ddac029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/12/2022] Open
|
241
|
Computational Resources for the Interpretation of Variations in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:177-198. [DOI: 10.1007/978-3-030-91836-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
242
|
Pan Z, Tian H, Fang T, Liu Z, Liu X, Dou G, Huang G, Zhang Z, Chen G, Wang W, Zhuo C. OGDHL Variant rs2293239: A Potential Genetic Driver of Chinese Familial Depressive Disorder. Front Psychiatry 2022; 13:771950. [PMID: 35370858 PMCID: PMC8971628 DOI: 10.3389/fpsyt.2022.771950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/21/2022] [Indexed: 12/16/2022] Open
Abstract
Depressive disorders are a severe psychiatric and social problem that affect more than 4% of the global population. Depressive disorders have explicit hereditary characteristics; however, the precise driving genetic force behind these disorders has not yet been clearly illustrated. In the present study, we recruited a three-generation Chinese pedigree in which 5 of 17 members had long-term depression. We conducted whole-exome sequencing to identify the genetic mutation profiles of the family, and a list of susceptible genetic variations that were highly associated with depression onset was revealed via multiple omics analysis. In particular, a non-synonymous single nucleotide variation in the oxoglutarate dehydrogenase-like (OGDHL) gene, rs2293239 (p.Asn725Ser), was identified as one of the major driving genetic forces for depression onset in the family. This variant causes an important conformational change in the transketolase domain of OGDHL, thus reducing its binding affinity with the cofactor thiamine pyrophosphate and eventually resulting in the abnormal accumulation of glutamate in the brain. Brain imaging analysis further linked the rs2293239 variant with an enlarged amygdala and cerebellum in depressive family members. In summary, the present study enhances the current genetic understanding of depressive disorders. It also provides new options for prioritizing better clinical therapeutic regimens, as well as identifying a new protein target for the design of highly specific drugs to treat depressive disorders.
Collapse
Affiliation(s)
- Zhi Pan
- Key Laboratory of Genetic Psychiatry, Wenzhou Seventh People Hospital, Wenzhou, China
| | - Hongjun Tian
- Key Laboratory of Real Time Tracing of Brain Circuits of Neurology and Psychiatry, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin Fourth Center Hospital, Tianjin, China
| | - Tao Fang
- Key Laboratory of Real Time Tracing of Brain Circuits of Neurology and Psychiatry, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin Fourth Center Hospital, Tianjin, China
| | - Zhidong Liu
- Key Laboratory of Real Time Tracing of Brain Circuits of Neurology and Psychiatry, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin Fourth Center Hospital, Tianjin, China
| | - Xiangdong Liu
- Key Laboratory of Real Time Tracing of Brain Circuits of Neurology and Psychiatry, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin Fourth Center Hospital, Tianjin, China
| | - Guangqian Dou
- Key Laboratory of Real Time Tracing of Brain Circuits of Neurology and Psychiatry, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin Fourth Center Hospital, Tianjin, China
| | - Guoyong Huang
- Key Laboratory of Genetic Psychiatry, Wenzhou Seventh People Hospital, Wenzhou, China
| | - Zhenqing Zhang
- Department of Psychiatry, Xiamen Xianyue Hospital, Xiamen, China
| | - Guangdong Chen
- Key Laboratory of Genetic Psychiatry, Wenzhou Seventh People Hospital, Wenzhou, China
| | - Wenqiang Wang
- Department of Psychiatry, Xiamen Xianyue Hospital, Xiamen, China
| | - Chuanjun Zhuo
- Key Laboratory of Real Time Tracing of Brain Circuits of Neurology and Psychiatry, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin Fourth Center Hospital, Tianjin, China.,Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
243
|
Novo I, Santiago E, Caballero A. The estimates of effective population size based on linkage disequilibrium are virtually unaffected by natural selection. PLoS Genet 2022; 18:e1009764. [PMID: 35077457 PMCID: PMC8815936 DOI: 10.1371/journal.pgen.1009764] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/04/2022] [Accepted: 12/21/2021] [Indexed: 11/19/2022] Open
Abstract
The effective population size (Ne) is a key parameter to quantify the magnitude of genetic drift and inbreeding, with important implications in human evolution. The increasing availability of high-density genetic markers allows the estimation of historical changes in Ne across time using measures of genome diversity or linkage disequilibrium between markers. Directional selection is expected to reduce diversity and Ne, and this reduction is modulated by the heterogeneity of the genome in terms of recombination rate. Here we investigate by computer simulations the consequences of selection (both positive and negative) and recombination rate heterogeneity in the estimation of historical Ne. We also investigate the relationship between diversity parameters and Ne across the different regions of the genome using human marker data. We show that the estimates of historical Ne obtained from linkage disequilibrium between markers (NeLD) are virtually unaffected by selection. In contrast, those estimates obtained by coalescence mutation-recombination-based methods can be strongly affected by it, which could have important consequences for the estimation of human demography. The simulation results are supported by the analysis of human data. The estimates of NeLD obtained for particular genomic regions do not correlate, or they do it very weakly, with recombination rate, nucleotide diversity, proportion of polymorphic sites, background selection statistic, minor allele frequency of SNPs, loss of function and missense variants and gene density. This suggests that NeLD measures mainly reflect demographic changes in population size across generations.
Collapse
Affiliation(s)
- Irene Novo
- Centro de Investigación Mariña, Universidade de Vigo, Facultade de Bioloxía, Vigo, Spain
| | - Enrique Santiago
- Departamento de Biología Funcional, Facultad de Biología, Universidad de Oviedo, Oviedo, Spain
| | - Armando Caballero
- Centro de Investigación Mariña, Universidade de Vigo, Facultade de Bioloxía, Vigo, Spain
| |
Collapse
|
244
|
Application of deep learning algorithm on whole genome sequencing data uncovers structural variants associated with multiple mental disorders in African American patients. Mol Psychiatry 2022; 27:1469-1478. [PMID: 34997195 PMCID: PMC9095459 DOI: 10.1038/s41380-021-01418-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/11/2021] [Accepted: 12/01/2021] [Indexed: 11/08/2022]
Abstract
Mental disorders present a global health concern, while the diagnosis of mental disorders can be challenging. The diagnosis is even harder for patients who have more than one type of mental disorder, especially for young toddlers who are not able to complete questionnaires or standardized rating scales for diagnosis. In the past decade, multiple genomic association signals have been reported for mental disorders, some of which present attractive drug targets. Concurrently, machine learning algorithms, especially deep learning algorithms, have been successful in the diagnosis and/or labeling of complex diseases, such as attention deficit hyperactivity disorder (ADHD) or cancer. In this study, we focused on eight common mental disorders, including ADHD, depression, anxiety, autism, intellectual disabilities, speech/language disorder, delays in developments, and oppositional defiant disorder in the ethnic minority of African Americans. Blood-derived whole genome sequencing data from 4179 individuals were generated, including 1384 patients with the diagnosis of at least one mental disorder. The burden of genomic variants in coding/non-coding regions was applied as feature vectors in the deep learning algorithm. Our model showed ~65% accuracy in differentiating patients from controls. Ability to label patients with multiple disorders was similarly successful, with a hamming loss score less than 0.3, while exact diagnostic matches are around 10%. Genes in genomic regions with the highest weights showed enrichment of biological pathways involved in immune responses, antigen/nucleic acid binding, chemokine signaling pathway, and G-protein receptor activities. A noticeable fact is that variants in non-coding regions (e.g., ncRNA, intronic, and intergenic) performed equally well as variants in coding regions; however, unlike coding region variants, variants in non-coding regions do not express genomic hotspots whereas they carry much more narrow standard deviations, indicating they probably serve as alternative markers.
Collapse
|
245
|
McCreary D, Omoyinmi E, Hong Y, Jensen B, Burleigh A, Price-Kuehne F, Gilmour K, Eleftheriou D, Brogan P. A rapid turnaround gene panel for severe autoinflammation: Genetic results within 48 hours. Front Immunol 2022; 13:998967. [PMID: 36203604 PMCID: PMC9531256 DOI: 10.3389/fimmu.2022.998967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
There is an important unmet clinical need for fast turnaround next generation sequencing (NGS) to aid genetic diagnosis of patients with acute and sometimes catastrophic inflammatory presentations. This is imperative for patients who require precise and targeted treatment to prevent irreparable organ damage or even death. Acute and severe hyper- inflammation may be caused by primary immunodeficiency (PID) with immune dysregulation, or more typical autoinflammatory diseases in the absence of obvious immunodeficiency. Infectious triggers may be present in either immunodeficiency or autoinflammation. We compiled a list of 25 genes causing monogenetic immunological diseases that are notorious for their acute first presentation with fulminant inflammation and which may be amenable to specific treatment, including hemophagocytic lymphohistiocytosis (HLH); and autoinflammatory diseases that can present with early-onset stroke or other irreversible neurological inflammatory complications. We designed and validated a pipeline that enabled return of clinically actionable results in hours rather than weeks: the Rapid Autoinflammation Panel (RAP). We demonstrated accuracy of this new pipeline, with 100% sensitivity and 100% specificity. Return of results to clinicians was achieved within 48-hours from receiving the patient's blood or saliva sample. This approach demonstrates the potential significant diagnostic impact of NGS in acute medicine to facilitate precision medicine and save "life or limb" in these critical situations.
Collapse
Affiliation(s)
- Dara McCreary
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ebun Omoyinmi
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom.,National Amyloidosis Centre, Royal Free Hospital, London, United Kingdom
| | - Ying Hong
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Barbara Jensen
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Alice Burleigh
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom.,Centre for Adolescent Rheumatology, University College London, London, United Kingdom
| | - Fiona Price-Kuehne
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Kimberly Gilmour
- Camelia Botnar Laboratory, Great Ormond Street Hospital National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Despina Eleftheriou
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom.,Centre for Adolescent Rheumatology, University College London, London, United Kingdom.,Rheumatology Department, Great Ormond Street Hospital National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Paul Brogan
- Inflammation and Rheumatology Section, University College London Great Ormond Street Institute of Child Health, London, United Kingdom.,Rheumatology Department, Great Ormond Street Hospital National Health Service (NHS) Foundation Trust, London, United Kingdom
| |
Collapse
|
246
|
Plym A, Dióssy M, Szallasi Z, Sartor O, Silberstein J, Powell IJ, Rebbeck TR, Penney KL, Mucci LA, Pomerantz MM, Kibel AS. DNA Repair Pathways and Their Association With Lethal Prostate Cancer in African American and European American Men. JNCI Cancer Spectr 2021; 6:pkab097. [PMID: 35079693 PMCID: PMC8784166 DOI: 10.1093/jncics/pkab097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/25/2021] [Accepted: 11/11/2021] [Indexed: 11/23/2022] Open
Abstract
Background Altered DNA damage response (DDR) has emerged as an important mechanism for the development of aggressive prostate cancer among men of European ancestry but not other ancestry groups. Because common mechanisms for aggressive disease are expected, we explored a large panel of DDR genes and pathways to demonstrate that DDR alterations contribute to development of aggressive prostate cancer in both African American and European American men. Methods We performed a case-case study of 764 African American and European American men with lethal or indolent prostate cancer treated at 4 US hospitals. We calculated carrier frequencies of germline pathogenic or likely pathogenic sequence variants within 306 DDR genes, summarized by DDR pathway, and compared lethal cases against indolent cases using 2-sided Fisher’s exact tests. Secondary analysis examined if carrier frequencies differed by ancestry. Results Lethal cases were more likely to carry a pathogenic sequence variant in a DDR gene compared with indolent cases (18.5% vs 9.6%, P = 4.30 × 10−4), even after excluding BRCA2 (14.6% vs 9.6%, P = .04). The carrier frequency was similar among lethal cases of African (16.7% including and 15.8% excluding BRCA2) and lethal cases of European (19.3% including and 14.2% excluding BRCA2) ancestry. Three DDR pathways were statistically significantly associated with lethal disease: homologous recombination (P = .003), Fanconi anemia (P = .002), and checkpoint factor (P = .02). Conclusions Our findings suggest that altered DDR is an important mechanism for aggressive prostate cancer not only in men of European but also of African ancestry. Therefore, interrogation of entire DDR pathways is needed to fully characterize and better define genetic risk of lethal disease.
Collapse
Affiliation(s)
- Anna Plym
- Urology Division, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Miklós Dióssy
- Translational Cancer Genomics, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Zoltan Szallasi
- Translational Cancer Genomics, Danish Cancer Society Research Center, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, USA
- 2nd Department of Pathology, SE NAP, Brain Metastasis Research Group, Semmelweis University, Budapest, Hungary
| | - Oliver Sartor
- Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jonathan Silberstein
- Section of Urology and Uro-Oncology, Memorial Healthcare System, Broward, FL, USA
| | - Isaac J Powell
- Department of Urology, Wayne State University, Detroit, MI, USA
| | - Timothy R Rebbeck
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Adam S Kibel
- Urology Division, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
247
|
Zhan Q, Wen C, Zhao Y, Fang L, Jin Y, Zhang Z, Zou S, Li F, Yang Y, Wu L, Jin J, Lu X, Xie J, Cheng D, Xu Z, Zhang J, Wang J, Deng X, Chen H, Peng C, Li H, Zhang H, Fang H, Wang C, Shen B. Identification of copy number variation-driven molecular subtypes informative for prognosis and treatment in pancreatic adenocarcinoma of a Chinese cohort. EBioMedicine 2021; 74:103716. [PMID: 34839264 PMCID: PMC8628206 DOI: 10.1016/j.ebiom.2021.103716] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is one of the most lethal carcinomas, and the current histopathological classifications are of limited use in clinical decision-making. There is an unmet need to identify new biomarkers for prognosis-informative molecular subtyping and ultimately for precision medicine. METHODS We profiled genomic alterations for 608 PAAD patients in a Chinese cohort, including somatic mutations, pathogenic germline variants and copy number variations (CNV). Using the CNV information, we performed unsupervised consensus clustering of these patients, differential CNV analysis and functional/pathway enrichment analysis. Cox regression was conducted for progression-free survival analysis, the elastic net algorithm used for prognostic model construction, and rank-based gene set enrichment analysis for exploring tumor microenvironments. FINDINGS Our data did not support prognostic value of point mutations in either highly mutated genes (such as KRAS, TP53, CDKN2A and SMAD4) or homologous recombination repair genes. Instead, associated with worse prognosis were amplified genes involved in DNA repair and receptor tyrosine kinase (RTK) related signalings. Motivated by this observation, we categorized patients into four molecular subtypes (namely repair-deficient, proliferation-active, repair-proficient and repair-enhanced) that differed in prognosis, and also constructed a prognostic model that can stratify patients with low or high risk of relapse. Finally, we analyzed publicly available datasets, not only reinforcing the prognostic value of our identified genes in DNA repair and RTK related signalings, but also identifying tumor microenvironment correlates with prognostic risks. INTERPRETATION Together with the evidence from genomic footprint analysis, we suggest that repair-deficient and proliferation-active subtypes are better suited for DNA damage therapies, while immunotherapy is highly recommended for repair-proficient and repair-enhanced subtypes. Our results represent a significant step in molecular subtyping, diagnosis and management for PAAD patients. FUNDING This work was supported by the National Natural Science Foundation of China (grant numbers 81470894, 81502695, 81672325, 81871906, 82073326, 82103482 and 32170663), the Shanghai Sailing Program (grant number 20YF1426900), and the Program for Professor of Special Appointment (Eastern Scholar) at Shanghai Institutions of Higher Learning (awarded to H.F.).
Collapse
Affiliation(s)
- Qian Zhan
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Chenlei Wen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Yi Zhao
- Genecast Biotechnology Co., Ltd, Wuxi, China
| | - Lu Fang
- Genecast Biotechnology Co., Ltd, Wuxi, China
| | - Yangbing Jin
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Zehui Zhang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Siyi Zou
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Fanlu Li
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Ying Yang
- Genecast Biotechnology Co., Ltd, Wuxi, China
| | - Lijia Wu
- Genecast Biotechnology Co., Ltd, Wuxi, China
| | - Jiabin Jin
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Xiongxiong Lu
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Junjie Xie
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Dongfeng Cheng
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Zhiwei Xu
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Jun Zhang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Jiancheng Wang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - XiaXing Deng
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Chenghong Peng
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | - Hongwei Li
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China
| | | | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China..
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.; State Key Laboratory of Oncogenes and Related Genes, National Research Center for Translational Medicine (Shanghai), Shanghai, China..
| |
Collapse
|
248
|
Ware SM. Pediatric cardiomyopathy and the PCM Genes study: A summary with insights on genetic testing, variant interpretation, race and ethnicity. PROGRESS IN PEDIATRIC CARDIOLOGY 2021. [DOI: 10.1016/j.ppedcard.2021.101468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
249
|
A Novel WT1 Mutation Identified in a 46,XX Testicular/Ovotesticular DSD Patient Results in the Retention of Intron 9. BIOLOGY 2021; 10:biology10121248. [PMID: 34943163 PMCID: PMC8698877 DOI: 10.3390/biology10121248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/24/2021] [Accepted: 11/27/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary Disorders/differences of sexual development are very diverse. Among them is a condition characterized by the presence of testicular tissue in people with female chromosomes, which is typically manifested by male or ambiguous genitalia. While genetic counseling is beneficial for these people and their families, the genetic causes of these cases are only partially understood. We describe a new mutation in the WT1 gene that results in the presence of testicular tissue in a child with a female karyotype. We propose molecular mechanisms disrupted by this mutation. This finding widened our understanding of processes that govern sexual development and can be used to develop diagnostic tests for disorders/differences of sexual development. Abstract The 46,XX testicular DSD (disorder/difference of sexual development) and 46,XX ovotesticular DSD (46,XX TDSD and 46,XX OTDSD) phenotypes are caused by genetic rearrangements or point mutations resulting in imbalance between components of the two antagonistic, pro-testicular and pro-ovarian pathways; however, the genetic causes of 46,XX TDSD/OTDSD are not fully understood, and molecular diagnosis for many patients with the conditions is unavailable. Only recently few mutations in the WT1 (WT1 transcription factor; 11p13) gene were described in a group of 46,XX TDSD and 46,XX OTDSD individuals. The WT1 protein contains a DNA/RNA binding domain consisting of four zinc fingers (ZnF) and a three-amino acid (KTS) motif that is present or absent, as a result of alternative splicing, between ZnF3 and ZnF4 (±KTS isoforms). Here, we present a patient with 46,XX TDSD/OTDSD in whom whole exome sequencing revealed a heterozygous de novo WT1 c.1437A>G mutation within an alternative donor splice site which is used for −KTS WT1 isoform formation. So far, no mutation in this splice site has been identified in any patient group. We demonstrated that the mutation results in the retention of intron 9 in the mature mRNA of the 46,XX TDSD/OTDSD patient. In cases when the erroneous mRNA is translated, exclusively the expression of a truncated WT1 +KTS protein lacking ZnF4 and no −KTS protein occurs from the mutated allele of the patient. We discuss potential mechanisms and pathways which can be disturbed upon two conditions: Absence of Zn4F and altered +KTS/−KTS ratio.
Collapse
|
250
|
Thakur S, Jain M, Zhang C, Major C, Bielamowicz KJ, Lacayo NJ, Vaske O, Lewis V, Murguia-Favela L, Narendran A. Identification and in vitro validation of neoantigens for immune activation against high-risk pediatric leukemia cells. Hum Vaccin Immunother 2021; 17:5558-5562. [PMID: 34844524 DOI: 10.1080/21645515.2021.2001243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
There is experimental and clinical data to indicate the contribution of immune-escape mechanisms in relapsed/refractory pediatric leukemia. Studies have shown the accumulation of mutations that translate to peptides containing tumor-specific epitopes (neoantigens). The effectiveness of neoantigen-based vaccines has been shown in several clinical trials in adults. Though the initial results are encouraging, this knowledge must be developed to account for the uniqueness of pediatric cancer biology. We have completed the initial proof-of-concept analysis on a high-risk pediatric leukemia specimen and identified usable neoantigen sequences. We describe this approach, including the bioinformatics method and experimental model to verify their function that can be further broadened for personalized neoantigen prediction and testing for the generation of anticancer vaccines against high-risk pediatric leukemias.
Collapse
Affiliation(s)
- Satbir Thakur
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Mohit Jain
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Chunfen Zhang
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Candice Major
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Kevin J Bielamowicz
- Division of Hematology/Oncology, Arkansas Children's Hospital, Little Rock, Arkansas
| | - Norman J Lacayo
- Department of Oncology, Stanford University School of Medicine, Stanford, California
| | - Olena Vaske
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California
| | - Victor Lewis
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Luis Murguia-Favela
- Section of Hematology and Immunology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Aru Narendran
- Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| |
Collapse
|