51
|
Vecchio D, Macchiaiolo M, Gonfiantini MV, Panfili FM, Petrizzelli F, Liorni N, Cortellessa F, Sinibaldi L, Rana I, Agolini E, Cocciadiferro D, Colantoni N, Semeraro M, Rizzo C, Deodati A, Cotugno N, Caggiano S, Verrillo E, Nucci CG, Alkan S, Saraiva JM, De Sá J, Almeida PM, Krishna J, Buonuomo PS, Martinelli D, Dionisi Vici C, Caputo V, Bartuli A, Novelli A, Mazza T. Widening the infantile hypotonia with psychomotor retardation and characteristic Facies-1 Syndrome's clinical and molecular spectrum through NALCN in-silico structural analysis. Front Genet 2024; 15:1477940. [PMID: 39722796 PMCID: PMC11668739 DOI: 10.3389/fgene.2024.1477940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Infantile hypotonia with psychomotor retardation and characteristic facies-1 (IHPRF1, MIM#615419) is a rare, birth onset, autosomal recessive disorder caused by homozygous or compound heterozygous truncating variants in NALCN gene (MIM#611549) resulting in a loss-of-function effect. Methods We enrolled a new IHPRF1 patients' cohort in the framework of an international multicentric collaboration study. Using specialized in silico pathogenicity predictors and ad hoc structural analyses, we assessed the mechanistic consequences of the deleterious variants retrieved on NALCN structure and function. Results To date 38 different NALCN variants have been retrieved from 33 different families, 26 from unrelated and 22 from related patients. We report on five new IHPRF1 patients from four different families, harboring four newly identified and one previously retrieved variant that exhibited a markedly significant functional impact, thereby compromising the functionality of the protein complex. Discussion By widening the functional spectrum of biallelic variants affecting the NALCN gene, this article broadens the IHPRF1 syndrome's genotype-phenotype correlation and gives new insight into its pathogenic mechanism, diagnosis, and clinical management.
Collapse
Affiliation(s)
- Davide Vecchio
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Michaela V. Gonfiantini
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Filippo M. Panfili
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesco Petrizzelli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Niccolò Liorni
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabiana Cortellessa
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenzo Sinibaldi
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Ippolita Rana
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Emanuele Agolini
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Dario Cocciadiferro
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicole Colantoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Michela Semeraro
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Cristiano Rizzo
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Annalisa Deodati
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Diabetology and Growth Disorders Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicola Cotugno
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Research Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Serena Caggiano
- Pediatric Pulmonology and Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Elisabetta Verrillo
- Pediatric Pulmonology and Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Carlotta G. Nucci
- Neurosurgery Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Serpil Alkan
- Department of Pediatrics, Centre Hospitalier Universitaire, CHU, Liège, Belgium
| | - Jorge M. Saraiva
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
- University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Joaquim De Sá
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Pedro M. Almeida
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Jayanth Krishna
- Krishna Institute of Medical Sciences (KIMS Hospital), Hyderabad, India
| | - Paola S. Buonuomo
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Diego Martinelli
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Carlo Dionisi Vici
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Viviana Caputo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Bartuli
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
52
|
Gagliardi D, Wade C, Tucci A, Houlden H, Chataway J, Barkhof F, Lynch DS. Analysis of GFAP variants in UK Biobank suggests underdiagnosis or incomplete penetrance of adult-onset Alexander disease. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-335089. [PMID: 39643430 DOI: 10.1136/jnnp-2024-335089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Alexander disease is an autosomal dominant leukodystrophy caused by heterozygous pathogenic variants in the glial fibrillar acidic protein (GFAP) gene. Although increasingly recognised, there is evidence that Alexander disease, particularly later-onset disease, is significantly underdiagnosed and its true prevalence is unknown (the only population-based prevalence was estimated at one in 2.7 million). Using the extensive UK Biobank dataset, we analysed the frequency of pathogenic and likely pathogenic variants, GFAP variants, within the UK population and identified clinical and radiological phenotypes linked to these variants. METHODS Pathogenic, likely pathogenic and GFAP variants of uncertain significance were identified in the UK Biobank whole-exome sequencing data (n=4 70 000). Demographic information, previous medical history-including symptoms associated with Alexander disease-collected from self-reported data and hospital records, family history and various MRI metrics were compared between variant carriers and controls. RESULTS We identified 36 unique pathogenic and likely pathogenic GFAP variants in 106 carriers, yielding a carrier frequency of approximately 1 in 4435. Modelling based on the UK population estimated a prevalence of 6.8 per 100 000. Carriers of pathogenic and likely pathogenic GFAP variants had higher odds of bladder dysfunction (OR 3.17, p<0.0001), upper airway dysfunction (OR 7.82, p=0.004) and psychiatric conditions (OR 1.51, p=0.04). Additionally, carriers were more likely to report a paternal history of dementia (OR 2.79, p<0.0001). MRI data revealed significant atrophy in brainstem regions among variant carriers. CONCLUSION Pathogenic and likely pathogenic GFAP variants are more prevalent in the general population than previously expected and are associated with clinical and radiological characteristics of Alexander disease. This study indicates that Alexander disease may be under-reported, misdiagnosed, or exhibit reduced penetrance.
Collapse
Affiliation(s)
- Delia Gagliardi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Charles Wade
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Arianna Tucci
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Henry Houlden
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- National Hospital for Neurology and Neurosurgery, London, UK
| | - Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
- Biomedical Research Centre, National Institute for Health Research, University College London Hospitals Foundation Trust, London, UK
| | - Frederik Barkhof
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, London, UK
| | - David S Lynch
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
- National Hospital for Neurology and Neurosurgery, London, UK
- Biomedical Research Centre, National Institute for Health Research, University College London Hospitals Foundation Trust, London, UK
| |
Collapse
|
53
|
Sharma P, McFadden JR, Frost FG, Markello TC, Grange DK, Introne WJ, Gahl WA, Malicdan MCV. Biallelic germline DDX41 variants in a patient with bone dysplasia, ichthyosis, and dysmorphic features. Hum Genet 2024; 143:1445-1457. [PMID: 39453476 PMCID: PMC11576897 DOI: 10.1007/s00439-024-02708-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
DDX41 (DEAD‑box helicase 41) is a member of the largest family of RNA helicases. The DEAD-box RNA helicases share a highly conserved core structure and regulate all aspects of RNA metabolism. The functional role of DDX41 in innate immunity is also highly conserved. DDX41 acts as a sensor of viral DNA and activates the STING-TBK1-IRF3-type I IFN signaling pathway. Germline heterozygous variants in DDX41 have been reported in familial myelodysplasia syndrome (MDS)/acute myeloid leukemia (AML) patients; most patients also acquired a somatic variant in the second DDX41 allele. Here, we report a patient who inherited compound heterozygous DDX41 variants and presented with bone dysplasia, ichthyosis, and dysmorphic features. Functional analyses of the patient-derived dermal fibroblasts revealed a reduced abundance of DDX41 and abrogated activation of the IFN genes through the STING-type I interferon pathway. Genome-wide transcriptome analyses in the patient's fibroblasts revealed significant gene dysregulation and changes in the RNA splicing events. The patient's fibroblasts also displayed upregulation of periostin mRNA expression. Using an RNA binding protein assay, we identified DDX41 as a novel regulator of periostin expression. Our results suggest that functional impairment of DDX41, along with dysregulated periostin expression, likely contributes to this patient's multisystem disorder.
Collapse
Affiliation(s)
- Prashant Sharma
- NIH Undiagnosed Diseases Program, Common Fund, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Jason R McFadden
- NIH Undiagnosed Diseases Program, Common Fund, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - F Graeme Frost
- NIH Undiagnosed Diseases Program, Common Fund, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Thomas C Markello
- NIH Undiagnosed Diseases Program, Common Fund, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dorothy K Grange
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Wendy J Introne
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - William A Gahl
- NIH Undiagnosed Diseases Program, Common Fund, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - May Christine V Malicdan
- NIH Undiagnosed Diseases Program, Common Fund, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
54
|
Krishnaswamy SM, Arunachal G, Singh KG, Thomson VS, George P, Rao S, Danda S. Investigation of mutation spectrum amongst patients with familial primary cardiomyopathy using targeted NGS in Indian population. J Appl Genet 2024; 65:809-822. [PMID: 38551768 DOI: 10.1007/s13353-024-00855-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 11/14/2024]
Abstract
Genetic cardiomyopathies (CM) are disorders that affect morphology and function of cardiac muscle. Significant number of genes have been implicated in causing the phenotype. It is one of the leading genetic causes of death in young. We performed a study to understand the genetic variants in primary cardiomyopathies in an Indian cohort. Study comprised of 22 probands (13 with family history) representing hypertrophic (n = 10), dilated (n = 7), restrictive (n = 2) and arrhythmogenic ventricular(n = 3) cardiomyopathies. Genomic DNA was target captured with a panel of 46 genes and libraries sequenced on Illumina platform. Analysis identified, reported pathogenic as well as novel pathogenic (n = 6) variants in 16 probands. Of the 10 HCM patients, candidate variants were identified in nine of them involving sarcomere genes (62%, MYBPC3, MYH6, MYH7, MYL3, TTN), Z-disc (10%, ACTN2, LDB3, NEXN,), desmosome (10%, DSG2, DSP, PKP2) cytoskeletal (4%, DTNA) and ion channel (10% RYR2). In four DCM patients, variants were identified in genes NEXN, LMNA and TTN. Three arrhythmogenic right ventricular cardiomyopathy (ARVD) patients carried mutations in desmosome genes. Rare TTN variants were identified in multiple patients. Targeted capture and sequencing resulted in identification of candidate variants in about 70% of the samples which will help in management of disease in affected individual as well as in screening and early diagnosis in asymptomatic family members. Amongst the analysed cases, 22% were inconclusive without any significant variant identified. Study illustrates the utility of next-generation multi-gene panel as a cost-effective genetic testing to screen all forms of primary cardiomyopathies.
Collapse
Affiliation(s)
| | | | | | | | - Paul George
- Department of Cardiology, Christian Medical College, Vellore, India
| | - Sudha Rao
- Genotypic Technology Pvt. Ltd., Bangalore, Karnataka, 560094, India.
| | - Sumita Danda
- Department of Medical Genetics, Christian Medical College, Vellore, 632004, India.
| |
Collapse
|
55
|
Kovach AE, Komova D, Itov A, Gaskova M, Kalinina I, Voronin K, Rumiantseva Y, Karachunskii A, Maschan M, Maschan A, Novichkova G, Olshanskaya Y, Bhojwani D, Raca G, Zerkalenkova E. Pediatric therapy-related hematologic neoplasms show enrichment for KMT2A rearrangement and lymphoblastic phenotype. Leuk Lymphoma 2024; 65:1840-1852. [PMID: 39116419 DOI: 10.1080/10428194.2024.2376166] [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: 11/10/2023] [Revised: 06/25/2024] [Accepted: 06/29/2024] [Indexed: 08/10/2024]
Abstract
In children, therapy-related hematologic neoplasms (t-HN) are uncommon. Many are driven by genetic events independent of clonal hematopoiesis. We sought to understand the clinical and genetic factors of pediatric t-HN in a large independent cohort. Fifty-six t-HN were retrospectively identified. Chromosome microarray, next-generation and/or RNA sequencing were performed. Patients had primary hematologic, solid, or central nervous system tumors. t-HN included myeloid (t-MN) and lymphoblastic (t-ALL) phenotypes. Approximately half of the cases harbored KMTA2A rearrangement (KMT2Ar). Among t-HN without KMT2Ar, genetic drivers were heterogeneous, including diverse fusions or aneuploidy. Approximately 18% harbored 17p deletions and/or TP53 mutations. EFS/OS was not associated with t-HN lineage or KMT2Ar, but HSCT was associated with improved EFS and OS. We detail one of the largest cohorts to date of pediatric t-HN, confirming frequent KMT2Ar and t-ALL.
Collapse
Affiliation(s)
- Alexandra E Kovach
- Hematopathology, Division of Laboratory Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daria Komova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Albert Itov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Maria Gaskova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Irina Kalinina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Kirill Voronin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Yulia Rumiantseva
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexander Karachunskii
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Michael Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexey Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Galina Novichkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Yulia Olshanskaya
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Deepa Bhojwani
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Children's Hospital Los Angeles, CA, USA
| | - Gordana Raca
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Cytogenomics, Division of Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, CA, USA
| | - Elena Zerkalenkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| |
Collapse
|
56
|
Ilinca A, Kafantari E, Wallenius J, Kristoffersson U, Englund E, Puschmann A, Lindgren AG. Diagnosing Monogenic Stroke at Younger Age. Stroke 2024; 55:2846-2855. [PMID: 39498567 DOI: 10.1161/strokeaha.124.048044] [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: 06/02/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND An increasing number of monogenic conditions underlying stroke are being identified. We explored the possibilities of increasing the diagnostic yield of monogenic stroke in a population under 56 years of age. METHODS Fifty probands ≤55 years at their first stroke episode were characterized clinically and investigated by whole genome sequencing. Probands had one or more of: (1) one or more first to second degree relatives with stroke under 60 years or same stroke-causing condition/disease; (2) no hypertension, hypercholesterolemia, diabetes, heart disease, or smoking; or (3) either multiple stroke episodes or multiple arterial dissections. Variants with minor allele frequency under 0.01, identified by using our stroke gene panels, were assessed. The stroke subtypes, including large artery atherosclerotic, large artery nonatherosclerotic (tortuosity, dolichoectasia, aneurysm, nonatherosclerotic dissection, or occlusion), cerebral small vessel disease, cardioembolic (arrhythmia, heart defect, or cardiomyopathy), coagulation dysfunctions (venous thrombosis, arterial thrombosis, or bleeding tendency), intracerebral hemorrhage, vascular malformations (cavernoma or arteriovenous malformations), metabolic disorders, or cryptogenic embolic, were used for genotype-phenotype correlation. In a final step, we combined genetic and clinical information to determine if the genetic variant likely was the cause of stroke in the patients. RESULTS Whole genome sequencing of younger patients with stroke identified 17 clinically matching genetic variants in 15 of 50 (30%) patients, while a stronger clinical correlation with stroke was established in only 6 (12%) of them. Stroke-related genetic variants were identified in 4 of 5 (80%) patients with cardioembolic stroke subtype, 3 of 4 (75%) with intracerebral hemorrhage, 7 of 18 (39%) with cryptogenic embolic stroke, 1 of 6 (17%) with small vessel disease, and 3 of 15 (20%) of patients with nonatherosclerotic large artery stroke, including 1 of 11 (9%) with cervical dissection stroke. CONCLUSIONS Careful clinical interpretation of whole genome data using stroke gene panels can detect monogenic causes of early stroke, allowing individualized follow-up and opening new possibilities for potential treatment.
Collapse
Affiliation(s)
- Andreea Ilinca
- Department of Clinical Sciences Lund, Neurology, Lund University; Department of Neurology, Skåne University Hospital, Lund, Sweden (A.I., E.K., J.W., A.P., A.G.L.)
| | - Efthymia Kafantari
- Department of Clinical Sciences Lund, Neurology, Lund University; Department of Neurology, Skåne University Hospital, Lund, Sweden (A.I., E.K., J.W., A.P., A.G.L.)
| | - Joel Wallenius
- Department of Clinical Sciences Lund, Neurology, Lund University; Department of Neurology, Skåne University Hospital, Lund, Sweden (A.I., E.K., J.W., A.P., A.G.L.)
| | - Ulf Kristoffersson
- Department of Laboratory Medicine, Clinical Genetics, Lund University; Regional Laboratories, Region Skåne, Sweden (U.K.)
| | - Elisabet Englund
- Department of Clinical Sciences Lund, Pathology, Lund University; Regional Laboratories, Region Skåne, Sweden (E.E.)
| | - Andreas Puschmann
- Department of Clinical Sciences Lund, Neurology, Lund University; Department of Neurology, Skåne University Hospital, Lund, Sweden (A.I., E.K., J.W., A.P., A.G.L.)
- SciLifeLab National Research Infrastructure, Lund University, Sweden (A.P.)
| | - Arne G Lindgren
- Department of Clinical Sciences Lund, Neurology, Lund University; Department of Neurology, Skåne University Hospital, Lund, Sweden (A.I., E.K., J.W., A.P., A.G.L.)
| |
Collapse
|
57
|
Morandi A, Fornari E, Corradi M, Umano GR, Olivieri F, Piona C, Maguolo A, Panzeri C, Emiliani F, Cirillo G, Cavarzere P, Miraglia Del Giudice E, Maffeis C. Variant reclassification over time decreases the level of diagnostic uncertainty in monogenic obesity: Experience from two centres. Pediatr Obes 2024; 19:e13183. [PMID: 39462520 DOI: 10.1111/ijpo.13183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND The diagnosis of monogenic obesity is burdened by frequent variants of uncertain significance (VUS). We describe our real-life approach of variant reassessment over time and we assess whether inconclusive variants are decreasing in monogenic obesity. METHODS We tested for monogenic obesity (genes: LEPR, POMC, ADCY3, PCSK1, CARTPT, SIM1, MRAP2, LEP, NTRK2, BDNF, KSR2, MAGEL2, SH2B1, MC4R, MC3R) in 101 children/adolescents (11.7 [7.3-13.7] years, 3.6 [3.3-4.0] z-BMI) in Verona and 183 (11.3 [8.4-12.2] years, 3.2 [2.7-3.9] z-BMI) in Naples from January 2020 to February 2023. In March-July 2024 we reassessed the baseline variants by updated software interpretation and literature renavigation. RESULTS We initially found 20 VUS, 4 Likely Pathogenic (LP), 5 Likely Benign (LB) and 1 benign variant in 33 individuals. At follow-up, 6 VUS were reclassified as benign/LB, one LP as pathogenic and 3 LB as benign. Overall, 10/30 variants (6/18 in Verona, 3/11 in Naples and a variant found in both centres) were reclassified, leading to a less uncertain report for 13 of 33 variant-carrying patients. Monogenic obesity was diagnosed in 3 probands in Verona and 4 in Naples, carrying variants at MC4R or NTRK2. CONCLUSION Our variant reassessment was effective to improve classification certainty for the 39% of patients and suggested that the molecular diagnosis of monogenic obesity is becoming more accurate over time.
Collapse
Affiliation(s)
- Anita Morandi
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Elena Fornari
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Massimiliano Corradi
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Giuseppina Rosaria Umano
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesca Olivieri
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Claudia Piona
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Alice Maguolo
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Carola Panzeri
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Federica Emiliani
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Grazia Cirillo
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paolo Cavarzere
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Emanuele Miraglia Del Giudice
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Claudio Maffeis
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| |
Collapse
|
58
|
Wang Y, Liu M, Gao Z, Hua C, Jiang J, Zheng Y, Dong Z, Cao Y, Choy KW, Zhu X, Kong X. Detection of genomic variants by genome sequencing in foetuses with central nervous system abnormalities. Ann Med 2024; 56:2399317. [PMID: 39239799 PMCID: PMC11382719 DOI: 10.1080/07853890.2024.2399317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 09/07/2024] Open
Abstract
OBJECTIVE Clinical validity of genome sequencing (GS) (>30×) has been preliminarily verified in the post-natal setting. This study is to investigate the potential utility of trio-GS as a prenatal test for diagnosis of central nervous system (CNS) anomalies. METHODS We performed trio-based GS on a prospective cohort of 17 foetuses with CNS abnormalities. Single nucleotide variation (SNV), small insertion and deletion (Indel), copy number variation (CNV), structural variant (SV), and regions with absence of heterozygosity (AOH) were analyzed and classified according to ACMG guidelines. RESULTS Trio-GS identified diagnostic findings in 29.4% (5/17) of foetuses, with pathogenic variants found in SON, L1CAM, KMT2D, and ASPM. Corpus callosum (CC) and cavum septum pellucidum (CSP) abnormalities were the most frequent CNS abnormalities (47.1%, 8/17) with a diagnostic yield of 50%. A total of 29.4% (5/17) foetuses had variants of uncertain significance (VUS). Particularly, maternal uniparental disomy 16 and a de novo mosaic 4p12p11 duplication were simultaneously detected in one foetus with abnormal sulcus development. In addition, parentally inherited chromosomal inversions were identified in two foetuses. CONCLUSION GS demonstrates its feasibility in providing genetic diagnosis for foetal CNS abnormalities and shows the potential to expand the application to foetuses with other ultrasound anomalies in prenatal diagnosis.
Collapse
Affiliation(s)
- Yanfei Wang
- Genetics and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meimei Liu
- Prenatal Diagnosis Center, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhi Gao
- Genetics and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunxiao Hua
- Genetics and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinna Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yuting Zheng
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Zirui Dong
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Ye Cao
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Kwong Wai Choy
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaofan Zhu
- Genetics and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangdong Kong
- Genetics and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
59
|
Buianova AA, Bazanova MV, Belova VA, Ilyina GA, Samitova AF, Shmitko AO, Balakina AV, Pavlova AS, Suchalko ON, Korostin DO, Machalov AS, Daikhes NA, Rebrikov DV. Heterogeneous Group of Genetically Determined Auditory Neuropathy Spectrum Disorders. Int J Mol Sci 2024; 25:12554. [PMID: 39684270 DOI: 10.3390/ijms252312554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Auditory neuropathy spectrum disorder (ANSD) is often missed by standard hearing tests, accounting for up to 10% of hearing impairments (HI) and commonly linked to variants in 23 genes. We assessed 122 children with HI, including 102 with sensorineural hearing loss (SNHL) and 20 with ANSD. SNHL patients were genotyped for common GJB2 variants using qPCR, while ANSD patients underwent whole exome sequencing, with variants analyzed across 249 genes. Homozygous GJB2 variants were found in 54.9% of SNHL patients. In 60% of ANSD patients, variants were detected in OTOF (25%), CDH23, TMC1, COL11A1, PRPS1, TWNK, and HOMER2 genes, including eight novel variants. Transient evoked otoacoustic emissions testing revealed differences at 4000 Hz (p = 0.0084) between the ANSD and SNHL groups. The auditory steady-state response (ASSR) test showed significant differences at 500 Hz (p = 2.69 × 10-4) and 1000 Hz (p = 0.0255) compared to pure-tone audiometry (PTA) in ANSD patients. Our questionnaire shows that the parents of children with SNHL often report an improved quality of life with hearing aids or cochlear implants, while parents of children with ANSD frequently experience uncertainty about outcomes (p = 0.0026), leading to lower satisfaction.
Collapse
Affiliation(s)
- Anastasiia A Buianova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Marina V Bazanova
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
| | - Vera A Belova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Galit A Ilyina
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Alina F Samitova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Anna O Shmitko
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Anna V Balakina
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
| | - Anna S Pavlova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Oleg N Suchalko
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Dmitriy O Korostin
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Anton S Machalov
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Nikolai A Daikhes
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Denis V Rebrikov
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
- FSBI 'National Medical Research Center for Obstetrics, Gynecology, and Perinatology Named After Academician V.I. Kulakov', 117198 Moscow, Russia
| |
Collapse
|
60
|
Yarali O, Gündoğdu Öğütlü ÖB, Saritaş S, Guler MC, Keskin F, Türkyilmaz A. Epilepsy genetics in the paediatric population of the Eastern Anatolia region of Turkey. J Neurogenet 2024:1-10. [PMID: 39551975 DOI: 10.1080/01677063.2024.2424777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/29/2024] [Indexed: 11/19/2024]
Abstract
This study investigates the genetic causes of epilepsy in 166 paediatric patients under the age of 16 from the East Anatolian region of Turkey, who were treated at Erzurum City Hospital between 2018 and 2023. Patients with early-onset seizures, a family history of epilepsy or intellectual disability was selected for genetic analysis using a next-generation sequencing (NGS) gene panel targeting 449 genes associated with epilepsy and epileptic encephalopathy. The analysis revealed that pathogenic or probable pathogenic mutations were present in 14.8% (32 patients), highlighting the significant role of genetic factors in the aetiology of epilepsy in this population. In addition, 30.6% (66 patients) carried variants of uncertain significance (VUS), which, although not classified as pathogenic, have potential clinical relevance. Many epilepsy-related genes follow an autosomal dominant inheritance pattern, meaning that VUSs may gain pathogenic significance as more data and global studies accumulate, emphasising the evolving nature of genetic research. In addition to genetic factors, other aetiological causes such as perinatal insults (15.3%) and infections (7.9%) were identified, highlighting the multifactorial origin of epilepsy. While pathogenic mutations currently serve as important diagnostic and therapeutic markers, the role of VUS should not be underestimated. Genetic testing has proven to be essential for understanding the complex causes of epilepsy, providing opportunities for personalised treatment and genetic counselling. This study highlights the importance of genetic testing in regions such as Eastern Anatolia, where both environmental and genetic factors may influence the prevalence of epilepsy. As genetic databases expand, it is likely that the understanding of VUS will evolve, improving the clinical management of epilepsy through more targeted therapies and improved outcomes.
Collapse
Affiliation(s)
- Oğuzhan Yarali
- Department of Medical Genetics, Erzurum Regional Training and Research Hospital, Erzurum, Turkey
| | | | - Serdar Saritaş
- Department of Pediatric Neurology, Erzurum Regional Training and Research Hospital, Erzurum, Turkey
| | - Mustafa Can Guler
- Department of Physiology, Ataturk University Training and Research Hospital, Erzurum, Turkey
| | - Filiz Keskin
- Department of Pediatric Neurology, Erzurum Regional Training and Research Hospital, Erzurum, Turkey
| | - Ayberk Türkyilmaz
- Department of Medical Genetics, Karadeniz Technical University Faculty of Medicine, Trabzon, Turkey
| |
Collapse
|
61
|
Wang C, Chen J, Wang Y, Luo N, Han T, Yin X, Song Y, Chen D, Gong J. Genetic and clinical characteristics of genetic tumor syndromes in the central nervous system cancers: Implications for clinical practice. iScience 2024; 27:111073. [PMID: 39493880 PMCID: PMC11530818 DOI: 10.1016/j.isci.2024.111073] [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: 05/28/2024] [Revised: 08/02/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Recognizing individuals with Genetic tumor syndromes (GTS) in the primary central nervous system (CNS) tumors is crucial for optimizing proper genetic counseling and improving therapeutics and clinical care. We retrospectively analyzed the GTS in a Chinese CNS tumor cohort and examined the molecular characteristics and their clinical significance for diagnostic and therapeutic purposes. Our study identified 34 categories of GTS in 258 patients with CNS tumors. The gene with the highest germline pathogenic or likely pathogenic mutation frequency was TP53, followed by MSH2, NF1, and BRCA2. The top five GTS in CNS tumors showed high genetic heterogeneity GTS analysis reclassifies CNS tumors as "NEC." 53.88% of patients diagnosed with GTS harbor potential precision oncology therapy target mutations. The results of our study deepen our understanding of CNS tumors, provide a reference direction for the future design of clinical trials, and further expect to improve disease entire process management in CNS tumors.
Collapse
Affiliation(s)
- Chuanwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, Shandong 250012, China
| | - Jian Chen
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Yanzhao Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, Shandong 250012, China
| | - Ningning Luo
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, Jiangsu 210000, China
| | - Tiantian Han
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, Jiangsu 210000, China
| | - Xiangyu Yin
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, Jiangsu 210000, China
| | - Yunjie Song
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, Jiangsu 210000, China
| | - Dongsheng Chen
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, Jiangsu 210000, China
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, China
- Center of Translational Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Jie Gong
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, Shandong 250012, China
| |
Collapse
|
62
|
Yin Z, Zhang C, Dong R, Zhang X, Song Y, Hao S, Gai Z, Zhou B, Hui L, Wang S, Xue H, Cao Z, Liu Y, Ma X. Improving methylmalonic acidemia (MMA) screening and MMA genotype prediction using random forest classifier in two Chinese populations. Eur J Med Res 2024; 29:540. [PMID: 39523381 PMCID: PMC11552112 DOI: 10.1186/s40001-024-02115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Methylmalonic acidemia (MMA) is one of the most common hereditary organic acid metabolism disorders that endangers the lives and health of infants and children. Early detection and intervention before the appearance of a newborn's clinical symptoms can control disease progression and prevent or mitigate its serious consequences. METHODS 42,004 newborns from two Chinese populations were included in the study. The small molecular metabolite analytes were detected from the dried blood spot (DBS) samples by MS/MS. Genetic analysis of 68 Chinese MMA cases were performed by whole-exome sequencing and Sanger sequencing. Random forest classifiers (RFC) were constructed to improve the MMA screening performance and genotype prediction in two Chinese populations. Meanwhile, other six machine learning models were trained to separate MMA patients from normal newborns. Model performance was assessed using accuracy, sensitivity, specificity, false positive rate (FPR), and positive predictive value (PPV) and the area under the receiver operating characteristic curve (AUC). RESULTS In the total 42,004 newborn samples, 68 MMA cases were identified by genetic analysis, 42 cases of which were caused by variants in MMACHC, 24 cases by variants in MMUT, and two cases by variants in MMAA. Three novel variants including c.449T>G (p.I150R) of MMACHC, c.1151C>T (p.S384F) and c.1091_1108delins (p.Y364Sfs*4) in MMUT were identified in the MMA patients. RFC for newborn screening of MMA performed best as compared to several other classification models based on machine learning with 100% sensitivity, low FPR, excellent PPV and AUC. In addition, the subdivision RFC for MMA genotype prediction was constructed with superior performance. CONCLUSIONS It can be seen that RFC is extremely helpful for detection and genotype prediction in the newborn MMA screening. In addition, our findings extend the variant spectrum of genes related to MMA.
Collapse
Affiliation(s)
- Zhe Yin
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Chuan Zhang
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
- Gansu Province Medical Genetics Center, Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Rui Dong
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Xinyuan Zhang
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Yingnan Song
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Shengju Hao
- Gansu Province Medical Genetics Center, Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China
| | - Zhongtao Gai
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Bingbo Zhou
- Gansu Province Medical Genetics Center, Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China
| | - Ling Hui
- Gansu Province Medical Genetics Center, Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China
| | - Shifan Wang
- Gansu Province Medical Genetics Center, Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China
| | - Huiqin Xue
- Department of Cytogenetic Laboratory, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Zongfu Cao
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China.
- Graduate School of Peking Union Medical College, Beijing, China.
| | - Yi Liu
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, China.
| | - Xu Ma
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China.
- Graduate School of Peking Union Medical College, Beijing, China.
| |
Collapse
|
63
|
Lafraoui I, Heddar A, Cantalloube A, Braham I, Peigné M, Beneteau C, Gricourt S, Poirsier C, Legrand S, Stoeva R, Metayer-Amelot L, Lobersztajn A, Lebrun S, Gruchy N, Abdennebi I, Cedrin-Durnerin I, Fernandez H, Luton D, Torre A, Zagdoun L, Chevalier N, Khrouf M, Mahmoud K, Epelboin S, Catteau-Jonard S, Misrahi M. Genetic Landscape of a Cohort of 120 Patients with Diminished Ovarian Reserve: Correlation with Infertility. Int J Mol Sci 2024; 25:11915. [PMID: 39595984 PMCID: PMC11593603 DOI: 10.3390/ijms252211915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI) are major causes of female infertility. We recently found a monogenic etiology in 29.3% of POI, leading to personalized medicine. The genetic landscape of DOR is unknown. A prospective study (2018-2023) of an international cohort of 120 patients with unexplained DOR was performed using a large custom targeted next-generation sequencing panel including all known POI-causing genes. The diagnostic yield, based on the American College of Medical Genetics, was 24, 2%. Genes belong to different pathways: metabolism and mitochondria (29.7%), follicular growth (24.3%), DNA repair/meiosis (18.9%), aging (16.2%), ovarian development (8.1%), and autophagy (2.7%). Five genes were recurrently found: LMNA, ERCC6, SOX8, POLG, and BMPR1B. Six genes identified in single families with POI were involved in DOR, GNAS, TGFBR3, XPNPEP2, EXO1, BNC1, ATG, highlighting their role in maintaining ovarian reserve. In our cohort, 26 pregnancies were recorded, but no pregnancy was observed when meiosis/DNA repair genes were involved, suggesting severely impaired oocyte quality. Additional studies should confirm these preliminary results. This study with a large NGS panel defines the genetic landscape of a large cohort of DOR. It supports routine genetic diagnosis. Genetics could be a biomarker predicting infertility and progression to POI.
Collapse
Affiliation(s)
- Imène Lafraoui
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| | - Abdelkader Heddar
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| | - Adèle Cantalloube
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Inès Braham
- Service d’Endocrinologie, Diabétologie et Médecine de la Reproduction, CHU de Nice, 06000 Nice, France; (I.B.); (N.C.)
| | - Maëliss Peigné
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean-Verdier, Université Sorbonne Paris Nord, 93430 Bondy, France; (M.P.); (I.C.-D.)
| | - Claire Beneteau
- Service de Génétique Médicale, CHU de Nantes, 44000 Nantes, France;
| | - Solenne Gricourt
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Claire Poirsier
- Departement de Genetique, Centre Hospitalier Universitaire de Reims, 51092 Reims, France;
| | - Stéphanie Legrand
- Centre de Fertilité, Clinique de l’Atlantique, 17138 La Rochelle, France;
| | - Radka Stoeva
- Laboratoire de Génétique Médicale et Cytogénétique, CH Le Mans, 72037 Le Mans, France;
| | - Laure Metayer-Amelot
- Service d’Endocrinologie et Médecine de la Reproduction, CH Le Mans, 72037 Le Mans, France;
| | - Annina Lobersztajn
- Centre de la Fertilité—Paris Est, Nogent sur Marne, 94130 Nogent-sur-Marne, France;
| | - Soizic Lebrun
- Service de Génétique, FHU GenOMedS, CHRU de Tours, 37000 Tours, France;
| | - Nicolas Gruchy
- EA 7450 BioTARGen, FHU G4 Genomics, Service de Génétique Clinique, Departement de Genetique, CHU Côte de Nacre, Université de Caen Normandie UNICAEN, 14000 Caen, France;
| | - Inès Abdennebi
- Centre d’Aide Médicale à la Procréation, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France;
| | - Isabelle Cedrin-Durnerin
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean-Verdier, Université Sorbonne Paris Nord, 93430 Bondy, France; (M.P.); (I.C.-D.)
| | - Hervé Fernandez
- Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicêtre, France; (H.F.); (D.L.)
| | - Dominique Luton
- Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicêtre, France; (H.F.); (D.L.)
| | - Antoine Torre
- Centre d’Assistance Médicale à la Procréation Clinico-Biologique, Centre Hospitalier Sud Francilien Corbeil-Essonnes, 91100 Corbeil-Essonnes, France;
| | - Léonore Zagdoun
- Service de Diabétologie et Endocrinologie, Centre Hospitalier de Mont de Marsan et Pays des Sources, 40024 Mont de Marsan, France;
| | - Nicolas Chevalier
- Service d’Endocrinologie, Diabétologie et Médecine de la Reproduction, CHU de Nice, 06000 Nice, France; (I.B.); (N.C.)
| | - Mohamed Khrouf
- Centre FERTILLIA de Médecine de la Reproduction-Clinique la Rose, Tunis 1053, Tunisia; (M.K.); (K.M.)
| | - Khaled Mahmoud
- Centre FERTILLIA de Médecine de la Reproduction-Clinique la Rose, Tunis 1053, Tunisia; (M.K.); (K.M.)
| | - Sylvie Epelboin
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Sophie Catteau-Jonard
- Service de Gynécologie Endocrinienne, CHU de Lille, Hôpital Jeanne-de-Flandre, 59000 Lille, France;
| | - Micheline Misrahi
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| |
Collapse
|
64
|
Toreli ACM, Toni I, de Albuquerque DM, Lanaro C, Maues JH, Fertrin KY, Campos PDM, Costa FF. Investigation of BMP6 mutations in Brazilian patients with iron overload. Hematol Transfus Cell Ther 2024; 46 Suppl 5:S197-S200. [PMID: 38719717 PMCID: PMC11670610 DOI: 10.1016/j.htct.2024.04.001] [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: 12/05/2024] Open
Abstract
BACKGROUND Iron overload (IO) is a complex condition in which clinical, behavioral and genetic factors contribute to the phenotype. In multiethnic and non-Caucasian populations, mutations in HFE gene alone cannot explain IO in most of the cases, and additional genetic and environmental factors must be investigated. Bone Morphogenetic Proteins (BMPs) play a central role in iron homeostasis by modulating HAMP transcription through the signaling pathway that includes SMAD and HJV. In this study, we aimed to explore the clinical relevance of BMP6 mutations in a cohort of Brazilian patients with IO. METHODS 41 patients with IO were evaluated. Blood samples were collected to analyze BMP6 mutations through New Sequence Generations (NGS). Frequency of variants and mutations were analyzed and correlated with clinical and environmental characteristics. RESULTS We identified BMP6 mutations in three patients with IO. The p.Arg257His mutation was identified in two patients and the p.Leu71Val mutation was identified in one patient. Two of these patients had additional risk factors for IO (HFE mutations and diabetes mellitus). CONCLUSION BMP6 mutations, when combined to other genetic and clinical risk factors, may contribute to IO. Functional studies and THE evaluation of large cohorts are necessary to fully address BMP6 role in IO.
Collapse
Affiliation(s)
| | - Isabella Toni
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, SP, Brazil
| | | | - Carolina Lanaro
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, SP, Brazil
| | - Jersey Heitor Maues
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, SP, Brazil
| | | | - Paula de Melo Campos
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas, SP, Brazil
| | | |
Collapse
|
65
|
Awasthi S, Kumar R, Pradhan D, Rawal N, Goel H, Sahu P, Sisodiya S, Rana R, Kumar S, Dash NR, Das P, Agrawal U, Rath GK, Kaur T, Dhaliwal RS, Hussain S, Saluja SS, Tanwar P. Genomic landscape of gallbladder cancer: insights from whole exome sequencing. Int J Surg 2024; 110:6883-6897. [PMID: 39166960 PMCID: PMC11573093 DOI: 10.1097/js9.0000000000002031] [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: 05/14/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Gallbladder cancer (GBC) is a common gastrointestinal malignancy noted for its aggressive characteristics and poor prognosis, which is mostly caused by delayed detection. However, the scarcity of information regarding somatic mutations in Indian patients with GBC has hampered the development of efficient therapeutic options. In the present study, the authors attempted to bridge this gap by revealing the mutational profile of GBC. MATERIALS AND METHODS To evaluate the somatic mutation profile, whole exome sequencing (WES) was performed on 66 tumor and matched blood samples from individuals with GBC. Somatic variant calling was performed using GATK pipeline. Variants were annotated at pathogenic and oncogenic levels, using ANNOVAR, VEP tools and the OncoKB database. Mutational signature analysis, oncogenic pathway analysis and cancer driver genes identification were performed at the functional level by using the maftools package. RESULTS Our findings focused on the eight most altered genes with pathogenic and oncogenic mutations: TP53, SMAD4, ERBB3, KRAS, ARID1A, PIK3CA, RB1, and AXIN1. Genes with pathogenic single nucleotide variations (SNVs) were enriched in oncogenic signaling pathways, particularly RTK-RAS, WNT, and TP53 pathways. Furthermore, our research related certain mutational signatures, such as cosmic 1, cosmic 6, and cosmic 18, 29, to known characteristics including patient age and tobacco smoking, providing important insights into disease etiology. CONCLUSIONS Given the scarcity of exome-based sequencing studies focusing on the Indian population, this study represents a significant step forward in providing a framework for additional in-depth mutational analysis. Genes with substantial oncogenic and pathogenic mutations are promising candidates for developing targeted mutation panels, particularly for GBC detection.
Collapse
Affiliation(s)
| | - Rahul Kumar
- Laboratory Oncology Unit, Dr. BRA-IRCH, AIIMS
| | | | - Neetu Rawal
- Laboratory Oncology Unit, Dr. BRA-IRCH, AIIMS
| | - Harsh Goel
- Laboratory Oncology Unit, Dr. BRA-IRCH, AIIMS
| | - Parameswar Sahu
- Department of Gastrointestinal Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research
| | - Sandeep Sisodiya
- Division of Molecular Oncology, ICMR-National Institute of Cancer Prevention Research, Noida, India
| | - Rashmi Rana
- Department of Biotechnology and Research, GRIPMER
| | - Sunil Kumar
- Department of Surgical Oncology, Dr. BRA-IRCH, AIIMS
| | | | | | - Usha Agrawal
- Ex Director, ICMR National Institute of Pathology
| | - GK Rath
- Ex Chief & Professor, Department of Radiotherapy, Dr. BRA-IRCH, AIIMS, New Delhi
| | - Tanvir Kaur
- Division of Non-Communicable Diseases, Indian Council of Medical Research
| | - RS Dhaliwal
- Division of Non-Communicable Diseases, Indian Council of Medical Research
| | - Showket Hussain
- Division of Molecular Oncology, ICMR-National Institute of Cancer Prevention Research, Noida, India
| | - Sundeep Singh Saluja
- Department of Gastrointestinal Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research
| | | |
Collapse
|
66
|
Acosta-Uribe J, Piña Escudero SD, Cochran JN, Taylor JW, Castruita PA, Jonson C, Barinaga EA, Roberts K, Levine AR, George DS, ÁvilaFunes JA, Behrens MI, Bruno MA, Brusco LI, Custodio N, Duran-Aniotz C, Lopera F, Matallana DL, Slachevsky A, Takada LT, Zapata-Restrepo LM, Durón-Reyes DE, de Paula França Resende E, Gelvez N, Godoy ME, Maito MA, Javandel S, Miller BL, Nalls MA, Leonard H, Vitale D, Bandres-Ciga S, Koretsky MJ, Singleton AB, Pantazis CB, Valcour V, Ibañez A, Kosik KS, Yokoyama JS, Multi-Partner Consortium to Expand Dementia Research in Latin America (ReDLat). Genetic Contributions to Alzheimer's Disease and Frontotemporal Dementia in Admixed Latin American Populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.29.24315197. [PMID: 39574875 PMCID: PMC11581085 DOI: 10.1101/2024.10.29.24315197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Background Latin America's diverse genetic makeup, shaped by centuries of admixture, presents a unique opportunity to study Alzheimer's disease dementia (AD) and frontotemporal dementia (FTD). Our aim is to identify genetic variations associated with AD and FTD within this population. Methods The Multi-Partner Consortium to Expand Dementia Research in Latin America (ReDLat) recruited 2,162 participants with AD, FTD, and healthy controls from six Latin American countries (Argentina, Brazil, Chile, Colombia, Mexico, and Peru). All participants underwent array, exome, and/or whole-genome sequencing. Population structure was analyzed using Principal Component Analysis and ADMIXTURE, projecting the ReDLat population onto the 1000 Genomes Project database. To identify genes associated with autosomal dominant, autosomal recessive, or X-linked forms of adult-onset dementia, we searched the Online Mendelian Inheritance in Man database and analyzed pedigree information. Variant interpretation followed guidelines from the American College of Medical Genetics and Genomics, and the Guerreiro algorithm was applied for the PSEN1 and PSEN2 genes. Results Global ancestry analysis of the ReDLat cohort revealed a predominant mix of American, African, and European ancestries. Uniquely, Brazil displayed an additional East Asian component accurately reflecting the historical admixture patterns from this region. We identified 17 pathogenic variants, a pathogenic C9orf72 expansion, and 44 variants of uncertain significance. Among our cohort, 70 families exhibited autosomal dominant inheritance of neurodegenerative diseases, with 48 families affected by AD and 22 by FTD. In families with AD, We discovered a novel variant in the PSEN1 gene, c.519G>T (p.Leu173Phe), along with other previously described variants seen in the region, such as c.356C>T (p.Thr119Ile). In families with FTD, the most commonly associated gene was GRN, followed by MAPT. Notably, we identified a patient meeting criteria for FTD who carried a pathogenic variant in SOD1, c.388G>A (p.Phe21Leu), which had previously been reported in another FTD patient from the same geographical region. Conclusions This study provides the first snapshot of genetic contributors to AD and FTD in a multisite cohort across Latin America. It will be critical to evaluate the generalizability of genetic risk factors for AD and FTD across diverse ancestral backgrounds, considering distinct social determinants of health and accounting for modifiable risk factors that may influence disease risk and resilience across different cultures.
Collapse
Affiliation(s)
- Juliana Acosta-Uribe
- Grupo de Neurociencias de Antioquia (GNA), Facultad de Medicina, Universidad de Antioquia. Medellín, Colombia
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara. Santa Barbara, USA
| | - Stefanie D. Piña Escudero
- Global Brain Health Institute (GBHI), Department of Neurology, University of California San Francisco. San Francisco, USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco. San Francisco, USA
| | | | | | - P. Alejandra Castruita
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco. San Francisco, USA
| | - Caroline Jonson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco. San Francisco, USA
- DataTecnica LLC, Washington DC, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
| | | | - Kevin Roberts
- HudsonAlpha Institute for Biotechnology. Huntsville, USA
| | - Alexandra R. Levine
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara. Santa Barbara, USA
| | - Dawwod S. George
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara. Santa Barbara, USA
| | - José Alberto ÁvilaFunes
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Ciudad de México, México
- Bordeaux Population Health Research Center, University of Bordeaux. Bordeaux, France
| | - María I. Behrens
- Departamento de Neurología y Psiquiatría, Clínica Alemana-Universidad del Desarrollo. Santiago, Chile
- Centro de Investigación Clínica Avanza (CICA), Departamento de Neurología y Neurocirugía and Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile. Santiago, Chile
| | - Martín A. Bruno
- Instituto de Ciencias Biomédicas (ICBM) Facultad de Ciencias Médicas, Universidad Católica de Cuyo. San Juan, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). San Juan, Argentina
| | - Luis I. Brusco
- Asociación Alzheimer Argentina (ALZAR). Buenos Aires, Argentina
- Universidad de Buenos Aires. Buenos Aires, Argentina
| | | | - Claudia Duran-Aniotz
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia (GNA), Facultad de Medicina, Universidad de Antioquia. Medellín, Colombia
| | - Diana L. Matallana
- Instituto de Envejecimiento, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
- Centro de Memoria y Cognición Intellectus, Hospital Universitario San Ignacio. Bogotá, Colombia
- Departamento de Salud Mental, Hospital Universitario Fundación Santa Fe. Bogotá, Colombia
| | - Andrea Slachevsky
- Departamento de Neurología y Psiquiatría, Clínica Alemana-Universidad del Desarrollo. Santiago, Chile
- Laboratorio de Neuropsicología y Neurociencias Cognitivas (LANNEC) del Programa de Fisiopatología del Instituto de Ciencias Biomédicas (ICBM), Departamento de Ciencias Neurológicas Oriente y Departamento de Neurociencias de la Facultad de Medicina de la Universidad de Chile. Santiago, Chile
- Centro de Gerociencia, Salud Mental y Metabolismo (GERO). Santiago, Chile
- Centro de Memoria y Neuropsiquiatría (CMYN), Departamento de Neurología. Hospital del Salvador, Chile
| | - Leonel T. Takada
- Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo. São Paulo, Brasil
| | - Lina M. Zapata-Restrepo
- Facultad de Ciencias de la Salud, Universidad Icesi. Cali, Colombia
- Fundación Valle de Lili. Cali, Colombia
| | - Dafne E. Durón-Reyes
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Ciudad de México, México
| | - Elisa de Paula França Resende
- Global Brain Health Institute (GBHI), Department of Neurology, University of California San Francisco. San Francisco, USA
- Faculdade de Medicina da Universidade Federal de Minas Gerais. Belo Horizonte, Brasil
| | - Nancy Gelvez
- Instituto de genética humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Maria E. Godoy
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
| | - Marcelo A. Maito
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
| | - Shireen Javandel
- Global Brain Health Institute (GBHI), Department of Neurology, University of California San Francisco. San Francisco, USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco. San Francisco, USA
| | - Bruce L. Miller
- Global Brain Health Institute (GBHI), Department of Neurology, University of California San Francisco. San Francisco, USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco. San Francisco, USA
| | - Mike A. Nalls
- DataTecnica LLC, Washington DC, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
| | - Hampton Leonard
- DataTecnica LLC, Washington DC, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
| | - Dan Vitale
- DataTecnica LLC, Washington DC, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
| | - Mathew J. Koretsky
- DataTecnica LLC, Washington DC, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
| | - Andrew B. Singleton
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
- National Institute on Aging, National Institutes of Health. Bethesda, USA
| | - Caroline B. Pantazis
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health. Bethesda, USA
| | - Victor Valcour
- Global Brain Health Institute (GBHI), Department of Neurology, University of California San Francisco. San Francisco, USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco. San Francisco, USA
- Division of Geriatric Medicine, University of California San Francisco. San Francisco, USA
| | - Agustin Ibañez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez. Santiago, Chile
- Global Brain Health Institute, Trinity College Dublin. Dublin, Ireland
| | - Kenneth S. Kosik
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara. Santa Barbara, USA
| | - Jennifer S. Yokoyama
- Global Brain Health Institute (GBHI), Department of Neurology, University of California San Francisco. San Francisco, USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco. San Francisco, USA
- Department of Radiology and Biomedical Imaging, University of California. San Francisco, USA
| | | |
Collapse
|
67
|
Li X, Yao Y, Xing S, Zheng YH, Zhou Y, Yu X, Su J, Chen S, Jin ZB. Trio-based whole-exome sequencing of 200 Chinese patients with keratoconus. Exp Eye Res 2024; 248:110109. [PMID: 39326774 DOI: 10.1016/j.exer.2024.110109] [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: 08/12/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Keratoconus (KC) is a complex corneal disorder with a well-recognized genetic component. In this study, we aimed to expand the genetic spectrum of 200 Chinese patients with keratoconus and their unaffected parents. Trio-based whole-exome sequencing was performed in 200 patients with sporadic keratoconus and their unaffected parents. The variants identified in candidate genes for keratoconus were analyzed using multiple bioinformatics tools. Finally, we identified 7 variants in 5 candidate genes for keratoconus in 5 patients. The c.T464C variant in the IMPDH1 gene was defined as likely pathogenic according to the guidelines of the American College of Medical Genetics and Genomics, and the remaining variants in candidate genes (TRANK1, SLC4A11, CERKL, IFT172) were defined as uncertain significance. Our results expand the genetic spectrum in KC, highlight the genetic heterogeneity of this disease and provide important clues for future functional validation.
Collapse
Affiliation(s)
- Xingyong Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; Taizhou Ophthalmology and Optometry Hospital, Taizhou, 318001, China
| | - Yinghao Yao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), The Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Shilai Xing
- Institute of PSI Genomics, Wenzhou Global Eye & Vision Innovation Center, Wenzhou, 325024, Zhejiang, China
| | - Yi-Han Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yang Zhou
- Taizhou Ophthalmology and Optometry Hospital, Taizhou, 318001, China
| | - Xiaoguang Yu
- Institute of PSI Genomics, Wenzhou Global Eye & Vision Innovation Center, Wenzhou, 325024, Zhejiang, China
| | - Jianzhong Su
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), The Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Shihao Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; Taizhou Ophthalmology and Optometry Hospital, Taizhou, 318001, China.
| | - Zi-Bing Jin
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China; Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, 100005, China
| |
Collapse
|
68
|
Shao YHJ, Liao CS, Hsu YC, Chiu YC, Lu TJ, Ou YC, Hsiao TH. Clinical relevance of protein-truncating variants of germline DNA repair genes in prostate cancer. BMC Cancer 2024; 24:1319. [PMID: 39455978 PMCID: PMC11520037 DOI: 10.1186/s12885-024-13045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Interpreting genetic variants remains a challenge in prostate cancer (PCa). Although many annotation tools are available for prioritizing causal variants, the clinical relevance of these variants is rarely studied. METHODS We collected a cohort study that included 274 PCa patients from June 2017 to December 2020 and sequenced 19 DNA damage repair (DDR) genes in these patients and explored the clinical consequence of these different approaches. We also examined all-cause and PCa-specific survival in DDR gene mutation carriers compared to non-carriers after androgen receptor (AR)-directed therapy. RESULTS We identified 13 variants from 19 DDR genes in a total of 14 (5.1%) patients who had at least one presumed pathogenic mutation using different annotation methods. Four variants were annotated as pathogenic, 11 variants were predicted as protein-truncating variants (PTVs), four variants received proxy-deleterious (Combined Annotation-Dependent Depletion scores of > 30), and only one variant was identified as a pathogenic variant or as having a functional effect by all three methods. PCa patients with PTVs were significantly associated with early onset, high cancer stage, and a worse response to AR-directed treatment. However, patients carrying a proxy-deleterious variant were only associated with a higher T (tumor) stage and N (node) stage than those without such a variant, but not associated with other clinical characteristics. In patients treated with AR-directed therapy, patients with a PTV showed an increased risk of all-cause death (adjusted hazard ratio (aHR) = 3.51, 95% confidence interval (CI): 1.06 ~ 11.56) and PCa-specific death (aHR = 4.49, 95% CI: 1.87 ~ 10.77) compared to non-PTV carriers after adjustment. We were unable to examine gene-specific risks due to the small number of patients. CONCLUSIONS PTVs may assist in guiding treatment and early screening in PCa, while population-specific data for pathogenic variants are still being amassed.
Collapse
Affiliation(s)
- Yu-Hsuan Joni Shao
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, 10675, Taiwan
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei, 10675, Taiwan
| | - Cai-Sian Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
| | - Yu-Ching Hsu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
- Bioinformatics Program, Taiwan International Graduate Program, National Taiwan University, Taipei, 106319, Taiwan
| | - Yu-Chiao Chiu
- Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Tsai-Jung Lu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
| | - Yen-Chuan Ou
- Department of Urology, Tungs' Taichung MetroHarbor Hospital, Taichung, 43503, Taiwan.
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 40705, Taiwan.
- Department of Public Health, Fu Jen Catholic University, New Taipei City, 24205, Taiwan.
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, 402202, Taiwan.
| |
Collapse
|
69
|
Kryukova OV, Islanov IO, Zaklyazminskaya EV, Korostin DO, Belova VA, Cheranev VV, Repinskaia ZA, Tonevitskaya SA, Petukhov PA, Dudek SM, Kost OA, Rebrikov DV, Danilov SM. Effects of Angiotensin-I-Converting Enzyme (ACE) Mutations Associated with Alzheimer's Disease on Blood ACE Phenotype. Biomedicines 2024; 12:2410. [PMID: 39457722 PMCID: PMC11504702 DOI: 10.3390/biomedicines12102410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUNDS Our recent analysis of 1200+ existing missense ACE mutations revealed that 400+ mutations are damaging and led us to hypothesize that carriers of heterozygous loss-of-function (LoF) ACE mutations (which result in low ACE levels) could be at risk for the development of late-onset Alzheimer's disease (AD). METHODS Here, we quantified blood ACE levels in EDTA plasma from 41 subjects with 10 different heterozygous ACE mutations, as well as 33 controls, and estimated the effect of these mutations on ACE phenotype using a set of mAbs to ACE and two ACE substrates. RESULTS We found that relatively frequent (~1%) AD-associated ACE mutations in the N domain of ACE, Y215C, and G325R are truly damaging and likely transport-deficient, with the ACE levels in plasma at only ~50% of controls. Another AD-associated ACE mutation, R1250Q, in the cytoplasmic tail, did not cause a decrease in ACE and likely did not affect surface ACE expression. We have also developed a method to identify patients with anti-catalytic mutations in the N domain. These mutations may result in reduced degradation of amyloid beta peptide Aβ42, an important component for amyloid deposition. Consequently, these could pose a risk factor for the development of AD. CONCLUSIONS Therefore, a systematic analysis of blood ACE levels in patients with all ACE mutations has the potential to identify individuals at an increased risk of late-onset AD. These individuals may benefit from future preventive or therapeutic interventions involving a combination of chemical and pharmacological chaperones, as well as proteasome inhibitors, aiming to enhance ACE protein traffic. This approach has been previously demonstrated in our cell model of the transport-deficient ACE mutation Q1069R.
Collapse
Affiliation(s)
- Olga V. Kryukova
- Faculty of Chemistry, M.V. Lomonosov Moscow University, 119991 Moscow, Russia; (O.V.K.); (O.A.K.)
| | - Igor O. Islanov
- Medical Genetics Department, Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia; (I.O.I.); (E.V.Z.)
| | - Elena V. Zaklyazminskaya
- Medical Genetics Department, Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia; (I.O.I.); (E.V.Z.)
| | - Dmitry O. Korostin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.O.K.); (V.A.B.); (V.V.C.); (Z.A.R.); (D.V.R.)
| | - Vera A. Belova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.O.K.); (V.A.B.); (V.V.C.); (Z.A.R.); (D.V.R.)
| | - Valery V. Cheranev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.O.K.); (V.A.B.); (V.V.C.); (Z.A.R.); (D.V.R.)
| | - Zhanna A. Repinskaia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.O.K.); (V.A.B.); (V.V.C.); (Z.A.R.); (D.V.R.)
| | - Svetlana A. Tonevitskaya
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 117418 Moscow, Russia;
| | - Pavel A. Petukhov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60612, USA;
| | - Steven M. Dudek
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois, Chicago, IL 60612, USA;
| | - Olga A. Kost
- Faculty of Chemistry, M.V. Lomonosov Moscow University, 119991 Moscow, Russia; (O.V.K.); (O.A.K.)
| | - Denis V. Rebrikov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.O.K.); (V.A.B.); (V.V.C.); (Z.A.R.); (D.V.R.)
| | - Sergei M. Danilov
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois, Chicago, IL 60612, USA;
| |
Collapse
|
70
|
Roos-Mattila M, Kallio P, Luck TJ, Polso M, Kumari R, Mikkonen P, Välimäki K, Malmstedt M, Ellonen P, Pellinen T, Heckman CA, Mustonen H, Puolakkainen PA, Alitalo K, Kallioniemi O, Mirtti T, Rannikko AS, Pietiäinen VM, Seppänen HE. Distinct molecular profiles and shared drug vulnerabilities in pancreatic metastases of renal cell carcinoma. Commun Biol 2024; 7:1355. [PMID: 39427059 PMCID: PMC11490566 DOI: 10.1038/s42003-024-07004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
Clear-cell renal cell carcinoma (ccRCC) is the most common origin of pancreatic metastases (PM). Distinct genomic aberrations, favorable prognosis, and clinical observations on high angiogenesis, and succeeding tyrosine kinase inhibitor (TKI) sensitivity have been reported in PM-ccRCC. However, no functional or single-cell studies have been conducted thus far. We recruited five PM-ccRCC patients and investigated the genomic, single-cell transcriptomic, and drug sensitivity profiles of their patient-derived cells (PDCs). The PM depicted both expected and novel genomic alterations. Further, the transcriptomics differed from both primary and metastatic ccRCC, with upregulations of the PI3K/mTOR and - supporting the clinical observations - angiogenesis pathways. Data integration at pathway level showed that transcriptomics explained drug sensitivities the best. Accordingly, PM-ccRCC PDCs shared sensitivity to many PI3K/mTOR inhibitors. Altogether, we show distinct genomic and transcriptomic signatures in PM-ccRCC, highlight the superiority of transcriptomics in interpreting drug sensitivities, and encourage the use of TKIs and PI3K/mTOR inhibitors in PM-ccRCC.
Collapse
Affiliation(s)
- Matilda Roos-Mattila
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland.
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.
| | - Pauliina Kallio
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Tamara J Luck
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Minttu Polso
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Romika Kumari
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Piia Mikkonen
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Katja Välimäki
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Minna Malmstedt
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- ONCOSYS Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Caroline A Heckman
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Harri Mustonen
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Pauli A Puolakkainen
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Kari Alitalo
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
- Science for Life Laboratory (SciLifeLab), Department of Oncology and Pathology, Karolinska Institutet, Solna, 17165, Sweden
| | - Tuomas Mirtti
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- ONCOSYS Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| | - Antti S Rannikko
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- ONCOSYS Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Vilja M Pietiäinen
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland -FIMM, Helsinki Institute for Life Sciences -HiLIFE, University of Helsinki, Helsinki, Finland
| | - Hanna E Seppänen
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| |
Collapse
|
71
|
Hu Y, He Z, Liu S, Ying W, Chen Y, Zhao M, He M, Wu X, Tang Y, Gu W, Ying M, Wang J, Tao T. Patient-derived rhabdomyosarcoma cells recapitulate the genetic and transcriptomic landscapes of primary tumors. iScience 2024; 27:110862. [PMID: 39319271 PMCID: PMC11417342 DOI: 10.1016/j.isci.2024.110862] [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: 05/13/2024] [Revised: 07/29/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in childhood and adolescence. The availability of appropriate and well-characterized preclinical models for RMS is limited, posing a challenge for investigating the molecular mechanisms and evaluating new targeted compounds in preclinical settings. Here, we collected 51 RMS specimens (referred to as ZJUCH-RMS cohort) and established 9 patient-derived cells (PDCs) and validated the identity of these cells by the expression of RMS-specific markers. Whole-transcriptome analysis identified high-confidence mutations in ZJUCH-RMS cohort including RAS, TP53, ARID1A, MYOD1, and MYCN. Further studies showed that RMS PDCs retained the genetic alterations and the expression of RMS hallmark and dependency genes in matched primary tumors and acted as valuable tools to assess drug responses and pharmacogenomic interactions. Our study provides unique PDCs that are available for preclinical studies of RMS and further advances the feasibility of RMS PDCs as valuable tools for developing personalized treatments for patients.
Collapse
Affiliation(s)
- Yuxiang Hu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- The First Clinical Institute, Zunyi Medical University, Zunyi 563000, China
| | - Ziqi He
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- The First Clinical Institute, Zunyi Medical University, Zunyi 563000, China
| | - Shuangai Liu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- The First Clinical Institute, Zunyi Medical University, Zunyi 563000, China
| | - Wenwen Ying
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yifan Chen
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Manli Zhao
- Department of Pathology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Min He
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Xuan Wu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Yinbing Tang
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Weizhong Gu
- Department of Pathology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Meidan Ying
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Jinhu Wang
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou 310052, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Ting Tao
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Department of Surgical Oncology, Children’s Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou 310052, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
72
|
Buianova A, Yukina M, Cheranev V, Suchalko O, Shmitko A, Samitova A, Nuralieva N, Kulagina E, Savvateeva E, Troshina E, Rebrikov D, Gryadunov D, Korostin D. Trio-based exome sequencing and high-resolution HLA typing in families of patients with autoimmune adrenal insufficiency and autoimmune polyglandular syndrome. PLoS One 2024; 19:e0312335. [PMID: 39423205 PMCID: PMC11488712 DOI: 10.1371/journal.pone.0312335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024] Open
Abstract
Autoimmune adrenal insufficiency (AAI) is a rare disease. This research evaluates three patients with AAI, including autoimmune polyglandular syndrome (APS) type 2. Two patients had APS or AAI during childhood, and one had a history of endocrine autoimmune disease, indicating a possible hereditary basis of the condition. Trio-based exome sequencing and high-resolution HLA typing were employed to analyze patients and their parents. Benign or likely benign variants of the AIRE gene were identified in all participants of the study. These variants, coupled with clinical data and the results of antibody studies to type I interferons, helped to exclude APS-1. Patients with APS-2, in contrast to patient with AAI, inherited distinct variants of unknown significance in the CLEC16A gene, which is associated with autoimmune diseases, including AAI. Various risk alleles in other genes associated with autoimmunity were identified in all patients. HLA typing of class II loci revealed alleles related to APS. Nevertheless, the frequencies of the haplotypes identified are substantial in the healthy Russian population. Immunological tests can detect antibody carriers and assess the risk of autoimmune disease development. In the future, to identify genetic predictors of autoimmune endocrinopathies, it is recommended to analyze the whole genome of patients and their relatives, examining clinically relevant variants in non-coding regions.
Collapse
Affiliation(s)
- Anastasiia Buianova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Marina Yukina
- Endocrinology Research Centre, Ministry of Health of Russia, Moscow, Russia
| | - Valery Cheranev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Oleg Suchalko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Anna Shmitko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alina Samitova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Nurana Nuralieva
- Endocrinology Research Centre, Ministry of Health of Russia, Moscow, Russia
| | - Elena Kulagina
- Engelhardt Institute of Molecular Biology (EIMB), Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Russian Academy of Sciences, Moscow, Russia
| | - Elena Savvateeva
- Engelhardt Institute of Molecular Biology (EIMB), Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina Troshina
- Endocrinology Research Centre, Ministry of Health of Russia, Moscow, Russia
| | - Denis Rebrikov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Dmitry Gryadunov
- Engelhardt Institute of Molecular Biology (EIMB), Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Russian Academy of Sciences, Moscow, Russia
| | - Dmitriy Korostin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
73
|
Skrypnyk C, AlHarmi R, Mathur A, AlHafnawi HH, Chandan Appikonda SH, Matsa LS. Expanding families: a pilot study on preconception expanded carrier screening in Bahrain. BMC Pregnancy Childbirth 2024; 24:684. [PMID: 39425040 PMCID: PMC11490169 DOI: 10.1186/s12884-024-06878-1] [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: 07/18/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Preconception expanded carrier screening (ECS) is a genetic test that enables the identification of at-risk carriers of recessive disorders by screening for up to hundreds of genes. Next-generation sequencing (NGS) development has paved the way for its integration into ECS. This study aims to identify the carrier genetic status of couples experiencing or anticipating conception challenges through NGS-based ECS and to gain an overview of the rare genetic disorders in a population with increased consanguinity. METHODS Thirty couples who presented to the Genetic Disease Clinic between 2015 and 2024 with failed reproductive outcomes or with a positive personal or family history of genetic disorders and underwent ECS were included and retrospectively analyzed. RESULTS Fifty-four individuals (90.00%) were found to carry at least one variant of 95 identified genes, totaling 174 variants. Six individuals (10.00%) tested negative for any variant. Seven individuals had one variant (11.67%), 13 had two variants (21.67%), and 34 had 3 or more variants (56.67%). The most common variants identified were of HBA, HBB, CYP21A2, and G6PD genes. Most of the detected variants were unknown or unexpected (n = 143, 82.18%). Eight couples carried two or more variants in common. Consanguinity was reported in 14 couples (46.67%). CONCLUSIONS Preconception ECS is crucial for reproductive planning, permitting couples to evaluate their combined genetic risks and make informed decisions, reducing the chance of having children with genetic disorders.
Collapse
Affiliation(s)
- Cristina Skrypnyk
- Department of Molecular Medicine, AlJawhara Center, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain.
- Genetic Disease Clinic, University Medical Center, King Abdullah Medical City, Manama, Bahrain.
| | - Rawan AlHarmi
- Regenerative Medicine Unit, AlJawhara Center, Arabian Gulf University, Manama, Bahrain
| | - Aanchal Mathur
- Regenerative Medicine Unit, AlJawhara Center, Arabian Gulf University, Manama, Bahrain
| | - Hussein Hifnawi AlHafnawi
- Clinical Research Center, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
| | | | | |
Collapse
|
74
|
Lesurf R, Breckpot J, Bouwmeester J, Hanafi N, Jain A, Liang Y, Papaz T, Lougheed J, Mondal T, Alsalehi M, Altamirano-Diaz L, Oechslin E, Audain E, Dombrowsky G, Postma AV, Woudstra OI, Bouma BJ, Hitz MP, Bezzina CR, Blue GM, Winlaw DS, Mital S. A validated heart-specific model for splice-disrupting variants in childhood heart disease. Genome Med 2024; 16:119. [PMID: 39402625 PMCID: PMC11476204 DOI: 10.1186/s13073-024-01383-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Congenital heart disease (CHD) is the most common congenital anomaly. Almost 90% of isolated cases have an unexplained genetic etiology after clinical testing. Non-canonical splice variants that disrupt mRNA splicing through the loss or creation of exon boundaries are not routinely captured and/or evaluated by standard clinical genetic tests. Recent computational algorithms such as SpliceAI have shown an ability to predict such variants, but are not specific to cardiac-expressed genes and transcriptional isoforms. METHODS We used genome sequencing (GS) (n = 1101 CHD probands) and myocardial RNA-Sequencing (RNA-Seq) (n = 154 CHD and n = 43 cardiomyopathy probands) to identify and validate splice disrupting variants, and to develop a heart-specific model for canonical and non-canonical splice variants that can be applied to patients with CHD and cardiomyopathy. Two thousand five hundred seventy GS samples from the Medical Genome Reference Bank were analyzed as healthy controls. RESULTS Of 8583 rare DNA splice-disrupting variants initially identified using SpliceAI, 100 were associated with altered splice junctions in the corresponding patient myocardium affecting 95 genes. Using strength of myocardial gene expression and genome-wide DNA variant features that were confirmed to affect splicing in myocardial RNA, we trained a machine learning model for predicting cardiac-specific splice-disrupting variants (AUC 0.86 on internal validation). In a validation set of 48 CHD probands, the cardiac-specific model outperformed a SpliceAI model alone (AUC 0.94 vs 0.67 respectively). Application of this model to an additional 947 CHD probands with only GS data identified 1% patients with canonical and 11% patients with non-canonical splice-disrupting variants in CHD genes. Forty-nine percent of predicted splice-disrupting variants were intronic and > 10 bp from existing splice junctions. The burden of high-confidence splice-disrupting variants in CHD genes was 1.28-fold higher in CHD cases compared with healthy controls. CONCLUSIONS A new cardiac-specific in silico model was developed using complementary GS and RNA-Seq data that improved genetic yield by identifying a significant burden of non-canonical splice variants associated with CHD that would not be detectable through panel or exome sequencing.
Collapse
Affiliation(s)
- Robert Lesurf
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Jade Bouwmeester
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Nour Hanafi
- The Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anjali Jain
- The Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yijing Liang
- The Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Tanya Papaz
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Jane Lougheed
- Division of Cardiology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Tapas Mondal
- Division of Cardiology, Department of Pediatrics, McMaster Children's Hospital, Hamilton, ON, Canada
| | - Mahmoud Alsalehi
- Division of Cardiology, Department of Pediatrics, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Luis Altamirano-Diaz
- Division of Cardiology, Department of Pediatrics, London Health Sciences Centre, London, ON, Canada
| | - Erwin Oechslin
- Division of Cardiology, Department of Medicine, Toronto Adult Congenital Heart Disease Program at Peter Munk Cardiac Centre, University Health Network, and University of Toronto, Toronto, ON, Canada
| | - Enrique Audain
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Kiel, Germany
| | - Gregor Dombrowsky
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Alex V Postma
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Odilia I Woudstra
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Berto J Bouma
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Marc-Phillip Hitz
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Kiel, Germany
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Gillian M Blue
- Heart Centre for Children, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - David S Winlaw
- Heart Center, Ann and Robert H. Lurie Children's Hospital of Chicago and Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Seema Mital
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.
- Ted Rogers Centre for Heart Research, Toronto, ON, Canada.
- Division of Cardiology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
75
|
Kim J, Ney G, Frone MN, Haley JS, Mirshahi UL, Astiazaran-Symonds E, Shandrina M, Urban G, Rao HS, Stahl R, Golden A, Yohe ME, Gross AM, Ding Y, Carey DJ, Gelb BD, Stewart DR. Genomic ascertainment to quantify prevalence and cancer risk in adults with pathogenic and likely pathogenic germline variants in RASopathy genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.09.24314324. [PMID: 39802765 PMCID: PMC11722494 DOI: 10.1101/2024.10.09.24314324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Purpose Genomic ascertainment of electronic health record-linked exome data in two large biobanks was used to quantify germline pathogenic/likely pathogenic (P/LP) variant prevalence, cancer prevalence, and survival in adults with non-NF1 RAS/mitogen-activated protein kinase genes (RASopathies). Patients and Methods Germline RASopathy variants were examined from adult participants in UK Biobank (UKBB; n=469,802), Geisinger MyCode (n=167,050) and Mount Sinai BioMe (n=30,470). Variants were classified as per American College of Medical Genetics/Association for Molecular Pathology criteria and reviewed by a RASopathy variant expert. Heterozygotes harbored a RASopathy pathogenic/likely pathogenic variant; controls harbored wild type or benign/likely benign RASopathy variation. To distinguish germline variants from clonal hematopoiesis, benign tissues were Sanger sequenced. Tumor phenotype and demographic data were retrieved from MyCode and UKBB. Results Pathogenic variants in Noonan syndrome-associated genes (excluding known Noonan syndrome with multiple lentigines variants) were the most common with an estimated prevalence that ranged between 1:1,772-1:3,330 in the three cohorts. Pathogenic variants in cardiofaciocutaneous syndrome-associated genes had an estimated prevalence of 1:41,762-1:55,683 in two cohorts. Pathogenic variants in SPRED1 (Legius syndrome) were more frequent in UKBB (1:19,567 [95%CI: 1:13,150-1:29,116]) compared to MyCode (1:41,762 [95%CI: 1:15,185-1:130,367]). In SPRED1-heterozygotes, cancer prevalence was significantly increased in UKBB (OR:3.8 [95% CI: 2.48-8.64]; p=1.2×10-3) but not in the MyCode cohort. Pathogenic variants in HRAS (Costello syndrome) were not identified. In MyCode and UKBB cohorts, there was no significant increase in cancer prevalence in individuals with Noonan-, CBL- and CFC syndrome-associated pathogenic variants. Conclusion Genomic ascertainment from two large biobanks did not show evidence of elevated cancer risk in adult Noonan syndrome heterozygotes. There may be an increased cancer risk for adult SPRED1 heterozygotes.
Collapse
Affiliation(s)
- Jung Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Gina Ney
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Megan N Frone
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Jeremy S Haley
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | | | | | - Mariya Shandrina
- Mindich Child Health and Development Institute and the Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gretchen Urban
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - H Shanker Rao
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Rick Stahl
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Alicia Golden
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Marielle E Yohe
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Andrea M Gross
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Yi Ding
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - David J Carey
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and the Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| |
Collapse
|
76
|
Carpentieri G, Cecchetti S, Bocchinfuso G, Radio FC, Leoni C, Onesimo R, Calligari P, Pietrantoni A, Ciolfi A, Ferilli M, Calderan C, Cappuccio G, Martinelli S, Messina E, Caputo V, Hüffmeier U, Mignot C, Auvin S, Capri Y, Lourenco CM, Russell BE, Neustad A, Brunetti Pierri N, Keren B, Reis A, Cohen JS, Heidlebaugh A, Smith C, Thiel CT, Salviati L, Zampino G, Campeau PM, Stella L, Tartaglia M, Flex E. Dominantly acting variants in ATP6V1C1 and ATP6V1B2 cause a multisystem phenotypic spectrum by altering lysosomal and/or autophagosome function. HGG ADVANCES 2024; 5:100349. [PMID: 39210597 PMCID: PMC11465052 DOI: 10.1016/j.xhgg.2024.100349] [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: 06/06/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is a functionally conserved multimeric complex localized at the membranes of many organelles where its proton-pumping action is required for proper lumen acidification. The V-ATPase complex is composed of several subunits, some of which have been linked to human disease. We and others previously reported pathogenic dominantly acting variants in ATP6V1B2, the gene encoding the V1B2 subunit, as underlying a clinically variable phenotypic spectrum including dominant deafness-onychodystrophy (DDOD) syndrome, Zimmermann-Laband syndrome (ZLS), and deafness, onychodystrophy, osteodystrophy, intellectual disability, and seizures (DOORS) syndrome. Here, we report on an individual with features fitting DOORS syndrome caused by dysregulated ATP6V1C1 function, expand the clinical features associated with ATP6V1B2 pathogenic variants, and provide evidence that these ATP6V1C1/ATP6V1B2 amino acid substitutions result in a gain-of-function mechanism upregulating V-ATPase function that drives increased lysosomal acidification. We demonstrate a disruptive effect of these ATP6V1B2/ATP6V1C1 variants on lysosomal morphology, localization, and function, resulting in a defective autophagic flux and accumulation of lysosomal substrates. We also show that the upregulated V-ATPase function affects cilium biogenesis, further documenting pleiotropy. This work identifies ATP6V1C1 as a new gene associated with a neurodevelopmental phenotype resembling DOORS syndrome, documents the occurrence of a phenotypic continuum between ZLS, and DDOD and DOORS syndromes, and classify these conditions as lysosomal disorders.
Collapse
Affiliation(s)
- Giovanna Carpentieri
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy; Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Serena Cecchetti
- Confocal Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Gianfranco Bocchinfuso
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome 00168, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome 00168, Italy
| | - Paolo Calligari
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Agostina Pietrantoni
- Electron Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Marco Ferilli
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Cristina Calderan
- Department of Women and Children's Health, University of Padua, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padua, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, "Federico II" University, 80131 Naples, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Elena Messina
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Viviana Caputo
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Ulrike Hüffmeier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Cyril Mignot
- Department of Genetics, La Pitié-Salpêtrière Hospital, Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France
| | - Stéphane Auvin
- Service de Neurologie Pediatrique, Hopital Universitaire Robert Debré, Université Paris Cité, 75935 Paris, France
| | - Yline Capri
- Department of Genetics, Robert-Debré University Hospital, Assistance Publique-Hopitaux de Paris, 75935 Paris, France
| | - Charles Marques Lourenco
- Faculdade de Medicina, Centro Universitario Estácio de Ribeirão Preto, Ribeirão Preto 14096-160, São Paulo, Brazil
| | - Bianca E Russell
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ahna Neustad
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Brunetti Pierri
- Department of Translational Medicine, "Federico II" University, 80131 Naples, Italy; Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy; Scuola Superiore Meridionale, Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| | - Boris Keren
- Department of Genetics, La Pitié-Salpêtrière Hospital, Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Julie S Cohen
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alexis Heidlebaugh
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Clay Smith
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Christian T Thiel
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg FAU, 91054 Erlangen, Germany
| | - Leonardo Salviati
- Department of Women and Children's Health, University of Padua, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padua, Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome 00168, Italy; Facoltà di Medicina e Chirurgia, Università Cattolica del S. Cuore, 00168 Rome, Italy
| | | | - Lorenzo Stella
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy.
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
77
|
Peirone S, Tirtei E, Campello A, Parlato C, Guarrera S, Mareschi K, Marini E, Asaftei SD, Bertero L, Papotti M, Priante F, Perrone S, Cereda M, Fagioli F. Impaired neutrophil-mediated cell death drives Ewing's Sarcoma in the background of Down syndrome. Front Oncol 2024; 14:1429833. [PMID: 39421445 PMCID: PMC11484044 DOI: 10.3389/fonc.2024.1429833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Ewing Sarcoma (EWS) has been reported in seven children with Down syndrome (DS). To date, a detailed assessment of this solid tumour in DS patients is yet to be made. Methods Here, we characterise a chemo-resistant mediastinal EWS in a 2-year-old DS child, the youngest ever reported case, by exploiting sequencing approaches. Results The tumour showed a neuroectodermal development driven by the EWSR1-FLI1 fusion. The inherited myeloperoxidase deficiency of the patient caused failure of neutrophil-mediated cell death and promoted genomic instability. Discussion In this context, the tumour underwent genome-wide near haploidisation resulting in a massive overexpression of pro-inflammatory cytokines. Recruitment of defective neutrophils fostered rapid evolution of this EWS.
Collapse
Affiliation(s)
- Serena Peirone
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
- Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
| | - Elisa Tirtei
- Paediatric Oncology Department, Regina Margherita Children’s Hospital, Turin, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Anna Campello
- Paediatric Oncology Department, Regina Margherita Children’s Hospital, Turin, Italy
| | - Caterina Parlato
- Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Simonetta Guarrera
- Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Katia Mareschi
- Paediatric Oncology Department, Regina Margherita Children’s Hospital, Turin, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Elena Marini
- Paediatric Oncology Department, Regina Margherita Children’s Hospital, Turin, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | | | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Mauro Papotti
- Pathology Unit, Department of Oncology, University of Turin, Turin, Italy
| | - Francesca Priante
- Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
- Department of Oncology, University of Torino, Candiolo, Italy
| | - Sarah Perrone
- Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
- Department of Oncology, University of Torino, Candiolo, Italy
| | - Matteo Cereda
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
- Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
| | - Franca Fagioli
- Paediatric Oncology Department, Regina Margherita Children’s Hospital, Turin, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| |
Collapse
|
78
|
Gianferante DM, Moore A, Spector LG, Wheeler W, Yang T, Hubbard A, Gorlick R, Patiño-Garcia A, Lecanda F, Flanagan AM, Amary F, Andrulis IL, Wunder JS, Thomas DM, Ballinger ML, Serra M, Hattinger C, Demerath E, Johnson W, Birmann BM, De Vivo I, Giles G, Teras LR, Arslan A, Vermeulen R, Sample J, Freedman ND, Huang WY, Chanock SJ, Savage SA, Berndt SI, Mirabello L. Genetically inferred birthweight, height, and puberty timing and risk of osteosarcoma. Cancer Epidemiol 2024; 92:102432. [PMID: 37596165 PMCID: PMC10869637 DOI: 10.1016/j.canep.2023.102432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/14/2023] [Indexed: 08/20/2023]
Abstract
INTRODUCTION Several studies have linked increased risk of osteosarcoma with tall stature, high birthweight, and early puberty, although evidence is inconsistent. We used genetic risk scores (GRS) based on established genetic loci for these traits and evaluated associations between genetically inferred birthweight, height, and puberty timing with osteosarcoma. METHODS Using genotype data from two genome-wide association studies, totaling 1039 cases and 2923 controls of European ancestry, association analyses were conducted using logistic regression for each study and meta-analyzed to estimate pooled odds ratios (ORs) and 95% confidence intervals (CIs). Subgroup analyses were conducted by case diagnosis age, metastasis status, tumor location, tumor histology, and presence of a known pathogenic variant in a cancer susceptibility gene. RESULTS Genetically inferred higher birthweight was associated with an increased risk of osteosarcoma (OR =1.59, 95% CI 1.07-2.38, P = 0.02). This association was strongest in cases without metastatic disease (OR =2.46, 95% CI 1.44-4.19, P = 9.5 ×10-04). Although there was no overall association between osteosarcoma and genetically inferred taller stature (OR=1.06, 95% CI 0.96-1.17, P = 0.28), the GRS for taller stature was associated with an increased risk of osteosarcoma in 154 cases with a known pathogenic cancer susceptibility gene variant (OR=1.29, 95% CI 1.03-1.63, P = 0.03). There were no significant associations between the GRS for puberty timing and osteosarcoma. CONCLUSION A genetic propensity to higher birthweight was associated with increased osteosarcoma risk, suggesting that shared genetic factors or biological pathways that affect birthweight may contribute to osteosarcoma pathogenesis.
Collapse
Affiliation(s)
| | - Amy Moore
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA
| | - Logan G Spector
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Tianzhong Yang
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Aubrey Hubbard
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA
| | - Richard Gorlick
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Patiño-Garcia
- Department of Pediatrics and Solid Tumor Division CIMA, IdiSNA, Clínica Universidad de Navarra, Pamplona, Spain
| | - Fernando Lecanda
- Center for Applied Medical Research (CIMA)-University of Navarra, IdiSNA, and CIBERONC, Pamplona, Spain
| | - Adrienne M Flanagan
- UCL Cancer Institute, Huntley Street, London WC1E 6BT, UK; Royal National Orthopaedic Hospital NHS Trust, Stanmore, Middlesex HA7 4LP, UK
| | - Fernanda Amary
- Royal National Orthopaedic Hospital NHS Trust, Stanmore, Middlesex HA7 4LP, UK
| | - Irene L Andrulis
- Litwin Centre for Cancer Genetics, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Jay S Wunder
- Litwin Centre for Cancer Genetics, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - David M Thomas
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Mandy L Ballinger
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Massimo Serra
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, Pharmacogenomics and Pharmacogenetics Research Unit, Bologna, Italy
| | - Claudia Hattinger
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, Pharmacogenomics and Pharmacogenetics Research Unit, Bologna, Italy
| | - Ellen Demerath
- Division of Epidemiology and Clinical Research, School of Public Health, UMN, USA
| | - Will Johnson
- School of Sport, Exercise, and Health Sciences, University of Loughborough, UK
| | - Brenda M Birmann
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Graham Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Lauren R Teras
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Alan Arslan
- Department of Obstetrics and Gynecology, New York School of Medicine, New York, NY, USA; Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jeannette Sample
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, MD, USA.
| |
Collapse
|
79
|
Mianesaz H, Ghalamkari S, Abbasi F, Razzaghy-Azar M, Sayarifard F, Vakili R, Sedghi M, Noroozi Asl S, Hosseini S, Amoli MM, Yaghootkar H. Genetic variant profiling of neonatal diabetes mellitus in Iranian patients: Unveiling 58 distinct variants in 14 genes. J Diabetes Investig 2024; 15:1390-1402. [PMID: 38970407 PMCID: PMC11442839 DOI: 10.1111/jdi.14254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 05/11/2024] [Accepted: 06/04/2024] [Indexed: 07/08/2024] Open
Abstract
INTRODUCTION Neonatal diabetes mellitus (NDM) is a rare non-immunological monogenic disorder characterized by hyperglycemic conditions primarily occurring within the first 6 months of life. The majority of cases are attributed to pathogenic variants in genes affecting beta-cell survival, insulin regulation, and secretion. This study aims to investigate the genetic landscape of NDM in Iran. METHODS We recruited a total of 135 patients who were initially diagnosed with diabetes at <12 months of age in Iran and referred to pediatric endocrinology clinics across the country. These patients underwent genetic diagnostic tests conducted by the Exeter Molecular Genetics Laboratory in the UK. The pathogenic variants identified were sorted and described based on type, pathogenicity (according to ACMG/AMP criteria), novelty, and the affected protein domain. RESULTS Genetic defects were identified in 93 probands, presenting various pathogenic abnormalities associated with NDM and its associated syndromes. 76% of the patients were born as a result of consanguineous marriage, and a familial history of diabetes was found in 43% of the cases. A total of 58 distinct variants in 14 different genes were discovered, including 20 variants reported for the first time. Causative variants were most frequently identified in EIF2AK3, KCNJ11, and ABCC8, respectively. Notably, EIF2AK3 and ABCC8 exhibited the highest number of novel variants. DISCUSSION These findings provide valuable insights into the genetic landscape of NDM in the Iranian population and contribute to the knowledge of novel pathogenic variants within known causative genes.
Collapse
Affiliation(s)
- Hamidreza Mianesaz
- Department of Human Genetics, Medical School, University of Debrecen, Debrecen, Hungary
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Safoura Ghalamkari
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Farzaneh Abbasi
- Growth and Development Research Center, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Razzaghy-Azar
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular - Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sayarifard
- Growth and Development Research Center, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Rahim Vakili
- Department of Pediatric Endocrinology and Metabolism, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Sedghi
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Noroozi Asl
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sousan Hosseini
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa M Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular - Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
80
|
Uchida M, Horisawa S, Azuma K, Akagawa H, Tokushige S, Kawamata T, Taira T. Thalamic Deep Brain Stimulation for SPG56-Related Focal Hand Dystonia. J Mov Disord 2024; 17:447-449. [PMID: 38932635 PMCID: PMC11540542 DOI: 10.14802/jmd.24022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/19/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024] Open
Affiliation(s)
- Momo Uchida
- Department of Neurosurgery, Tokyo Women’s Medical University, Tokyo, Japan
| | - Shiro Horisawa
- Department of Neurosurgery, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kenkou Azuma
- Tokyo Women’s Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
| | - Hiroyuki Akagawa
- Tokyo Women’s Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
| | | | - Takakazu Kawamata
- Department of Neurosurgery, Tokyo Women’s Medical University, Tokyo, Japan
| | - Takaomi Taira
- Department of Neurosurgery, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
81
|
Park Y, Kim Y, Koh I, Lee JY. Whole-Exome Sequencing Improves Understanding of Inherited Retinal Dystrophies in Korean Patients. Curr Issues Mol Biol 2024; 46:11021-11030. [PMID: 39451534 PMCID: PMC11506058 DOI: 10.3390/cimb46100654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Retinitis pigmentosa (RP) encompasses a diverse range of hereditary, degenerative retinal ailments, presenting notable obstacles to molecular genetic diagnoses due to the intricate array of variants in different genes involved. This study enrolled 21 probands and their families who have been diagnosed with nonsyndromic RP but without a previous molecular diagnosis. We employed whole-exome sequencing (WES) to detect possible harmful gene variations in individuals with unknown-cause RP at the molecular level. WES allowed the identification of ten potential disease-causing variants in eight different genes. In 8 out of the total 21 patients, this method successfully identified the underlying molecular causes, such as putative pathogenic variants in genes including CRB1, KLHL7, PDE6B, RDH12, RP1, RPE65, USH2A, and RHO. A novel variant was identified in one of these genes, specifically PDE6B, providing valuable information on prospective targets for future enhanced gene therapeutic approaches.
Collapse
Affiliation(s)
- Youngchan Park
- Department of Biomedical Informatics, Hanyang University, Seoul 04763, Republic of Korea;
- Division of Bio Bigdata, Department of Precision Medicine, Korea National Institution of Health, KCDC, Cheongju 28159, Republic of Korea
| | - Youngjin Kim
- Elite Eye Hospital, Seoul 03779, Republic of Korea;
| | - Insong Koh
- Department of Biomedical Informatics, Hanyang University, Seoul 04763, Republic of Korea;
| | | |
Collapse
|
82
|
Sargen MR, Kim J, Haley JS, Barker HP, Mundra PA, Ballinger ML, Thomas DM, Carey DJ, Goldstein AM, Stewart DR. Increased frequency of CHEK2 germline pathogenic variants among individuals with dermatofibrosarcoma protuberans. GENETICS IN MEDICINE OPEN 2024; 2:101895. [PMID: 39669616 PMCID: PMC11613564 DOI: 10.1016/j.gimo.2024.101895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 12/14/2024]
Abstract
Purpose To identify candidate susceptibility genes for dermatofibrosarcoma protuberans (DFSP). Methods All individuals with DFSP from the International Sarcoma Kindred Study (n = 3767 individuals with sarcoma diagnoses from Australia, Europe, New Zealand, and United States) and cohorts that were not ascertained based on sarcoma status or other phenotypes (Geisinger MyCode, n = 170,503 individuals, United States; UK Biobank, n = 469,789 individuals, United Kingdom) were evaluated for germline pathogenic or likely pathogenic (P/LP) variants in 156 cancer genes. Results There were 92 unrelated individuals with DFSP across the 3 cohorts. The mean age at diagnosis (standard deviation) in the International Sarcoma Kindred Study, Geisinger, and UK Biobank was 40.8 (14.5), 50.3 (9.4), and 49.4 (13.2) years, respectively. Germline P/LP variants were most common in the CHEK2 gene (4/92 [4.3%]). CHEK2-related cases were often associated with early onset disease (age at diagnosis: 30-39 years) and were observed in all 3 cohorts. Among 640,292 individuals in Geisinger and UK Biobank who were not ascertained based on phenotype, there was a significantly increased frequency of CHEK2 P/LP variants among individuals with DFSP (n = 3/65 [4.6%]) compared to those without (n = 6388/640,227 [1.0%]) (Fisher exact, P = .03). Additional genes with P/LP variation (1 case for each gene) included ACD, ERCC5, ERCC1, DOCK8, GBA1, ATM, MUTYH, TP53, RECQL4, and COL7A1. Conclusion This study of multiple cohorts identifies CHEK2 as a candidate susceptibility gene for DFSP. Additional epidemiologic and functional studies are needed to further characterize this potential gene-tumor relationship.
Collapse
Affiliation(s)
- Michael R. Sargen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Jung Kim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Jeremy S. Haley
- Department of Genomic Health, Geisinger Clinic, Geisinger Health System, Danville, PA
| | | | - Piyushkumar A. Mundra
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Mandy L. Ballinger
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - David M. Thomas
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Centre for Molecular Oncology, University of New South Wales, Sydney, NSW, Australia
| | - David J. Carey
- Department of Genomic Health, Geisinger Clinic, Geisinger Health System, Danville, PA
| | - Alisa M. Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Douglas R. Stewart
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD
| |
Collapse
|
83
|
Yamamoto N, Urabe Y, Nakahara H, Nakamura T, Shimizu D, Konishi H, Ishibashi K, Ariyoshi M, Miyamoto R, Mizuno J, Takasago T, Ishikawa A, Tsuboi A, Tanaka H, Yamashita K, Hiyama Y, Kishida Y, Takigawa H, Kuwai T, Arihiro K, Shimamoto F, Oka S. Genetic Analysis of Biopsy Tissues from Colorectal Tumors in Patients with Ulcerative Colitis. Cancers (Basel) 2024; 16:3271. [PMID: 39409892 PMCID: PMC11475702 DOI: 10.3390/cancers16193271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal neoplasia developing from ulcerative colitis mucosa (CRNUC) can be divided into ulcerative colitis-associated neoplasia (UCAN) and non-UCAN; however, it is often difficult to distinguish UCAN from non-UCAN during a biopsy diagnosis. We investigated whether a genomic analysis could improve the diagnostic accuracy of UCAN using biopsy specimens. METHODS In step 1, 14 CRNUCs were used to examine whether the genomic landscape of biopsy and resection specimens matched. In step 2, we investigated the relationship between the genomic landscapes and the pathological diagnosis of 26 CRNUCs. The cancer genome was analyzed by deep sequencing using a custom panel of 27 genes found to be mutated in our previous CRNUC analysis. RESULTS In step 1, of the 27 candidate genes, 14 were mutated. The concordance rate of the pathogenic mutations in these 14 genes between the biopsy and resection specimens was 29% (4/14), while that of the pathogenic mutations in TP53 and KRAS was 79% (11/14). In step 2, the pathological diagnosis of biopsy specimens using only hematoxylin and eosin (HE) staining had a sensitivity of 33% and an accuracy of 38% for UCAN diagnosis. On the other hand, the combination of the HE pathology and p53 immunohistochemical staining had a sensitivity of 73% and an accuracy of 85% for UCAN diagnosis, while the combination of HE staining and a TP53 mutation had a sensitivity of 87% and an accuracy of 88% for UCAN diagnosis. CONCLUSIONS An evaluation of TP53 mutations in biopsy specimens may be useful for diagnosing UCAN. However, further studies with larger sample sizes are required before this can be applied in clinical practice.
Collapse
Affiliation(s)
- Noriko Yamamoto
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Yuji Urabe
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Hikaru Nakahara
- Department of Clinical and Molecular Genetics, Hiroshima University Hospital, Hiroshima 734-8551, Japan;
| | - Takeo Nakamura
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Daisuke Shimizu
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Hirona Konishi
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Kazuki Ishibashi
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Misa Ariyoshi
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Ryo Miyamoto
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Junichi Mizuno
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Takeshi Takasago
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Akira Ishikawa
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Akiyoshi Tsuboi
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Hidenori Tanaka
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Ken Yamashita
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Yuichi Hiyama
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, Hiroshima 734-8551, Japan;
| | - Yoshihiro Kishida
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Hidehiko Takigawa
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| | - Toshio Kuwai
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
- Gastrointestinal Endoscopy and Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima 734-8551, Japan;
| | - Fumio Shimamoto
- Faculty of Health Sciences, Hiroshima Cosmopolitan University, Hiroshima 734-0014, Japan;
| | - Shiro Oka
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (N.Y.); (T.N.); (D.S.); (H.K.); (K.I.); (M.A.); (R.M.); (J.M.); (T.T.); (A.T.); (H.T.); (K.Y.); (Y.K.); (H.T.); (T.K.); (S.O.)
| |
Collapse
|
84
|
Wang D, Scalici A, Wang Y, Lin H, Pitsillides A, Heard-Costa N, Cruchaga C, Ziegemeier E, Bis JC, Fornage M, Boerwinkle E, De Jager PL, Wijsman E, Dupuis J, Renton AE, Seshadri S, Goate AM, DeStefano AL, Peloso GM. Frequency of Variants in Mendelian Alzheimer's Disease Genes within the Alzheimer's Disease Sequencing Project (ADSP). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.24.23297227. [PMID: 37961373 PMCID: PMC10635182 DOI: 10.1101/2023.10.24.23297227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
BACKGROUND Prior studies using the ADSP data examined variants within presenilin-2 (PSEN2), presenilin-1 (PSEN1), and amyloid precursor protein (APP) genes. However, previously-reported clinically-relevant variants and other predicted damaging missense (DM) variants have not been characterized in a newer release of the Alzheimer's Disease Sequencing Project (ADSP). OBJECTIVE To characterize previously-reported clinically-relevant variants and DM variants in PSEN2, PSEN1, APP within the participants from the ADSP. METHODS We identified rare variants (MAF <1%) previously-reported in PSEN2, PSEN1, and APP in the available ADSP sample of 14,641 individuals with whole genome sequencing and 16,849 individuals with whole exome sequencing available for research-use (Ntotal = 31,490). We additionally curated variants in these three genes from ClinVar, OMIM, and Alzforum and report carriers of variants in clinical databases as well as predicted DM variants in these genes. RESULTS We detected 31 previously-reported clinically-relevant variants with alternate alleles observed within the ADSP: 4 variants in PSEN2, 25 in PSEN1, and 2 in APP. The overall variant carrier rate for the 31 clinically-relevant variants in the ADSP was 0.3%. We observed that 79.5% of the variant carriers were cases compared to 3.9% were controls. In those with AD, the mean age of onset of AD among carriers of these clinically-relevant variants was 19.6 ± 1.4 years earlier compared with noncarriers (p-value=7.8×10-57). CONCLUSION A small proportion of individuals in the ADSP are carriers of a previously-reported clinically-relevant variant allele for AD and these participants have significantly earlier age of AD onset compared to noncarriers.
Collapse
|
85
|
Diossy M, Tisza V, Li H, Sahgal P, Zhou J, Sztupinszki Z, Young D, Nousome D, Kuo C, Jiang J, Chen Y, Ebner R, Sesterhenn IA, Moncur JT, Chesnut GT, Petrovics G, Klus GT, Valcz G, Nuzzo PV, Ribli D, Börcsök J, Prosz A, Krzystanek M, Ried T, Szuts D, Rizwan K, Kaochar S, Pathania S, D'Andrea AD, Csabai I, Srivastava S, Freedman ML, Dobi A, Spisak S, Szallasi Z. Frequent CHD1 deletions in prostate cancers of African American men is associated with rapid disease progression. NPJ Precis Oncol 2024; 8:208. [PMID: 39294262 PMCID: PMC11411125 DOI: 10.1038/s41698-024-00705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
We analyzed genomic data from the prostate cancer of African- and European American men to identify differences contributing to racial disparity of outcome. We also performed FISH-based studies of Chromodomain helicase DNA-binding protein 1 (CHD1) loss on prostate cancer tissue microarrays. We created CHD1-deficient prostate cancer cell lines for genomic, drug sensitivity and functional homologous recombination (HR) activity analysis. Subclonal deletion of CHD1 was nearly three times as frequent in prostate tumors of African American than in European American men and it associates with rapid disease progression. CHD1 deletion was not associated with HR deficiency associated mutational signatures or HR deficiency as detected by RAD51 foci formation. This was consistent with the moderate increase of olaparib and talazoparib sensitivity with several CHD1 deficient cell lines showing talazoparib sensitivity in the clinically relevant concentration range. CHD1 loss may contribute to worse disease outcome in African American men.
Collapse
Affiliation(s)
- Miklos Diossy
- Danish Cancer Institute, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Viktoria Tisza
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hua Li
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Pranshu Sahgal
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - Jia Zhou
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Zsofia Sztupinszki
- Danish Cancer Institute, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Denise Young
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Darryl Nousome
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Claire Kuo
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Jiji Jiang
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Yongmei Chen
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | | | | | | | - Gregory T Chesnut
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Gregory T Klus
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Gabor Valcz
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary
| | - Pier Vitale Nuzzo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dezso Ribli
- Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | | | - Aurel Prosz
- Danish Cancer Institute, Copenhagen, Denmark
| | | | - Thomas Ried
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David Szuts
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Kinza Rizwan
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Salma Kaochar
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Shailja Pathania
- Center for Personalized Cancer Therapy, University of Massachusetts, Boston, MA, USA
- Department of Biology, University of Massachusetts, Boston, MA, USA
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Istvan Csabai
- Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | - Shiv Srivastava
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Biochemistry and Molecular & Cell Biology, Georgetown University School of Medicine, Washington, DC, USA
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Albert Dobi
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA.
| | - Sandor Spisak
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - Zoltan Szallasi
- Danish Cancer Institute, Copenhagen, Denmark.
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- 2nd Department of Pathology and Department of Bioinformatics, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
86
|
Zhang Q, Yang Q, Shen F, Wang L, Luo J. Identification of a novel FERMT1 variant causing kindler syndrome and a review of the clinical and molecular genetic features in Chinese patients. Front Pediatr 2024; 12:1425030. [PMID: 39309641 PMCID: PMC11415864 DOI: 10.3389/fped.2024.1425030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Background Kindler Syndrome (KS, OMIM #173650), a rare autosomal recessive genetic disorder, is characterized by a spectrum of symptoms such as cutaneous fragility, blistering, photosensitivity, and mucosal involvement. These symptoms result from variations in the FERMT1 gene (Fermitin family member 1, OMIM: 607900), encoding kindlin-1, an essential component of focal adhesions. Objective This study aims to ascertain the potential pathogenicity of a FERMT1 variant identified in a Chinese patient and to explore the phenotypic and molecular genetic characteristics of all reported cases of Kindler Syndrome in the Chinese population. Methods Whole-exome sequencing (WES) was performed on the patient to identify candidate variants associated with KS, and Sanger sequencing was utilized to authenticate their presence and origin. To further assess the potential impact of these genetic variants, we employed a variety of in silico prediction tools. Concurrently, a review of various databases was undertaken to ascertain and consolidate information regarding cases of KS in Chinese families. Results We identified a novel likely pathogenic frameshift variant in the FERMT1 gene, specifically c.567_579delTATATATGACCCC (p.Ile190Serfs*10). The clinical presentation of this patient aligns with the diagnostic criteria for KS. The literature review reveals that the core clinical features of KS reported in the Chinese population include skin abnormalities (100%), as well as hyperkeratosis of the palms and soles (91.70%). Other clinical phenotypes encompass nail abnormalities (77.78%), abnormalities of the fingers/toes (75.00%), oral damage (70.00%), eye abnormalities (57.14%), and constipation (50.00%). Conclusion Our study enriches the genetic landscape of KS in the Chinese population and augments the understanding of phenotypic variability resulting from FERMT1 gene variants. The findings hold considerable significance for refining variant-based screening, genetic diagnosis, and comprehending the molecular pathogenesis underlying FERMT1-related disorders.
Collapse
Affiliation(s)
- Qiang Zhang
- Laboratory of Genetic Metabolism Center, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Hematology Laboratory, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Qi Yang
- Laboratory of Genetic Metabolism Center, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Fei Shen
- Laboratory of Genetic Metabolism Center, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Linlin Wang
- Laboratory of Genetic Metabolism Center, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jingsi Luo
- Laboratory of Genetic Metabolism Center, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
87
|
Gusakova M, Dzhumaniiazova I, Zelenova E, Kashtanova D, Ivanov M, Mamchur A, Rumyantseva A, Terekhov M, Mitrofanov S, Golubnikova L, Akinshina A, Grammatikati K, Kalashnikova I, Yudin V, Makarov V, Keskinov A, Yudin S. Prevalence of the cancer-associated germline variants in Russian adults and long-living individuals: using the ACMG recommendations and computational interpreters for pathogenicity assessment. Front Oncol 2024; 14:1420176. [PMID: 39301547 PMCID: PMC11410565 DOI: 10.3389/fonc.2024.1420176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
Background Population studies are essential for gathering critical disease prevalence data. Automated pathogenicity assessment tools enhance the capacity to interpret and annotate large amounts of genetic data. In this study, we assessed the prevalence of cancer-associated germline variants in Russia using a semiautomated variant interpretation algorithm. Methods We examined 74,996 Russian adults (Group 1) and 2,872 long-living individuals aged ≥ 90 years (Group 2) for variants in 28 ACMG-recommended cancer-associated genes in three steps: InterVar annotation; ClinVar interpretation; and a manual review of the prioritized variants based on the available data. Using the data on the place of birth and the region of residence, we determined the geographical distribution of the detected variants and tracked the migration dynamics of their carriers. Results We report 175 novel del-VUSs. We detected 232 pathogenic variants, 46 likely pathogenic variants, and 216 del-VUSs in Group 1 and 19 pathogenic variants, 2 likely pathogenic variants, and 16 del-VUSs in Group 2. For each detected variant, we provide a description of its functional significance and geographical distribution. Conclusion The present study offers extensive genetic data on the Russian population, critical for future genetic research and improved primary cancer prevention and genetic screening strategies. The proposed hybrid assessment algorithm streamlines variant prioritization and pathogenicity assessment and offers a reliable and verifiable way of identifying variants of uncertain significance that need to be manually reviewed.
Collapse
Affiliation(s)
- Mariia Gusakova
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Irina Dzhumaniiazova
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Elena Zelenova
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Daria Kashtanova
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Mikhail Ivanov
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Aleksandra Mamchur
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Antonina Rumyantseva
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Mikhail Terekhov
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Sergey Mitrofanov
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Liliya Golubnikova
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Aleksandra Akinshina
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Konstantin Grammatikati
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Irina Kalashnikova
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Vladimir Yudin
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Valentin Makarov
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Anton Keskinov
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Sergey Yudin
- The Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| |
Collapse
|
88
|
Martino S, D’Addabbo P, Turchiano A, Radio FC, Bruselles A, Cordeddu V, Mancini C, Stella A, Laforgia N, Capodiferro D, Simonetti S, Bagnulo R, Palumbo O, Marzano F, Tabaku O, Garganese A, Stasi M, Tartaglia M, Pesole G, Resta N. Deep Intronic ETFDH Variants Represent a Recurrent Pathogenic Event in Multiple Acyl-CoA Dehydrogenase Deficiency. Int J Mol Sci 2024; 25:9637. [PMID: 39273584 PMCID: PMC11395610 DOI: 10.3390/ijms25179637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Multiple acyl-CoA dehydrogenase deficiency (MADD) is a rare inborn error of metabolism affecting fatty acid and amino acid oxidation with an incidence of 1 in 200,000 live births. MADD has three clinical phenotypes: severe neonatal-onset with or without congenital anomalies, and a milder late-onset form. Clinical diagnosis is supported by urinary organic acid and blood acylcarnitine analysis using tandem mass spectrometry in newborn screening programs. MADD is an autosomal recessive trait caused by biallelic mutations in the ETFA, ETFB, and ETFDH genes encoding the alpha and beta subunits of the electron transfer flavoprotein (ETF) and ETF-coenzyme Q oxidoreductase enzymes. Despite significant advancements in sequencing techniques, many patients remain undiagnosed, impacting their access to clinical care and genetic counseling. In this report, we achieved a definitive molecular diagnosis in a newborn by combining whole-genome sequencing (WGS) with RNA sequencing (RNA-seq). Whole-exome sequencing and next-generation gene panels fail to detect variants, possibly affecting splicing, in deep intronic regions. Here, we report a unique deep intronic mutation in intron 1 of the ETFDH gene, c.35-959A>G, in a patient with early-onset lethal MADD, resulting in pseudo-exon inclusion. The identified variant is the third mutation reported in this region, highlighting ETFDH intron 1 vulnerability. It cannot be excluded that these intronic sequence features may be more common in other genes than is currently believed. This study highlights the importance of incorporating RNA analysis into genome-wide testing to reveal the functional consequences of intronic mutations.
Collapse
Affiliation(s)
- Stefania Martino
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.T.); (A.S.); (R.B.); (O.T.); (M.S.)
| | - Pietro D’Addabbo
- Department of Biosciences, Biotechnologies & Environment, University of Bari “Aldo Moro”, Via Edoardo Orabona 4, 70125 Bari, Italy; (P.D.); (G.P.)
| | - Antonella Turchiano
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.T.); (A.S.); (R.B.); (O.T.); (M.S.)
| | - Francesca Clementina Radio
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale di San Paolo 15, 00146 Rome, Italy; (F.C.R.); (C.M.); (M.T.)
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (V.C.)
| | - Viviana Cordeddu
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (V.C.)
| | - Cecilia Mancini
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale di San Paolo 15, 00146 Rome, Italy; (F.C.R.); (C.M.); (M.T.)
| | - Alessandro Stella
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.T.); (A.S.); (R.B.); (O.T.); (M.S.)
| | - Nicola Laforgia
- Section of Neonatology and Neonatal Intensive Care Unit, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (N.L.); (D.C.)
| | - Donatella Capodiferro
- Section of Neonatology and Neonatal Intensive Care Unit, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (N.L.); (D.C.)
| | - Simonetta Simonetti
- Clinical Pathology and Neonatal Screening, Hospital “Giovanni XXIII”, University Hospital Consortium Corporation Polyclinics of Bari, 70124 Bari, Italy;
| | - Rosanna Bagnulo
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.T.); (A.S.); (R.B.); (O.T.); (M.S.)
| | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS—Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Flaviana Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Via Amendola 122/O, 70126 Bari, Italy;
| | - Ornella Tabaku
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.T.); (A.S.); (R.B.); (O.T.); (M.S.)
| | - Antonella Garganese
- Medical Genetic Unit, University Hospital Consortium Corporation Polyclinics of Bari, 70124 Bari, Italy;
| | - Michele Stasi
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.T.); (A.S.); (R.B.); (O.T.); (M.S.)
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale di San Paolo 15, 00146 Rome, Italy; (F.C.R.); (C.M.); (M.T.)
| | - Graziano Pesole
- Department of Biosciences, Biotechnologies & Environment, University of Bari “Aldo Moro”, Via Edoardo Orabona 4, 70125 Bari, Italy; (P.D.); (G.P.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Via Amendola 122/O, 70126 Bari, Italy;
| | - Nicoletta Resta
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.M.); (A.T.); (A.S.); (R.B.); (O.T.); (M.S.)
| |
Collapse
|
89
|
He D, He X, Shen D, Liu L, Yang X, Hao M, Wang Y, Li Y, Liu Q, Liu M, Wang J, Zhang X, Cui L. Loss-of-function variants in RNA binding motif protein X-linked induce neuronal defects contributing to amyotrophic lateral sclerosis pathogenesis. MedComm (Beijing) 2024; 5:e712. [PMID: 39263607 PMCID: PMC11387721 DOI: 10.1002/mco2.712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 09/13/2024] Open
Abstract
Despite being one of the most prevalent RNA modifications, the role of N6-methyladenosine (m6A) in amyotrophic lateral sclerosis (ALS) remains ambiguous. In this investigation, we explore the contribution of genetic defects of m6A-related genes to ALS pathogenesis. We scrutinized the mutation landscape of m6A genes through a comprehensive analysis of whole-exome sequencing cohorts, encompassing 508 ALS patients and 1660 population-matched controls. Our findings reveal a noteworthy enrichment of RNA binding motif protein X-linked (RBMX) variants among ALS patients, with a significant correlation between pathogenic m6A variants and adverse clinical outcomes. Furthermore, Rbmx knockdown in NSC-34 cells overexpressing mutant TDP43Q331K results in cell death mediated by an augmented p53 response. Similarly, RBMX knockdown in ALS motor neurons derived from induced pluripotent stem cells (iPSCs) manifests morphological defects and activation of the p53 pathway. Transcriptional analysis using publicly available single-cell sequencing data from the primary motor cortex indicates that RBMX-regulated genes selectively influence excitatory neurons and exhibit enrichment in ALS-implicated pathways. Through integrated analyses, our study underscores the emerging roles played by RBMX in ALS, suggesting a potential nexus between the disease and dysregulated m6A-mediated mRNA metabolism.
Collapse
Affiliation(s)
- Di He
- Department of Neurology Beijing Tiantan Hospital, Capital Medical University Beijing China
- Department of Neurology Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xinyi He
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute Fudan University Shanghai China
| | - Dongchao Shen
- Department of Neurology Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Liyang Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College Beijing China
| | - Xunzhe Yang
- Department of Neurology Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Meng Hao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute Fudan University Shanghai China
| | - Yi Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute Fudan University Shanghai China
| | - Yi Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute Fudan University Shanghai China
| | - Qing Liu
- Department of Neurology Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Mingsheng Liu
- Department of Neurology Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute Fudan University Shanghai China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases Chinese Academy of Medical Sciences (2019RU058) Shanghai China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College Beijing China
- Neuroscience Center Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS) Beijing China
| | - Liying Cui
- Department of Neurology Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
90
|
Ahmad S, Muurinen M, Loid P, Ali MZ, Muzammal M, Fatima S, Khan J, Khan MA, Mäkitie O. A clinical and molecular characterization of a Pakistani family with multicentric osteolysis, nodulosis and arthropathy (MONA) syndrome. Bone Rep 2024; 22:101789. [PMID: 39540058 PMCID: PMC11558256 DOI: 10.1016/j.bonr.2024.101789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 11/16/2024] Open
Abstract
Multicentric osteolysis nodulosis and arthropathy (MONA) is a rare skeletal dysplasia characterized primarily by progressive osteolysis, particularly affecting the carpal and tarsal bones, accompanied by osteoporosis. In addition, it features subcutaneous nodules on the palms and soles, along with the progressive onset of arthropathy, encompassing joint contractures, pain, swelling and stiffness. It is caused by a deficiency of the Matrix Metalloproteinase-2 (MMP2). In the current study we present a comprehensive clinical, radiological, genetic and in silico analysis of MONA in a consanguineous Pakistani family. Clinical and radiological examinations of the three severely affected siblings demonstrated a progressive MONA syndrome with phenotypic variability. The patients presented unusual facial appearance, thickened skin, severe short stature, short hands and feet. Radiographs revealed extensive bone deformities affecting upper and lower arms, legs, vertebrae and hip. Genetic analysis revealed a homozygous missense variant [c.539 A > T p.(Asp180Val)] in the MMP2 gene. In silico findings suggested a mutant MMP2 protein with a decreased stability and an altered pattern of interactions. Our findings add to the existing literature on the skeletal phenotype of MONA syndrome, including the specific clinical and radiological patterns observed. Moreover, the study will aid in genetic counseling and accurate diagnosis of families affected by the same disorder within the Pakistani population.
Collapse
Affiliation(s)
- Safeer Ahmad
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mari Muurinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Genetics Research Program, Helsinki, Finland
| | - Petra Loid
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Genetics Research Program, Helsinki, Finland
| | - Muhammad Zeeshan Ali
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| | - Muhammad Muzammal
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| | - Sana Fatima
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| | - Jabbar Khan
- Institute of Biological Sciences, Gomal University, Dera Ismail Khan, Pakistan
| | - Muzammil Ahmad Khan
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| | - Outi Mäkitie
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Genetics Research Program, Helsinki, Finland
| |
Collapse
|
91
|
Shi F, Liu Y, Chen Z, Li D, Yao Y, Zhou M, Zhuo Y, Ma X, Cao D. An integrated approach for improving clinical management of non-obstructive azoospermia. Andrology 2024; 12:1312-1323. [PMID: 38221731 DOI: 10.1111/andr.13587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/06/2023] [Accepted: 12/19/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND Non-obstructive azoospermia is the most severe form of male infertility. A testicular biopsy is required for the diagnosis of non-obstructive azoospermia, and the causal factors for non-obstructive azoospermia remain unknown. OBJECTIVES To reduce the risk of multiple biopsies and identify factors that contribute to non-obstructive azoospermia, we proposed an integrated approach for the preoperative diagnosis and clinical management of non-obstructive azoospermia by applying the chromosome-spreading technique and whole-exome sequencing. MATERIALS AND METHODS Between July 2020 and December 2022, after ruling out definitive obstructive azoospermia and non-obstructive azoospermia patients with testicular volume < 6 mL, 20 patients with non-obstructive azoospermia who underwent preoperative testicular diagnostic biopsy using testicular sperm aspiration were subjected to retrospective analysis. RESULTS Microscopic examination identified four patients with sperm cells, and 16 without sperm cells. Routine pathological analysis classified one patient as normal spermatogenesis, three as hypospermatogenesis, five as maturation arrest, nine as Sertoli cell-only, and two as unable to judge. With chromosome-spreading technology using routine cell suspension samples for microscopic examination, 18 patient diagnoses were validated, and two patients without a definitive diagnosis were supplemented. Detection of the Y chromosome and a well-organized whole-exome sequencing analysis revealed potential genetic factors. DISCUSSION AND CONCLUSION The full use of testicular biopsy is beneficial for the diagnosis of azoospermia, as it avoids the risk of multiple biopsies. Moreover, in combination with whole-exome sequencing, clinicians can obtain more information regarding the pathogenesis of non-obstructive azoospermia, which may guide treatment.
Collapse
Affiliation(s)
- Fu Shi
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Ye Liu
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Zheng Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongliang Li
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuanqing Yao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Meixun Zhou
- Department of Pathology, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Yumin Zhuo
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xin Ma
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Senior Department of Urology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Dandan Cao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
92
|
Lin YJ, Menon AS, Hu Z, Brenner SE. Variant Impact Predictor database (VIPdb), version 2: trends from three decades of genetic variant impact predictors. Hum Genomics 2024; 18:90. [PMID: 39198917 PMCID: PMC11360829 DOI: 10.1186/s40246-024-00663-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Variant interpretation is essential for identifying patients' disease-causing genetic variants amongst the millions detected in their genomes. Hundreds of Variant Impact Predictors (VIPs), also known as Variant Effect Predictors (VEPs), have been developed for this purpose, with a variety of methodologies and goals. To facilitate the exploration of available VIP options, we have created the Variant Impact Predictor database (VIPdb). RESULTS The Variant Impact Predictor database (VIPdb) version 2 presents a collection of VIPs developed over the past three decades, summarizing their characteristics, ClinGen calibrated scores, CAGI assessment results, publication details, access information, and citation patterns. We previously summarized 217 VIPs and their features in VIPdb in 2019. Building upon this foundation, we identified and categorized an additional 190 VIPs, resulting in a total of 407 VIPs in VIPdb version 2. The majority of the VIPs have the capacity to predict the impacts of single nucleotide variants and nonsynonymous variants. More VIPs tailored to predict the impacts of insertions and deletions have been developed since the 2010s. In contrast, relatively few VIPs are dedicated to the prediction of splicing, structural, synonymous, and regulatory variants. The increasing rate of citations to VIPs reflects the ongoing growth in their use, and the evolving trends in citations reveal development in the field and individual methods. CONCLUSIONS VIPdb version 2 summarizes 407 VIPs and their features, potentially facilitating VIP exploration for various variant interpretation applications. VIPdb is available at https://genomeinterpretation.org/vipdb.
Collapse
Affiliation(s)
- Yu-Jen Lin
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Arul S Menon
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- College of Computing, Data Science, and Society, University of California, Berkeley, CA, 94720, USA
| | - Zhiqiang Hu
- Department of Plant and Microbial Biology, University of California, 111 Koshland Hall #3102, Berkeley, CA, 94720-3102, USA
- Illumina, Foster City, CA, 94404, USA
| | - Steven E Brenner
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA.
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA.
- College of Computing, Data Science, and Society, University of California, Berkeley, CA, 94720, USA.
- Department of Plant and Microbial Biology, University of California, 111 Koshland Hall #3102, Berkeley, CA, 94720-3102, USA.
| |
Collapse
|
93
|
Del Valle-Peréz M, Mejía-García A, Echeverri-López D, Gallo-Bonilla K, Tejada-Moreno JA, Villegas-Lanau A, Chvatal-Medina M, Restrepo JE, Cuartas-Montoya G, Zapata-Builes W. Urofacial (Ochoa) syndrome with a founder pathogenic variant in the HPSE2 gene: a case report and mutation origin. J Appl Genet 2024:10.1007/s13353-024-00896-7. [PMID: 39150614 DOI: 10.1007/s13353-024-00896-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024]
Abstract
Urofacial syndrome or Ochoa syndrome (UFS or UFOS) is a rare disease characterized by inverted facial expression and bladder dysfunction that was described for the first time in Colombia. It is an autosomal recessive pathology with mutations in the HPSE2 and LRIG2 genes. However, 16% of patients do not have any mutations associated with the syndrome. Despite the importance of neurobiology in its pathophysiology, there are no neurological, neuropsychological, or psychological studies in these patients. A 30-year-old male from Medellín, Colombia, with a significant perinatal history, was diagnosed with grade 4 hydronephrosis on his first ultrasound test. At 4 months of age, symptoms such as hypomimia, lagophthalmos, and recurrent urinary tract infections started to manifest. Imaging studies revealed urinary tract dilatation, vesicoureteral reflux, and a double collector system on his left side, which led to the diagnosis of UFS. Multiple procedures, including vesicostomy, ureterostomy, and enterocystoplasty, were performed. At 20 years of age, he achieved urinary sphincter control. Genetic analysis revealed a founder pathogenic variant, c.1516C > T (p.Arg506Ter), in the HPSE2 gene, which produces a truncated protein that lacks 86 amino acids. This variant is classified as pathogenic according to the ClinVar database for UFS. The mutation age is approximately 260-360 years, and the two alleles share a 7.2-7.4 Mb IBD segment. Moreover, we detected European local ancestry in the IBD segment, which is consistent with a Spanish introduction. Neurological examination, neuropsychological assessment, and psychological testing revealed no abnormalities, except for high stress levels. Clinical analysis of this patient revealed distorted facial expression and detrusor-sphincter dyssynergia, which are typical of patients with UFS. Genetic analysis revealed a pathogenic variant in the HPSE2 gene of European origin and a mutation age of 260-360 years. From a neurological, neuropsychological, and psychological (emotional and personality) perspective, the patient showed no signs or symptoms of clinical interest.
Collapse
Affiliation(s)
- Manuela Del Valle-Peréz
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Alejandro Mejía-García
- Grupo de Genética Molecular (GENMOL), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Antioquia, Medellín, Colombia
| | - Dayana Echeverri-López
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Katherine Gallo-Bonilla
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Johanna A Tejada-Moreno
- Grupo de Genética Molecular (GENMOL), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Antioquia, Medellín, Colombia
| | - Andrés Villegas-Lanau
- Grupo de Genética Molecular (GENMOL), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Antioquia, Medellín, Colombia
| | - Mateo Chvatal-Medina
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Jorge E Restrepo
- Grupo OBSERVATOS, Facultad de Educación Y Ciencias Sociales, Tecnológico de Antioquia - Institución Universitaria, Medellín, Colombia
| | - Gina Cuartas-Montoya
- Facultad de Psicología, Grupo Neurociencia Y Cognición, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Wildeman Zapata-Builes
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia.
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
94
|
Kuehn JC, Metzger P, Neidert N, Matysiak U, Gräßel L, Philipp U, Bleul S, Pauli T, Falkenstein J, Bertemes H, Cysar S, Hess ME, Frey AV, Duque-Afonso J, Schorb E, Machein M, Beck J, Schnell O, von Bubnoff N, Illert AL, Peters C, Brummer T, Prinz M, Miething C, Becker H, Lassmann S, Werner M, Börries M, Duyster J, Heiland DH, Sankowski R, Scherer F. Comprehensive genetic profiling and molecularly guided treatment for patients with primary CNS tumors. NPJ Precis Oncol 2024; 8:180. [PMID: 39143272 PMCID: PMC11324882 DOI: 10.1038/s41698-024-00674-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
Despite major advances in molecular profiling and classification of primary brain tumors, personalized treatment remains limited for most patients. Here, we explored the feasibility of individual molecular profiling and the efficacy of biomarker-guided therapy for adult patients with primary brain cancers in the real-world setting within the molecular tumor board Freiburg, Germany. We analyzed genetic profiles, personalized treatment recommendations, and clinical outcomes of 102 patients with 21 brain tumor types. Alterations in the cell cycle, BRAF, and mTOR pathways most frequently led to personalized treatment recommendations. Molecularly informed therapies were recommended in 71% and implemented in 32% of patients with completed molecular diagnostics. The disease control rate following targeted treatment was 50% and the overall response rate was 30%, with a progression-free survival 2/1 ratio of at least 1.3 in 31% of patients. This study highlights the efficacy of molecularly guided treatment and the need for biomarker-stratified trials in brain cancers.
Collapse
Affiliation(s)
- Julia C Kuehn
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicolas Neidert
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Uta Matysiak
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Linda Gräßel
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrike Philipp
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Bleul
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Pauli
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Falkenstein
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Henriette Bertemes
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stepan Cysar
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Elena Hess
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Anna Verena Frey
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jesús Duque-Afonso
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elisabeth Schorb
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcia Machein
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
| | - Jürgen Beck
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Anna L Illert
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Department of Medicine III, Faculty of Medicine, Klinikum Rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Christoph Peters
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tilman Brummer
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Institute of Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius Miething
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Becker
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Silke Lassmann
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Werner
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Börries
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Justus Duyster
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Dieter H Heiland
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine Freiburg, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Roman Sankowski
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian Scherer
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Heidelberg, Germany.
| |
Collapse
|
95
|
Kim SY, Kim J, Ramos M, Haley J, Smelser D, Rao HS, Mirshahi UL, Geisinger-Regeneron DiscovEHR Collaboration, Graubard BI, Katki HA, Carey D, Stewart DR. Genomic ascertainment of CHEK2-related cancer predisposition. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311613. [PMID: 39371170 PMCID: PMC11451703 DOI: 10.1101/2024.08.07.24311613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Purpose There is clear evidence that deleterious germline variants in CHEK2 increases risk for breast and prostate cancers; there is limited or conflicting evidence for other cancers. Genomic ascertainment was used to quantify cancer risk in CHEK2 germline pathogenic variant heterozygotes. Patients and Methods Germline CHEK2 variants were extracted from two exome-sequenced biobanks linked to the electronic health record: UK Biobank (n= 469,765) and Geisinger MyCode (n=170,503). Variants were classified as per American College of Medical Genetics and Genomics (ACMG)/Association for Molecular Pathology (AMP) criteria. Heterozygotes harbored a CHEK2 pathogenic/likely pathogenic (P/LP) variant; controls harbored benign/likely benign CHEK2 variation or wildtype CHEK2. Tumor phenotype and demographic data were retrieved; to adjust for relatedness, association analysis was performed with SAIGE-GENE+ with Bonferroni correction. Results In CHEK2 heterozygotes in both MyCode and UK Biobank, there was a significant excess risk of all cancers tested, including breast cancer (C50; OR=1.54 and 1.84, respectively), male genital organ cancer (C60-C63; OR=1.61 and 1.77 respectively), urinary tract cancer (C64-C68; OR=1.56 and 1.75, respectively) and lymphoid, hematopoietic, and related tissue cancer (C81-C96; OR=1.42 and 2.11, respectively). Compared to controls, age-dependent cancer penetrance in CHEK2 heterozygotes was significantly younger in both cohorts; no significant difference was observed between the penetrance of truncating and missense variants for cancer in either cohort. Overall survival was significantly decreased in CHEK2 heterozygotes in UK Biobank but there was no statistical difference in MyCode. Conclusion Using genomic ascertainment in two population-scale cohorts, this investigation quantified the prevalence, penetrance, cancer phenotype and survival in CHEK2 heterozygotes. Tailored treatment options and surveillance strategies to manage those risks are warranted.
Collapse
Affiliation(s)
- Sun Young Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jung Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Mark Ramos
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Jeremy Haley
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Diane Smelser
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - H. Shanker Rao
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | | | | | - Barry I. Graubard
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Hormuzd A. Katki
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - David Carey
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Douglas R. Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| |
Collapse
|
96
|
Yuan J, Zhuang YY, Liu X, Zhang Y, Li K, Chen ZJ, Li D, Chen H, Liang J, Yao Y, Yu X, Zhuo R, Zhao F, Zhou X, Yu X, Qu J, Su J. Exome-wide association study identifies KDELR3 mutations in extreme myopia. Nat Commun 2024; 15:6703. [PMID: 39112444 PMCID: PMC11306401 DOI: 10.1038/s41467-024-50580-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Extreme myopia (EM), defined as a spherical equivalent (SE) ≤ -10.00 diopters (D), is one of the leading causes of sight impairment. Known EM-associated variants only explain limited risk and are inadequate for clinical decision-making. To discover risk genes, we performed a whole-exome sequencing (WES) on 449 EM individuals and 9606 controls. We find a significant excess of rare protein-truncating variants (PTVs) in EM cases, enriched in the retrograde vesicle-mediated transport pathway. Employing single-cell RNA-sequencing (scRNA-seq) and a single-cell polygenic burden score (scPBS), we pinpointed PI16 + /SFRP4+ fibroblasts as the most relevant cell type. We observed that KDELR3 is highly expressed in scleral fibroblast and involved in scleral extracellular matrix (ECM) organization. The zebrafish model revealed that kdelr3 downregulation leads to elongated ocular axial length and increased lens diameter. Together, our study provides insight into the genetics of EM in humans and highlights KDELR3's role in EM pathogenesis.
Collapse
Affiliation(s)
- Jian Yuan
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - You-Yuan Zhuang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyu Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yue Zhang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kai Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Zhen Ji Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dandan Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - He Chen
- School of Biomedical Engineering, Hainan University, Haikou, China
| | - Jiacheng Liang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yinghao Yao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Xiangyi Yu
- Institute of PSI Genomics, Wenzhou, China
| | - Ran Zhuo
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fei Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiangtian Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | | | - Jia Qu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China.
- School of Biomedical Engineering, Hainan University, Haikou, China.
| | - Jianzhong Su
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| |
Collapse
|
97
|
Saidin A, Papazovska Cherepnalkovski A, Shaukat Z, Arsov T, Hussain R, Roberts BJ, Bucat M, Cogelja K, Ricos MG, Dibbens LM. A Novel Pathogenic TUBA1A Variant in a Croatian Infant Is Linked to a Severe Tubulinopathy with Walker-Warburg-like Features. Genes (Basel) 2024; 15:1031. [PMID: 39202391 PMCID: PMC11353499 DOI: 10.3390/genes15081031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Tubulinopathies are associated with malformations of cortical development but not Walker-Warburg Syndrome. Intensive monitoring of a Croatian infant presenting as Walker-Warburg Syndrome in utero began at 21 weeks due to increased growth of cerebral ventricles and foetal biparietal diameter. Monitoring continued until Caesarean delivery at 34 weeks where the infant was eutrophic. Clinical assessment of a progressive neurological disorder of unknown aetiology found a macrocephalic head and markedly hypoplastic genitalia with a micropenis. Neurological examination showed generalized hypotonia with very rare spontaneous movements, hypotonia-induced respiratory insufficiency and ventilator dependence, and generalized myoclonus intensifying during manipulation. With clinical features of hypotonia, lissencephaly, and brain malformations, Walker-Warburg Syndrome was suspected; however, eye anomalies were absent. Genetic trio analysis via whole-exome sequencing only identified a novel de novo mutation in the TUBA1A gene (NM_006009.4:c.848A>G; NP_006000.2:p.His283Arg) in the infant, who died at 2 months of age, as the likely cause. We report a previously unpublished, very rare heterozygous TUBA1A mutation with clinical features of macrocephaly and hypoplastic genitalia which have not previously been associated with the gene. The absence of eye phenotypes or mutations in Walker-Warburg-associated genes confirm this as not a new presentation of Walker-Warburg Syndrome but a novel TUBA1A tubulinopathy for neonatologists to be aware of.
Collapse
Affiliation(s)
- Akzam Saidin
- Epilepsy Research Group, Clinical and Health Sciences, Australian Centre for Precision Health, University of South Australia, Adelaide, SA 5000, Australia; (A.S.); (Z.S.); (R.H.); (M.G.R.)
- Novocraft Technologies, Petaling Jaya 46300, Malaysia
| | - Anet Papazovska Cherepnalkovski
- Department of Neonatology, Clinic for Gynecology and Obstetrics, Clinical Hospital Center Split, 21000 Split, Croatia; (A.P.C.); (M.B.); (K.C.)
- Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Zeeshan Shaukat
- Epilepsy Research Group, Clinical and Health Sciences, Australian Centre for Precision Health, University of South Australia, Adelaide, SA 5000, Australia; (A.S.); (Z.S.); (R.H.); (M.G.R.)
| | - Todor Arsov
- Faculty of Medical Sciences, University Goce Delcev in Shtip, 2000 Shtip, North Macedonia;
| | - Rashid Hussain
- Epilepsy Research Group, Clinical and Health Sciences, Australian Centre for Precision Health, University of South Australia, Adelaide, SA 5000, Australia; (A.S.); (Z.S.); (R.H.); (M.G.R.)
| | - Ben J. Roberts
- Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA 5000, Australia;
| | - Marija Bucat
- Department of Neonatology, Clinic for Gynecology and Obstetrics, Clinical Hospital Center Split, 21000 Split, Croatia; (A.P.C.); (M.B.); (K.C.)
| | - Klara Cogelja
- Department of Neonatology, Clinic for Gynecology and Obstetrics, Clinical Hospital Center Split, 21000 Split, Croatia; (A.P.C.); (M.B.); (K.C.)
| | - Michael G. Ricos
- Epilepsy Research Group, Clinical and Health Sciences, Australian Centre for Precision Health, University of South Australia, Adelaide, SA 5000, Australia; (A.S.); (Z.S.); (R.H.); (M.G.R.)
| | - Leanne M. Dibbens
- Epilepsy Research Group, Clinical and Health Sciences, Australian Centre for Precision Health, University of South Australia, Adelaide, SA 5000, Australia; (A.S.); (Z.S.); (R.H.); (M.G.R.)
| |
Collapse
|
98
|
Lee S, Kang M, So KH, Jang R, Shin YW, Jang SS, Yoon JG, Kim S, Kim M, Chu K, Lee SK, Kim KJ, Baek ST, Lim BC, Moon J. Broadening the scope of multigene panel analysis for adult epilepsy patients. Epilepsia Open 2024; 9:1538-1549. [PMID: 38946282 PMCID: PMC11296137 DOI: 10.1002/epi4.12993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024] Open
Abstract
OBJECTIVE Epilepsy is a suitable target for gene panel sequencing because a considerable portion of epilepsy is now explained by genetic components, especially in syndromic cases. However, previous gene panel studies on epilepsy have mostly focused on pediatric patients. METHODS We enrolled adult epilepsy patients meeting any of the following criteria: family history of epilepsy, seizure onset age ≤ 19 years, neuronal migration disorder, and seizure freedom not achieved by dual anti-seizure medications. We sequenced the exonic regions of 211 epilepsy genes in these patients. To confirm the pathogenicity of a novel MTOR truncating variant, we electroporated vectors with different MTOR variants into developing mouse brains. RESULTS A total of 92 probands and 4 affected relatives were tested, and the proportion of intellectual disability (ID) and/or developmental disability (DD) was 21.7%. As a result, twelve probands (13.0%) had pathogenic or likely pathogenic variants in the following genes or regions: DEPDC5, 15q12-q13 duplication (n = 2), SLC6A1, SYNGAP1, EEF1A2, LGI1, MTOR, KCNQ2, MEF2C, and TSC1 (n = 1). We confirmed the functional impact of a novel truncating mutation in the MTOR gene (c.7570C > T, p.Gln2524Ter) that disrupted neuronal migration in a mouse model. The diagnostic yield was higher in patients with ID/DD or childhood-onset seizures. We also identified additional candidate variants in 20 patients that could be reassessed by further studies. SIGNIFICANCE Our findings underscore the clinical utility of gene panel sequencing in adult epilepsy patients suspected of having genetic etiology, especially those with ID/DD or early-onset seizures. Gene panel sequencing could not only lead to genetic diagnosis in a substantial portion of adult epilepsy patients but also inform more precise therapeutic decisions based on their genetic background. PLAIN LANGUAGE SUMMARY This study demonstrated the effectiveness of gene panel sequencing in adults with epilepsy, revealing pathogenic or likely pathogenic variants in 13.0% of patients. Higher diagnostic yields were observed in those with neurodevelopmental disorders or childhood-onset seizures. Additionally, we have shown that expanding genetic studies into adult patients would uncover new types of pathogenic variants for epilepsy, contributing to the advancement of precision medicine for individuals with epilepsy. In conclusion, our results highlight the practical value of employing gene panel sequencing in adult epilepsy patients, particularly when genetic etiology is clinically suspected.
Collapse
Affiliation(s)
- Seungbok Lee
- Department of Genomic MedicineSeoul National University HospitalSeoulKorea
- Department of PediatricsSeoul National University College of Medicine, Seoul National University Children's HospitalSeoulKorea
| | - Mi‐Kyoung Kang
- Department of NeurologySeoul National University College of Medicine, Seoul National University HospitalSeoulKorea
| | - Ki Hurn So
- Department of Life SciencesPohang University of Science and Technology (POSTECH)PohangKorea
| | - Riyul Jang
- Department of Life SciencesPohang University of Science and Technology (POSTECH)PohangKorea
| | - Yong Woo Shin
- Department of NeurologySeoul National University College of Medicine, Seoul National University HospitalSeoulKorea
| | - Se Song Jang
- Department of PediatricsSeoul National University College of Medicine, Seoul National University Children's HospitalSeoulKorea
| | - Jihoon G. Yoon
- Department of Genomic MedicineSeoul National University HospitalSeoulKorea
| | - Sheehyun Kim
- Department of Genomic MedicineSeoul National University HospitalSeoulKorea
| | - Manjin Kim
- Department of Genomic MedicineSeoul National University HospitalSeoulKorea
- Department of Laboratory MedicineSeoul National University College of Medicine, Seoul National University HospitalSeoulKorea
| | - Kon Chu
- Department of NeurologySeoul National University College of Medicine, Seoul National University HospitalSeoulKorea
| | - Sang Kun Lee
- Department of NeurologySeoul National University College of Medicine, Seoul National University HospitalSeoulKorea
| | - Ki Joong Kim
- Department of PediatricsSeoul National University College of Medicine, Seoul National University Children's HospitalSeoulKorea
| | - Seung Tae Baek
- Department of Life SciencesPohang University of Science and Technology (POSTECH)PohangKorea
| | - Byung Chan Lim
- Department of PediatricsSeoul National University College of Medicine, Seoul National University Children's HospitalSeoulKorea
| | - Jangsup Moon
- Department of Genomic MedicineSeoul National University HospitalSeoulKorea
- Department of NeurologySeoul National University College of Medicine, Seoul National University HospitalSeoulKorea
| |
Collapse
|
99
|
Stawiński P, Płoski R. Genebe.net: Implementation and validation of an automatic ACMG variant pathogenicity criteria assignment. Clin Genet 2024; 106:119-126. [PMID: 38440907 DOI: 10.1111/cge.14516] [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: 11/17/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/06/2024]
Abstract
We present GeneBe, an online platform streamlining the automated application of American College of Medical Genetics and Genomics (ACMG), Association for Molecular Pathology (AMP), and the College of American Pathologists (CAP) criteria for assessment of pathogenicity of genetic variants. GeneBe utilizes automated algorithms that evaluate 17 criteria from 28, closely aligning with current guidelines and leveraging data from diverse sources, including ClinVar. The user-friendly web interface enables manual refinement of assignments for specific criteria based on site-collected data. Our algorithm demonstrates a high correlation (r = 0.90) of assigned pathogenicity scores compared to expert assessments from the ClinGen Evidence Repository and substantial concordance with ClinVar verdict assignments (κ = 0.69). Comparative analysis with other published tools reveals that GeneBe performs similarly to VarSome while being superior over TAPES and InterVar. In contrast to some other tools, GeneBe's web implementation is tracker-free and third-party request-free, safeguarding user privacy. Additionally, GeneBe offers an Application Programming Interface (API) for enhanced flexibility and integration into existing workflows and is provided free of charge for research purposes. GeneBe is available at https://genebe.net.
Collapse
Affiliation(s)
- Piotr Stawiński
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
100
|
Bonetti E, Tini G, Mazzarella L. Accuracy of renovo predictions on variants reclassified over time. J Transl Med 2024; 22:713. [PMID: 39085881 PMCID: PMC11293099 DOI: 10.1186/s12967-024-05508-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/14/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Interpreting the clinical consequences of genetic variants is the central problem in modern clinical genomics, for both hereditary diseases and oncology. However, clinical validation lags behind the pace of discovery, leading to distressing uncertainty for patients, physicians and researchers. This "interpretation gap" changes over time as evidence accumulates, and variants initially deemed of uncertain (VUS) significance may be subsequently reclassified in pathogenic/benign. We previously developed RENOVO, a random forest-based tool able to predict variant pathogenicity based on publicly available information from GnomAD and dbNFSP, and tested on variants that have changed their classification status over time. Here, we comprehensively evaluated the accuracy of RENOVO predictions on variants that have been reclassified over the last four years. METHODS we retrieved 16 retrospective instances of the ClinVar database, every 3 months since March 2020 to March 2024, and analyzed time trends of variant classifications. We identified variants that changed their status over time and compared RENOVO predictions generated in 2020 with the actual reclassifications. RESULTS VUS have become the most represented class in ClinVar (44.97% vs. 9.75% (likely) pathogenic and 40,33% (likely) benign). The rate of VUS reclassification is linear and slow compared to the rate of VUS reporting, exponential and currently ~ 30x faster, creating a growing divide between what can be sequenced vs. what can be interpreted. Out of 10,196 VUS variants in January 2020 that have undergone a clinically meaningful reclassification to march 2024, RENOVO correctly classified 82.6% in 2020. In addition, RENOVO correctly identified the majority of the few variants that switched clinically meaningful classes (e.g., from benign to pathogenic and vice versa). We highlight variant classes and clinically relevant genes for which RENOVO provides particularly accurate estimates. In particularly, genes characterized by large prevalence of high- or low-impact variants (e.g., POLE, NOTCH1, FANCM etc.). Suboptimal RENOVO predictions mostly concern genes validated through dedicated consortia (e.g., BRCA1/2), in which RENOVO would anyway have a limited impact. CONCLUSIONS Time trend analysis demonstrates that the current model of variant interpretation cannot keep up with variant discovery. Machine learning-based tools like RENOVO confirm high accuracy that can aid in clinical practice and research.
Collapse
Affiliation(s)
- Emanuele Bonetti
- Department of Experimental Oncology, European Institute of Oncology, IEO-IRCCS, Milan, 20139, Italy
| | - Giulia Tini
- Department of Experimental Oncology, European Institute of Oncology, IEO-IRCCS, Milan, 20139, Italy
| | - Luca Mazzarella
- Department of Experimental Oncology, European Institute of Oncology, IEO-IRCCS, Milan, 20139, Italy.
| |
Collapse
|