1
|
Knowles EEM, Peralta JM, Rodrigue AL, Mathias SR, Mollon J, Leandro AC, Curran JE, Blangero J, Glahn DC. Differential gene expression study in whole blood identifies candidate genes for psychosis in African American individuals. Schizophr Res 2025; 280:85-94. [PMID: 40267851 DOI: 10.1016/j.schres.2025.04.018] [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: 06/26/2023] [Revised: 03/10/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association has identified regions of the genome that mediate risk for psychosis. It is possible that variants in these regions confer risk by altering gene expression. This work has predominantly been conducted in individuals of European descent and has focused narrowly on schizophrenia rather than psychosis as a syndrome. In the present study we investigated alterations in gene expression in African American individuals with a range of psychotic diagnoses to increase understanding of the etiology in an underserved population. We performed RNA-seq in whole bloody to survey the transcriptome in 126 patients with a psychosis-spectrum disorder and 217 healthy controls and applied differential gene expression analyses across the genome while controlling for age, sex, population stratification and batch. We found 18 differentially expressed genes (DEGs), some of the locations of the corresponding genes overlap with previously implicated regions for psychosis, but many of which were novel associations. Enrichment analysis of nominally significant genes (p < 0.05) revealed overrepresentation of biological processes relating to platelet, immune and cellular function, and sensory perception. Weighted gene co-expression network analysis, applied to identify modules of co-expressed genes associated with psychosis, revealed 10 modules, one of which was significantly associated with psychosis. This module was significantly enriched for DEGs, and for platelet function. These results support the potential role of immune function in the etiology of psychosis, identify novel candidate gene expression phenotypes that correspond to both established and new genomic regions, in individuals of African American ancestry.
Collapse
Affiliation(s)
- E E M Knowles
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | - J M Peralta
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - A L Rodrigue
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - S R Mathias
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - J Mollon
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - A C Leandro
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - J E Curran
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - J Blangero
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - D C Glahn
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| |
Collapse
|
2
|
Joshi N, Lango-Allen H, Downes K, Simeoni I, Vladescu C, Paul D, Hart A, Ademokun C, Cooper N. The role of genetic sequencing in the diagnostic workup for chronic immune thrombocytopenia. Blood Adv 2025; 9:1497-1507. [PMID: 39808791 PMCID: PMC11985033 DOI: 10.1182/bloodadvances.2024014639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRACT Immune thrombocytopenia (ITP) is a heterogenous autoimmune disorder diagnosed by excluding other conditions. Misdiagnosis of primary ITP occurs in patients with inherited thrombocytopenia and primary immunodeficiency syndromes. This study investigates whether genetic testing for inherited thrombocytopenia or primary immunodeficiency can enhance diagnostic accuracy in ITP, and guide treatment strategies. We performed whole genome sequencing or targeted panel sequencing on peripheral blood samples in a cohort of 80 participants with chronic ITP, utilizing the ThromboGenomics panel (n = 72) and the Genomics of Rare Immune Disorders panel (n = 50) consisting of genes known to cause bleeding and platelet disorders (BPDG) or primary immuodeficiency genes (PIDG) respectively. A replication cohort of 73 patients underwent clinical genomics testing with either the R90 (BPDG, n = 35) or R15 (PIDG, n = 50) National Health Service Genomics panels. Known pathogenic or likely pathogenic, disease-causing, variants were identified in 9 patients in the first cohort (11%, 95% confidence interval [CI]: 5-20); 7 patients (10%, 95% CI: 4-19) in BPDG and 2 patients (4% CI,1-14) in PIDG. In addition, 26 patients (32.5%) carried variants of uncertain significance. In the replication cohort, 8% (95% CI, 2-20) and 9% (95% CI, 2-23) of patients had a pathogenic variant identified on the R15 (PIDG) or R90 panel (BPDG), respectively. The findings impacted clinical management such as avoidance of immunosuppression (ANKRD26, GP1BB, ETV6, TUBB1, and ITGB3) and eligibility for allogeneic stem cell transplantation (UNC13D). Our findings demonstrate that genomic sequencing identifies diagnostically relevant variants in patients with chronic ITP. Identification of these variants can guide treatment decisions and improve patient outcomes.
Collapse
Affiliation(s)
- Nehal Joshi
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Hana Lango-Allen
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Ilenia Simeoni
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Camelia Vladescu
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | - Deena Paul
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | - Alice Hart
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | - Christine Ademokun
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
| | - Nichola Cooper
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Li J, Li J, Li X, Wang W, Ding Y, Zhou J, Wang W, Xi Y, Gou R, Liu S, Zhou Z, Gao M. Identification of coagulation diagnostic biomarkers related to the severity of spinal cord injury. Int Immunopharmacol 2024; 137:112505. [PMID: 38908081 DOI: 10.1016/j.intimp.2024.112505] [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: 01/17/2024] [Revised: 04/02/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Blood always shows coagulation changes after spinal cord injury (SCI), and identifying these blood changes may be helpful for diagnosis and treatment of SCI. Nevertheless, studies to date on blood coagulation changes after SCI in humans are not comprehensive. Therefore, this study aims to identify blood coagulation diagnostic biomarkers and immune changes related to SCI and its severity levels. METHODS Human blood sequencing datasets were obtained from public databases. Differentially expressed coagulation-related genes were analyzed (DECRGs). Enrichment analysis and assessment of immune changes were conducted. Weighted gene co-expression network analysis, least absolute shrinkage and selection operator logistic regression were used to identify biomarkers. Validation for these biomarkers was performed. The correlation between biomarkers and immune cells was evaluated. Transcription factors, miRNA, lncRNA, and drugs that can regulate biomarkers were analyzed. RESULTS DECRGs associated with SCI and its different grades were identified, showing enrichment in altered coagulation and immune-related signaling pathways. ADAM9, CD55, and STAT4 were identified as coagulation diagnostic biomarkers for SCI. IRF4 and PABPC4 were identified as coagulation diagnostic biomarkers for American Spinal Injury Association Impairment Scale (AIS) A grade of SCI. GP9 was designated as a diagnostic biomarker for AIS D grade of SCI. Immune changes in blood of SCI and its different grades were observed. Correlation between diagnostic biomarkers and immune cells were identified. Transcription factors, miRNA, lncRNA, and drugs that can regulate diagnostic biomarker expression were discovered. CONCLUSION Therefore, detecting the expression of these putative diagnostic biomarkers and related immune changes may be helpful for predicting the severity of SCI. Uncovering potential regulatory mechanisms for biomarkers may be beneficial for further research.
Collapse
Affiliation(s)
- Jianfeng Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China, 518107; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China, 510080
| | - Junhong Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China, 518107; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China, 510080
| | - Xianlong Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China, 518107; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China, 510080
| | - Wei Wang
- Linyi Central Hospital, Linyi, Shandong, China, 276000
| | - Yi Ding
- Department of Spine Surgery, Ganzhou People's Hospital, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi, China, 341000
| | - Jiaxiang Zhou
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China, 266000
| | - Wentao Wang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China, 518107; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China, 510080
| | - Yongming Xi
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China, 266000
| | - Ruijie Gou
- Department of Orthopedics and Trauma, The Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China, 650091
| | - Shaoyu Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China, 510080
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China, 518107; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China, 510080.
| | - Manman Gao
- Department of Pediatric Orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian, China, 350007.
| |
Collapse
|
4
|
Stefanucci L, Collins J, Sims MC, Barrio-Hernandez I, Sun L, Burren OS, Perfetto L, Bender I, Callahan TJ, Fleming K, Guerrero JA, Hermjakob H, Martin MJ, Stephenson J, Paneerselvam K, Petrovski S, Porras P, Robinson PN, Wang Q, Watkins X, Frontini M, Laskowski RA, Beltrao P, Di Angelantonio E, Gomez K, Laffan M, Ouwehand WH, Mumford AD, Freson K, Carss K, Downes K, Gleadall N, Megy K, Bruford E, Vuckovic D. The effects of pathogenic and likely pathogenic variants for inherited hemostasis disorders in 140 214 UK Biobank participants. Blood 2023; 142:2055-2068. [PMID: 37647632 PMCID: PMC10733830 DOI: 10.1182/blood.2023020118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023] Open
Abstract
Rare genetic diseases affect millions, and identifying causal DNA variants is essential for patient care. Therefore, it is imperative to estimate the effect of each independent variant and improve their pathogenicity classification. Our study of 140 214 unrelated UK Biobank (UKB) participants found that each of them carries a median of 7 variants previously reported as pathogenic or likely pathogenic. We focused on 967 diagnostic-grade gene (DGG) variants for rare bleeding, thrombotic, and platelet disorders (BTPDs) observed in 12 367 UKB participants. By association analysis, for a subset of these variants, we estimated effect sizes for platelet count and volume, and odds ratios for bleeding and thrombosis. Variants causal of some autosomal recessive platelet disorders revealed phenotypic consequences in carriers. Loss-of-function variants in MPL, which cause chronic amegakaryocytic thrombocytopenia if biallelic, were unexpectedly associated with increased platelet counts in carriers. We also demonstrated that common variants identified by genome-wide association studies (GWAS) for platelet count or thrombosis risk may influence the penetrance of rare variants in BTPD DGGs on their associated hemostasis disorders. Network-propagation analysis applied to an interactome of 18 410 nodes and 571 917 edges showed that GWAS variants with large effect sizes are enriched in DGGs and their first-order interactors. Finally, we illustrate the modifying effect of polygenic scores for platelet count and thrombosis risk on disease severity in participants carrying rare variants in TUBB1 or PROC and PROS1, respectively. Our findings demonstrate the power of association analyses using large population datasets in improving pathogenicity classifications of rare variants.
Collapse
Affiliation(s)
- Luca Stefanucci
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Janine Collins
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
| | - Matthew C. Sims
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Inigo Barrio-Hernandez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Luanluan Sun
- Department of Public Health and Primary Care, BHF Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Oliver S. Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Livia Perfetto
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
- Department of Biology and Biotechnology “C.Darwin,” Sapienza University of Rome, Rome, Italy
| | - Isobel Bender
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Tiffany J. Callahan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY
| | - Kathryn Fleming
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Jose A. Guerrero
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Maria J. Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - James Stephenson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - NIHR BioResource
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
- Department of Public Health and Primary Care, BHF Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Department of Biology and Biotechnology “C.Darwin,” Sapienza University of Rome, Rome, Italy
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, United Kingdom
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
- Genomic Medicine, The Jackson Laboratory, Farmington, CT
- Institute for Systems Genomics, University of Connecticut, Farmington, CT
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences RILD Building, University of Exeter Medical School, Exeter, United Kingdom
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
- Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Health Data Science Centre, Human Technopole, Milan, Italy
- Haemophilia Centre and Thrombosis Unit, Royal Free London NHS Foundation Trust, London, United Kingdom
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, London, United Kingdom
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KULeuven, Leuven, Belgium
- Cambridge Genomics Laboratory, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Kalpana Paneerselvam
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, United Kingdom
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
| | - Pablo Porras
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Peter N. Robinson
- Genomic Medicine, The Jackson Laboratory, Farmington, CT
- Institute for Systems Genomics, University of Connecticut, Farmington, CT
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Xavier Watkins
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences RILD Building, University of Exeter Medical School, Exeter, United Kingdom
| | - Roman A. Laskowski
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Pedro Beltrao
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Emanuele Di Angelantonio
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Public Health and Primary Care, BHF Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
- Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Health Data Science Centre, Human Technopole, Milan, Italy
| | - Keith Gomez
- Haemophilia Centre and Thrombosis Unit, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Mike Laffan
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, London, United Kingdom
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Andrew D. Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KULeuven, Leuven, Belgium
| | - Keren Carss
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Cambridge Genomics Laboratory, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nick Gleadall
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Karyn Megy
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Elspeth Bruford
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Dragana Vuckovic
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Barozzi S, Pecci A, Marinoni M, Fontana G, Zanchetta ME, Noris P, Savoia A, Faleschini M. GP1BB c.179C > T is the most frequent cause of monoallelic Bernard-Soulier syndrome in the Italian population after the Bolzano variant: a report of two new families. Ann Hematol 2023; 102:677-679. [PMID: 36539614 DOI: 10.1007/s00277-022-05079-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Serena Barozzi
- Medicina Generale 1, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessandro Pecci
- Medicina Generale 1, IRCCS Policlinico San Matteo Foundation, Pavia, Italy.,Department of Internal Medicine, University of Pavia, Pavia, Italy
| | | | - Giorgia Fontana
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | | | - Patrizia Noris
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Anna Savoia
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Michela Faleschini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
6
|
Zhang F, Zhang Q, Zhu J, Yao B, Ma C, Qiao N, He S, Ye Z, Wang Y, Han R, Feng J, Wang Y, Qin Z, Ma Z, Li K, Zhang Y, Tian S, Chen Z, Tan S, Wu Y, Ran P, Wang Y, Ding C, Zhao Y. Integrated proteogenomic characterization across major histological types of pituitary neuroendocrine tumors. Cell Res 2022; 32:1047-1067. [PMID: 36307579 PMCID: PMC9715725 DOI: 10.1038/s41422-022-00736-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 09/30/2022] [Indexed: 02/07/2023] Open
Abstract
Pituitary neuroendocrine tumor (PitNET) is one of the most common intracranial tumors. Due to its extensive tumor heterogeneity and the lack of high-quality tissues for biomarker discovery, the causative molecular mechanisms are far from being fully defined. Therefore, more studies are needed to improve the current clinicopathological classification system, and advanced treatment strategies such as targeted therapy and immunotherapy are yet to be explored. Here, we performed the largest integrative genomics, transcriptomics, proteomics, and phosphoproteomics analysis reported to date for a cohort of 200 PitNET patients. Genomics data indicate that GNAS copy number gain can serve as a reliable diagnostic marker for hyperproliferation of the PIT1 lineage. Proteomics-based classification of PitNETs identified 7 clusters, among which, tumors overexpressing epithelial-mesenchymal transition (EMT) markers clustered into a more invasive subgroup. Further analysis identified potential therapeutic targets, including CDK6, TWIST1, EGFR, and VEGFR2, for different clusters. Immune subtyping to explore the potential for application of immunotherapy in PitNET identified an association between alterations in the JAK1-STAT1-PDL1 axis and immune exhaustion, and between changes in the JAK3-STAT6-FOS/JUN axis and immune infiltration. These identified molecular markers and alternations in various clusters/subtypes were further confirmed in an independent cohort of 750 PitNET patients. This proteogenomic analysis across traditional histological boundaries improves our current understanding of PitNET pathophysiology and suggests novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Fan Zhang
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qilin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. .,National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jiajun Zhu
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Boyuan Yao
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chi Ma
- grid.462338.80000 0004 0605 6769State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan center for outstanding overseas scientists of pulmonary fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan China
| | - Nidan Qiao
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shiman He
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhao Ye
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunzhi Wang
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rui Han
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinwen Feng
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongfei Wang
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhaoyu Qin
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zengyi Ma
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kai Li
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yichao Zhang
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sha Tian
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengyuan Chen
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Subei Tan
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Wu
- grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Ran
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Wang
- grid.8547.e0000 0001 0125 2443Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. .,National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. .,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China. .,Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China. .,Neurosurgical Institute of Fudan University, Shanghai, China. .,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Dib F, Quéméner A, Bayart S, Boisseau P, Babuty A, Trossaërt M, Sigaud M, Ternisien C, Drillaud N, Eveillard M, Guillet B, Béné MC, Fouassier M. Biological, clinical features and modelling of heterozygous variants of glycoprotein Ib platelet subunit alpha (GP1BA) and glycoprotein Ib platelet subunit beta (GP1BB) genes responsible for constitutional thrombocytopenia. Br J Haematol 2022; 199:744-753. [PMID: 36173017 DOI: 10.1111/bjh.18462] [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: 03/04/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022]
Abstract
Constitutional thrombocytopenias are rare disorders, often difficult to discriminate from acquired thrombocytopenias. More than 80 genes have been described as being at the origin of these diseases. Among them, several variants of the glycoprotein Ib platelet subunit alpha (GP1BA) and glycoprotein Ib platelet subunit beta (GP1BB) genes, coding for the GpIb-IX-V glycoprotein complex, have been reported in the literature. The study reported here aimed at describing newly identified monoallelic anomalies affecting the GP1BA and GP1BB genes on a clinical, biological and molecular level. In a cohort of nine patients with macrothrombocytopenia, eight heterozygous variants of the GP1BA or GP1BB genes were identified. Five of them had never been described in the heterozygous state. Computer modelling disclosed structure/function relationships of these five variants.
Collapse
Affiliation(s)
- Fatema Dib
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France
| | - Agnès Quéméner
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France
| | | | - Pierre Boisseau
- Service de Génétique Médicale, CHU de Nantes, Nantes, France
| | - Antoine Babuty
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,CRC-MH, CHU de Nantes, Nantes, France
| | - Marc Trossaërt
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,CRC-MH, CHU de Nantes, Nantes, France
| | - Marianne Sigaud
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,CRC-MH, CHU de Nantes, Nantes, France
| | - Catherine Ternisien
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,CRC-MH, CHU de Nantes, Nantes, France
| | - Nicolas Drillaud
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,CRC-MH, CHU de Nantes, Nantes, France
| | - Marion Eveillard
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France
| | - Benoit Guillet
- CRC-MH, CHU de Rennes, Rennes, France.,Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Marie C Béné
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France
| | - Marc Fouassier
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France.,CRC-MH, CHU de Nantes, Nantes, France
| |
Collapse
|
8
|
Warren JT, Di Paola J. Genetics of inherited thrombocytopenias. Blood 2022; 139:3264-3277. [PMID: 35167650 PMCID: PMC9164741 DOI: 10.1182/blood.2020009300] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/04/2022] [Indexed: 01/19/2023] Open
Abstract
The inherited thrombocytopenia syndromes are a group of disorders characterized primarily by quantitative defects in platelet number, though with a variety demonstrating qualitative defects and/or extrahematopoietic findings. Through collaborative international efforts applying next-generation sequencing approaches, the list of genetic syndromes that cause thrombocytopenia has expanded significantly in recent years, now with over 40 genes implicated. In this review, we focus on what is known about the genetic etiology of inherited thrombocytopenia syndromes and how the field has worked to validate new genetic discoveries. We highlight the important role for the clinician in identifying a germline genetic diagnosis and strategies for identifying novel causes through research-based endeavors.
Collapse
Affiliation(s)
- Julia T Warren
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Jorge Di Paola
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
9
|
Skalníková M, Staňo Kozubík K, Trizuljak J, Vrzalová Z, Radová L, Réblová K, Holbová R, Kurucová T, Svozilová H, Štika J, Blaháková I, Dvořáčková B, Prudková M, Stehlíková O, Šmída M, Křen L, Smejkal P, Pospíšilová Š, Doubek M. A GP1BA Variant in a Czech Family with Monoallelic Bernard-Soulier Syndrome. Int J Mol Sci 2022; 23:ijms23020885. [PMID: 35055070 PMCID: PMC8777725 DOI: 10.3390/ijms23020885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Bernard-Soulier syndrome (BSS) is a rare inherited disorder characterized by unusually large platelets, low platelet count, and prolonged bleeding time. BSS is usually inherited in an autosomal recessive (AR) mode of inheritance due to a deficiency of the GPIb-IX-V complex also known as the von Willebrand factor (VWF) receptor. We investigated a family with macrothrombocytopenia, a mild bleeding tendency, slightly lowered platelet aggregation tests, and suspected autosomal dominant (AD) inheritance. We have detected a heterozygous GP1BA likely pathogenic variant, causing monoallelic BSS. A germline GP1BA gene variant (NM_000173:c.98G > A:p.C33Y), segregating with the macrothrombocytopenia, was detected by whole-exome sequencing. In silico analysis of the protein structure of the novel GPIbα variant revealed a potential structural defect, which could impact proper protein folding and subsequent binding to VWF. Flow cytometry, immunoblot, and electron microscopy demonstrated further differences between p.C33Y GP1BA carriers and healthy controls. Here, we provide a detailed insight into its clinical presentation and phenotype. Moreover, the here described case first presents an mBSS patient with two previous ischemic strokes.
Collapse
Affiliation(s)
- Magdalena Skalníková
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
- Correspondence: (M.S.); (M.D.); Tel.: +421-54-949-8293 (M.S.)
| | - Kateřina Staňo Kozubík
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
| | - Jakub Trizuljak
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Zuzana Vrzalová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
| | - Lenka Radová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
| | - Kamila Réblová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Radka Holbová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
| | - Terézia Kurucová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
| | - Hana Svozilová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Jiří Štika
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
| | - Ivona Blaháková
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
| | - Barbara Dvořáčková
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
| | - Marie Prudková
- Department of Clinical Hematology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (M.P.); (P.S.)
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Olga Stehlíková
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
| | - Michal Šmída
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
| | - Leoš Křen
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic;
| | - Petr Smejkal
- Department of Clinical Hematology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (M.P.); (P.S.)
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Šárka Pospíšilová
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Michael Doubek
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (K.S.K.); (J.T.); (Z.V.); (L.R.); (K.R.); (R.H.); (T.K.); (H.S.); (J.Š.); (I.B.); (M.Š.); (Š.P.)
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; (B.D.); (O.S.)
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
- Correspondence: (M.S.); (M.D.); Tel.: +421-54-949-8293 (M.S.)
| |
Collapse
|
10
|
Collins J, Astle WJ, Megy K, Mumford AD, Vuckovic D. Advances in understanding the pathogenesis of hereditary macrothrombocytopenia. Br J Haematol 2021; 195:25-45. [PMID: 33783834 DOI: 10.1111/bjh.17409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/19/2021] [Indexed: 12/14/2022]
Abstract
Low platelet count, or thrombocytopenia, is a common haematological abnormality, with a wide differential diagnosis, which may represent a clinically significant underlying pathology. Macrothrombocytopenia, the presence of large platelets in combination with thrombocytopenia, can be acquired or hereditary and indicative of a complex disorder. In this review, we discuss the interpretation of platelet count and volume measured by automated haematology analysers and highlight some important technical considerations relevant to the analysis of blood samples with macrothrombocytopenia. We review how large cohorts, such as the UK Biobank and INTERVAL studies, have enabled an accurate description of the distribution and co-variation of platelet parameters in adult populations. We discuss how genome-wide association studies have identified hundreds of genetic associations with platelet count and mean platelet volume, which in aggregate can explain large fractions of phenotypic variance, consistent with a complex genetic architecture and polygenic inheritance. Finally, we describe the large genetic diagnostic and discovery programmes, which, simultaneously to genome-wide association studies, have expanded the repertoire of genes and variants associated with extreme platelet phenotypes. These have advanced our understanding of the pathogenesis of hereditary macrothrombocytopenia and support a future clinical diagnostic strategy that utilises genotype alongside clinical and laboratory phenotype data.
Collapse
Affiliation(s)
- Janine Collins
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, Barts Health NHS Trust, London, UK
| | - William J Astle
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge Institute of Public Health, Forvie Site, Robinson Way, Cambridge, UK
| | - Karyn Megy
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrew D Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Dragana Vuckovic
- Department of Biostatistics and Epidemiology, Faculty of Medicine, Imperial College London, London, UK
- Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| |
Collapse
|
11
|
Gomez K, Anderson J, Baker P, Biss T, Jennings I, Lowe G, Platton S. Clinical and laboratory diagnosis of heritable platelet disorders in adults and children: a British Society for Haematology Guideline. Br J Haematol 2021; 195:46-72. [PMID: 34435350 DOI: 10.1111/bjh.17690] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Keith Gomez
- Haemophilia Centre and Thrombosis Unit, Royal Free London NHS Foundation Trust, London
| | - Julia Anderson
- Haemophilia Thrombosis and Immunology Centre, Royal Infirmary, NHS Lothian, Edinburgh
| | - Peter Baker
- Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, Oxford
| | - Tina Biss
- Haemophilia Comprehensive Care Centre, Royal Victoria Infirmary, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne
| | - Ian Jennings
- UK NEQAS for Blood Coagulation, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield
| | - Gillian Lowe
- Haemophilia Comprehensive Care Centre, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Sean Platton
- Haemophilia Centre, The Royal London Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
12
|
A Novel Mutation in GP1BB Reveals the Role of the Cytoplasmic Domain of GPIbβ in the Pathophysiology of Bernard-Soulier Syndrome and GPIb-IX Complex Assembly. Int J Mol Sci 2021; 22:ijms221910190. [PMID: 34638529 PMCID: PMC8508601 DOI: 10.3390/ijms221910190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 12/24/2022] Open
Abstract
Bernard-Soulier syndrome (BSS) is an autosomal-recessive bleeding disorder caused by biallelic variants in the GP1BA, GP1BB, and GP9 genes encoding the subunits GPIbα, GPIbβ, and GPIX of the GPIb-IX complex. Pathogenic variants usually affect the extracellular or transmembrane domains of the receptor subunits. We investigated a family with BSS caused by the homozygous c.528_550del (p.Arg177Serfs*124) variant in GP1BB, which is the first mutation ever identified that affects the cytoplasmic domain of GPIbβ. The loss of the intracytoplasmic tail of GPIbβ results in a mild form of BSS, characterized by only a moderate reduction of the GPIb-IX complex expression and mild or absent bleeding tendency. The variant induces a decrease of the total platelet expression of GPIbβ; however, all of the mutant subunit expressed in platelets is correctly assembled into the GPIb-IX complex in the plasma membrane, indicating that the cytoplasmic domain of GPIbβ is not involved in assembly and trafficking of the GPIb-IX receptor. Finally, the c.528_550del mutation exerts a dominant effect and causes mild macrothrombocytopenia in heterozygous individuals, as also demonstrated by the investigation of a second unrelated pedigree. The study of this novel GP1BB variant provides new information on pathophysiology of BSS and the assembly mechanisms of the GPIb-IX receptor.
Collapse
|
13
|
Tsai FD, Battinelli EM. Inherited Platelet Disorders. Hematol Oncol Clin North Am 2021; 35:1069-1084. [PMID: 34391603 DOI: 10.1016/j.hoc.2021.07.003] [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] [Indexed: 01/19/2023]
Abstract
Bleeding disorders due to platelet dysfunction are a common hematologic complication affecting patients, and typically present with mucocutaneous bleeding or hemorrhage. An inherited platelet disorder should be suspected in individuals with a suggestive family history and no identified secondary causes of bleeding. Genetic defects have been described at all levels of platelet activation, including receptor binding, signaling, granule release, cytoskeletal remodeling, and platelet hematopoiesis. Management of these disorders is typically supportive, with an emphasis on awareness, patient education, and anticipatory guidance to prevent future episodes of bleeding.
Collapse
Affiliation(s)
- Frederick D Tsai
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA; Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Elisabeth M Battinelli
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Inherited Platelet Disorders: An Updated Overview. Int J Mol Sci 2021; 22:ijms22094521. [PMID: 33926054 PMCID: PMC8123627 DOI: 10.3390/ijms22094521] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Platelets play a major role in hemostasis as ppwell as in many other physiological and pathological processes. Accordingly, production of about 1011 platelet per day as well as appropriate survival and functions are life essential events. Inherited platelet disorders (IPDs), affecting either platelet count or platelet functions, comprise a heterogenous group of about sixty rare diseases caused by molecular anomalies in many culprit genes. Their clinical relevance is highly variable according to the specific disease and even within the same type, ranging from almost negligible to life-threatening. Mucocutaneous bleeding diathesis (epistaxis, gum bleeding, purpura, menorrhagia), but also multisystemic disorders and/or malignancy comprise the clinical spectrum of IPDs. The early and accurate diagnosis of IPDs and a close patient medical follow-up is of great importance. A genotype-phenotype relationship in many IPDs makes a molecular diagnosis especially relevant to proper clinical management. Genetic diagnosis of IPDs has been greatly facilitated by the introduction of high throughput sequencing (HTS) techniques into mainstream investigation practice in these diseases. However, there are still unsolved ethical concerns on general genetic investigations. Patients should be informed and comprehend the potential implications of their genetic analysis. Unlike the progress in diagnosis, there have been no major advances in the clinical management of IPDs. Educational and preventive measures, few hemostatic drugs, platelet transfusions, thrombopoietin receptor agonists, and in life-threatening IPDs, allogeneic hematopoietic stem cell transplantation are therapeutic possibilities. Gene therapy may be a future option. Regular follow-up by a specialized hematology service with multidisciplinary support especially for syndromic IPDs is mandatory.
Collapse
|
15
|
Dunstan-Harrison C, Morison IM, Ledgerwood EC. A novel frameshift GP1BB mutation causes autosomal dominant macrothrombocytopenia with decreased vWF receptor expression but normal platelet aggregation. Platelets 2021; 33:324-327. [PMID: 33813986 DOI: 10.1080/09537104.2021.1909716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
GP1bβ is a component of the von Willebrand factor (vWF) receptor complex that is necessary for platelet formation and activation. A novel frameshift variant in GP1BB has been identified in a family with macrothrombocytopenia. The variant leads to a protein that is 101 amino acids longer than wild type with loss of the transmembrane domain. As there is no defect in platelet aggregation, the family are classified as heterozygous carriers of a Bernard-Soulier syndrome-related mutation. The levels of the vWF receptor on platelets are reduced to 50% of the controls, with the presence of large platelets but normal platelet aggregation demonstrating that decreased vWF receptor expression impacts proplatelet formation but not platelet function.
Collapse
Affiliation(s)
| | - Ian M Morison
- Departments of Pathology, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
16
|
Quach ME, Li R. Structure-function of platelet glycoprotein Ib-IX. J Thromb Haemost 2020; 18:3131-3141. [PMID: 32735697 PMCID: PMC7854888 DOI: 10.1111/jth.15035] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/20/2022]
Abstract
The glycoprotein (GP)Ib-IX receptor complex plays a critical role in platelet physiology and pathology. Its interaction with von Willebrand factor (VWF) on the subendothelial matrix instigates platelet arrest at the site of vascular injury and is vital to primary hemostasis. Its reception to other ligands and counter-receptors in the bloodstream also contribute to various processes of platelet biology that are still being discovered. While its basic composition and its link to congenital bleeding disorders were well documented and firmly established more than 25 years ago, recent years have witnessed critical advances in the organization, dynamics, activation, regulation, and functions of the GPIb-IX complex. This review summarizes important findings and identifies questions that remain about this unique platelet mechanoreceptor complex.
Collapse
Affiliation(s)
- M Edward Quach
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
17
|
Pecci A, Balduini CL. Inherited thrombocytopenias: an updated guide for clinicians. Blood Rev 2020; 48:100784. [PMID: 33317862 DOI: 10.1016/j.blre.2020.100784] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
Abstract
The great advances in the knowledge of inherited thrombocytopenias (ITs) made since the turn of the century have significantly changed our view of these conditions. To date, ITs encompass 45 disorders with different degrees of complexity of the clinical picture and very wide variability in the prognosis. They include forms characterized by thrombocytopenia alone, forms that present with other congenital defects, and conditions that predispose to acquire additional diseases over the course of life. In this review, we recapitulate the clinical features of ITs with emphasis on the forms predisposing to additional diseases. We then discuss the key issues for a rational approach to the diagnosis of ITs in clinical practice. Finally, we aim to provide an updated and comprehensive guide to the treatment of ITs, including the management of hemostatic challenges, the treatment of severe forms, and the approach to the manifestations that add to thrombocytopenia.
Collapse
Affiliation(s)
- Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy.
| | | |
Collapse
|
18
|
Busonero F, Steri M, Orrù V, Sole G, Olla S, Marongiu M, Maschio A, Sidore C, Lai S, Mulas A, Zoledziewska M, Floris M, Pala M, Forabosco P, Asunis I, Pitzalis M, Deidda F, Masala M, Caria CA, Barella S, Abecasis GR, Schlessinger D, Sanna S, Fiorillo E, Cucca F. A Sardinian founder mutation in glycoprotein Ib platelet subunit beta (GP1BB) that impacts thrombocytopenia. Br J Haematol 2020; 191:e124-e128. [PMID: 33216977 DOI: 10.1111/bjh.17090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Fabio Busonero
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Gabriella Sole
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Stefania Olla
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Michele Marongiu
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Andrea Maschio
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Carlo Sidore
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Sandra Lai
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Magdalena Zoledziewska
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Matteo Floris
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - Mauro Pala
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Paola Forabosco
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Isadora Asunis
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Maristella Pitzalis
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Francesca Deidda
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Marco Masala
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Cristian Antonio Caria
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Susanna Barella
- Ospedale Pediatrico Microcitemico 'Antonio Cao' (A.O.Brotzu), Cagliari, Italy
| | - Goncalo R Abecasis
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - David Schlessinger
- Laboratory of Genetics and Genomics, National Institute on Aging, US National Institutes of Health, Baltimore, MD, USA
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (Cagliari), Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| |
Collapse
|
19
|
Van Hout CV, Tachmazidou I, Backman JD, Hoffman JD, Liu D, Pandey AK, Gonzaga-Jauregui C, Khalid S, Ye B, Banerjee N, Li AH, O'Dushlaine C, Marcketta A, Staples J, Schurmann C, Hawes A, Maxwell E, Barnard L, Lopez A, Penn J, Habegger L, Blumenfeld AL, Bai X, O'Keeffe S, Yadav A, Praveen K, Jones M, Salerno WJ, Chung WK, Surakka I, Willer CJ, Hveem K, Leader JB, Carey DJ, Ledbetter DH, Cardon L, Yancopoulos GD, Economides A, Coppola G, Shuldiner AR, Balasubramanian S, Cantor M, Nelson MR, Whittaker J, Reid JG, Marchini J, Overton JD, Scott RA, Abecasis GR, Yerges-Armstrong L, Baras A. Exome sequencing and characterization of 49,960 individuals in the UK Biobank. Nature 2020; 586:749-756. [PMID: 33087929 PMCID: PMC7759458 DOI: 10.1038/s41586-020-2853-0] [Citation(s) in RCA: 331] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
Abstract
The UK Biobank is a prospective study of 502,543 individuals, combining extensive phenotypic and genotypic data with streamlined access for researchers around the world1. Here we describe the release of exome-sequence data for the first 49,960 study participants, revealing approximately 4 million coding variants (of which around 98.6% have a frequency of less than 1%). The data include 198,269 autosomal predicted loss-of-function (LOF) variants, a more than 14-fold increase compared to the imputed sequence. Nearly all genes (more than 97%) had at least one carrier with a LOF variant, and most genes (more than 69%) had at least ten carriers with a LOF variant. We illustrate the power of characterizing LOF variants in this population through association analyses across 1,730 phenotypes. In addition to replicating established associations, we found novel LOF variants with large effects on disease traits, including PIEZO1 on varicose veins, COL6A1 on corneal resistance, MEPE on bone density, and IQGAP2 and GMPR on blood cell traits. We further demonstrate the value of exome sequencing by surveying the prevalence of pathogenic variants of clinical importance, and show that 2% of this population has a medically actionable variant. Furthermore, we characterize the penetrance of cancer in carriers of pathogenic BRCA1 and BRCA2 variants. Exome sequences from the first 49,960 participants highlight the promise of genome sequencing in large population-based studies and are now accessible to the scientific community.
Collapse
Affiliation(s)
| | | | | | - Joshua D Hoffman
- GlaxoSmithKline, Collegeville, PA, USA
- Foresite Labs, Cambridge, MA, USA
| | - Daren Liu
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | - Bin Ye
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | | | | | - Claudia Schurmann
- Regeneron Genetics Center, Tarrytown, NY, USA
- Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - John Penn
- Regeneron Genetics Center, Tarrytown, NY, USA
- DNANexus, Mountain View, CA, USA
| | | | | | | | | | | | | | | | | | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | - Kristian Hveem
- Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | | | | | | | | | | | | | | | | | - Matthew R Nelson
- GlaxoSmithKline, Collegeville, PA, USA
- Deerfield, New York, NY, USA
| | | | | | | | | | | | | | | | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, USA.
| |
Collapse
|
20
|
Ver Donck F, Downes K, Freson K. Strengths and limitations of high-throughput sequencing for the diagnosis of inherited bleeding and platelet disorders. J Thromb Haemost 2020; 18:1839-1845. [PMID: 32521110 DOI: 10.1111/jth.14945] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/27/2020] [Accepted: 05/31/2020] [Indexed: 12/23/2022]
Abstract
Inherited bleeding and platelet disorders (BPD) are highly heterogeneous and their diagnosis involves a combination of clinical investigations, laboratory tests, and genetic screening. This review will outline some of the challenges that geneticists and experts in clinical hemostasis face when implementing high-throughput sequencing (HTS) for patient care. We will provide an overview of the strengths and limitations of the different HTS techniques that can be used to diagnose BPD. An HTS test is cost-efficient and expected to increase the diagnostic rate with a possibility to detect unexpected diagnoses and decrease the turnaround time to diagnose patients. On the other hand, technical shortcomings, variant interpretation difficulties, and ethical issues related to HTS for BPD will also be documented. Delivering a genetic diagnosis to patients is highly desirable to improve clinical management and allow family counseling, but making incorrect assumptions about variants and providing insufficient information to patients before initiating the test could be harmful. Data-sharing and improved HTS guidelines are essential to limit these major drawbacks of HTS.
Collapse
Affiliation(s)
- Fabienne Ver Donck
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Kate Downes
- East Midlands and East of England Genomics Laboratory Hub, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Turro E, Astle WJ, Megy K, Gräf S, Greene D, Shamardina O, Allen HL, Sanchis-Juan A, Frontini M, Thys C, Stephens J, Mapeta R, Burren OS, Downes K, Haimel M, Tuna S, Deevi SVV, Aitman TJ, Bennett DL, Calleja P, Carss K, Caulfield MJ, Chinnery PF, Dixon PH, Gale DP, James R, Koziell A, Laffan MA, Levine AP, Maher ER, Markus HS, Morales J, Morrell NW, Mumford AD, Ormondroyd E, Rankin S, Rendon A, Richardson S, Roberts I, Roy NBA, Saleem MA, Smith KGC, Stark H, Tan RYY, Themistocleous AC, Thrasher AJ, Watkins H, Webster AR, Wilkins MR, Williamson C, Whitworth J, Humphray S, Bentley DR, Kingston N, Walker N, Bradley JR, Ashford S, Penkett CJ, Freson K, Stirrups KE, Raymond FL, Ouwehand WH. Whole-genome sequencing of patients with rare diseases in a national health system. Nature 2020; 583:96-102. [PMID: 32581362 PMCID: PMC7610553 DOI: 10.1038/s41586-020-2434-2] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 05/05/2020] [Indexed: 02/02/2023]
Abstract
Most patients with rare diseases do not receive a molecular diagnosis and the aetiological variants and causative genes for more than half such disorders remain to be discovered1. Here we used whole-genome sequencing (WGS) in a national health system to streamline diagnosis and to discover unknown aetiological variants in the coding and non-coding regions of the genome. We generated WGS data for 13,037 participants, of whom 9,802 had a rare disease, and provided a genetic diagnosis to 1,138 of the 7,065 extensively phenotyped participants. We identified 95 Mendelian associations between genes and rare diseases, of which 11 have been discovered since 2015 and at least 79 are confirmed to be aetiological. By generating WGS data of UK Biobank participants2, we found that rare alleles can explain the presence of some individuals in the tails of a quantitative trait for red blood cells. Finally, we identified four novel non-coding variants that cause disease through the disruption of transcription of ARPC1B, GATA1, LRBA and MPL. Our study demonstrates a synergy by using WGS for diagnosis and aetiological discovery in routine healthcare.
Collapse
Affiliation(s)
- Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK.
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK.
| | - William J Astle
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Karyn Megy
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Stefan Gräf
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Daniel Greene
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Olga Shamardina
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Hana Lango Allen
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Alba Sanchis-Juan
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- British Heart Foundation Cambridge Centre of Excellence, University of Cambridge, Cambridge, UK
| | - Chantal Thys
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Jonathan Stephens
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Rutendo Mapeta
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Oliver S Burren
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthias Haimel
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Salih Tuna
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Sri V V Deevi
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Timothy J Aitman
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London, UK
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - David L Bennett
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
| | - Paul Calleja
- High Performance Computing Service, University of Cambridge, Cambridge, UK
| | - Keren Carss
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Mark J Caulfield
- Genomics England Ltd, London, UK
- William Harvey Research Institute, NIHR Biomedical Research Centre at Barts, Queen Mary University of London, London, UK
| | - Patrick F Chinnery
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, Cambridge Biomedical Campus, Cambridge, UK
| | - Peter H Dixon
- Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
- Rare Renal Disease Registry, UK Renal Registry, Bristol, UK
| | - Roger James
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Ania Koziell
- King's College London, London, UK
- Department of Paediatric Nephrology, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Michael A Laffan
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
- Centre for Haematology, Imperial College London, London, UK
| | - Adam P Levine
- Department of Renal Medicine, University College London, London, UK
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Bristol, UK
| | - Joannella Morales
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Nicholas W Morrell
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrew D Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
- University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Elizabeth Ormondroyd
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Department of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stuart Rankin
- High Performance Computing Service, University of Cambridge, Cambridge, UK
| | - Augusto Rendon
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Genomics England Ltd, London, UK
| | - Sylvia Richardson
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Irene Roberts
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Noemi B A Roy
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Moin A Saleem
- Bristol Renal and Children's Renal Unit, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Royal Hospital for Children, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Kenneth G C Smith
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Hannah Stark
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Rhea Y Y Tan
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Bristol, UK
| | - Andreas C Themistocleous
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | - Hugh Watkins
- Department of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Andrew R Webster
- Moorfields Eye Hospital NHS Trust, London, UK
- UCL Institute of Opthalmology, University College London, London, UK
| | | | - Catherine Williamson
- Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - James Whitworth
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | | | | | - Nathalie Kingston
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Neil Walker
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - John R Bradley
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Renal Medicine, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sofie Ashford
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Christopher J Penkett
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Kathleen E Stirrups
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - F Lucy Raymond
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK.
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
- British Heart Foundation Cambridge Centre of Excellence, University of Cambridge, Cambridge, UK.
- Wellcome Sanger Institute, Cambridge, UK.
| |
Collapse
|
22
|
Nurden AT, Nurden P. Inherited thrombocytopenias: history, advances and perspectives. Haematologica 2020; 105:2004-2019. [PMID: 32527953 PMCID: PMC7395261 DOI: 10.3324/haematol.2019.233197] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022] Open
Abstract
Over the last 100 years the role of platelets in hemostatic events and their production by megakaryocytes have gradually been defined. Progressively, thrombocytopenia was recognized as a cause of bleeding, first through an acquired immune disorder; then, since 1948, when Bernard-Soulier syndrome was first described, inherited thrombocytopenia became a fascinating example of Mendelian disease. The platelet count is often severely decreased and platelet size variable; associated platelet function defects frequently aggravate bleeding. Macrothrombocytopenia with variable proportions of enlarged platelets is common. The number of circulating platelets will depend on platelet production, consumption and lifespan. The bulk of macrothrombocytopenias arise from defects in megakaryopoiesis with causal variants in transcription factor genes giving rise to altered stem cell differentiation and changes in early megakaryocyte development and maturation. Genes encoding surface receptors, cytoskeletal and signaling proteins also feature prominently and Sanger sequencing associated with careful phenotyping has allowed their early classification. It quickly became apparent that many inherited thrombocytopenias are syndromic while others are linked to an increased risk of hematologic malignancies. In the last decade, the application of next-generation sequencing, including whole exome sequencing, and the use of gene platforms for rapid testing have greatly accelerated the discovery of causal genes and extended the list of variants in more common disorders. Genes linked to an increased platelet turnover and apoptosis have also been identified. The current challenges are now to use next-generation sequencing in first-step screening and to define bleeding risk and treatment better.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut Hospitalo-Universitaire LIRYC, Pessac, France
| | | |
Collapse
|
23
|
Fouassier M, Babuty A, Debord C, Béné MC. Platelet immunophenotyping in health and inherited bleeding disorders, a review and practical hints. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:464-475. [PMID: 32516490 DOI: 10.1002/cyto.b.21892] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/16/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022]
Abstract
Inherited platelet function disorders are rare hemorrhagic diseases. The gold standard for their exploration is optical aggregometry; however, investigations by flow cytometry (FCM) are being increasingly used. In this review, the physiology of platelets is first recalled, setting the stage for the compartments of platelets that can be apprehended by specific and appropriate labeling. As this requires some pre-analytical precautions and specific analytical settings, a second part focuses on these characteristic aspects, based on literature and on the authors' experience in the field, for qualitative or quantitative explorations. Membrane labeling with antibodies to CD42a or CD41, respectively, useful to assess the genetic-related defects of Glanzmann thrombocytopenia and Bernard Soulier syndrome are then described. Platelet degranulation disorders are detailed in the next section, as they can be explored, upon platelet activation, by measuring the expression of surface P-Selectin (CD62P) or CD63. Mepacrin uptake and release after activation is another test allowing to explore the function of dense granules. Finally, the flip-flop anomaly related to Scott syndrome is depicted. Tables summarizing possible FCM assays, and characteristic histograms are provided as reference for flow laboratories interested in developing platelet exploration.
Collapse
Affiliation(s)
- Marc Fouassier
- Hematology Biology Department, Nantes University Hospital and CRCINA, Nantes, France
| | - Antoine Babuty
- Hematology Biology Department, Nantes University Hospital and CRCINA, Nantes, France
| | - Camille Debord
- Hematology Biology Department, Nantes University Hospital and CRCINA, Nantes, France
| | - Marie C Béné
- Hematology Biology Department, Nantes University Hospital and CRCINA, Nantes, France
| |
Collapse
|
24
|
Diagnostic high-throughput sequencing of 2396 patients with bleeding, thrombotic, and platelet disorders. Blood 2020; 134:2082-2091. [PMID: 31064749 DOI: 10.1182/blood.2018891192] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/22/2019] [Indexed: 12/17/2022] Open
Abstract
A targeted high-throughput sequencing (HTS) panel test for clinical diagnostics requires careful consideration of the inclusion of appropriate diagnostic-grade genes, the ability to detect multiple types of genomic variation with high levels of analytic sensitivity and reproducibility, and variant interpretation by a multidisciplinary team (MDT) in the context of the clinical phenotype. We have sequenced 2396 index patients using the ThromboGenomics HTS panel test of diagnostic-grade genes known to harbor variants associated with rare bleeding, thrombotic, or platelet disorders (BTPDs). The molecular diagnostic rate was determined by the clinical phenotype, with an overall rate of 49.2% for all thrombotic, coagulation, platelet count, and function disorder patients and a rate of 3.2% for patients with unexplained bleeding disorders characterized by normal hemostasis test results. The MDT classified 745 unique variants, including copy number variants (CNVs) and intronic variants, as pathogenic, likely pathogenic, or variants of uncertain significance. Half of these variants (50.9%) are novel and 41 unique variants were identified in 7 genes recently found to be implicated in BTPDs. Inspection of canonical hemostasis pathways identified 29 patients with evidence of oligogenic inheritance. A molecular diagnosis has been reported for 894 index patients providing evidence that introducing an HTS genetic test is a valuable addition to laboratory diagnostics in patients with a high likelihood of having an inherited BTPD.
Collapse
|
25
|
De Kesel PM, Vantilborgh A, Dierick J, Luyckx A, Debussche S, Freson K, Devreese KMJ. Autosomal dominant macrothrombocytopenia caused by a rare GPIBB variant: The importance of DNA sequencing. Int J Lab Hematol 2019; 42:e98-e100. [PMID: 31793234 DOI: 10.1111/ijlh.13136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 11/27/2022]
Affiliation(s)
- Pieter M De Kesel
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Anna Vantilborgh
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Jan Dierick
- Department of Laboratory Medicine, AZ Maria Middelares, Ghent, Belgium
| | - Ariane Luyckx
- Department of Laboratory Medicine, AZ Maria Middelares, Ghent, Belgium
| | - Sarah Debussche
- Department of Hematology, AZ Maria Middelares, Ghent, Belgium
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | | |
Collapse
|
26
|
Gentilini F, Turba ME, Giancola F, Chiocchetti R, Bernardini C, Dajbychova M, Jagannathan V, Drögemüller M, Drögemüller C. A large deletion in the GP9 gene in Cocker Spaniel dogs with Bernard-Soulier syndrome. PLoS One 2019; 14:e0220625. [PMID: 31484196 PMCID: PMC6726462 DOI: 10.1371/journal.pone.0220625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/20/2019] [Indexed: 12/16/2022] Open
Abstract
Inherited bleeding disorders including abnormalities of platelet number and function rarely occur in a variety of dog breeds, but are probably underdiagnosed. Genetically characterized canine forms of platelet disorders provide valuable large animal models for understanding similar platelet disorders in people. Breed-specific disease associated genetic variants in only eight different genes are known to cause intrinsic platelet disorders in dogs. However, the causative genetic variant in many dog breeds has until now remained unknown. Four cases of a mild to severe bleeding disorder in Cocker Spaniel dogs are herein presented. The affected dogs showed a platelet adhesion defect characterized by macrothrombocytopenia with variable platelet counts resembling human Bernard-Soulier syndrome (BSS). Furthermore, the lack of functional GPIb-IX-V was demonstrated by immunocytochemistry. Whole genome sequencing of one affected dog and visual inspection of the candidate genes identified a deletion in the glycoprotein IX platelet (GP9) gene. The GP9 gene encodes a subunit of a platelet surface membrane glycoprotein complex; this functions as a receptor for von Willebrand factor, which initiates the maintenance of hemostasis after injury. Variants in human GP9 are associated with Bernard-Soulier syndrome, type C. The deletion spanned 2460 bp, and included a significant part of the single coding exon of the canine GP9 gene on dog chromosome 20. The variant results in a frameshift and premature stop codon which is predicted to truncate almost two-thirds of the encoded protein. PCR-based genotyping confirmed recessive inheritance. The homozygous variant genotype seen in affected dogs did not occur in 98 control Cocker Spaniels. Thus, it was concluded that the structural variant identified in the GP9 gene was most likely causative for the BSS-phenotype in the dogs examined. These findings provide the first large animal GP9 model for this group of inherited platelet disorders and greatly facilitate the diagnosis and identification of affected and/or normal carriers in Cocker Spaniels.
Collapse
Affiliation(s)
- Fabio Gentilini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
- * E-mail:
| | | | - Fiorella Giancola
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Roberto Chiocchetti
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | | | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Cord Drögemüller
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
27
|
Li X, Wang S, Wu J, Wang H, Wang J, Dong X, Zhang N. A Case of Bernard-Soulier Syndrome due to a Novel Homozygous Missense Mutation in an Exon of the GP1BA Gene. Acta Haematol 2019; 143:60-64. [PMID: 31302646 DOI: 10.1159/000500797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 05/07/2019] [Indexed: 11/19/2022]
Abstract
Bernard-Soulier syndrome (BSS) is an extremely rare autosomal recessive bleeding disorder clinically characterized by macrothrombocytopenia and a mucocutaneous bleeding tendency. A 1-year-old Chinese patient who was born to consanguineous parents was diagnosed with early onset of BSS. Gene sequencing and bioinformatics analysis were conducted. We identified a novel homozygous missense mutation (c.790T>C) in the GP1BAgene that causes an amino acid residue substitution of a cysteine with an arginine that might have a deleterious effect on the protein function as predicted by bioinformatics analysis. If a patient has clinical manifestations that include recurrent mucocutaneous bleeding, a mean platelet volume and platelet-large cell ratio above normal levels, and giant platelets on a peripheral smear and has consanguineous parents, a diagnosis of BSS can be suspected. In these situations, gene sequencing for mutations in the GPIb-IX-V complex is necessary.
Collapse
Affiliation(s)
- Xingchuan Li
- Department of Pediatrics, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Song Wang
- Department of Radiotherapy, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jiusi Wu
- Department of Pediatrics, Dunhuang City Hospital, Dunhuang, China
| | - Haidong Wang
- Department of Pediatrics, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jing Wang
- Department of Radiotherapy, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiangyu Dong
- Department of Pediatrics, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Ni Zhang
- Department of Pediatrics, The Second Hospital of Lanzhou University, Lanzhou, China,
| |
Collapse
|
28
|
Almazni I, Stapley R, Morgan NV. Inherited Thrombocytopenia: Update on Genes and Genetic Variants Which may be Associated With Bleeding. Front Cardiovasc Med 2019; 6:80. [PMID: 31275945 PMCID: PMC6593073 DOI: 10.3389/fcvm.2019.00080] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/31/2019] [Indexed: 01/10/2023] Open
Abstract
Inherited thrombocytopenia (IT) is comprised of a group of hereditary disorders characterized by a reduced platelet count as the main feature, and often with abnormal platelet function, which can subsequently lead to impaired haemostasis. Inherited thrombocytopenia results from genetic mutations in genes implicated in megakaryocyte differentiation and/or platelet formation and clearance. The identification of the underlying causative gene of IT is challenging given the high degree of heterogeneity, but important due to the presence of various clinical presentations and prognosis, where some defects can lead to hematological malignancies. Traditional platelet function tests, clinical manifestations, and hematological parameters allow for an initial diagnosis. However, employing Next-Generation Sequencing (NGS), such as Whole Genome and Whole Exome Sequencing (WES) can be an efficient method for discovering causal genetic variants in both known and novel genes not previously implicated in IT. To date, 40 genes and their mutations have been implicated to cause many different forms of inherited thrombocytopenia. Nevertheless, despite this advancement in the diagnosis of IT, the molecular mechanism underlying IT in some patients remains unexplained. In this review, we will discuss the genetics of thrombocytopenia summarizing the recent advancement in investigation and diagnosis of IT using phenotypic approaches, high-throughput sequencing, targeted gene panels, and bioinformatics tools.
Collapse
Affiliation(s)
- Ibrahim Almazni
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rachel Stapley
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
29
|
Khoriaty R, Ozel AB, Ramdas S, Ross C, Desch K, Shavit JA, Everett L, Siemieniak D, Li JZ, Ginsburg D. Genome-wide linkage analysis and whole-exome sequencing identifies an ITGA2B mutation in a family with thrombocytopenia. Br J Haematol 2019; 186:574-579. [PMID: 31119735 DOI: 10.1111/bjh.15961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/19/2019] [Indexed: 11/27/2022]
Abstract
Hereditary thrombocytopenias can be subclassified based on mode of inheritance and platelet size. Here we report a family with autosomal dominant (AD) thrombocytopenia with normal platelet size. Linkage analysis and whole exome sequencing identified the R1026W substitution in ITGA2B as the causative defect. The same mutation has been previously reported in 7 Japanese families/patients with AD thrombocytopenia, but all of these patients had macrothrombocytopenia. This is the first report of a family with AD thrombocytopenia with normal platelet size resulting from mutation in ITGA2B. ITGA2B mutations should therefore be included in the differential diagnosis of this latter disorder.
Collapse
Affiliation(s)
- Rami Khoriaty
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA.,University of Michigan Rogel Cancer Center, Univeristy of Michigan, Ann Arbor, MI, USA
| | - Ayse B Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Shweta Ramdas
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Charles Ross
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Karl Desch
- Department of Pediatrics and Communicable Disease, University of Michigan, Ann Arbor, MI, USA
| | - Jordan A Shavit
- Department of Pediatrics and Communicable Disease, University of Michigan, Ann Arbor, MI, USA
| | - Lesley Everett
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - David Siemieniak
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - David Ginsburg
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.,Department of Pediatrics and Communicable Disease, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Lambert MP. Inherited Platelet Disorders: A Modern Approach to Evaluation and Treatment. Hematol Oncol Clin North Am 2019; 33:471-487. [PMID: 31030814 DOI: 10.1016/j.hoc.2019.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The inherited platelet disorders are a heterogeneous group of disorders that can be pleotropic in their clinical presentations. They may present with variable platelet counts and bleeding, making their diagnosis difficult. New diagnostic tools range from flow cytometric platelet function assessments to next-generation sequencing. Several platelet disorders may now be treated with gene therapy or bone marrow transplant. Improved understanding of the molecular and biologic mechanisms of the inherited platelet disorders may lead to novel targeted therapies.
Collapse
Affiliation(s)
- Michele P Lambert
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Special Coagulation Laboratory, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Frontier Program in Immune Dysregulation, Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Zwifelhofer NMJ, Bercovitz RS, Weik LA, Moroi A, LaRose S, Newman PJ, Newman DK. Hemizygosity for the gene encoding glycoprotein Ibβ is not responsible for macrothrombocytopenia and bleeding in patients with 22q11 deletion syndrome. J Thromb Haemost 2019; 17:295-305. [PMID: 30549403 PMCID: PMC6410711 DOI: 10.1111/jth.14357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Indexed: 12/17/2022]
Abstract
Essentials How thrombocytopenia relates to bleeding in 22q11 deletion syndrome (22q11DS) is not clear. Bleeding severity, platelet count and volume, and GPIBB were examined in patients with 22q11DS. Macrothrombocytopenia and bleeding typified imperfectly overlapping subsets of 22q11DS patients. GPIBB hemizygosity does not cause macrothrombocytopenia or bleeding in patients with 22q11DS. SUMMARY: Background and objectives Macrothrombocytopenia and bleeding are frequently associated with 22q11 deletion syndrome (22q11DS). GPIBB, which encodes the glycoprotein (GP) Ibβ subunit of GPIb-IX-V, is commonly deleted in patients with 22q11DS. Absence of functional GPIb-IX-V causes Bernard-Soulier syndrome, which is a severe bleeding disorder characterized by macrothrombocytopenia. Patients with 22q11DS are often obligate hemizygotes for GPIBB, and those with only a pathogenically disrupted copy of GPIBB present with Bernard-Soulier syndrome. The objective of this study was to determine how GPIBB hemizygosity and sequence variation relate to macrothrombocytopenia and bleeding in patients with 22q11DS who do not have Bernard-Soulier syndrome. Patients/methods We thoroughly characterized bleeding severity, mean platelet volume, platelet count and GPIBB copy number and sequence in patients with 22q11DS. Results and conclusions Macrothrombocytopenia and mild bleeding were observed in incompletely overlapping subsets of patients, and GPIBB copy number and sequence variation did not correlate with either macrothrombocytopenia or bleeding in patients with 22q11DS. These findings indicate that GPIBB hemizygosity does not result in either macrothrombocytopenia or bleeding in these patients. Alternative genetic causes of macrothrombocytopenia, potential causes of acquired thrombocytopenia and bleeding and ways in which platelet size, platelet count and GPIBB sequence information can be used to aid in the diagnosis and management of patients with 22q11DS are discussed.
Collapse
Affiliation(s)
- N M J Zwifelhofer
- Blood Research Institute, BloodCenter of Wisconsin - part of Versiti, Milwaukee, WI, USA
| | - R S Bercovitz
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - L A Weik
- Children's Hospital of Wisconsin, Milwaukee, WI, USA
| | - A Moroi
- Blood Research Institute, BloodCenter of Wisconsin - part of Versiti, Milwaukee, WI, USA
| | - S LaRose
- Blood Research Institute, BloodCenter of Wisconsin - part of Versiti, Milwaukee, WI, USA
| | - P J Newman
- Blood Research Institute, BloodCenter of Wisconsin - part of Versiti, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - D K Newman
- Blood Research Institute, BloodCenter of Wisconsin - part of Versiti, Milwaukee, WI, USA
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
32
|
Babuty A, Boisseau P, Eveillard M, Ternisien C, Debord C, Sigaud M, Giraud M, Gillet B, Trossaert M, Béné M, Fouassier M. A new case of heterozygous variant of the
GP
1
BB
gene responsible for macrothrombocytopenia. Br J Haematol 2019; 186:157-159. [DOI: 10.1111/bjh.15739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Antoine Babuty
- Centre de Traitement de l'Hémophilie CHU de Nantes – Hôtel DieuNantesCedex 1France
| | - Pierre Boisseau
- Service de Génétique Médicale CHU de Nantes – Hôtel DieuNantesCedex 1France
| | - Marion Eveillard
- Service d'Hématologie Biologique CHU de Nantes – Hôtel Dieu Nantes Cedex 1 France
| | - Catherine Ternisien
- Centre de Traitement de l'Hémophilie CHU de Nantes – Hôtel DieuNantesCedex 1France
| | - Camille Debord
- Service d'Hématologie Biologique CHU de Nantes – Hôtel Dieu Nantes Cedex 1 France
| | - Marianne Sigaud
- Centre de Traitement de l'Hémophilie CHU de Nantes – Hôtel DieuNantesCedex 1France
| | - Mathilde Giraud
- Service de Génétique Médicale CHU de Nantes – Hôtel DieuNantesCedex 1France
| | - Benjamin Gillet
- Centre de Traitement de l'Hémophilie CHU de Nantes – Hôtel DieuNantesCedex 1France
| | - Marc Trossaert
- Centre de Traitement de l'Hémophilie CHU de Nantes – Hôtel DieuNantesCedex 1France
| | - Marie‐Christine Béné
- Service d'Hématologie Biologique CHU de Nantes – Hôtel Dieu Nantes Cedex 1 France
| | - Marc Fouassier
- Centre de Traitement de l'Hémophilie CHU de Nantes – Hôtel DieuNantesCedex 1France
| |
Collapse
|
33
|
The Glycoprotein Ib-IX-V Complex. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
34
|
Ferrari S, Lombardi AM, Cortella I, Businaro MA, Bertomoro A, Di Pasquale I, Fabris F. New heterozygous variant in GP1BB gene is responsible for an inherited form of macrothrombocytopenia. Br J Haematol 2018. [PMID: 29527674 DOI: 10.1111/bjh.15176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Silvia Ferrari
- Department of Medicine, University of Padua Medical School, Padua, Italy
| | - Anna M Lombardi
- Department of Medicine, University of Padua Medical School, Padua, Italy
| | - Irene Cortella
- Department of Medicine, University of Padua Medical School, Padua, Italy
| | - Maria A Businaro
- Department of Medicine, University of Padua Medical School, Padua, Italy
| | | | - Irene Di Pasquale
- Department of Medicine, University of Padua Medical School, Padua, Italy
| | - Fabrizio Fabris
- Department of Medicine, University of Padua Medical School, Padua, Italy
| |
Collapse
|
35
|
|
36
|
Bastida JM, Lozano ML, Benito R, Janusz K, Palma-Barqueros V, Del Rey M, Hernández-Sánchez JM, Riesco S, Bermejo N, González-García H, Rodriguez-Alén A, Aguilar C, Sevivas T, López-Fernández MF, Marneth AE, van der Reijden BA, Morgan NV, Watson SP, Vicente V, Hernández-Rivas JM, Rivera J, González-Porras JR. Introducing high-throughput sequencing into mainstream genetic diagnosis practice in inherited platelet disorders. Haematologica 2017; 103:148-162. [PMID: 28983057 PMCID: PMC5777202 DOI: 10.3324/haematol.2017.171132] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/29/2017] [Indexed: 12/30/2022] Open
Abstract
Inherited platelet disorders are a heterogeneous group of rare diseases, caused by inherited defects in platelet production and/or function. Their genetic diagnosis would benefit clinical care, prognosis and preventative treatments. Until recently, this diagnosis has usually been performed via Sanger sequencing of a limited number of candidate genes. High-throughput sequencing is revolutionizing the genetic diagnosis of diseases, including bleeding disorders. We have designed a novel high-throughput sequencing platform to investigate the unknown molecular pathology in a cohort of 82 patients with inherited platelet disorders. Thirty-four (41.5%) patients presented with a phenotype strongly indicative of a particular type of platelet disorder. The other patients had clinical bleeding indicative of platelet dysfunction, but with no identifiable features. The high-throughput sequencing test enabled a molecular diagnosis in 70% of these patients. This sensitivity increased to 90% among patients suspected of having a defined platelet disorder. We found 57 different candidate variants in 28 genes, of which 70% had not previously been described. Following consensus guidelines, we qualified 68.4% and 26.3% of the candidate variants as being pathogenic and likely pathogenic, respectively. In addition to establishing definitive diagnoses of well-known inherited platelet disorders, high-throughput sequencing also identified rarer disorders such as sitosterolemia, filamin and actinin deficiencies, and G protein-coupled receptor defects. This included disease-causing variants in DIAPH1 (n=2) and RASGRP2 (n=3). Our study reinforces the feasibility of introducing high-throughput sequencing technology into the mainstream laboratory for the genetic diagnostic practice in inherited platelet disorders.
Collapse
Affiliation(s)
- José M Bastida
- Servicio de Hematología, Hospital Universitario de Salamanca-IBSAL-USAL, Spain .,On behalf of the Project "Functional and Molecular Characterization of Patients with Inherited Platelet Disorders" of the Hemorrhagic Diathesis Working Group of the Spanish Society of Thrombosis and Haemostasis
| | - María L Lozano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CB15/00055-CIBERER, Spain.,On behalf of the Project "Functional and Molecular Characterization of Patients with Inherited Platelet Disorders" of the Hemorrhagic Diathesis Working Group of the Spanish Society of Thrombosis and Haemostasis
| | - Rocío Benito
- IBSAL, IBMCC, CIC, Universidad de Salamanca-CSIC, Spain
| | - Kamila Janusz
- IBSAL, IBMCC, CIC, Universidad de Salamanca-CSIC, Spain
| | - Verónica Palma-Barqueros
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CB15/00055-CIBERER, Spain
| | | | | | - Susana Riesco
- Servicio de Pediatría, Hospital Universitario de Salamanca-IBSAL, Spain
| | - Nuria Bermejo
- Servicio de Hematología, Complejo Hospitalario San Pedro Alcántara, Cáceres, Spain
| | | | - Agustín Rodriguez-Alén
- Servicio de Hematología y Hemoterapia, Hospital Virgen de la Salud, Complejo Hospitalario de Toledo, Spain
| | - Carlos Aguilar
- Servicio de Hematología, Complejo Asistencial de Soria, Spain
| | - Teresa Sevivas
- Serviço de Imunohemoterapia, Sangue e Medicina Transfusional do Centro Hospitalar e Universitário de Coimbra, EPE, Portugal
| | | | - Anna E Marneth
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Bert A van der Reijden
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Neil V Morgan
- Birmingham Platelet Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Steve P Watson
- Birmingham Platelet Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Vicente Vicente
- On behalf of the Project "Functional and Molecular Characterization of Patients with Inherited Platelet Disorders" of the Hemorrhagic Diathesis Working Group of the Spanish Society of Thrombosis and Haemostasis
| | - Jesús M Hernández-Rivas
- Servicio de Hematología, Hospital Universitario de Salamanca-IBSAL-USAL, Spain.,IBSAL, IBMCC, CIC, Universidad de Salamanca-CSIC, Spain
| | - José Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CB15/00055-CIBERER, Spain.,On behalf of the Project "Functional and Molecular Characterization of Patients with Inherited Platelet Disorders" of the Hemorrhagic Diathesis Working Group of the Spanish Society of Thrombosis and Haemostasis
| | | |
Collapse
|
37
|
Greene D, Richardson S, Turro E, Turro E. A Fast Association Test for Identifying Pathogenic Variants Involved in Rare Diseases. Am J Hum Genet 2017; 101:104-114. [PMID: 28669401 PMCID: PMC5501869 DOI: 10.1016/j.ajhg.2017.05.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/22/2017] [Indexed: 11/26/2022] Open
Abstract
We present a rapid and powerful inference procedure for identifying loci associated with rare hereditary disorders using Bayesian model comparison. Under a baseline model, disease risk is fixed across all individuals in a study. Under an association model, disease risk depends on a latent bipartition of rare variants into pathogenic and non-pathogenic variants, the number of pathogenic alleles that each individual carries, and the mode of inheritance. A parameter indicating presence of an association and the parameters representing the pathogenicity of each variant and the mode of inheritance can be inferred in a Bayesian framework. Variant-specific prior information derived from allele frequency databases, consequence prediction algorithms, or genomic datasets can be integrated into the inference. Association models can be fitted to different subsets of variants in a locus and compared using a model selection procedure. This procedure can improve inference if only a particular class of variants confers disease risk and can suggest particular disease etiologies related to that class. We show that our method, called BeviMed, is more powerful and informative than existing rare variant association methods in the context of dominant and recessive disorders. The high computational efficiency of our algorithm makes it feasible to test for associations in the large non-coding fraction of the genome. We have applied BeviMed to whole-genome sequencing data from 6,586 individuals with diverse rare diseases. We show that it can identify multiple loci involved in rare diseases, while correctly inferring the modes of inheritance, the likely pathogenic variants, and the variant classes responsible.
Collapse
Affiliation(s)
| | | | | | - Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK; NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK; Medical Research Council Biostatistics Unit, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| |
Collapse
|
38
|
Freson K, Turro E. High-throughput sequencing approaches for diagnosing hereditary bleeding and platelet disorders. J Thromb Haemost 2017; 15:1262-1272. [PMID: 28671349 DOI: 10.1111/jth.13681] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hereditary bleeding and platelet disorders (BPDs) are characterized by marked genetic heterogeneity, far greater than previously appreciated. The list of genes involved in the regulation of megakaryopoiesis, platelet formation, platelet function and bleeding has been growing rapidly since the introduction of high-throughput sequencing (HTS) approaches in research. Thanks to the gradual adoption of HTS in diagnostic practice, these discoveries are improving the diagnostic yield for BPD patients, who may or may not present with bleeding problems and often have other clinical symptoms unrelated to the blood system. However, it was previously found that screening for all known etiologies gives a diagnostic yield of over 90% when the phenotype closely matches a known BPD but drops to 10% when the phenotype is indicative of a novel disorder. Thus, further research is needed to identify currently unknown etiologies for BPDs. Novel genes are likely to be found to be implicated in BPDs. New modes of inheritance, including digenic inheritance, are likely to play a role in some cases. Additionally, identifying and interpreting pathogenic variants outside exons is a looming challenge that can only be tackled with an improved understanding of the regulatory landscape of relevant cell types and with the transition from targeted sequencing to whole-genome sequencing in the clinic.
Collapse
Affiliation(s)
- K Freson
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - E Turro
- Department of Haematology and MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|