1
|
Ramanan R, Verstraete A, Van Laer C, Freson K. Implementation and clinical utility of multigene panels for bleeding, platelet, and thrombotic disorders. J Thromb Haemost 2025:S1538-7836(25)00277-6. [PMID: 40345666 DOI: 10.1016/j.jtha.2025.04.026] [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: 02/24/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
High-throughput sequencing, with its capacity to simultaneously sequence large volumes of genomic data, has evolved from a research-focused technology to a clinical tool. This review outlines key steps in the development of a clinical hemostasis and thrombosis genetics service leveraging a multigene panel. We discuss its value across inherited bleeding, platelet, and thrombotic disorders (BPTDs) in the context of published studies utilizing multigene panels for these conditions. Benefits of sequencing include establishing a diagnosis through the simultaneous assessment of multiple candidate genes, exclusion of genocopies, and predictions of phenotype to deliver targeted therapy. The presence of concomitant variants may modify phenotype; however, predictions on disease course from oligogenic modifiers are not yet used to guide patient care or counseling. Limitations in the widespread roll-out of multigene panels for clinical diagnosis of BPTDs exist. These challenges relate to detection of structural variants, variable diagnostic hit rates, and management of incidental findings. Variants of uncertain significance also frustrate diagnostic yield. Family segregation studies, in vitro characterization, and protein modeling aid interpretation of variant pathogenicity. Although multigene panels offer substantial opportunities to improve BPTD diagnostics, their implementation should be guided by appropriate expertise and in conjunction with clinical research to ensure safe and ethical care.
Collapse
Affiliation(s)
- Radha Ramanan
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia; Department of Human Molecular Pathology, Alfred Hospital, Melbourne, Victoria, Australia.
| | - Andreas Verstraete
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Christine Van Laer
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Kathleen Freson
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Chen G, Pei Y, Ye Q, Xie Z, Gyawali L, Liang X. NEDD4L-mediated RASGRP2 suppresses high-glucose and oxLDL-induced vascular endothelial cell dysfunctions by activating Rap1 and R-Ras. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119844. [PMID: 39260747 DOI: 10.1016/j.bbamcr.2024.119844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Ras guanyl-releasing protein 2 (RASGRP2) is an important regulator mediating endothelial cell function. However, whether RASGRP2 mediates diabetes mellitus (DM)-related atherosclerosis (AS) progression by regulating endothelial cell functions is unknown. METHODS Human cardiac microvascular endothelial cells (HCMECs) were treated with high-glucose (HG) and oxidized low-density lipoprotein (oxLDL). The expression of RASGRP2 and neural precursor cell expressed developmentally downregulated 4-like (NEDD4L) was examined by quantitative real-time PCR and western blot (WB). Cell viability, apoptosis, migration, angiogenesis were detected by CCK8 assay, flow cytometry, transwell assay and tube formation assay. ROS production and cell permeability were tested to assess cell function. Rap1 and R-Ras protein levels were examined using WB. The interaction between RASGRP2 and NEDD4L was confirmed by Co-IP assay and ubiquitination assay. Exosomes were isolated from adipose-derived MSC (ADMSC)-transfected RASGRP2 overexpression vector, and then co-cultured with HG + oxLDL-induced HCMECs. RESULTS RASGRP2 was lowly expressed in HG + oxLDL-induced HCMECs. RASGRP2 overexpression inhibited HG + oxLDL-induced HCMECs permeability, apoptosis and ROS production, while accelerated cell viability, migration and angiogenesis. NEDD4L could interact with RASGRP2 by ubiquitination, thus inhibiting RASGRP2 protein stability to degrade its expression. Functional experiments showed that NEDD4L knockdown suppressed HG + oxLDL-induced HCMECs dysfunction, while these effects were reversed by RASGRP2 downregulation. ADMSC-Exo overexpressed RASGRP2 could promote cell viability, migration and angiogenesis, while suppress permeability, apoptosis and ROS production in HG + oxLDL-induced HCMECs. CONCLUSION Our data showed that targeting NEDD4L/RASGRP2 axis or inducing RASGRP2-modified ADMSC-Exo might be the efficient strategy for alleviating DM-related AS.
Collapse
Affiliation(s)
- Guozhu Chen
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China.
| | - Yisong Pei
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China
| | - Qiaoling Ye
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China
| | - Zulong Xie
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China
| | | | - Xing Liang
- The Second Affiliated Hospital of Chongqing Medical University Cardiology Department, Chongqing, China.
| |
Collapse
|
3
|
Shi ZY, Lei QL, Duan SQ, Zhou Y, Cui TT, Lin YB, Yang CH, Song CY, Fang CL, Tian X, Zhang XW, Huang TL. A splice mutation in RASGRP2 gene in the patient with recurrent epistaxis and nasal vascular malformation. Platelets 2024; 35:2425664. [PMID: 39512176 DOI: 10.1080/09537104.2024.2425664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/08/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024]
Abstract
Platelet type bleeding disorder-18 (BDPLT18) caused by mutations of Ras guanyl releasing protein 2 (RASGRP2) is a relatively rare, new autosomal recessive disorder. Here, we reported a splice mutation in RASGRP2 gene in the patient with recurrent epistaxis and nasal vascular malformation. The patient, an 8-year-old girl, suffered from anemia due to frequently severe recurrent epistaxis, requiring regular blood transfusions every 2-3 months. Hematological investigations showed moderate anemia (Hb: 89 g/L), normal platelet count, morphology, and platelet glycoproteins. Arachidonic acid and adenosine diphosphate induced platelet aggregation was markedly reduced in the patient. A homozygous splice variant (C.74-1 G>C) in RASGRP2 gene, located within the exon 3, was detected by next-generation sequencing. Interestingly, we identified nasal vascular malformation by percutaneous super-selective angiography during the treatment of an intractable epistaxis. Our case further support that genetic testing should be performed for some unexplained bleeding diseases.
Collapse
Affiliation(s)
- Zhong-Yu Shi
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Qing-Ling Lei
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Shao-Qin Duan
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Yan Zhou
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Ting-Ting Cui
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Yun-Bi Lin
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Chun-Hui Yang
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Chun-Yan Song
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Chun-Lian Fang
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Xin Tian
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| | - Xian-Wen Zhang
- Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Ti-Long Huang
- Department of Hematology, Kunming Children's Hospital, Kunming, China
| |
Collapse
|
4
|
Frelinger AL. Flow Cytometry and Platelets. Clin Lab Med 2024; 44:511-526. [PMID: 39089755 DOI: 10.1016/j.cll.2024.04.011] [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] [Indexed: 08/04/2024]
Abstract
Clinical assessment of platelet activation by flow cytometry is useful in the characterization and diagnosis of platelet-specific disorders and as a measure of risk for thrombosis or bleeding. Platelets circulate in a resting, "unactivated" state, but when activated they undergo alterations in surface glycoprotein function and/or expression level, exposure of granule membrane proteins, and exposure of procoagulant phospholipids. Flow cytometry provides the means to detect these changes and, unlike other platelet tests, is appropriate for measuring platelet function in samples from patients with low platelet counts. The present review will focus on flow cytometric tests for platelet activation markers.
Collapse
Affiliation(s)
- Andrew L Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115-5737, USA.
| |
Collapse
|
5
|
Trégouët DA, Morange PE. Next-generation sequencing strategies in venous thromboembolism: in whom and for what purpose? J Thromb Haemost 2024; 22:1826-1834. [PMID: 38641321 DOI: 10.1016/j.jtha.2024.04.004] [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: 02/13/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/21/2024]
Abstract
This invited review follows the oral presentation "To Sequence or Not to Sequence, That Is Not the Question; But 'When, Who, Which and What For?' Is" given during the State of the Art session "Translational Genomics in Thrombosis: From OMICs to Clinics" of the International Society on Thrombosis and Haemostasis 2023 Congress. Emphasizing the power of next-generation sequencing technologies and the diverse strategies associated with DNA variant analysis, this review highlights the unresolved questions and challenges in their implementation both for the clinical diagnosis of venous thromboembolism and in translational research.
Collapse
Affiliation(s)
- David-Alexandre Trégouët
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale, Bordeaux Population Health Research Center, Unité Mixte de Recherche 1219, Bordeaux, France.
| | - Pierre-Emmanuel Morange
- Cardiovascular and Nutrition Research Center (Centre de Recherche en CardioVasculaire et Nutrition), Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l' Alimentation et l'Environnement, Aix-Marseille University, Marseille, France
| |
Collapse
|
6
|
O'Donoghue L, Smolenski A. Roles of G proteins and their GTPase-activating proteins in platelets. Biosci Rep 2024; 44:BSR20231420. [PMID: 38808367 PMCID: PMC11139668 DOI: 10.1042/bsr20231420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Platelets are small anucleate blood cells supporting vascular function. They circulate in a quiescent state monitoring the vasculature for injuries. Platelets adhere to injury sites and can be rapidly activated to secrete granules and to form platelet/platelet aggregates. These responses are controlled by signalling networks that include G proteins and their regulatory guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Recent proteomics studies have revealed the complete spectrum of G proteins, GEFs, and GAPs present in platelets. Some of these proteins are specific for platelets and very few have been characterised in detail. GEFs and GAPs play a major role in setting local levels of active GTP-bound G proteins in response to activating and inhibitory signals encountered by platelets. Thus, GEFs and GAPs are highly regulated themselves and appear to integrate G protein regulation with other cellular processes. This review focuses on GAPs of small G proteins of the Arf, Rab, Ras, and Rho families, as well as of heterotrimeric G proteins found in platelets.
Collapse
Affiliation(s)
- Lorna O'Donoghue
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| | - Albert Smolenski
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| |
Collapse
|
7
|
Rudran T, Antoniak S, Flick MJ, Ginsberg MH, Wolberg AS, Bergmeier W, Lee RH. Protease-activated receptors and glycoprotein VI cooperatively drive the platelet component in thromboelastography. J Thromb Haemost 2023; 21:2236-2247. [PMID: 37068592 PMCID: PMC10824270 DOI: 10.1016/j.jtha.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/27/2023] [Accepted: 04/09/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Thromboelastography (TEG) is used for real-time determination of hemostatic status in patients with acute risk of bleeding. Thrombin is thought to drive clotting in TEG through generation of polymerized fibrin and activation of platelets through protease-activated receptors (PARs). However, the specific role of platelet agonist receptors and signaling in TEG has not been reported. OBJECTIVES Here, we investigated the specific receptors and signaling pathways required for platelet function in TEG using genetic and pharmacologic inhibition of platelet proteins in mouse and human blood samples. METHODS Clotting parameters (R time, α-angle [α], and maximum amplitude [MA]), were determined in recalcified, kaolin-triggered citrated blood samples using a TEG 5000 analyzer. RESULTS We confirmed the requirement of platelets, platelet contraction, and αIIbβ3 integrin function for normal α and MA. Loss of the integrin adaptor Talin1 in megakaryocytes/platelets (Talin1mKO) also reduced α and MA, but only minimal defects were observed in samples from mice lacking Rap1 GTPase signaling. PAR4mKO samples showed impaired α but normal MA. However, impaired TEG traces similar to those in platelet-depleted samples were observed with samples from PAR4mKO mice depleted of glycoprotein VI on platelets or with addition of a Syk inhibitor. We reproduced these results in human blood with combined inhibition of PAR1, PAR4, and Syk. CONCLUSION Our results demonstrate that standard TEG is not sensitive to platelet signaling pathways critical for integrin inside-out activation and platelet hemostatic function. Furthermore, we provide the first evidence that PARs and glycoprotein VI play redundant roles in platelet-mediated clot contraction in TEG.
Collapse
Affiliation(s)
- Tanvi Rudran
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Silvio Antoniak
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew J Flick
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mark H Ginsberg
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Alisa S Wolberg
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Robert H Lee
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
8
|
Horitani S, Ueda Y, Kamioka Y, Kondo N, Ikeda Y, Naganuma M, Kinashi T. The critical role of Rap1-GAPs Rasa3 and Sipa1 in T cells for pulmonary transit and egress from the lymph nodes. Front Immunol 2023; 14:1234747. [PMID: 37545505 PMCID: PMC10399222 DOI: 10.3389/fimmu.2023.1234747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Rap1-GTPase activates integrins and plays an indispensable role in lymphocyte trafficking, but the importance of Rap1 inactivation in this process remains unknown. Here we identified the Rap1-inactivating proteins Rasa3 and Sipa1 as critical regulators of lymphocyte trafficking. The loss of Rasa3 and Sipa1 in T cells induced spontaneous Rap1 activation and adhesion. As a consequence, T cells deficient in Rasa3 and Sipa1 were trapped in the lung due to firm attachment to capillary beds, while administration of LFA1 antibodies or loss of talin1 or Rap1 rescued lung sequestration. Unexpectedly, mutant T cells exhibited normal extravasation into lymph nodes, fast interstitial migration, even greater chemotactic responses to chemokines and sphingosine-1-phosphate, and entrance into lymphatic sinuses but severely delayed exit: mutant T cells retained high motility in lymphatic sinuses and frequently returned to the lymph node parenchyma, resulting in defective egress. These results reveal the critical trafficking processes that require Rap1 inactivation.
Collapse
Affiliation(s)
- Shunsuke Horitani
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
- Division of Gastroenterology and Hepatology, the Third Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Yoshihiro Ueda
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yuji Kamioka
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Naoyuki Kondo
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yoshiki Ikeda
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Makoto Naganuma
- Division of Gastroenterology and Hepatology, the Third Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Tatsuo Kinashi
- The Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
9
|
Mitchell A, Frontini M, Islam S, Sivapalaratnam S, Krishnan A. Increased bleeding and thrombosis in myeloproliferative neoplasms mediated through altered expression of inherited platelet disorder genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541977. [PMID: 37292725 PMCID: PMC10245891 DOI: 10.1101/2023.05.23.541977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
An altered thrombo-hemorrhagic profile has long been observed in patients with myeloproliferative neoplasms (MPNs). We hypothesized that this observed clinical phenotype may result from altered expression of genes known to harbor genetic variants in bleeding, thrombotic, or platelet disorders. Here, we identify 32 genes from a clinically validated gene panel that were also significantly differentially expressed in platelets from MPN patients as opposed to healthy donors. This work begins to unravel previously unclear mechanisms underlying an important clinical reality in MPNs. Knowledge of altered platelet gene expression in MPN thrombosis/bleeding diathesis opens opportunities to advance clinical care by: (1) enabling risk stratification, in particular, for patients undergoing invasive procedures, and (2) facilitating tailoring of treatment strategies for those at highest risk, for example, in the form of antifibrinolytics, desmopressin or platelet transfusions (not current routine practice). Marker genes identified in this work may also enable prioritization of candidates in future MPN mechanistic as well as outcome studies.
Collapse
Affiliation(s)
- Alan Mitchell
- Department of Clinical Haematology, Barts Health NHS Trust, University of Exeter Medical School, Faculty of Health and Life Sciences, RILD Building, Barrack Road, Exeter, EX2 5DW
| | - Mattia Frontini
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, RILD Building, Barrack Road, Exeter, EX2 5DW
| | | | - Suthesh Sivapalaratnam
- Department of Clinical Haematology, Barts Health NHS Trust, University of Exeter Medical School, Faculty of Health and Life Sciences, RILD Building, Barrack Road, Exeter, EX2 5DW
- Blizard Institute, Queen Mary University London
| | - Anandi Krishnan
- Department of Pathology, Stanford University School of Medicine
| |
Collapse
|
10
|
Roos D, van Leeuwen K, Madkaikar M, Kambli PM, Gupta M, Mathews V, Rawat A, Kuhns DB, Holland SM, de Boer M, Kanegane H, Parvaneh N, Lorenz M, Schwarz K, Klein C, Sherkat R, Jafari M, Wolach B, den Dunnen JT, Kuijpers TW, Köker MY. Hematologically important mutations: Leukocyte adhesion deficiency (second update). Blood Cells Mol Dis 2023; 99:102726. [PMID: 36696755 DOI: 10.1016/j.bcmd.2023.102726] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Leukocyte adhesion deficiency (LAD) is an immunodeficiency caused by defects in the adhesion of leukocytes (especially neutrophils) to the blood vessel wall. As a result, patients with LAD suffer from severe bacterial infections and impaired wound healing, accompanied by neutrophilia. In LAD-I, characterized directly after birth by delayed separation of the umbilical cord, mutations are found in ITGB2, the gene that encodes the β subunit (CD18) of the β2 integrins. In the rare LAD-II disease, the fucosylation of selectin ligands is disturbed, caused by mutations in SLC35C1, the gene that encodes a GDP-fucose transporter of the Golgi system. LAD-II patients lack the H and Lewis Lea and Leb blood group antigens. Finally, in LAD-III, the conformational activation of the hematopoietically expressed β integrins is disturbed, leading to leukocyte and platelet dysfunction. This last syndrome is caused by mutations in FERMT3, encoding the kindlin-3 protein in all blood cells, involved in the regulation of β integrin conformation. This article contains an update of the mutations that we consider to be relevant for the various forms of LAD.
Collapse
Affiliation(s)
- Dirk Roos
- Sanquin Research, and Landsteiner Laboratory, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Karin van Leeuwen
- Sanquin Research, and Landsteiner Laboratory, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Manisha Madkaikar
- Pediatric Immunology and Leukocyte Biology Lab CMR, National Institute of Immunohaematology, K E M Hospital, Parel, Mumbai, India
| | - Priyanka M Kambli
- Pediatric Immunology and Leukocyte Biology Lab CMR, National Institute of Immunohaematology, K E M Hospital, Parel, Mumbai, India
| | - Maya Gupta
- Pediatric Immunology and Leukocyte Biology Lab CMR, National Institute of Immunohaematology, K E M Hospital, Parel, Mumbai, India
| | - Vikram Mathews
- Dept of Hematology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Amit Rawat
- Paediatric Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Chandigarh, India
| | - Douglas B Kuhns
- Neutrophil Monitoring Laboratory, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Martin de Boer
- Sanquin Research, and Landsteiner Laboratory, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Nima Parvaneh
- Infectious Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Myriam Lorenz
- Institute for Transfusion Medicine, University Ulm, Ulm, Germany
| | - Klaus Schwarz
- Institute for Transfusion Medicine, University Ulm, Ulm, Germany; Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Württemberg - Hessen, Ulm, Germany
| | - Christoph Klein
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Roya Sherkat
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahbube Jafari
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Baruch Wolach
- Pediatric Immunology Service, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Johan T den Dunnen
- Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Taco W Kuijpers
- Sanquin Research, and Landsteiner Laboratory, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Emma Children's Hospital, Amsterdam University Medical Centre, location AMC, Amsterdam, the Netherlands
| | - M Yavuz Köker
- Department of Immunology, Erciyes Medical School, University of Erciyes, Kayseri, Türkiye
| |
Collapse
|
11
|
Liu Y, Ouyang Y, Feng Z, Jiang Z, Ma J, Zhou X, Cai C, Han Y, Zeng S, Liu S, Shen H. RASGRP2 is a potential immune-related biomarker and regulates mitochondrial-dependent apoptosis in lung adenocarcinoma. Front Immunol 2023; 14:1100231. [PMID: 36817422 PMCID: PMC9936229 DOI: 10.3389/fimmu.2023.1100231] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Ras guanine nucleotide-releasing protein 2 (RASGRP2), one of the guanine nucleotide exchange factors (GEFs), has attracted much attention in recent years. However, the correlation between RASGRP2 and immune infiltration and malignant features in lung adenocarcinoma (LUAD) has rarely been mentioned. METHODS The Limma package and the LASSO regression model were performed to screen for differentially expressed genes. Data from the TCGA and 5 GEO databases were used to explore the expression level of RASGRP2 in LUAD patients. A weighted co-expression network and LinkFinder module were established to find the related genes of RASGRP2. The ESTIMATE algorithm was used to analyze the correlation between RASGRP2 and immune infiltration in LUAD. Tumor-infiltrating immune cells were sorted and sequenced at the single-cell level to analyze differences in RASGRP2. Real-time PCR and immunohistochemistry were performed in the real-world cohort to verify the expression of RASGRP2 and its correlation with immune-related genes. Clone formation and EdU assays were used to verify the proliferation ability. The proportion of apoptotic cells was analyzed by flow cytometry. Observation of mitochondrial membrane potential (MMP) changes by fluorescence microscopy. RESULTS Our results suggested that decreased RASGRP2 was associated with worse clinical parameters and prognosis in LUAD patients. And we constructed a FLI1-HSA-miR-1976-RASGRP2 transcriptional network to support the role of RASGRP2. Enrichment analysis revealed that RASGRP2 was involved in lymphocyte activation and leukocyte adhesion. RASGRP2 was found to be positively correlated with the infiltration of most immune cells, immunoregulators, and chemokines in a subsequent study. Meanwhile, the real-world cohort confirmed that the expression levels of PDCD1, CTLA4, CD40LG, CCL14, CXCR5, and CCR7 were higher in the high-RASGRP2 expression group. Cytological experiments proved that RASGRP2 inhibited cell proliferation in LUAD by regulating mitochondrial-dependent apoptosis. CONCLUSION RASGRP2 was a potential immune-related biomarker of LUAD. In addition, RASGRP2 was involved in the malignant progression of LUAD through the regulation of mitochondrial-dependent apoptosis.
Collapse
Affiliation(s)
- Yongting Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanhong Ouyang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Ziyang Feng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Jiang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiayao Ma
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shanshan Liu
- Department of Radiotherapy, Tianjin First Central Hospital, Tianjin, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Zieger B, Boeckelmann D. Inherited Platelet Disorders: A Short Introduction. Hamostaseologie 2023; 43:52-59. [PMID: 36807820 DOI: 10.1055/a-1987-3310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Platelets play an important role regarding coagulation by contributing to thrombus formation by platelet adhesion, aggregation, and α-/δ-granule secretion. Inherited platelet disorders (IPDs) are a very heterogeneous group of disorders that are phenotypically and biochemically diverse. Platelet dysfunction (thrombocytopathy) can be accompanied by a reduction in the number of thrombocytes (thrombocytopenia). The extent of the bleeding tendency can vary greatly. Symptoms comprise mucocutaneous bleeding (petechiae, gastrointestinal bleeding and/or menorrhagia, epistaxis) and increased hematoma tendency. Life-threatening bleeding can occur after trauma or surgery. In the last years, next-generation sequencing had a great impact on unrevealing the underlying genetic cause of individual IPDs. Because IPDs are so diverse, a comprehensive analysis of platelet function and genetic testing is indispensable.
Collapse
Affiliation(s)
- Barbara Zieger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Doris Boeckelmann
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Glanzmann Thrombasthenia in Pakistani Patients: Identification of 7 Novel Pathogenic Variants in the Fibrinogen Receptor αIIbβ3. Cells 2023; 12:cells12020213. [PMID: 36672149 PMCID: PMC9856889 DOI: 10.3390/cells12020213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/12/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Glanzmann thrombasthenia (GT) is a rare autosomal recessive inherited platelet disorder occurring frequently in populations with high incidence of consanguineous marriages. GT is characterized by quantitative and/or qualitative defect of the platelet αIIbβ3 (GPIIb/IIIa) receptor caused by pathogenic variants of the encoding genes: ITGA2B and ITGB3. Patients present with a moderate to severe bleeding tendency with normal platelet count. Platelets show reduced/absent aggregation for all agonists except ristocetin in light transmission aggregometry and reduced/absent αIIbβ3 expression in flow cytometry (FC). In this study, we investigated a cohort of 20 Pakistani patients and 2 families collected from the National Institute of Blood Disease, Karachi and Chughtai's Lab, Lahore. Platelet aggregation studies, FC (platelet CD41, CD61, CD42a, CD42b) and direct sequencing of the candidate genes were performed. All patients showed altered platelet aggregation, but normal agglutination after stimulation with ristocetin. Absent/reduced αIIbβ3 receptor expression was present in the platelets of 16 patients, in 4 patients expression was borderline/normal. Candidate gene sequencing identified pathogenic/likely pathogenic variants in 15 patients. Seven variants are novel. One patient with absent receptor expression remained without genetic finding. 13 (86.7%) of 15 patients stated consanguinity reflected by homozygosity finding in 14 (93.3%) patients.
Collapse
|
14
|
Influence of genetic polymorphisms in P2Y12 receptor signaling pathway on antiplatelet response to clopidogrel in coronary heart disease. BMC Cardiovasc Disord 2022; 22:575. [PMID: 36581799 PMCID: PMC9801627 DOI: 10.1186/s12872-022-02988-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 08/18/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUNDS Remarkable interindividual variability in clopidogrel response is observed, genetic polymorphisms in P2RY12 and its signal pathway is supposed to affect clopidogrel response in CHD patients. METHODS 539 CHD patients treated with clopidogrel were recruited. The platelet reaction index (PRI) indicated by VASP-P level were detected in 12-24 h after clopidogrel loading dose or within 5-7 days after initiation of maintain dose clopidogrel. A total of 13 SNPs in relevant genes were genotyped in sample A (239 CHD patients). The SNPs which have significant differences in PRI will be validated in another sample (sample B, 300 CHD patients). RESULTS CYP2C19*2 increased the risk of clopidogrel resistance significantly. When CYP2C19*2 and CYP2C19*3 were considered, CYP2C19 loss of function (LOF) alleles were associated with more obviously increased the risk of clopidogrel resistance; P2RY12 rs6809699C > A polymorphism was also associated with increased risk of clopidogrel resistance (AA vs CC: P = 0.0398). This difference still existed after stratification by CYP2C19 genotypes. It was also validated in sample B. The association was also still significant even in the case of stratification by CYP2C19 genotypes in all patients (sample A + B). CONCLUSION Our data suggest that P2RY12 rs6809699 is associated with clopidogrel resistance in CHD patients. Meanwhile, the rs6809699 AA genotype can increase on-treatment platelet activity independent of CYP2C19 LOF polymorphisms.
Collapse
|
15
|
Huang S, Wang J, Zhang L, Tian S, Wang Y, Shao X, Zhou S, Yu P. Ras guanine nucleotide-releasing protein-4 promotes renal inflammatory injury in type 2 diabetes mellitus. Metabolism 2022; 131:155177. [PMID: 35218794 DOI: 10.1016/j.metabol.2022.155177] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/31/2022] [Accepted: 02/18/2022] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Ras guanine nucleotide-releasing protein-4 (RasGRP4) is an activator of Ras protein, which plays significant roles in both the inflammatory response and immune activation. This study determined the role of RasGRP4 in diabetic kidney disease (DKD) progression. METHODS CRISPR/Cas9 technology was used to establish RasGRP4 knockout (KO) mice. Diabetes was induced by a high-fat diet combined with five consecutive daily intraperitoneal injections of streptozotocin (60 mg/kg) in C57BL/6J wild-type (WT) mice and RasGRP4 KO mice. Hematoxylin and eosin, periodic acid-Schiff, and Masson's trichrome staining were used to observe the histology of pathological injury. Immunohistochemical staining was used to analyze inflammatory cell infiltration. Quantitative PCR and Western blotting were used to detect the expression of inflammatory mediators and the activation of signaling pathways in renal tissues. In vitro cell co-culture experiments were performed to explore the interactions between peripheral blood mononuclear cells (PBMCs) and glomerular endothelial cells (GEnCs). RESULTS RasGRP4 KO mice developed less severe diabetic kidney injury compared to WT mice, exhibiting lower proteinuria, reduced CD3+ T lymphocyte and F4/80+ macrophage infiltration, less inflammatory mediator expression including interleukin 6, tumor necrosis alpha, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1, and lower expression levels of critical signal transduction molecules in the NLR family pyrin domain-containing 3 inflammasome and mitogen-activated protein kinase (MAPK)/nuclear factor kappa B (NF-κB) signaling pathways in the diabetic kidney. In vitro experiments showed that the adhesion function of PBMCs of RasGRP4 KO mice was reduced compared to that of WT mice. Moreover, the expression of adhesion molecules and critical signal transduction molecules in the NLRP3 inflammasome and MAPK/NF-κB signaling pathways in GEnCs was stimulated by the supernatant of PBMCs, which were derived from RasGRP4 KO mice treated with high glucose and were also significantly reduced compared to those derived from WT mice. CONCLUSION RasGRP4 promotes the inflammatory injury mediated by PBMCs in diabetes, probably by regulating the interaction between PBMCs and GEnCs and further activating the NLRP3 inflammasome and MAPK/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Shuai Huang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Junmei Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Li Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Shasha Tian
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Yao Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xian Shao
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Saijun Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Pei Yu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
16
|
Sun H, Lagarrigue F, Ginsberg MH. The Connection Between Rap1 and Talin1 in the Activation of Integrins in Blood Cells. Front Cell Dev Biol 2022; 10:908622. [PMID: 35721481 PMCID: PMC9198492 DOI: 10.3389/fcell.2022.908622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/25/2022] [Indexed: 01/13/2023] Open
Abstract
Integrins regulate the adhesion and migration of blood cells to ensure the proper positioning of these cells in the environment. Integrins detect physical and chemical stimuli in the extracellular matrix and regulate signaling pathways in blood cells that mediate their functions. Integrins are usually in a resting state in blood cells until agonist stimulation results in a high-affinity conformation ("integrin activation"), which is central to integrins' contribution to blood cells' trafficking and functions. In this review, we summarize the mechanisms of integrin activation in blood cells with a focus on recent advances understanding of mechanisms whereby Rap1 regulates talin1-integrin interaction to trigger integrin activation in lymphocytes, platelets, and neutrophils.
Collapse
Affiliation(s)
- Hao Sun
- Department of Medicine, University of California San Diego, San Diego, CA, United States
| | - Frederic Lagarrigue
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Mark H. Ginsberg
- Department of Medicine, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
17
|
Lee RH, Ghalloussi D, Harousseau GL, Kenny JP, Kramer PA, Proamer F, Nieswandt B, Flick MJ, Gachet C, Casari C, Eckly A, Bergmeier W. Rasa3 deficiency minimally affects thrombopoiesis but promotes severe thrombocytopenia due to integrin-dependent platelet clearance. JCI Insight 2022; 7:e155676. [PMID: 35290242 PMCID: PMC9089782 DOI: 10.1172/jci.insight.155676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/09/2022] [Indexed: 01/26/2023] Open
Abstract
Platelet homeostasis is dependent on a tight regulation of both platelet production and clearance. The small GTPase Rap1 mediates platelet adhesion and hemostatic plug formation. However, Rap1 signaling is also critical for platelet homeostasis as both Rap1 deficiency and uninhibited Rap1 signaling lead to marked thrombocytopenia in mice. Here, we investigated the mechanism by which deficiency in Rasa3, a critical negative regulator of Rap1, causes macrothrombocytopenia in mice. Despite marked morphological and ultrastructural abnormalities, megakaryocytes in hypomorphic Rasa3hlb/hlb (R3hlb/hlb) or Rasa3-/- mice demonstrated robust proplatelet formation in vivo, suggesting that defective thrombopoiesis is not the main cause of thrombocytopenia. Rather, we observed that R3hlb/hlb platelets became trapped in the spleen marginal zone/red pulp interface, with evidence of platelet phagocytosis by macrophages. Clearance of mutant platelets was also observed in the liver, especially in splenectomized mice. Platelet count and platelet life span in Rasa3-mutant mice were restored by genetic or pharmacological approaches to inhibit the Rap1/talin1/αIIbβ3 integrin axis. A similar pattern of splenic clearance was observed in mice injected with anti-αIIbβ3 but not anti-glycoprotein Ibα platelet-depleting antibodies. In summary, we describe a potentially novel, integrin-based mechanism of platelet clearance that could be critical for our understanding of select inherited and acquired thrombocytopenias.
Collapse
Affiliation(s)
- Robert H. Lee
- Department of Biochemistry and Biophysics and
- Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Dorsaf Ghalloussi
- Department of Biochemistry and Biophysics and
- Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | | | | | | | - Fabienne Proamer
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67065 Strasbourg, France
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Matthew J. Flick
- Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67065 Strasbourg, France
| | - Caterina Casari
- Department of Biochemistry and Biophysics and
- HITh, UMR_S1176, INSERM, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, F-67065 Strasbourg, France
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics and
- Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| |
Collapse
|
18
|
Bourguignon A, Tasneem S, Hayward CP. Screening and diagnosis of inherited platelet disorders. Crit Rev Clin Lab Sci 2022; 59:405-444. [PMID: 35341454 DOI: 10.1080/10408363.2022.2049199] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inherited platelet disorders are important conditions that often manifest with bleeding. These disorders have heterogeneous underlying pathologies. Some are syndromic disorders with non-blood phenotypic features, and others are associated with an increased predisposition to developing myelodysplasia and leukemia. Platelet disorders can present with thrombocytopenia, defects in platelet function, or both. As the underlying pathogenesis of inherited thrombocytopenias and platelet function disorders are quite diverse, their evaluation requires a thorough clinical assessment and specialized diagnostic tests, that often challenge diagnostic laboratories. At present, many of the commonly encountered, non-syndromic platelet disorders do not have a defined molecular cause. Nonetheless, significant progress has been made over the past few decades to improve the diagnostic evaluation of inherited platelet disorders, from the assessment of the bleeding history to improved standardization of light transmission aggregometry, which remains a "gold standard" test of platelet function. Some platelet disorder test findings are highly predictive of a bleeding disorder and some show association to symptoms of prolonged bleeding, surgical bleeding, and wound healing problems. Multiple assays can be required to diagnose common and rare platelet disorders, each requiring control of preanalytical, analytical, and post-analytical variables. The laboratory investigations of platelet disorders include evaluations of platelet counts, size, and morphology by light microscopy; assessments for aggregation defects; tests for dense granule deficiency; analyses of granule constituents and their release; platelet protein analysis by immunofluorescent staining or flow cytometry; tests of platelet procoagulant function; evaluations of platelet ultrastructure; high-throughput sequencing and other molecular diagnostic tests. The focus of this article is to review current methods for the diagnostic assessment of platelet function, with a focus on contemporary, best diagnostic laboratory practices, and relationships between clinical and laboratory findings.
Collapse
Affiliation(s)
- Alex Bourguignon
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Subia Tasneem
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Catherine P Hayward
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.,Department of Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
19
|
Pluthero FG, Westbury SK, Kahr WHA. Inherited platelet disorders: From new variants to new knowledge. Br J Haematol 2022; 197:245-246. [PMID: 35187641 DOI: 10.1111/bjh.18074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Fred G Pluthero
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sarah K Westbury
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Walter H A Kahr
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Departments of Paediatrics and Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Boeckelmann D, Glonnegger H, Sandrock-Lang K, Zieger B. Pathogenic Aspects of Inherited Platelet Disorders. Hamostaseologie 2021; 41:460-468. [PMID: 34942659 DOI: 10.1055/a-1665-6249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Inherited platelet disorders (IPDs) constitute a large heterogeneous group of rare bleeding disorders. These are classified into: (1) quantitative defects, (2) qualitative disorders, or (3) altered platelet production rate disorders or increased platelet turnover. Classically, IPD diagnostic is based on clinical phenotype characterization, comprehensive laboratory analyses (platelet function analysis), and, in former times, candidate gene sequencing. Today, molecular genetic analysis is performed using next-generation sequencing, mostly by targeting enrichment of a gene panel or by whole-exome sequencing. Still, the biochemical and molecular genetic characterization of patients with congenital thrombocytopathias/thrombocytopenia is essential, since postoperative or posttraumatic bleeding often occurs due to undiagnosed platelet defects. Depending upon the kind of surgery or trauma, this bleeding may be life-threatening, e.g., after tonsillectomy or in brain surgery. Undiagnosed platelet defects may lead to additional surgery, hysterectomy, pulmonary bleeding, and even resuscitation. In addition, these increased bleeding symptoms can lead to wound healing problems. Only specialized laboratories can perform the special platelet function analyses (aggregometry, flow cytometry, or immunofluorescent microscopy of the platelets); therefore, many IPDs are still undetected.
Collapse
Affiliation(s)
- Doris Boeckelmann
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Hannah Glonnegger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Kirstin Sandrock-Lang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Barbara Zieger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
21
|
Ning W, Ma Y, Li S, Wang X, Pan H, Wei C, Zhang S, Bai D, Liu X, Deng Y, Acharya A, Pelekos G, Savkovic V, Li H, Gaus S, Haak R, Schmalz G, Ziebolz D, Ma Y, Xu Y. Shared Molecular Mechanisms between Atherosclerosis and Periodontitis by Analyzing the Transcriptomic Alterations of Peripheral Blood Monocytes. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:1498431. [PMID: 34899963 PMCID: PMC8664523 DOI: 10.1155/2021/1498431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/12/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVE This study investigated the nature of shared transcriptomic alterations in PBMs from periodontitis and atherosclerosis to unravel molecular mechanisms underpinning their association. METHODS Gene expression data from PBMs from patients with periodontitis and those with atherosclerosis were each downloaded from the GEO database. Differentially expressed genes (DEGs) in periodontitis and atherosclerosis were identified through differential gene expression analysis. The disease-related known genes related to periodontitis and atherosclerosis each were downloaded from the DisGeNET database. A Venn diagram was constructed to identify crosstalk genes from four categories: DEGs expressed in periodontitis, periodontitis-related known genes, DEGs expressed in atherosclerosis, and atherosclerosis-related known genes. A weighted gene coexpression network analysis (WGCNA) was performed to identify significant coexpression modules, and then, coexpressed gene interaction networks belonging to each significant module were constructed to identify the core crosstalk genes. RESULTS Functional enrichment analysis of significant modules obtained by WGCNA analysis showed that several pathways might play the critical crosstalk role in linking both diseases, including bacterial invasion of epithelial cells, platelet activation, and Mitogen-Activated Protein Kinases (MAPK) signaling. By constructing the gene interaction network of significant modules, the core crosstalk genes in each module were identified and included: for GSE23746 dataset, RASGRP2 in the blue module and VAMP7 and SNX3 in the green module, as well as HMGB1 and SUMO1 in the turquoise module were identified; for GSE61490 dataset, SEC61G, PSMB2, SELPLG, and FIBP in the turquoise module were identified. CONCLUSION Exploration of available transcriptomic datasets revealed core crosstalk genes (RASGRP2, VAMP7, SNX3, HMGB1, SUMO1, SEC61G, PSMB2, SELPLG, and FIBP) and significant pathways (bacterial invasion of epithelial cells, platelet activation, and MAPK signaling) as top candidate molecular linkage mechanisms between atherosclerosis and periodontitis.
Collapse
Affiliation(s)
- Wanchen Ning
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yihong Ma
- Department of Neurology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Simin Li
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xin Wang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hongying Pan
- School of Dentistry, University of Michigan, 1011 N University Ave, Ann Arbor, MI 48109, USA
| | - Chenxuan Wei
- School of Dentistry, University of Michigan, 1011 N University Ave, Ann Arbor, MI 48109, USA
| | - Shaochuan Zhang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dongying Bai
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
| | - Xiangqiong Liu
- Laboratory of Molecular Cell Biology, Beijing Tibetan Hospital, China Tibetology Research Center, 218 Anwaixiaoguanbeili Street, Chaoyang, Beijing 100029, China
| | - Yupei Deng
- Laboratory of Molecular Cell Biology, Beijing Tibetan Hospital, China Tibetology Research Center, 218 Anwaixiaoguanbeili Street, Chaoyang, Beijing 100029, China
| | - Aneesha Acharya
- Dr D Y Patil Dental College and Hospital, Dr D Y Patil Vidyapeeth, Pimpri, Pune, India
| | - George Pelekos
- Faculty of Dentistry, University of Hong Kong, Hong KongChina
| | - Vuk Savkovic
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Hanluo Li
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Sebastian Gaus
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Rainer Haak
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, 04103 Leipzig, Germany
| | - Gerhard Schmalz
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, 04103 Leipzig, Germany
| | - Dirk Ziebolz
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, 04103 Leipzig, Germany
| | - Yanbo Ma
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province 271000, China
| |
Collapse
|
22
|
CalDAG-GEFI Deficiency in a Family with Symptomatic Heterozygous and Homozygous Carriers of a Likely Pathogenic Variant in RASGRP2. Int J Mol Sci 2021; 22:ijms222212423. [PMID: 34830306 PMCID: PMC8618213 DOI: 10.3390/ijms222212423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 11/28/2022] Open
Abstract
RASGRP2 encodes the calcium and diacylglycerol (DAG)-regulated guanine nucleotide exchange factor I (CalDAG-GEFI) identified as a Rap1-activating molecule. Pathogenic variants previously identified in RASGRP2 allowed the characterization of CalDAG-GEFI deficiency as a non-syndromic, autosomal recessive platelet function disease. We report on the clinical manifestations and laboratory features of a Portuguese family with a likely pathogenic variant in RASGRP2 (c.999G>C leading to a p.Lys333Asn change in the CDC25 catalytic domain of CalDAG-GEFI) and discuss the contribution of this variant to the disease manifestations. Based on the study of this family with one homozygous patient and five heterozygous carriers and on a critical analysis of the literature, we challenge previous knowledge that CalDAG-GEFI deficiency only manifests in homozygous patients. Our data suggest that at least for the RASGRP2 variant reported herein, there is a phenotypic expression, albeit milder, in heterozygous carriers.
Collapse
|
23
|
Saultier P, Cabantous S, Puceat M, Peiretti F, Bigot T, Saut N, Bordet JC, Canault M, van Agthoven J, Loosveld M, Payet-Bornet D, Potier D, Falaise C, Bernot D, Morange PE, Alessi MC, Poggi M. GATA1 pathogenic variants disrupt MYH10 silencing during megakaryopoiesis. J Thromb Haemost 2021; 19:2287-2301. [PMID: 34060193 DOI: 10.1111/jth.15412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/24/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND GATA1 is an essential transcription factor for both polyploidization and megakaryocyte (MK) differentiation. The polyploidization defect observed in GATA1 variant carriers is not well understood. OBJECTIVE To extensively phenotype two pedigrees displaying different variants in the GATA1 gene and determine if GATA1 controls MYH10 expression levels, a key modulator of MK polyploidization. METHOD A total of 146 unrelated propositi with constitutional thrombocytopenia were screened on a multigene panel. We described the genotype-phenotype correlation in GATA1 variant carriers and investigated the effect of these novel variants on MYH10 transcription using luciferase constructs. RESULTS The clinical profile associated with the p.L268M variant localized in the C terminal zinc finger was unusual in that the patient displayed bleeding and severe platelet aggregation defects without early-onset thrombocytopenia. p.N206I localized in the N terminal zinc finger was associated, on the other hand, with severe thrombocytopenia (15G/L) in early life. High MYH10 levels were evidenced in platelets of GATA1 variant carriers. Analysis of MKs anti-GATA1 chromatin immunoprecipitation-sequencing data revealed two GATA1 binding sites, located in the 3' untranslated region and in intron 8 of the MYH10 gene. Luciferase reporter assays showed their respective role in the regulation of MYH10 gene expression. Both GATA1 variants significantly alter intron 8 driven MYH10 transcription. CONCLUSION The discovery of an association between MYH10 and GATA1 is a novel one. Overall, this study suggests that impaired MYH10 silencing via an intronic regulatory element is the most likely cause of GATA1-related polyploidization defect.
Collapse
Affiliation(s)
- Paul Saultier
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- Department of Pediatric Hematology, Immunology and Oncology, APHM, La Timone Children's Hospital, Marseille, France
| | | | | | | | - Timothée Bigot
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | - Noémie Saut
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | | | | | - Johannes van Agthoven
- Structural Biology Program, Division of Nephrology/Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Marie Loosveld
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
- Aix-Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | | - Céline Falaise
- Department of Pediatric Hematology, Immunology and Oncology, APHM, La Timone Children's Hospital, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Denis Bernot
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | - Pierre-Emmanuel Morange
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Marie-Christine Alessi
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
- APHM, CHU Timone, French Reference Center on Inherited Platelet Disorders, Marseille, France
| | - Marjorie Poggi
- Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| |
Collapse
|
24
|
Crittenden JR, Zhai S, Sauvage M, Kitsukawa T, Burguière E, Thomsen M, Zhang H, Costa C, Martella G, Ghiglieri V, Picconi B, Pescatore KA, Unterwald EM, Jackson WS, Housman DE, Caine SB, Sulzer D, Calabresi P, Smith AC, Surmeier DJ, Graybiel AM. CalDAG-GEFI mediates striatal cholinergic modulation of dendritic excitability, synaptic plasticity and psychomotor behaviors. Neurobiol Dis 2021; 158:105473. [PMID: 34371144 PMCID: PMC8486000 DOI: 10.1016/j.nbd.2021.105473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 01/19/2023] Open
Abstract
CalDAG-GEFI (CDGI) is a protein highly enriched in the striatum, particularly in the principal spiny projection neurons (SPNs). CDGI is strongly down-regulated in two hyperkinetic conditions related to striatal dysfunction: Huntington’s disease and levodopa-induced dyskinesia in Parkinson’s disease. We demonstrate that genetic deletion of CDGI in mice disrupts dendritic, but not somatic, M1 muscarinic receptors (M1Rs) signaling in indirect pathway SPNs. Loss of CDGI reduced temporal integration of excitatory postsynaptic potentials at dendritic glutamatergic synapses and impaired the induction of activity-dependent long-term potentiation. CDGI deletion selectively increased psychostimulant-induced repetitive behaviors, disrupted sequence learning, and eliminated M1R blockade of cocaine self-administration. These findings place CDGI as a major, but previously unrecognized, mediator of cholinergic signaling in the striatum. The effects of CDGI deletion on the self-administration of drugs of abuse and its marked alterations in hyperkinetic extrapyramidal disorders highlight CDGI’s therapeutic potential.
Collapse
Affiliation(s)
- Jill R Crittenden
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Shenyu Zhai
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Magdalena Sauvage
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Leibniz Institute for Neurobiology, Functional Architecture of Memory Dept., Magdeburg, Germany
| | - Takashi Kitsukawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Eric Burguière
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Brain and Spine Institute (ICM), CNRS UMR 7225, INSERM U 1127, UPMC-P6 UMR S, 1127, Hôpital de la Pitié-Salpêtrière, 47 boulevard de l'hôpital, Paris, France
| | - Morgane Thomsen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University, DK-2100, Copenhagen, Denmark; Basic Neuroscience Division, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Hui Zhang
- Departments of Psychiatry, Pharmacology, Neurology, Columbia University, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Cinzia Costa
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della misericordia, University of Perugia, 06100 Perugia, Italy
| | - Giuseppina Martella
- Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | | | | | - Karen A Pescatore
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Ellen M Unterwald
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Walker S Jackson
- Wallenberg Center for Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - David E Housman
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - S Barak Caine
- Basic Neuroscience Division, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - David Sulzer
- Departments of Psychiatry, Pharmacology, Neurology, Columbia University, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Paolo Calabresi
- Neurological Clinic, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; Department of Neuroscience, Faculty of Medicine, Università Cattolica del "Sacro Cuore", 00168 Rome, Italy
| | - Anne C Smith
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85724, USA
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
25
|
Montenont E, Bhatlekar S, Jacob S, Kosaka Y, Manne BK, Lee O, Parra-Izquierdo I, Tugolukova E, Tolley ND, Rondina MT, Bray PF, Rowley JW. CRISPR-edited megakaryocytes for rapid screening of platelet gene functions. Blood Adv 2021; 5:2362-2374. [PMID: 33944898 PMCID: PMC8114553 DOI: 10.1182/bloodadvances.2020004112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/09/2021] [Indexed: 01/07/2023] Open
Abstract
Human anucleate platelets cannot be directly modified using traditional genetic approaches. Instead, studies of platelet gene function depend on alternative models. Megakaryocytes (the nucleated precursor to platelets) are the nearest cell to platelets in origin, structure, and function. However, achieving consistent genetic modifications in primary megakaryocytes has been challenging, and the functional effects of induced gene deletions on human megakaryocytes for even well-characterized platelet genes (eg, ITGA2B) are unknown. Here we present a rapid and systematic approach to screen genes for platelet functions in CD34+ cell-derived megakaryocytes called CRIMSON (CRISPR-edited megakaryocytes for rapid screening of platelet gene functions). By using CRISPR/Cas9, we achieved efficient nonviral gene editing of a panel of platelet genes in megakaryocytes without compromising megakaryopoiesis. Gene editing induced loss of protein in up to 95% of cells for platelet function genes GP6, RASGRP2, and ITGA2B; for the immune receptor component B2M; and for COMMD7, which was previously associated with cardiovascular disease and platelet function. Gene deletions affected several select responses to platelet agonists in megakaryocytes in a manner largely consistent with those expected for platelets. Deletion of B2M did not significantly affect platelet-like responses, whereas deletion of ITGA2B abolished agonist-induced integrin activation and spreading on fibrinogen without affecting the translocation of P-selectin. Deletion of GP6 abrogated responses to collagen receptor agonists but not thrombin. Deletion of RASGRP2 impaired functional responses to adenosine 5'-diphosphate (ADP), thrombin, and collagen receptor agonists. Deletion of COMMD7 significantly impaired multiple responses to platelet agonists. Together, our data recommend CRIMSON for rapid evaluation of platelet gene phenotype associations.
Collapse
Affiliation(s)
- Emilie Montenont
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Seema Bhatlekar
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Shancy Jacob
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Yasuhiro Kosaka
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Bhanu K Manne
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Olivia Lee
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | | | - Emilia Tugolukova
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Neal D Tolley
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
- George E. Wahlen Department of Veterans Affairs Medical Center
- Department of Internal Medicine and Geriatric Research and Education Clinical Center, and
- Department of Pathology, The University of Utah, Salt Lake City, UT
| | - Paul F Bray
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
| | - Jesse W Rowley
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
| |
Collapse
|
26
|
Yang EJ, Shim YJ, Kim HS, Lim YT, Im HJ, Koh KN, Kim H, Suh JK, Park ES, Lee NH, Choi YB, Hah JO, Lee JM, Han JW, Lee JH, Lee YH, Jung HL, Ha JS, Ki CS, on behalf of the Benign Hematology Committee of the Korean Pediatric Hematology Oncology Group (KPHOG). Genetic Confirmation and Identification of Novel Variants for Glanzmann Thrombasthenia and Other Inherited Platelet Function Disorders: A Study by the Korean Pediatric Hematology Oncology Group (KPHOG). Genes (Basel) 2021; 12:genes12050693. [PMID: 34066320 PMCID: PMC8148153 DOI: 10.3390/genes12050693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/21/2021] [Accepted: 05/02/2021] [Indexed: 11/30/2022] Open
Abstract
The diagnosis of inherited platelet function disorders (IPFDs) is challenging owing to the unavailability of essential testing methods, including light transmission aggregometry and flow cytometry, in several medical centers in Korea. This study, conducted by the Korean Pediatric Hematology Oncology Group from March 2017 to December 2020, aimed to identify the causative genetic variants of IPFDs in Korean patients using next-generation sequencing (NGS). Targeted exome sequencing, followed by whole-genome sequencing, was performed for diagnosing IPFDs. Of the 11 unrelated patients with suspected IPFDs enrolled in this study, 10 patients and 2 of their family members were diagnosed with Glanzmann thrombasthenia (GT). The variant c.1913+5G>T of ITGB3 was the most common, followed by c.2333A>C (p.Gln778Pro) of ITGB2B. Known variants of GT, including c.917A>C (p.His306Pro) of ITGB3 and c.2975del (p.Glu992Glyfs*), c.257T>C (p.Leu86Pro), and c.1750C>T (p.Arg584*) of ITGA2B, were identified. Four novel variants of GT, c.1451G>T (p.Gly484Val) and c.1595G>T (p.Cys532Phe) of ITGB3 and c.1184G>T (p.Gly395Val) and c.2390del (p.Gly797Valfs*29) of ITGA2B, were revealed. The remaining patient was diagnosed with platelet type bleeding disorder 18 and harbored two novel RASGRP2 variants, c.1479dup (p.Arg494Alafs*54) and c.813+1G>A. We demonstrated the successful application of NGS for the accurate and differential diagnosis of heterogeneous IPFDs.
Collapse
Affiliation(s)
- Eu Jeen Yang
- Department of Pediatrics, Pusan National University School of Medicine, Pusan National University Children’s Hospital, Yangsan 50612, Korea; (E.J.Y.); (Y.T.L.)
| | - Ye Jee Shim
- Department of Pediatrics, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, Daegu 42601, Korea
- Correspondence: ; Tel.: +82-53-258-7824
| | - Heung Sik Kim
- Department of Pediatrics, Keimyung University School of Medicine, Keimyung University Daegu Dongsan Hospital, Daegu 41931, Korea;
| | - Young Tak Lim
- Department of Pediatrics, Pusan National University School of Medicine, Pusan National University Children’s Hospital, Yangsan 50612, Korea; (E.J.Y.); (Y.T.L.)
| | - Ho Joon Im
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul 05505, Korea; (H.J.I.); (K.-N.K.); (H.K.)
| | - Kyung-Nam Koh
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul 05505, Korea; (H.J.I.); (K.-N.K.); (H.K.)
| | - Hyery Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul 05505, Korea; (H.J.I.); (K.-N.K.); (H.K.)
| | - Jin Kyung Suh
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul 01812, Korea;
| | - Eun Sil Park
- Department of Pediatrics, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, Jinju 52727, Korea;
| | - Na Hee Lee
- Department of Pediatrics, Cha Bundang Medical Center, Cha University, Seongnam 13496, Korea;
| | - Young Bae Choi
- Department of Pediatrics, Ajou University School of Medicine, Ajou University Hospital, Suwon 16499, Korea;
| | - Jeong Ok Hah
- Department of Pediatrics, Daegu Fatima Hospital, Daegu 41199, Korea;
| | - Jae Min Lee
- Department of Pediatrics, Yeungnam University College of Medicine, Daegu 42415, Korea;
| | - Jung Woo Han
- Department of Pediatrics, Yonsei University College of Medicine, Yonsei University Health System, Seoul 03722, Korea;
| | - Jae Hee Lee
- Department of Pediatrics, Chungbuk National University School of Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea;
| | - Young-Ho Lee
- Department of Pediatrics, Hanyang University Seoul Hospital, Seoul 04763, Korea;
| | - Hye Lim Jung
- Deparment of Pediatrics, Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Seoul 03181, Korea;
| | - Jung-Sook Ha
- Department of Laboratory Medicine, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, Daegu 42601, Korea;
| | | | | |
Collapse
|
27
|
Rosenberg N, Dardik R, Hauschner H, Nakav S, Barel O, Luboshitz J, Yacobovich J, Tamary H, Kenet G. Mutations in RASGRP2 gene identified in patients misdiagnosed as Glanzmann thrombasthenia patients. Blood Cells Mol Dis 2021; 89:102560. [PMID: 33711653 DOI: 10.1016/j.bcmd.2021.102560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Glanzmann thrombasthenia (GT) is a severe inherited platelet function disorder (IPFD), presenting with bleeding diathesis and impaired platelet aggregation, is caused by mutations in the genes ITGA2B or ITGB3. AIM We aimed to study the genetic cause of IPFD mimicking GT. METHODS During 2017-2019, 16 patients were referred to our tertiary center with bleeding symptoms, impaired platelet aggregation and normal platelet count and size. RESULTS Using flow cytometry, 13/16 patients were diagnosed with GT, yet three patients displayed normal surface expression of the integrins αIIbβ3 and αvβ3, as well as normal integrin αIIbβ3 activation following incubation with the activating monoclonal antibody anti-LIBS6, while platelet activation following ADP or epinephrine was impaired. Whole exome sequencing detected 2 variants in RASGRP2 gene in all 3 patients. DISCUSSION Both RASGRP2 mutations predicted frameshift, premature stop codon (p. I427Mfs*92 and p. R494Afs*54, respectively) and truncated calcium-sensing guanine nucleotide exchange factor [CalDAG-GEFI]- the major signaling molecule that regulates integrin-mediated aggregation and granule secretion, causing IPFD-18. CONCLUSION Patients who suffer from bleeding diathesis without immune dysregulation, may be mistakenly diagnosed as GT. Further studies are required to confirm the diagnosis of specific IPFD.
Collapse
Affiliation(s)
- Nurit Rosenberg
- The Israeli National Hemophilia Center and Thrombosis Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer, and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rima Dardik
- The Israeli National Hemophilia Center and Thrombosis Institute, Sheba Medical Center, Tel Hashomer, Israel
| | - Hagit Hauschner
- The Israeli National Hemophilia Center and Thrombosis Institute, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer, and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sigal Nakav
- Coagulation and Hemostasis Laboratory, Hematology Laboratories, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Ortal Barel
- Bioinformatic Unit, Sheba Cancer Research Center, Tel-Hashomer, Israel
| | - Jacob Luboshitz
- The Israeli National Hemophilia Center and Thrombosis Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Joanne Yacobovich
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Pediatric Hematology, Schneider Children's Medical Center, Petach-Tikva; Israel
| | - Hannah Tamary
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Pediatric Hematology, Schneider Children's Medical Center, Petach-Tikva; Israel
| | - Gili Kenet
- The Israeli National Hemophilia Center and Thrombosis Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel Hashomer, and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
28
|
Nurden P, Stritt S, Favier R, Nurden AT. Inherited platelet diseases with normal platelet count: phenotypes, genotypes and diagnostic strategy. Haematologica 2021; 106:337-350. [PMID: 33147934 PMCID: PMC7849565 DOI: 10.3324/haematol.2020.248153] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
Inherited platelet disorders resulting from platelet function defects and a normal platelet count cause a moderate or severe bleeding diathesis. Since the description of Glanzmann thrombasthenia resulting from defects of ITGA2B and ITGB3, new inherited platelet disorders have been discovered, facilitated by the use of high throughput sequencing and genomic analyses. Defects of RASGRP2 and FERMT3 responsible for severe bleeding syndromes and integrin activation have illustrated the critical role of signaling molecules. Important are mutations of P2RY12 encoding the major ADP receptor causal for an inherited platelet disorder with inheritance characteristics that depend on the variant identified. Interestingly, variants of GP6 encoding the major subunit of the collagen receptor GPVI/FcRγ associate only with mild bleeding. The numbers of genes involved in dense granule defects including Hermansky-Pudlak and Chediak Higashi syndromes continue to progress and are updated. The ANO6 gene encoding a Ca2+-activated ion channel required for phospholipid scrambling is responsible for the rare Scott syndrome and decreased procoagulant activity. A novel EPHB2 defect in a familial bleeding syndrome demonstrates a role for this tyrosine kinase receptor independent of the classical model of its interaction with ephrins. Such advances highlight the large diversity of variants affecting platelet function but not their production, despite the difficulties in establishing a clear phenotype when few families are affected. They have provided insights into essential pathways of platelet function and have been at the origin of new and improved therapies for ischemic disease. Nevertheless, many patients remain without a diagnosis and requiring new strategies that are now discussed.
Collapse
Affiliation(s)
| | - Simon Stritt
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala
| | - Remi Favier
- French National Reference Center for Inherited Platelet Disorders, Armand Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Paris
| | | |
Collapse
|
29
|
Körholz J, Lucas N, Boiti F, Althaus K, Tiebel O, Fang M, Berner R, Lee-Kirsch MA, Knöfler R. Severe Bleeding Diathesis in Siblings with Platelet Dysfunction due to a Novel Nonsense RASGRP2 Mutation. TH OPEN 2020; 4:e413-e416. [PMID: 33376940 PMCID: PMC7762629 DOI: 10.1055/s-0040-1718910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 09/15/2020] [Indexed: 12/25/2022] Open
Abstract
Next-generation sequencing is increasingly applied during the diagnostic work-up of patients with bleeding diathesis and has facilitated the diagnosis of rare bleeding disorders such as inherited platelet function disorders. Mutations in RAS guanyl releasing protein 2 (RasGRP2), also known as calcium- and diacylglycerol-regulated guanine nucleotide exchange factor I (CalDAG-GEFI), underlie a recently described platelet signal transduction abnormality. Here we present the case of a consanguineous family originating from Afghanistan with two siblings affected by recurrent severe mucocutaneous bleedings. Platelet function testing demonstrated a marked reduction of aggregation induced by collagen and adenosine diphosphate. Whole exome sequencing revealed a novel homozygous nonsense
RASGRP2
mutation segregating with the bleeding disorder in the family.
Collapse
Affiliation(s)
- Julia Körholz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nadja Lucas
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Franziska Boiti
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Karina Althaus
- Institute for Transfusion Medicine, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Oliver Tiebel
- Institute for Clinical Chemistry and Laboratory Medicine, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mingyan Fang
- BGI-Shenzhen and China National GeneBank, Shenzhen, China
| | - Reinhard Berner
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ralf Knöfler
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
30
|
Tynngård N, Bell A, Gryfelt G, Cvetkovic S, Wikman A, Uhlin M, Sandgren P. Cryopreservation of buffy coat derived platelets: Paired in vitro characterization using uncontrolled versus controlled freezing rate protocols. Transfusion 2020; 61:546-556. [PMID: 33345368 PMCID: PMC7898315 DOI: 10.1111/trf.16227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/21/2020] [Accepted: 10/09/2020] [Indexed: 12/21/2022]
Abstract
Background Cryopreserved platelets show a reduced recovery and viability after freezing and thawing including several ultrastructural and phenotypic deteriorations compared with liquid‐stored platelets. It is suggested that using Controlled‐Rate Freezing (CRF) can reduce variability and optimize the functionality profile for cells. The objective of the study is to compare cellular, metabolic, phenotypic and functional effects on platelets after cryopreservation using different freezing rate protocols. Study Design and Methods To evaluate the possible effects of different freezing rate protocols a two‐experimental study comparing diverse combinations was tested with a pool and split design. Uncontrolled freezing of platelets in materials with different thermal conductivity (metal vs cardboard) was evaluated in experiment 1. Experiment 2 evaluated uncontrolled vs a controlled‐rate freezing protocol in metal boxes. All variables were assessed pre and post cryopreservation. Results Directly after thawing, no major differences in platelet recovery, LDH, ATP, Δψ, CD62P, CD42b, platelet endothelial cell adhesion molecule and sCD40L were seen between units frozen with different thermal conductivity for temperature. In contrast, we observed signs of increased activation after freezing using the CRF protocol, reflected by increased cell surface expression of CD62P, PAC‐1 binding and increased concentration of LDH. Agonist induced expression of a conformational epitope on the GPIIb/IIIa complex and contribution to blood coagulation in an experimental rotational thromboelastometry setup were not statistically different between the groups. Conclusion The use of a uncontrolled freezing rate protocol is feasible, creating a platelet product comparable to using a controlled rate freezing equipment during cryopreservation of platelets.
Collapse
Affiliation(s)
- Nahreen Tynngård
- Research and Development Unit in Region Östergötland and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Alice Bell
- Department of Laboratory Medicine, Karolinska Institutet, Solna, Sweden
| | - Gunilla Gryfelt
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Huddinge, Sweden
| | - Stefan Cvetkovic
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Huddinge, Sweden
| | - Agneta Wikman
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Huddinge, Sweden.,Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Huddinge, Sweden
| | - Michael Uhlin
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Huddinge, Sweden.,Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Huddinge, Sweden
| | - Per Sandgren
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Huddinge, Sweden.,Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Huddinge, Sweden
| |
Collapse
|
31
|
Al-Hebshi A. Inherited Platelet Function Disorder From Novel Mutations in RAS Guanyl-Releasing Protein-2 Confirmed by Sanger Sequencing. Cureus 2020; 12:e11708. [PMID: 33391941 PMCID: PMC7769792 DOI: 10.7759/cureus.11708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Inherited platelet disorders (IPDs) are genetically heterogeneous rare disorders due to quantitative and/or qualitative abnormalities of the platelet. IPDs are often predisposed to significant medical complications. RAS guanyl-releasing protein-2 (RASGRP2) was recently identified as a gene affected in patients with platelet function defects and a bleeding complication. RASGRP2 codes for the protein CalDAG-GEFI RAS (guanyl-releasing protein-2), a guanine nucleotide exchange factor for small guanosine triphosphate(GTP)ase Rap1. We used Sanger sequencing to identify a novel function-disrupting homozygous mutation in RASGRP2 responsible for bleeding diathesis and platelet dysfunction in a patient.
Collapse
Affiliation(s)
- Abdulqader Al-Hebshi
- Pediatric Hematology Oncology, Prince Mohammed Bin Abdulaziz Hospital, Medina, SAU.,Pediatric Hematology Oncology, Ministry of National Guard Health Affairs, Medina, SAU.,Pediatric Hematology Oncology, King Abdullah International Medical Research Center, Riyadh, SAU.,Pediatric Hematology Oncology, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| |
Collapse
|
32
|
Wang H, Wu Y, Fang R, Sa J, Li Z, Cao H, Cui Y. Time-Varying Gene Network Analysis of Human Prefrontal Cortex Development. Front Genet 2020; 11:574543. [PMID: 33304381 PMCID: PMC7701309 DOI: 10.3389/fgene.2020.574543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/19/2020] [Indexed: 11/13/2022] Open
Abstract
The prefrontal cortex (PFC) constitutes a large part of the human central nervous system and is essential for the normal social affection and executive function of humans and other primates. Despite ongoing research in this region, the development of interactions between PFC genes over the lifespan is still unknown. To investigate the conversion of PFC gene interaction networks and further identify hub genes, we obtained time-series gene expression data of human PFC tissues from the Gene Expression Omnibus (GEO) database. A statistical model, loggle, was used to construct time-varying networks and several common network attributes were used to explore the development of PFC gene networks with age. Network similarity analysis showed that the development of human PFC is divided into three stages, namely, fast development period, deceleration to stationary period, and recession period. We identified some genes related to PFC development at these different stages, including genes involved in neuronal differentiation or synapse formation, genes involved in nerve impulse transmission, and genes involved in the development of myelin around neurons. Some of these genes are consistent with findings in previous reports. At the same time, we explored the development of several known KEGG pathways in PFC and corresponding hub genes. This study clarified the development trajectory of the interaction between PFC genes, and proposed a set of candidate genes related to PFC development, which helps further study of human brain development at the genomic level supplemental to regular anatomical analyses. The analytical process used in this study, involving the loggle model, similarity analysis, and central analysis, provides a comprehensive strategy to gain novel insights into the evolution and development of brain networks in other organisms.
Collapse
Affiliation(s)
- Huihui Wang
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yongqing Wu
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Ruiling Fang
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Jian Sa
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Zhi Li
- Department of Hematology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongyan Cao
- Division of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The increasing use of high throughput sequencing and genomic analysis has facilitated the discovery of new causes of inherited platelet disorders. Studies of these disorders and their respective mouse models have been central to understanding their biology, and also in revealing new aspects of platelet function and production. This review covers recent contributions to the identification of genes, proteins and variants associated with inherited platelet defects, and highlights how these studies have provided insights into platelet development and function. RECENT FINDINGS Novel genes recently implicated in human platelet dysfunction include the galactose metabolism enzyme UDP-galactose-4-epimerase in macrothrombocytopenia, and erythropoietin-producing hepatoma-amplified sequence receptor transmembrane tyrosine kinase EPHB2 in a severe bleeding disorder with deficiencies in platelet agonist response and granule secretion. Recent studies of disease-associated variants established or clarified roles in platelet function and/or production for the membrane receptor G6b-B, the FYN-binding protein FYB1/ADAP, the RAS guanyl-releasing protein RASGRP2/CalDAG-GEFI and the receptor-like protein tyrosine phosphatase PTPRJ/CD148. Studies of genes associated with platelet disorders advanced understanding of the cellular roles of neurobeachin-like 2, as well as several genes influenced by the transcription regulator RUNT-related transcription factor 1 (RUNX1), including NOTCH4. SUMMARY The molecular bases of many hereditary platelet disorders have been elucidated by the application of recent advances in cell imaging and manipulation, genomics and protein function analysis. These techniques have also aided the detection of new disorders, and enabled studies of disease-associated genes and variants to enhance understanding of platelet development and function.
Collapse
|
34
|
Ibrahim-Kosta M, Alessi MC, Hezard N. Laboratory Techniques Used to Diagnose Constitutional Platelet Dysfunction. Hamostaseologie 2020; 40:444-459. [PMID: 32932546 DOI: 10.1055/a-1223-3306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Platelets play a major role in primary hemostasis, where activated platelets form plugs to stop hemorrhaging in response to vessel injuries. Defects in any step of the platelet activation process can cause a variety of platelet dysfunction conditions associated with bleeding. To make an accurate diagnosis, constitutional platelet dysfunction (CPDF) should be considered once von Willebrand disease and drug intake are ruled out. CPDF may be associated with thrombocytopenia or a genetic syndrome. CPDF diagnosis is complex, as no single test enables the analysis of all aspects of platelet function. Furthermore, the available tests lack standardization, and repeat tests must be performed in specialized laboratories especially for mild and moderate forms of the disease. In this review, we provide an overview of the laboratory tests used to diagnose CPDF, with a focus on light transmission platelet aggregation (LTA), flow cytometry (FC), and granules assessment. Global tests, mainly represented by LTA, are often initially performed to investigate the consequences of platelet activation on platelet aggregation in a single step. Global test results should be confirmed by additional analytical tests. FC represents an accurate, simple, and reliable test to analyze abnormalities in platelet receptors, and granule content and release. This technique may also be used to investigate platelet function by comparing resting- and activated-state platelet populations. Assessment of granule content and release also requires additional specialized analytical tests. High-throughput sequencing has become increasingly useful to diagnose CPDF. Advanced tests or external research laboratory techniques may also be beneficial in some cases.
Collapse
Affiliation(s)
- Manal Ibrahim-Kosta
- Aix Marseille University, INSERM, INRAE, Marseille Cedex 05, France.,Laboratory of Hematology, CHU Timone, Marseille Cedex 05, France
| | - Marie-Christine Alessi
- Aix Marseille University, INSERM, INRAE, Marseille Cedex 05, France.,Laboratory of Hematology, CHU Timone, Marseille Cedex 05, France
| | - Nathalie Hezard
- Laboratory of Hematology, CHU Timone, Marseille Cedex 05, France
| |
Collapse
|
35
|
Sprenkeler EGG, Webbers SDS, Kuijpers TW. When Actin is Not Actin' Like It Should: A New Category of Distinct Primary Immunodeficiency Disorders. J Innate Immun 2020; 13:3-25. [PMID: 32846417 DOI: 10.1159/000509717] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
An increasing number of primary immunodeficiencies (PIDs) have been identified over the last decade, which are caused by deleterious mutations in genes encoding for proteins involved in actin cytoskeleton regulation. These mutations primarily affect hematopoietic cells and lead to defective function of immune cells, such as impaired motility, signaling, proliferative capacity, and defective antimicrobial host defense. Here, we review several of these immunological "actinopathies" and cover both clinical aspects, as well as cellular mechanisms of these PIDs. We focus in particular on the effect of these mutations on human neutrophil function.
Collapse
Affiliation(s)
- Evelien G G Sprenkeler
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, The Netherlands, .,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, The Netherlands,
| | - Steven D S Webbers
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Alharby E, Bakhsh MA, Albalawi AM, Almutairi SO, Hashmi JA, Basit S. A novel missense variant in the RASGRP2 gene in patients with moderate to severe bleeding disorder. Platelets 2020; 31:646-651. [PMID: 32609603 DOI: 10.1080/09537104.2019.1663803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Inherited platelet function disorder-18 (IPD-18) is a relatively new non-syndromic autosomal recessive bleeding disorder. It is characterized by deficient or dysfunctional CalDAG-GEFI protein. The distinctive feature of the disease is impaired platelet aggregation in response to multiple physiologic agonists. We here report a family with a platelet-type bleeding disorder and a novel mutation in the RASGRP2 gene. The overall bleeding score for the affected individuals was 15 and 12. Based on the initial diagnosis of Glanzmann thrombasthenia, targeted sequencing of integrin subunit alpha 2b and integrin subunit beta 3 encoding genes ITGA2B and ITGB3 were carried out in both affected members of a family. Sequence alignment failed to identify the disease-causing variant(s) in both genes. Therefore, whole exome sequencing in one affected individual was performed. Data analysis detected a novel homozygous missense variant (c.956C>T; p.Pro319Leu) in the exon 9 of the RASGRP2 gene. Five additional individuals of a family including both parents, an affected individual and two asymptomatic individuals were Sanger sequenced for the variant (c.956C>T). The variant segregates in the family in an autosomal recessive manner. Several in silico tools predicted the variant as pathogenic. Protein modeling studies suggest that the mutation (p.Pro319Leu) cause a conformational change in the loop structure of the RasGEF domain of the CalDAG-GEFI protein. Reported variants in the RasGEF domain impair expression and/or nucleotide exchange activity of CalDAG-GEFI protein and thus inhibit the activation of Rap1 protein required for platelet adhesion and hemostatic plug formation.
Collapse
Affiliation(s)
- Essa Alharby
- Center for Genetics and Inherited Diseases, Taibah University Medina, Almadinah Almunawwarah , Saudi Arabia
| | - Mohammad A Bakhsh
- College of Applied Medical Science, Taibah University Medina, Almadinah Almunawwarah , Saudi Arabia
| | - Alia M Albalawi
- Center for Genetics and Inherited Diseases, Taibah University Medina, Almadinah Almunawwarah , Saudi Arabia
| | - Sultan O Almutairi
- College of Applied Medical Science, Taibah University Medina, Almadinah Almunawwarah , Saudi Arabia
| | - Jamil A Hashmi
- Center for Genetics and Inherited Diseases, Taibah University Medina, Almadinah Almunawwarah , Saudi Arabia
| | - Sulman Basit
- Center for Genetics and Inherited Diseases, Taibah University Medina, Almadinah Almunawwarah , Saudi Arabia
| |
Collapse
|
37
|
Martyanov AA, Morozova DS, Sorokina MA, Filkova AA, Fedorova DV, Uzueva SS, Suntsova EV, Novichkova GA, Zharkov PA, Panteleev MA, Sveshnikova AN. Heterogeneity of Integrin α IIbβ 3 Function in Pediatric Immune Thrombocytopenia Revealed by Continuous Flow Cytometry Analysis. Int J Mol Sci 2020; 21:ijms21093035. [PMID: 32344835 PMCID: PMC7246588 DOI: 10.3390/ijms21093035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/06/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune condition primarily induced by the loss of immune tolerance to the platelet glycoproteins. Here we develop a novel flow cytometry approach to analyze integrin αIIbβ3 functioning in ITP in comparison with Glanzmann thrombasthenia (GT) (negative control) and healthy pediatric donors (positive control). Continuous flow cytometry of Fura-Red-loaded platelets from whole hirudinated blood was used for the characterization of platelet responses to conventional activators. Calcium levels and fibrinogen binding were normalized to ionomycin-induced responses. Ex vivo thrombus formation on collagen was observed in parallel-plate flow chambers. Platelets from all ITP patients had significantly higher cytosolic calcium concentration in the quiescent state compared to healthy donors (15 ± 5 nM vs. 8 ± 5 nM), but calcium increases in response to all activators were normal. Clustering analysis revealed two subpopulations of ITP patients: the subgroup with high fibrinogen binding (HFB), and the subgroup with low fibrinogen binding (LFB) (8% ± 5% for LFB vs. 16% ± 3% for healthy donors in response to ADP). GT platelets had calcium mobilization (81 ± 23 nM), fibrinogen binding (5.1% ± 0.3%) and thrombus growth comparable to the LFB subgroup. Computational modeling suggested phospholipase C-dependent platelet pre-activation for the HFB subgroup and lower levels of functional integrin molecules for the LFB group.
Collapse
Affiliation(s)
- Alexey A. Martyanov
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia
- Institute for Biochemical Physics (IBCP), Russian Academy of Sciences (RAS), Russian Federation, Moscow, Kosyigina 4 119334, Russia
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
| | - Daria S. Morozova
- Faculty of Basic Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky av., Moscow 119991, Russia
| | - Maria A. Sorokina
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
| | - Aleksandra A. Filkova
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
| | - Daria V. Fedorova
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
| | - Selima S. Uzueva
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
| | - Elena V. Suntsova
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
| | - Galina A. Novichkova
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
| | - Pavel A. Zharkov
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
| | - Mikhail A. Panteleev
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
- Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, 9 Institutskii per., Dolgoprudnyi 141700, Russia
| | - Anastasia N. Sveshnikova
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
- Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
- Department of Normal Physiology, Sechenov First Moscow State Medical University, 8/2 Trubetskaya St., Moscow 119991, Russia
- Correspondence:
| |
Collapse
|
38
|
Alessi MC, Sié P, Payrastre B. Strengths and Weaknesses of Light Transmission Aggregometry in Diagnosing Hereditary Platelet Function Disorders. J Clin Med 2020; 9:jcm9030763. [PMID: 32178287 PMCID: PMC7141357 DOI: 10.3390/jcm9030763] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/17/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
Hereditary defects in platelet function are responsible for sometimes severe mucocutaneous hemorrhages. They are a heterogeneous group of abnormalities whose first-line diagnosis typically involves interpreting the results of in vitro light transmission aggregometry (LTA) traces. Interpretation of LTA is challenging. LTA is usually performed in specialized laboratories with expertise in platelet pathophysiology. This review updates knowledge on LTA, describing the various platelet aggregation profiles typical of hereditary platelet disorders to guide the physician in the diagnosis of functional platelet disorders.
Collapse
Affiliation(s)
- Marie-Christine Alessi
- Aix Marseille Univ, Inserm, Inrae, C2VN, 13385 Marseille CEDEX, France
- Correspondence: ; Tel.: +33-4-91-32-45-06
| | - Pierre Sié
- CHU de Toulouse, Laboratoire d’Hématologie, 31059 Toulouse CEDEX, France;
| | - Bernard Payrastre
- Inserm U1048, I2MC et Université Paul Sabatier, 31024 Toulouse CEDEX, France;
| |
Collapse
|
39
|
Sarker M, Goliaei A, Golesi F, Poggi M, Cook A, Khan MAI, Temple BR, Stefanini L, Canault M, Bergmeier W, Campbell SL. Subcellular localization of Rap1 GTPase activator CalDAG-GEFI is orchestrated by interaction of its atypical C1 domain with membrane phosphoinositides. J Thromb Haemost 2020; 18:693-705. [PMID: 31758832 PMCID: PMC7050387 DOI: 10.1111/jth.14687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/17/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The small GTPase Rap1 and its guanine nucleotide exchange factor, CalDAG-GEFI (CDGI), are critical for platelet function and hemostatic plug formation. CDGI function is regulated by a calcium binding EF hand regulatory domain and an atypical C1 domain with unknown function. OBJECTIVE Here, we investigated whether the C1 domain controls CDGI subcellular localization, both in vitro and in vivo. METHODS CDGI interaction with phosphoinositides was studied by lipid co-sedimentation assays and molecular dynamics simulations. Cellular localization of CDGI was studied in heterologous cells by immunofluorescence and subcellular fractionation assays. RESULTS Lipid co-sedimentation studies demonstrated that the CDGI C1 domain associates with membranes through exclusive recognition of phosphoinositides, phosphatidylinositol (4,5)-biphosphate (PIP2) and phosphatidylinositol (3,4,5)-triphosphate (PIP3). Molecular dynamics simulations identified a phospholipid recognition motif consisting of residues exclusive to the CDGI C1 domain. Mutation of those residues abolished co-sedimentation of the C1 domain with lipid vesicles and impaired membrane localization of CDGI in heterologous cells. CONCLUSION Our studies identify a novel interaction between an atypical C1 domain and phosphatidylinositol (4,5)-biphosphate and phosphatidylinositol (3,4,5)-triphosphate in cellular membranes, which is critical for Rap1 signaling in health and disease.
Collapse
Affiliation(s)
- Muzaddid Sarker
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ardeshir Goliaei
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Marjorie Poggi
- Aix Marseille University, INSERM, INRA, Marseille, France
| | - Aaron Cook
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mohammad A. I. Khan
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brenda R. Temple
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- RL Juliano Structural Bioinformatics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lucia Stefanini
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | | | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sharon L. Campbell
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
40
|
Integration of Rap1 and Calcium Signaling. Int J Mol Sci 2020; 21:ijms21051616. [PMID: 32120817 PMCID: PMC7084553 DOI: 10.3390/ijms21051616] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ is a universal intracellular signal. The modulation of cytoplasmic Ca2+ concentration regulates a plethora of cellular processes, such as: synaptic plasticity, neuronal survival, chemotaxis of immune cells, platelet aggregation, vasodilation, and cardiac excitation–contraction coupling. Rap1 GTPases are ubiquitously expressed binary switches that alternate between active and inactive states and are regulated by diverse families of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Active Rap1 couples extracellular stimulation with intracellular signaling through secondary messengers—cyclic adenosine monophosphate (cAMP), Ca2+, and diacylglycerol (DAG). Much evidence indicates that Rap1 signaling intersects with Ca2+ signaling pathways to control the important cellular functions of platelet activation or neuronal plasticity. Rap1 acts as an effector of Ca2+ signaling when activated by mechanisms involving Ca2+ and DAG-activated (CalDAG-) GEFs. Conversely, activated by other GEFs, such as cAMP-dependent GEF Epac, Rap1 controls cytoplasmic Ca2+ levels. It does so by regulating the activity of Ca2+ signaling proteins such as sarcoendoplasmic reticulum Ca2+-ATPase (SERCA). In this review, we focus on the physiological significance of the links between Rap1 and Ca2+ signaling and emphasize the molecular interactions that may offer new targets for the therapy of Alzheimer’s disease, hypertension, and atherosclerosis, among other diseases.
Collapse
|
41
|
Canault M, Alessi MC. RasGRP2 Structure, Function and Genetic Variants in Platelet Pathophysiology. Int J Mol Sci 2020; 21:E1075. [PMID: 32041177 PMCID: PMC7037602 DOI: 10.3390/ijms21031075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/18/2022] Open
Abstract
RasGRP2 is calcium and diacylglycerol-regulated guanine nucleotide exchange factor I that activates Rap1, which is an essential signaling-knot in "inside-out" αIIbβ3 integrin activation in platelets. Inherited platelet function disorder caused by variants of RASGRP2 represents a new congenital bleeding disorder referred to as platelet-type bleeding disorder-18 (BDPLT18). We review here the structure of RasGRP2 and its functions in the pathophysiology of platelets and of the other cellular types that express it. We will also examine the different pathogenic variants reported so far as well as strategies for the diagnosis and management of patients with BDPLT18.
Collapse
Affiliation(s)
- Matthias Canault
- Aix Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Marie-Christine Alessi
- Aix Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
- Hematology laboratory, APHM, CHU Timone, 13005 Marseille, France
| |
Collapse
|
42
|
Manukjan G, Wiegering VA, Reindl T, Strauß G, Klopocki E, Schulze H, Andres O. Novel variants in FERMT3 and RASGRP2-Genetic linkage in Glanzmann-like bleeding disorders. Pediatr Blood Cancer 2020; 67:e28078. [PMID: 31724816 DOI: 10.1002/pbc.28078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 11/07/2022]
Abstract
Defects of platelet intracellular signaling can result in severe platelet dysfunction. Several mutations in each of the linked genes FERMT3 and RASGRP2 on chromosome 11 causing a Glanzmann-like bleeding phenotype have been identified so far. We report on novel variants in two unrelated pediatric patients with severe bleeding diathesis-one with leukocyte adhesion deficiency type III due to a homozygous frameshift in FERMT3 and the other with homozygous variants in both, FERMT3 and RASGRP2. We focus on the challenging genetic and functional variant assessment and aim to accentuate the risk of obtaining misleading results due to the phenomenon of genetic linkage.
Collapse
Affiliation(s)
- Georgi Manukjan
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | | | - Tobias Reindl
- Department for Pediatric Hematology and Oncology, Charité-University Medicine Berlin, Berlin, Germany
| | - Gabriele Strauß
- Department for Pediatric Hematology and Oncology, Charité-University Medicine Berlin, Berlin, Germany.,Department for Pediatric Oncology and Hematology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Eva Klopocki
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Harald Schulze
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany.,Centre of Inherited Blood Cell Disorders, University Hospital Würzburg, Würzburg, Germany
| | - Oliver Andres
- Department of Pediatrics, University of Würzburg, Würzburg, Germany.,Centre of Inherited Blood Cell Disorders, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
43
|
Activation and suppression of hematopoietic integrins in hemostasis and immunity. Blood 2020; 135:7-16. [DOI: 10.1182/blood.2019003336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Nolte and Margadant review the current understanding of the activation and inactivation of integrin receptors expressed by hematopoietic cells and the role of these conformational changes in modulating platelet and leukocyte function.
Collapse
|
44
|
Lee RH, Piatt R, Dhenge A, Lozano ML, Palma-Barqueros V, Rivera J, Bergmeier W. Impaired hemostatic activity of healthy transfused platelets in inherited and acquired platelet disorders: Mechanisms and implications. Sci Transl Med 2019; 11:eaay0203. [PMID: 31826978 PMCID: PMC10824274 DOI: 10.1126/scitranslmed.aay0203] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022]
Abstract
Platelet transfusions can fail to prevent bleeding in patients with inherited platelet function disorders (IPDs), such as Glanzmann's thrombasthenia (GT; integrin αIIbβ3 dysfunction), Bernard-Soulier syndrome [BSS; glycoprotein (GP) Ib/V/IX dysfunction], and the more recently identified nonsyndromic RASGRP2 variants. Here, we used IPD mouse models and real-time imaging of hemostatic plug formation to investigate whether dysfunctional platelets impair the hemostatic function of healthy donor [wild-type (WT)] platelets. In Rasgrp2-/- mice or mice with platelet-specific deficiency in the integrin adaptor protein TALIN1 ("GT-like"), WT platelet transfusion was ineffective unless the ratio between mutant and WT platelets was ~2:1. In contrast, thrombocytopenic mice or mice lacking the extracellular domain of GPIbα ("BSS-like") required very few transfused WT platelets to normalize hemostasis. Both Rasgrp2-/- and GT-like, but not BSS-like, platelets effectively localized to the injury site. Mechanistic studies identified at least two mechanisms of interference by dysfunctional platelets in IPDs: (i) delayed adhesion of WT donor platelets due to reduced access to GPIbα ligands exposed at sites of vascular injury and (ii) impaired consolidation of the hemostatic plug. We also investigated the hemostatic activity of transfused platelets in the setting of dual antiplatelet therapy (DAPT), an acquired platelet function disorder (APD). "DAPT" platelets did not prolong the time to initial hemostasis, but plugs were unstable and frequent rebleeding was observed. Thus, we propose that the endogenous platelet count and the ratio of transfused versus endogenous platelets should be considered when treating select IPD and APD patients with platelet transfusions.
Collapse
Affiliation(s)
- Robert H Lee
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA.
- UNC Blood Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Raymond Piatt
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ankita Dhenge
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Blood Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - 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, Murcia 30003, 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, Murcia 30003, 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, Murcia 30003, Spain
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA.
- UNC Blood Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
45
|
Lunghi B, Lecchi A, Santacroce R, Scavone M, Paniccia R, Artoni A, Gachet C, Castaman G, Margaglione M, Bernardi F, Cattaneo M. Severe bleeding and absent ADP-induced platelet aggregation associated with inherited combined CalDAG-GEFI and P2Y 12 deficiencies. Haematologica 2019; 105:e361-e364. [PMID: 31649128 DOI: 10.3324/haematol.2019.232850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Barbara Lunghi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Anna Lecchi
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Rosa Santacroce
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Mariangela Scavone
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Rita Paniccia
- Center for Bleeding Disorders and Coagulation, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - Andrea Artoni
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Giancarlo Castaman
- Center for Bleeding Disorders and Coagulation, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - Maurizio Margaglione
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Marco Cattaneo
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy .,Unità di Medicina, ASST Santi Paolo e Carlo, Presidio San Paolo, Milan, Italy
| |
Collapse
|
46
|
Grosdidier C, Blanz KD, Deharo P, Bernot D, Poggi M, Bastelica D, Wolf D, Duerschmied D, Grino M, Cuisset T, Alessi M, Canault M. Platelet CD40 ligand and bleeding during P2Y12 inhibitor treatment in acute coronary syndrome. Res Pract Thromb Haemost 2019; 3:684-694. [PMID: 31624788 PMCID: PMC6781928 DOI: 10.1002/rth2.12244] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 06/20/2019] [Indexed: 01/06/2023] Open
Abstract
Antiplatelet therapy through inhibition of the adenosine diphosphate (ADP)/P2Y12 pathway is commonly used in the treatment of acute coronary syndrome (ACS). Although efficient in preventing platelet activation and thrombus formation, it increases the risk of bleeding complications. In patients with ACS receiving platelet aggregation inhibitors, that is, P2Y12 blockers (n = 923), we investigated the relationship between plasma and platelet-associated CD40L levels and bleeding events (n = 71). Treatment with P2Y12 inhibitors in patients with ACS did not affect plasma-soluble CD40L levels, but decreased platelet CD40L surface expression (pCD40L) and platelet-released CD40L (rCD40L) levels in response to stimulation as compared to healthy controls. In vitro inhibition of the ADP pathway in healthy control platelets reduced both pCD40L and rCD40L levels. In a multivariable analysis, the reduced pCD40L level observed in ACS patients was significantly associated with the risk of bleeding occurrence (adjusted odds ratio = 0.15; 95% confidence interval = 0.034-0.67). P2Y12 inhibitor-treated (ticagrelor) mice exhibited a 2.5-fold increase in tail bleeding duration compared with controls. A significant reduction in bleeding duration was observed on CD40L+/+ but not CD40L-/- platelet infusion. In addition, CD40L blockade in P2Y12 inhibitor-treated blood samples from a healthy human reduced thrombus growth over immobilized collagen under arterial flow. In conclusion, measurement of pCD40L may offer a novel approach to assessing bleeding risk in patients with ACS who are being treated with P2Y12 inhibitors.
Collapse
Affiliation(s)
- Charlotte Grosdidier
- Aix Marseille UniversityINSERM, INRA, C2VNMarseilleFrance
- Hematology LaboratoryAPHM, CHU TimoneMarseilleFrance
| | - Kelly D. Blanz
- Spemann Graduate School of Biology and MedicineUniversity of FreiburgFreiburg im BreisgauGermany
| | - Pierre Deharo
- Department of CardiologyAPHM, CHU TimoneMarseilleFrance
| | - Denis Bernot
- Hematology LaboratoryAPHM, CHU TimoneMarseilleFrance
| | - Marjorie Poggi
- Aix Marseille UniversityINSERM, INRA, C2VNMarseilleFrance
| | | | - Dennis Wolf
- Department of Cardiology and Angiology IHeart Center Freiburg UniversityFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburg im BreisgauGermany
| | - Daniel Duerschmied
- Department of Cardiology and Angiology IHeart Center Freiburg UniversityFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburg im BreisgauGermany
| | - Michel Grino
- Aix Marseille UniversityINSERM, INRA, C2VNMarseilleFrance
| | | | - Marie‐Christine Alessi
- Aix Marseille UniversityINSERM, INRA, C2VNMarseilleFrance
- Hematology LaboratoryAPHM, CHU TimoneMarseilleFrance
| | | |
Collapse
|
47
|
Mezzapesa A, Bastelica D, Crescence L, Poggi M, Grino M, Peiretti F, Panicot-Dubois L, Dupont A, Valero R, Maraninchi M, Bordet JC, Alessi MC, Dubois C, Canault M. Increased levels of the megakaryocyte and platelet expressed cysteine proteases stefin A and cystatin A prevent thrombosis. Sci Rep 2019; 9:9631. [PMID: 31270351 PMCID: PMC6610149 DOI: 10.1038/s41598-019-45805-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 06/07/2019] [Indexed: 11/09/2022] Open
Abstract
Increased platelet activity occurs in type 2 diabetes mellitus (T2DM) and such platelet dysregulation likely originates from altered megakaryopoiesis. We initiated identification of dysregulated pathways in megakaryocytes in the setting of T2DM. We evaluated through transcriptomic analysis, differential gene expressions in megakaryocytes from leptin receptor-deficient mice (db/db), exhibiting features of human T2DM, and control mice (db/+). Functional gene analysis revealed an upregulation of transcripts related to calcium signaling, coagulation cascade and platelet receptors in diabetic mouse megakaryocytes. We also evidenced an upregulation (7- to 9.7-fold) of genes encoding stefin A (StfA), the human ortholog of Cystatin A (CSTA), inhibitor of cathepsin B, H and L. StfA/CSTA was present in megakaryocytes and platelets and its expression increased during obesity and diabetes in rats and humans. StfA/CSTA was primarily localized at platelet membranes and granules and was released upon agonist stimulation and clot formation through a metalloprotease-dependent mechanism. StfA/CSTA did not affect platelet aggregation, but reduced platelet accumulation on immobilized collagen from flowing whole blood (1200 s-1). In-vivo, upon laser-induced vascular injury, platelet recruitment and thrombus formation were markedly reduced in StfA1-overexpressing mice without affecting bleeding time. The presence of CA-074Me, a cathepsin B specific inhibitor significantly reduced thrombus formation in-vitro and in-vivo in human and mouse, respectively. Our study identifies StfA/CSTA as a key contributor of platelet-dependent thrombus formation in both rodents and humans.
Collapse
Affiliation(s)
- Anna Mezzapesa
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| | | | - Lydie Crescence
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| | - Marjorie Poggi
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| | - Michel Grino
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| | - Franck Peiretti
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| | | | - Annabelle Dupont
- CHU Lille, Université de Lille, Inserm U1011 - EGID, Institut Pasteur de Lille, Lille, France
| | - René Valero
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| | - Marie Maraninchi
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| | - Jean-Claude Bordet
- Laboratoire d'Hémostase, Centre de Biologie Est, Hospices Civils de Lyon, Bron, France.,Laboratoire de Recherche sur l'Hémophilie, UCBL1, Lyon, France
| | | | | | - Matthias Canault
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, 13385, France
| |
Collapse
|
48
|
Palma-Barqueros V, Ruiz-Pividal J, Bohdan N, Vicente V, Bastida JM, Lozano M, Rivera J. RASGRP2 gene variations associated with platelet dysfunction and bleeding. Platelets 2019; 30:535-539. [PMID: 30849270 DOI: 10.1080/09537104.2019.1585528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This manuscript reviews pathogenic variants in RASGRP2, which are the cause of a relatively new autosomal recessive and nonsyndromic inherited platelet function disorder, referred to as platelet-type bleeding disorder-18 (BDPLT18)(OMIM:615888). To date, 18 unrelated BDPLT18 pedigrees have been reported, harboring 19 different homozygous or compound heterozygous RASGRP2 variants. Patients with this disease present with lifelong moderate to severe bleeding, with epistaxis as the most common and relevant bleeding symptom. Biologically, they exhibit normal platelet count and morphology, reduced aggregation responses to ADP, epinephrine and low-dose collagen, and impaired αIIbβ3 integrin activation (fibrinogen or PAC-1 binding) in response to most agonists except PMA. Diagnosis is confirmed by genetic analysis of RASGRP2.
Collapse
Affiliation(s)
- Verónica Palma-Barqueros
- a Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación , Universidad de Murcia , Murcia , Spain
| | - Juan Ruiz-Pividal
- a Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación , Universidad de Murcia , Murcia , Spain
| | - Natalia Bohdan
- a Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación , Universidad de Murcia , Murcia , Spain
| | - Vicente Vicente
- a Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación , Universidad de Murcia , Murcia , Spain
| | - Jose Maria Bastida
- b Department of Hematology , IBSAL-Hospital Universitario de Salamanca , Salamanca , Spain
| | - María Lozano
- a Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación , Universidad de Murcia , Murcia , Spain.,c On behalf of the "Inherited Platelet Disorders Project", Hemorrhagic Diathesis Working Group SETH
| | - José Rivera
- a Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación , Universidad de Murcia , Murcia , Spain.,c On behalf of the "Inherited Platelet Disorders Project", Hemorrhagic Diathesis Working Group SETH
| |
Collapse
|
49
|
Yun JW, Lee KO, Jung CW, Oh SY, Kim SH, Choi CW, Kim HJ. Hereditary platelet function disorder from RASGRP2 gene mutations encoding CalDAG-GEFI identified by whole-exome sequencing in a Korean woman with severe bleeding. Haematologica 2019; 104:e274-e276. [PMID: 30846498 DOI: 10.3324/haematol.2019.218487] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Jae Won Yun
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| | - Ki-O Lee
- Samsung Biomedical Research Institute, Samsung Medical Center
| | - Chul Won Jung
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Sun-Hee Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| | - Chul Won Choi
- Division of Hematology and Oncology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Hee-Jin Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul
| |
Collapse
|
50
|
|