Inherited platelet diseases with normal platelet count: phenotypes, genotypes and diagnostic strategy

Paquita Nurden, Simon Stritt, Remi Favier, Alan T Nurden, Paquita Nurden, Simon Stritt, Remi Favier, Alan T Nurden

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.

Figures

Figure 1.
Figure 1.
Integrin αIIbβ3 and Glanzmann thrombasthenia. The model illustrates the bent resting mature integrin and shows the many interactions between IIb (green) and 3 (violet). Much information has been obtained from variant forms (noted in blue) that block functional sites in the extracellular and cytoplasmic domains while allowing IIb3 expression. Other rare mutations (in red) give rise to activated integrin. Full information on mutations giving rise to Glanzmann thrombasthenia is found in Nurden & Pillois. Fg: fibrinogen; GT: Glanzmann thrombasthenia.
Figure 2.
Figure 2.
Loss of CalDAG-GEFI and kindlin-3 function abrogates αIIbβ3 activation. (A) A schematic representation showing examples of agonistic receptors and αIIbβ3 in the resting and activated forms. Intracytoplasmic signaling pathways induced by the binding of appropriate ligands lead to parallel roles of CalDAG-GEFI and kindlin-3 in promoting IIb3 activation. CalDAG-GEFI is critical for RAP1 activation both in the circulation and at sites of vascular injury. It responds to changes in cytoplasmic Ca2+ providing sensitivity and speed to the activation response; its absence leads to a Glanzmann thrombasthenia-like phenotype. Kindlin-3 binds directly to -integrin cytoplasmic tails in platelets but also in white blood cells, explaining the susceptibility to infections and immune disorders in its absence. (B) Key elements of the syndromes resulting from RASGRP2 and FERMT3 defects are summarized. Bleeding in both cases can be very severe but defects in FERMT3 are syndromic and life-threatening. TRAP: thrombin receptor agonist peptid; PAR: protease activated receptor; Cvx: convulxin; GPVI: glycoprotein VI; TXA2: thromboxane A2; PLC/: phospholipase C/γ; DAG: diacylglyerol: CalDAG-GEFI: calcium and diacylglycerol-regulated guanine nucleotide exchange factor I; RAP1: RAS-related protein 1; RD: related disease; AR: autosomal recessive; LTA: light transmission aggregometry; ADP: adenosine triphosphate; Col: collagen; Cvx: convulxin; AA: arachidonic acid; PMA: phorbol 12-myristate 13-acetate.
Figure 3.
Figure 3.
G-protein-coupled ADP receptors and signaling pathways highlighting P2RY12 variants and their different subgroups. (A) A schematic representation of ADP-induced signaling pathways. Binding of extracellular ADP to the Gq-coupled P2Y1 receptor initiates aggregation whereas the Gi-coupled P2Y12 receptor enhances and sustains the platelet response. Specific signaling pathways are involved with PLCβ and PI3K enabling both RAP1 activation and RASA3 inactivation; both are required for the formation of a stable hemostatic thrombus. The release of ADP from dense granule storage pools after protein kinase C (PKC) activation contributes to the stabilization of thrombi. (B) The schematic representation shows the structure of P2Y12; variants reported as causal for a bleeding syndrome are highlighted (red spots). (C) Summarizes the genotype/phenotype relationship when the variants are regrouped according to bleeding severity and their mode of transmission. The first group consists of patients with autosomal recessive transmission and platelets showing defects of both the binding of ADP analogs and/or receptor cycling. The p.H187Q variant located in the fifth transmembrane domain has normal receptor expression but suppressed function. Interestingly p.R256 has a side chain that inserts into a hydrophobic pocket and in docking models it potentially makes contact with the phosphate groups of ADP; p.R265W impairs receptor activation and probably alters the conformational state of the receptor. Significantly, a severe phenotype-monoallelic form has a different amino acid substitution on the same residue, p.R265P associated with an increased expression of mutated mRNA and an impaired wild-type homodimer formation, possibly accounting for the bleeding severity. The third subgroup has a mild phenotype and monoallelic expression of mutations that affect receptor function: note that p.P341A located in the PDZ intracellular domain is associated with both abnormal ligand binding and a defect of re-sensitization of the receptor after ADP agonist-induced desensitization. (D) Electron microscopy of platelet aggregates stimulated with 10 M ADP examined at the peak of aggregation. For the control the platelets are in close contact with some of the cells having lost their granule content. For our patient with the p.I240fs*29 mutation, platelets remain loosely bound and their granule contents are still present, as shown by the immunogold (black dots) localization of fibrinogen.
Figure 4.
Figure 4.
Hermansky-Pudlak syndrome. Syndromic platelet defect resulting from the abnormal biogenesis of dense granules with an autosomal recessive inheritance. (A) Summarizes the genotype/phenotype relationship of different types of Hermansky-Pudlak syndrome (HPS) regrouped according to the four multi-protein complexes involved in different steps of lysosome-related organelle (LRO) biogenesis including vesicle formation and/or trafficking: BLOC1, 2 and 3 and AP-3 with each containing several subunits. Variants in HPS 7, 8 and 9 are causal in BLOC1 but with mild clinical consequences. For BLOC2, variants in HPS 3, 5 or 6 give syndromes of moderate severity; they are rare but a variant of HPS 3 occurs in Puerto Rican communities. The most frequent causes of HPS are BLOC3 defects (variants in HPS 1 or 4). The syndromes are severe and are associated with lung fibrosis and gastro-intestinal defects; in Puerto Rico a community is affected by a common HPS1 variant. In the last group, defects of AP-3 are associated with neutropenia and infections. (B) A scheme illustrates the major steps of granule biogenesis. Dense granules may derive from the endolysosomal system, including either early or late endosomes. As secretory granules the membrane constituents including membrane transporters come from the Golgi complex. A network of interconnected and functionally distinct tubular subdomains transports their cargoes along microtubule tracks from the endosomes. Tubules ferry contents from the trans-Golgi-network and the plasma membrane to the LRO, a system that requires the coordination of numerous effectors. Components stored in dense granules or associated with their membranes are shown. (C) Platelets from a control and a patient with HPS3 examined by electron microscopy as whole mounts are shown. The dense granules observed in controls as black spots are absent from the patient’s platelet. AP-3: adaptor protein-3; BLOC: biogenesis of lysosome-related organelles complex; LRO: lysosome-related organelle; OCA: oculocutaneous albinism; MVB: multivesicular body.
Figure 5.
Figure 5.
Schema with management options for patients with inherited defects of platelet function. It is important to limit risks in daily life. It is also important to provide each patient with a card detailing the type of disorder and proposing the medication to be used according to the bleeding risk, in particular when platelet expert medical centers are unavailable to take charge of the patient’s needs. While the main therapeutic approaches for all disorders, including those patients with Glanzmann thrombasthenia (GT), RASGRP2 (CalDAG-GEFI)-related disease, and leukocyte adhesion deficiency III (LAD-III) syndrome for whom bleeding can be severe, are similar, some situations (e.g., isoantibody formation in GT) or infections in LAD-III syndrome need special care. Surgery requires a multidisciplinary consensus to evaluate the risk of bleeding, the benefit-risk ratio of prophylaxis, and to assess therapeutic efficiency. Pregnancy is always a challenge, from women with mild bleeding risks for whom minimal measures are needed to those with a severe bleeding history and for whom maximal prophylactic care is required. Here again, the management must be planned between obstetricians, anesthetists and hematologists. The contraindication of epidural or spinal anesthesia for those with mild risk remains a difficult problem, as is the choice of vaginal delivery or Cesarean section for the most severe forms. In the figure, it is noted that for mild bleeding risk, an antifibrinolytic medication or desmopressin is preferred as the first line of bleeding prevention and that platelet transfusions should be considered only when these approaches are insufficient. There is a risk of allo- or iso-immunization which, if it occurs, can be a lifelong handicap, and recombinant activated factor VII is increasingly recommended. Finally, stem cell transplantation is reserved for young patients with recurrent, severe bleeding that fails to respond to standard treatments and when the survival of the patient is at risk. Gene therapy is not yet an option for the disorders covered in this review but will probably become available in the future.

References

    1. Nurden P, Nurden AT. Congenital disorders associated with platelet dysfunctions. Thromb Haemost. 2008;99(2):253-263.
    1. Favier R, Raslova H. Progress in understanding the diagnosis and molecular genetics of macrothrombocytopenias. Br J Haematol. 2015;170(5):626-639.
    1. Heremans J, Freson K. High-throughput sequencing for diagnosing platelet disorders: lessons learned from exploring the causes of bleeding disorders. Int J Lab Hematol. 2018;40(Suppl):89-96.
    1. Lentaigne C, Freson K, Laffan MA, et al. Inherited platelet disorders: towards DNAbased diagnosis. Blood. 2016;127(33):2814-2823.
    1. Johnson B, Lowe GC, Futterer J, et al. Whole exome sequencing identifies genetic variants in inherited thrombocytopenia with secondary qualitative function defects. Haematologica. 2016;101(10):1170-1179.
    1. Simeoni I, Stephens JC, Hu F, et al. A highthroughput sequencing test for diagnosing inherited bleeding, thrombotic and platelet disorders. Blood. 2016;127(23):2791-2803.
    1. Downes K, Megy K, Duarte D, et al. Diagnostic high-throughput sequencing of 2,396 patients with bleeding, thrombotic and platelet disorders. Blood. 2019;134(23): 2082-2091.
    1. Megy K, Downes K, Simeoni I, et al. Curated disease-causing genes for bleeding, thrombotic, and platelet disorders: Communication from the SSC of the ISTH. J Thromb Haemost. 2019;17(8):1253-1260.
    1. Gunay-Aygum M, Zivony-Elboum Y, Gumruk F, et al. Gray platelet syndrome: natural history of a large patient cohort and locus assignment to chromosome 3p. Blood. 2010;116(23):4990-5001.
    1. Hollopeter G, Jantzen AM, Vincent D, et al. Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature. 2001;409(6817):202-207.
    1. Nurden A, Nurden P. Inherited thrombocytopenias: history, advances and perspectives. Haematologica. 2020;105(8):1-16.
    1. Nurden AT, Fiore M, Nurden P, Pillois X. Glanzmann thrombasthenia: a review of ITGA2B and ITGB3 defects with emphasis on variants, phenotypic variability, and mouse models. Blood. 2011;118(23):5996-6005.
    1. Coller BS, Shattil SA. The GPIIb/IIIa (integrin IIb3) odyssey: a technology-driven saga of a receptor with twists, turns, and even a bend. Blood. 2008;112(8):3011-3025.
    1. Mackman N, Bergmeier W, Stouffer GA, Weitz JI. Therapeutic strategies for thrombosis: new targets and approaches. Nat Rev Drug Disc. 2020;19(5):333-352.
    1. Botero JP, Lee K, Branchford BR, et al. ClinGen Platelet Disorder Variant Curation Expert Panel. Haematologica. 2020;105(4): 888-894.
    1. Nurden AT. Acquired Glanzmann thrombasthenia: from antibodies to anti-platelet drugs. Blood Rev. 2019;36:10-22.
    1. Nurden AT, Pillois X, Fiore M, et al. Expanding the mutation spectrum of the IIb3 integrin in Glanzmann thrombasthenia: screening of the ITGA2B and ITGB3 genes in a large international cohort. Hum Mutat. 2015;36(5):548-61.
    1. Nurden AT, Pillois X. ITGA2B and ITGB3 gene mutations associated with Glanzmann thrombasthenia. Platelets. 2018;29(1):98-101.
    1. Pillois X, Nurden AT. Linkage disequilibrium amongst ITGA2B and ITGB3 gene variants in patients with Glanzmann thrombasthenia confirms that most disease-causing mutations are recent. Br J Haematol. 2016;175(4):686-695.
    1. Takagi J, Petre BM, Walz T, Springer TA. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell. 2002;110(5): 599-611.
    1. Zhu J, Zhu J, Springer TA. Complete integrin headpiece opening in eight steps. J Cell Biol. 2013;201(7):1053-1068.
    1. Guillet B, Bayart S, Pillois X, Nurden P, Caen JP, Nurden AT. A Glanzmann thrombasthenia family associated with a TUBB1-related macrothrombasthenia. J Thromb Haemost. 2019;17(12):2211-2215.
    1. Jamasbi J, Ayabe K, Goto S, Nieswandt B, Peter K, Siess W. Platelet receptors as therapeutic targets: past, present and future. Thromb Haemost. 2017;117(7):1249-1257.
    1. Roule V, Agueznai M, Sabatier R, et al. Safety and efficacy of IIb/IIIa inhibitors in combination with highly active oral antiplatelet regimens in acute coronary syndromes: a meta-analysis of pivotal trials. Platelets. 2017;28(2):174-181.
    1. Canault M, Ghalloussi D, Grosdidier C, et al. Human CalDAG-GEFI gene (RASGRP2) mutation affects platelet function and causes severe bleeding. J Exp Med. 2014;211(7): 1349-1362.
    1. Canault M, Alessi MC. RASGRP2 structure, function and genetic variants in platelet pathophysiology. Int J Molec Sci. 2020; 21(3):1075.
    1. Stefanini L, Bergmeier W. RAP GTPAses and platelet integrin signaling. Platelets. 2019;30(1):41-47.
    1. Desai A, Bergmeier W, Canault M, et al. Phenotype analysis and clinical management in a large family with a novel truncating mutation in RASGRP2, the CalDAGGEFI encoding gene. Res Pract Thromb Haemost. 2017;1(1):128-133.
    1. Rognoni E, Ruppert R, Fassler R. The kindlin family: functions, signaling properties and implications in disease. J Cell Sci. 2016;129(1):17-27.
    1. Jurk K, Schulz AS, Kehrel BE, et al. Novelintegrin dependent platelet malfunction in siblings with leukocyte adhesion deficiency- III (LAD-III) caused by a point mutation in FERMT3. Thromb Haemost 2010;103(5): 1053-1054.
    1. Nagy M, Mastenbroek TG, Mattheij NJA, et al. Variable impairment of platelet functions in patients with severe genetically linked immune deficiencies. Haematologica. 2018;103(3):540-549.
    1. Moser M, Nieswandt B, Ussar S, Pozgajova M, Fassler R. Kindlin-3 is an essential cofactor for integrin activation and platelet aggregation. Nat Med. 2008;14(3):325-330.
    1. Kuijpers T.W, van de Vijver E, Weterman MA, et al. LAD-1/variant syndrome is caused by mutations in FERMT3. Blood. 2009;113(19):4740-4746.
    1. Malinin NL, Plow EF, Byzova TV. Kindlins in FERM adhesion. Blood. 2010;115(20):4011-4017.
    1. Weiss HJ. Scott syndrome: a disorder of platelet coagulant activity. Semin Hematol. 1994;31(4):312-319.
    1. Suzuki J, Umeda M, Sims PJ, Nagata S. Calcium-dependent phospholipid scrambling by TMEM16F. Nature. 2010;458(7325): 834-838.
    1. Toti F, Satta N, Fressinaud E, Meyer D, Freyssinet JM. Scott syndrome characterized by impaired transmembrane migration of procoagulant phosphatidylserine and hemorrhagic complications, is an inherited disorder. Blood. 1996;87(4):1409-1415.
    1. Lhermusier T, Chap H, Payrastre B. Platelet membrane phospholipid asymmetry: from the characterization of a scramblase activity to the identification of an essential protein mutated in Scott syndrome. J Thromb Haemost. 2011;9(10):1883-1891.
    1. Millington-Burgess SL, Harper MT. Gene of the issue: ANO6 and Scott syndrome. Platelets. 2020;31(7):964-967
    1. Reddy EC, Rand ML. Procoagulant phosphatidylserine- exposing platelets in vitro and in vivo. Front Cardiovasc Med. 2020;7:15.
    1. Boisseau P, Bene MC, Besnard T, et al. A new mutation of ANO6 in two familial cases of Scott syndrome. Br J Haematol. 2018;180(5):750-752.
    1. Castoldi E, Collins PW, Williamson PL, Bevers EM. Compound heterozygosity for 2 novel TMEM16F mutations in a patient with Scott syndrome. Blood. 2011;117(16): 4399-4400.
    1. Berrou E, Soukaseum C, Favier R, et al. A mutation of the human EPHB2 gene leads to a major platelet functional defect. Blood. 2018;132(19):2067-2077.
    1. Cattaneo M. The platelet P2Y12 receptor for adenosine diphosphate: congenital and drug-induced defects. Blood. 2011;117(7): 2102-2012.
    1. Cattaneo M, Lecchi A, Randi AM, McGregor JL, Mannucci PM. Identification of a new congenital detect of platelet function characterized by severe impairment of platelet responses to adenosine diphosphate. Blood. 1992;80(11):2787-2796.
    1. Nurden P, Savi P, Heilmann E, et al. An inherited bleeding disorder linked to a defective interaction between ADP and its receptor on platelets. Its influence on glycoprotein IIb– IIIa complex function. J Clin Invest. 1995;95(4):1612-1622.
    1. Mundell SJ, Rabbolini D, Gabrielli S, et al. Receptor homodimerization plays a critical role in a novel dominant negative P2RY12 variant identified in a family with severe bleeding. J Thromb Haemost. 2018;16(1):44-53.
    1. Lecchi A, Femia EA, Paoletta S, et al. Inherited dysfunctional platelet P2Y12 receptor mutations associated with bleeding disorders. Hamostaseologie. 2016;36(4):279-283.
    1. Cattaneo M, Zighetti ML, Lombardi R, et al. Molecular bases of defective signal transduction in the platelet P2Y12 receptor of a patient with congenital bleeding. Proc Natl Acad Sci U S A. 2003;100(4):1978-1983.
    1. Zhang K, Zhang J, Gao ZG, et al. Structure of the human P2Y12 receptor in complex with an antithrombotic drug. Nature. 2014;509(7498):115-118.
    1. Nisar S, Daly ME, Federici AB, et al. An intact PDZ motif is essential for correct P2Y12 purinoceptor traffic in human platelets. Blood. 2011;118(20):5641-5651.
    1. Patel YM, Lordkipanidze M, Lowe GC, et al. A novel mutation in the P2Y12 receptor and a function-reducing polymorphism in protease- activated receptor 1 in a patient with chronic bleeding. J Thromb Haemost. 2014;12(5):716-725.
    1. Daly ME, Dawood BB, Lester WA, et al. Identification and characterization of a novel P2Y12 variant in a patient diagnosed with type I von Willebrand disease in the European MCMDM-1VWD study Blood. 2009;113(17):4110-4113.
    1. Mammadova-Bach E, Ollivier V, Loyau S, et al. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood. 2015;126():683-691.
    1. Reyes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors CLEC-2 and GPVI. J Clin Invest. 2019;129(1):12-23.
    1. Moroi M, Jung SM, Okuma M, Shinmyozu K. A patient with platelets deficient in glycoprotein VI that lack both collagen-induced aggregation and adhesion. J Clin Invest. 1989;84(5):1440-1445.
    1. Nurden AT. Clinical significance of altered platelet collagen-receptor functioning in platelets with emphasis on glycoprotein VI. Blood Rev. 2019;38:100592.
    1. Jandrot-Perrus M, Hermans C, Mezzano D. Platelet glycoprotein VI genetic quantitative and qualitative defects. Platelets. 2019;30(6):708-713.
    1. Matus V, Valenzuela G, Saez CG, et al. An adenine insertion in exon 6 of human GP6 generates a truncated protein associated with a bleeding disorder in four Chilean families. J Thromb Haemost. 2013;11(9): 1751-1759.
    1. Nisar SP, Jones ML, Cunningham MR, Mumford AD, Mundell SJ. on behalf of the UK GAPP study group. Rare platelet GPCR variants: what can we learn? Br J Pharmacol. 2015;172(13):3242-3253.
    1. Habib A, FitzGerald GA, Maclouf J. Phosphorylation of the thromboxane receptor alpha, the predominant isoform expressed in human platelets. J Biol Chem. 1999;274(5):2645–265.
    1. Hirata T, Kakizuka A, Ushikubi F, Fuse I, Okuma M, Narumiya S. Arg60 to Leu mutation of the human thromboxane A2 receptor in a dominantly inherited bleeding disorder J Clin Invest. 1994;94(4):1662-1667.
    1. Fuse I, Hattori A, Mito M, et al. Pathogenetic analysis of five cases with a platelet disorder characterized by the absence of thromboxane A2 (TXA2)-induced platelet aggregation in spite of normal TXA2 binding activity. Thromb Haemost. 1996;76(6):1080-1085.
    1. Mundell SJ, Mumford A. TBXA2R gene variants associated with bleeding. Platelets. 2018;29(7):739-742.
    1. Lebozec K, Jandrot-Perrus M, Avenard G, Favre-Bulle O, Billiald P. Design, development and characterization of ACT017, a humanized Fab that blocks platelet’s glycoprotein VI function without causing bleeding risks. MAbs. 2017;9(6):945-958.
    1. Mammadova-Bach E, Gil-Pulido J, Sarukhanyan E, et al. Platelet glycoprotein VI promotes metastasis through interaction with cancer cell-derived galectin-3. Blood. 2020;135(14):1146-1160.
    1. Huizing M, Helip-Wooley A, Westbrook W, Gunay-Aygun M, Gahl WA. Disorders of lysosome-related organelle biogenesis: clinical and molecular genetics. Ann Rev Genomics Hum Genet. 2008;9:359-386.
    1. Bowman SL, Bi-Karchin J, Le L, Marks MS. The road to lysosome-related organelles: insights from Hermansky-Pudlak syndrome and other rare diseases. Traffic. 2019;20(6): 404-435.
    1. Ambrosio AL, Di Pietro SM. Storage pool diseases illuminate platelet dense granule biogenesis. Platelets. 2017;28(2):138-146.
    1. Huizing M, Malicdan MCV, Wang JA, et al. Hermansky-Pudlak syndrome: mutation update. Hum Mutat. 2020;42(3):543-580.
    1. Karampini E, Bierings R, Voorberg J. Orchestration of primary hemostasis by platelet and endothelial lysosome-related organelles. Arterioscler Thromb Vasc Biol. 2020;40(6):1441-1453.
    1. Enders A, Zieger B, Schwartz K, et al. Lethal hemophagocytic lymphohistiocytosis in Hermansky-Pudlak syndrome type II. Blood. 2006;108(1):81-87.
    1. Sharda A, Kim SH, Jasuja R, et al. Defective PDI release from platelets and endothelial cells impairs thrombus formation in Hermansky-Pudlak syndrome. Blood. 2015;125(10):1633-1642.
    1. Kaplan J, De Domenico I, McVey Ward D. Chediak-Higashi syndrome. Curr Opin Hematol. 2008;15(1):22-29.
    1. Dawood BB, Lowe GC, Lordkipanidze M, et al. Evaluation of participants with suspected heritable platelet function disorders including recommendation and validation of a streamlined agonist panel. Blood. 2012;120(5):5041-5049.
    1. Rabbolini D, Connor D, Morel-Kopp MC, et al. An integrated approach to inherited platelet disorders: results from a research collaborative, the Sydney Platelet Group. Pathology. 2020;52(2):243-255.
    1. Leo VC, Morgan NV, Bem D, et al. Use of next-generation sequencing and candidate gene analysis to identify underlying defects in patients with inherited platelet function disorders. J Thromb Haemost. 2015;13(4): 643-650.
    1. Gorski MM, Lecchi A, Femia EA, et al. Complications of whole-exome sequencing for causal gene discovery in primary platelet secretion defects. Haematologica. 2019;104(10):2084-2090.
    1. Sandrock K, Nakamura L, Vraetz T, Beutel K, Ehl S, Zieger B. Platelet secretion defect in patients with familial hemophagocytic lymphohistiocytosis type 5 (FLH5). Blood. 2010;116(26):6148-6150.
    1. Ye S, Karim ZA, Al Hawas R, Pessin JE, Filipovich AH, Whiteheart SW. Syntaxin-11 but not syntaxin-2 or syntaxin-4, is required for platelet secretion. Blood. 2012;120(12): 2484-2492.
    1. Nakamura L, Bertling A, Brodde MF, et al. First characterization of platelet secretion defect in patients with familial hemophagocytic lymphohistiocytosis type 3 (FHL-3). Blood. 2015;125(2):412-414.
    1. Fager Ferrari MF, Leinoe E, Rossing M, et al. Germline heterozygous variants in genes associated with familial hemophagocytic lymphohistiocytosis as a cause of increased bleeding. Platelets. 2017;29(1):56-64.
    1. Rao AK. Inherited defects in platelet signaling mechanisms. J Thromb Haemost. 2003;1(4):671-681.
    1. Adler DH, Cogan JD, Phillips JA III, et al. Inherited human cPLA2 deficiency is associated with impaired eicosanoid biosynthesis, small intestinal ulceration, and platelet dysfunction. J Clin Invest. 2008;118(6):2121-2131.
    1. Faioni EM, Razzari C, Zuleta A, et al. Bleeding diathesis and gastro-duodenal ulcers in inherited cytosolic phospholipase- A2 alpha deficiency. Thromb Haemost. 2014;112(6):1182-1189.
    1. Reed K, Tucker DE, Aloulou A, et al. Functional characterization of mutations in inherited human cPLA2 deficiency. Biochemistry. 2011;50(10):1731-1738.
    1. Rolf N, Knoefler R, Bugert P, et al. Clinical and laboratory phenotypes associated with the aspirin-like defect: a study in 17 unrelated families. Br J Haematol. 2009;144(3):416-424.
    1. Bastida JM, Lozano ML, Benito R, et al. Introducing high-throughput sequencing into mainstream genetic diagnosis practice in inherited platelet disorders. Haematologica. 2018;103(1):148-162.
    1. Chan MV, Hayman MA, Sivapalaratnam S, et al. Identification of a homozygous recessive variant in PTGS1 resulting in a congenital aspirin-like defect in platelet function. Haematologica. 2020. April 16. [Epub ahead of print]
    1. Geneviève D, Proulle V, Isidor B, et al. Thromboxane synthase mutations in an increased bone density disorder (Ghosal syndrome). Nat Genet. 2008;40(3):284-286.
    1. Seligsohn U. Treatment of inherited platelet disorders. Haemophilia. 2012;18 (Suppl 4:)161-165.
    1. Civaschi E, Klersy C, Melazzini F, et al. Analysis of 65 pregnacies in 24 women with five different forms of platelet function disorders. Br J Haematol. 2015;170(4):559-563.
    1. Lambert MP. Inherited platelet disorders: A modern approach to evaluation and treatment. Hematol Oncol Clin North Am. 2019;33(3):471-487.
    1. Freson K, Turro E. High-throughput sequencing approaches for diagnosing hereditary bleeding and platelet disorders. J Thromb Haemost. 2017;15(7):1262-1272.
    1. Thaventhiran JED, Lango Allen H, Burren OS, et al. Whole-genome sequencing of a sporadic primary immunodeficiency cohort. Nature. 2020;583(7814);90-95.
    1. Westbury SK, Turro E, Greene D, et al. Human Phenotype Ontology annotation and cluster analysis to unravel genetic defects in 707 cases with unexplained bleeding and platelet disorders. Genome Med. 2015;7(1):36.
    1. Greinacher A, Pecci A, Kunishima S, et al. Diagnosis of inherited platelet disorders on a blood smear: a tool to facilitate worldwide diagnosis of platelet disorders. J Thromb Haemost. 2017;15(7):1511-1521.
    1. Greinacher A, Eekels JJM. Simplifying the diagnosis of inherited platelet disorders? The new tools do not make it easier. Blood. 2019;133(23):2478-2483.
    1. Rodeghiero F, Pabinger I, Ragni M, et al. Fundamentals for a systematic approach to mild and inherited bleeding disorders: An EHA concensus report. Hemasphere. 2019;3(5):e286.

Source: PubMed

3
Abonnieren