Thromb Haemost 2009; 102(01): 90-96
DOI: 10.1160/TH09-01-0068
Platelets and Blood Cells
Schattauer GmbH

Megakaryocytes of patients with MYH9-related thrombocytopenia present an altered proplatelet formation

Alessandro Pecci
1   Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
,
Alessandro Malara
2   Department of Biochemistry, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
,
Stefania Badalucco
2   Department of Biochemistry, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
,
Valeria Bozzi
1   Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
,
Mauro Torti
2   Department of Biochemistry, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
,
Carlo L. Balduini
1   Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
,
Alessandra Balduini
2   Department of Biochemistry, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
› Author Affiliations
Financial support: This study was supported by grants from the Telethon Foundation (GGP06177) (CLB, MT), the Italian Ministero della Salute (CLB), the Italian Istituto Superiore di Sanità (CLB), the Cariplo Foudation (AB), and the Banca del Monte di Lombardia Foundation (AB).
Further Information

Publication History

Received: 30 January 2009

Accepted after minor revision: 17 April 2009

Publication Date:
24 November 2017 (online)

Summary

MYH9-related disease (MYH9-RD) is an autosomal-dominant thrombocytopenia caused by mutations of MYH9, the gene for the heavy chain of myosin-IIA. Pathogenesis of thrombocytopenia of MYH9-RD is unknown. Recent studies in mice demonstrated that myosin-IIA is an inhibitor of proplatelet formation (PPF), and suggested that it could be involved in the suppression of PPF exerted by megakaryocyte adhesion to type I collagen, which regulates the timing of platelet release within bone marrow. However, the consequences on PPF of the heterozygous mutations causative of the MYH9-RD have never been investigated. We studied the in-vitro PPF by megakaryocytes obtained from four patients carrying the p.D1424N or the p.R1933X mutations. We demonstrated that MYH9-RD megakaryocytes completely lose the physiologic suppression of proplatelet extension exerted by interaction with type I collagen, thus supporting the hypothesis that a premature platelet release within bone marrow contributes to pathogenesis of MYH9-related thrombocytopenia. Moreover, proplatelets extended by MYH9-RD megakaryocytes presented a significant defect in branching in secondary processes (p=0.001) and formed a significantly lower number of proplatelet tips (p=0.005). Since platelets are assembled at the level of proplatelet tips, this defect could further contribute to pathogenesis of thrombocytopenia of MYH9-RD patients.

 
  • References

  • 1 Seri M, Cusano R, Gangarossa S. et al. Mutations in MYH9 result in the May-Hegglin anomaly, and Fechtner and Sebastian syndromes. The May-Hegglin/Fechtner Syndrome Consortium. Nat Genet 2000; 26: 103-105.
  • 2 Heath KE, Campos-Barros A, Toren A. et al. Nonmuscle myosin heavy chain IIA mutations define a spectrum of autosomal dominant macrothrombocytopenias: May-Hegglin Anomaly and Fechtner, Sebastian, Epstein and Alport-like syndromes. Am J Hum Genet 2001; 69: 1033-1045.
  • 3 Seri M, Pecci A, Di Bari F. et al. MYH9-related disease: May Hegglin anomaly, Sebastian syndrome, Fechtner syndrome, and Epstein syndrome are not distinct entities but represent a variable expression of a single illness. Medicine (Baltimore) 2003; 82: 203-215.
  • 4 Pecci A, Panza E, Pujol-Moix N. et al. Position of nonmuscle myosin heavy chain IIA (NMMHC-IIA) mutations predicts the natural history of MYH9-related disease. Hum Mutat 2008; 29: 409-417.
  • 5 Italiano Jr JE, Lecine P, Shivdasani RA. et al. Blood platelets are assembled principally at the ends of proplatelet processes produced by differentiated megakaryocytes. J Cell Biol 1999; 147: 1299-1312.
  • 6 Avecilla ST, Hattori K, Heissig B. et al. Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med 2004; 10: 64-71.
  • 7 Patel SR, Hartwig JH, Italiano Jr JE. The biogenesis of platelets from megakaryocyte proplatelets. J Clin Invest 2005; 115: 3348-3354.
  • 8 Larson MK, Watson SP. A product of their environment: do megakaryocytes rely on extracellular cues for proplatelet formation?. Platelets 2006; 17: 435-440.
  • 9 Sabri S, Jandrot-Perrus M, Bertoglio J. et al. Differential regulation of actin stress fiber assembly and proplatelet formation by ?2?1 integrin and GPVI in human megakaryocytes. Blood 2004; 104: 3117-3125.
  • 10 Sabri S, Foudi A, Boukour S. et al. Deficiency in the Wiskott-Aldrich protein induces premature proplatelet formation and platelet production in the bone marrow compartment. Blood 2006; 108: 134-140.
  • 11 Larson MK, Watson SP. Regulation of proplatelet formation and platelet release by integrin αIIbβ3. Blood 2006; 108: 1509-1514.
  • 12 Chang Y, Auradé F, Larbret F. et al. Proplatelet formation is regulated by the Rho/ROCK pathway. Blood 2007; 109: 4229-4236.
  • 13 Chen Z, Naveiras O, Balduini A. et al. The May-Hegglin anomaly gene MYH9 is a negative regulator of platelet biogenesis modulated by the Rho-ROCK pathway. Blood 2007; 110: 171-179.
  • 14 Balduini A, Pallotta I, Malara A. et al. Adhesive receptors, extracellular proteins, and myosin IIA orchestrate proplatelet formation by human megakaryocytes. J Thromb Haemost 2008; 06: 1900-1907.
  • 15 Eckly A, Strassel C, Freund M. et al. Abnormal megakaryocyte morphology and proplatelet formation in mice with megakaryocyte-restricted MYH9 inactivation. Blood 2009; 113: 3182-3189.
  • 16 Balduini A, Malara A, Pecci A. et al. Proplatelet formation in heterozygous Bernard-Soulier syndrome type Bolzano. J Thromb Haemost 2009; 07: 478-484.
  • 17 Pecci A, Canobbio I, Balduini A. et al. Pathogenetic mechanisms of hematological abnormalities of patients with MYH9 mutations. Hum Mol Genet 2005; 14: 3169-3178.
  • 18 Williams N, Levine RF. The origin, development and regulation of megakaryocytes. Br J Haematol 1982; 52: 173-180.
  • 19 Balduini A, Pecci A, Lova P. et al. Expression, activation, and subcellular localization of the Rap1 GTPase in cord blood-derived human megakaryocytes. Exp Cell Res 2004; 300: 84-93.
  • 20 Heynen MJ, Blockmans D, Verwilghen RL. et al. Congenital macrothrombocytopenia, leukocyte inclusions, deafness and proteinuria: functional and electron microscopy observations on platelet and megakaryocytes. Br J Haematol 1988; 70: 441-448.
  • 21 Godwin HA, Ginsburg AD. May-Hegglin Anomaly: a defect of megakaryocyte fragmentation?. Br J Haematol 1974; 26: 117-128.
  • 22 Maupin P, Phillips CL, Adelstein RS. et al. Differential localization of myosin-II isozymes in human cultured cells and blood cells. J Cell Sci 1994; 107: 3077-3090.
  • 23 Kunishima S, Matsushita T, Kojima T. et al. Immunofluorescence analysis of neutrophil nonmuscle myosin heavy chain-A in MYH9 disorders: association of subcellular localization with MYH9 mutations. Lab Invest 2003; 83: 115-122.
  • 24 Kunishima S, Hamaguchi M, Saito H. Differential expression of wild-type and mutant NMMHC-IIA polypeptides in blood cells suggests cell-specific regulation mechanisms in MYH9 disorders. Blood 2008; 111: 3015-3023.
  • 25 Franke JD, Dong F, Rickoll WL. et al. Rod mutations associated with MYH9-related disorders disrupt non-muscle myosin-IIA assembly. Blood 2005; 105: 161-169.
  • 26 Rojnuckarin P, Kaushansky K. Actin reorganization and proplatelet formation in murine megakaryocytes: the role of protein kinase C alpha. Blood 2001; 97: 154-161.