Thromb Haemost 2011; 106(04): 693-704
DOI: 10.1160/TH11-02-0126
Platelets and Blood Cells
Schattauer GmbH

Mutations responsible for MYH9-related thrombocytopenia impair SDF-1-driven migration of megakaryoblastic cells

Alessandro Pecci
1   Department of Internal Medicine, 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
,
Emanuele Panza
2   Department of Gynaecological, Obstetric, and Paediatric Sciences, University of Bologna, Bologna, Italy
3   Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, USA
,
Serena Barozzi
1   Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
,
Cristian Gruppi
4   Department of Biochemistry, University of Pavia, Pavia, Italy
,
Marco Seri
2   Department of Gynaecological, Obstetric, and Paediatric Sciences, University of Bologna, Bologna, Italy
,
Carlo L. Balduini
1   Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
› Author Affiliations
Financial support: This study was supported by a grant from the Telethon Foundation (GGP06177).
Further Information

Publication History

Received: 23 February 2011

Accepted after major revision: 11 July 2011

Publication Date:
29 November 2017 (online)

Summary

MYH9-related disease (MYH9-RD) is an autosomal-dominant thrombocytopenia caused by mutations in the gene for the heavy chain of nonmuscle myosin-IIA (NMMHC-IIA). Recent in vitro studies led to the hypothesis that thrombocytopenia of MYH9-RD derives from an ectopic platelet release by megakaryocytes in the osteoblastic areas of bone marrow (BM), which are enriched in type I collagen, rather than in vascular spaces. SDF-1-driven migration of megakaryocytes within BM to reach the vascular spaces is a key mechanism for platelet biogenesis. Since myosin-IIA is implicated in polarised migration of different cell types, we hypothesised that MYH9 mutations could interfere with this mechanism. We therefore investigated the SDF-1-driven migration of a megakaryoblastic cell line, Dami cells, on type I collagen or fibrinogen by a modified transwell assay. Inhibition of myosin-IIA ATPase activity suppressed the SDF-1-driven migration of Dami cells, while over-expression of NMMHC-IIA increased the efficiency of chemotaxis, indicat- ing a role for NMMHC-IIA in this mechanism. Transfection of cells with three MYH9 mutations frequently responsible for MYH9-RD (p.R702C, p.D1424H, or p.R1933X) resulted in a defective SDF-1-driven migration with respect to the wild-type counterpart and in increased cell spreading onto collagen. Analysis of differential localisation of wild-type and mutant proteins suggested that mutant NMMHC-IIAs had an impaired cytoplasmic re-organisation in functional cytoskeletal structures after cell adhesion to collagen. These findings support the hypothesis that a defect of SDF-1-driven migration of megakaryocytes induced by MYH9 mutations contributes to ectopic platelet release in the BM osteoblastic areas, resulting in ineffective platelet production.

 
  • References

  • 1 The May-Hegglin/Fechtner Syndrome Consortium. Mutations in MYH9 result in the May Hegglin anomaly, and Fechtner and Sebastian syndromes. The May-Heggllin/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 Sellers JR. Myosins: a diverse superfamily. Biochim Biophys Acta 2000; 1496: 3-22.
  • 6 Kunishima S, Saito H. Advances in understanding of the MYH9 disorders. Curr Opin Hematol 2010; 17: 405-410.
  • 7 Balduini CL, Pecci A, Savoia A. Recent advances in the understanding and management of MYH9-related inherited thrombocytopenias. Br J Haematol. 2011 Epub ahead of print.
  • 8 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.
  • 9 Patel SR, Hartwig JH, Italiano Jr. JE. The biogenesis of platelets from megakaryocyte proplatelets. J Clin Invest 2005; 115: 3348-3354.
  • 10 Larson MK, Watson SP. A product of their environment: do megakaryocytes rely on extracellular cues for proplatelet formation?. Platelets 2006; 17: 435-440.
  • 11 Junt T, Schulze H, Chen Z. et al. Dynamic visualization of thrombopoiesis within bone marrow. Science 2007; 317: 1767-1770.
  • 12 Sabri S, Jadrot-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.
  • 13 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.
  • 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; 6: 1900-1907.
  • 15 Pallotta I, Lovett M, Rice W, Kaplan DL, Balduini A. Bone marrow osteoblastic niche: a new model to study physiological regulation of megakaryopoiesis. PLoS One 2009; 4: e8359.
  • 16 Chang Y, Auradé F, Larbret F. et al. Proplatelet formation is regulated by the Rho/ ROCK pathway. Blood 2007; 109: 4229-4236.
  • 17 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; 100: 171-179.
  • 18 Nilsson SK, Debatis ME, Dooner MS, Madri JA, Quesenberry PJ, Becker PS. Immunofluorescence characterization of key extracellular matrix proteins in murine bone marrow in situ. J Histochem Cytochem 1998; 46: 371-377.
  • 19 Reddi AH, Gay R, Gay S, Miller EJ. Transitions in collagen types during matrix-induced cartilage, bone, and bone marrow formation. Proc Natl Acad Sci USA 1977; 74: 5589-5592.
  • 20 Pecci A, Malara A, Badalucco S. et al. Megakaryocytes of patients with MYH9-related thrombocytopenia present an altered proplatelet formation. Thromb Haemost 2009; 102: 90-96.
  • 21 Dhanjal TS, Pendaries C, Ross EA. et al. A novel role for PECAM-1 in megakaryocytokinesis and recovery of platelet counts in thrombocytopenic mice. Blood 2007; 109: 4237-4244.
  • 22 Mathur A, Hong Y, Martin J, Erusalimsky J. Megakaryocytic differentiation is accompanied by a reduction in cell migratory potential. Br J Haematol 2001; 112: 459-465.
  • 23 Rivière C, Subra F, Cohen-Solal K. et al. Phenotypic and functional evidence for the expression of CXCR4 receptor during megakaryocytopoiesis. Blood 1999; 93: 1511-1523.
  • 24 Jacobelli J, Chmura SA, Buxton DB, Davis MM, Kummel MF. A single class II myosin modulates T cell motility and stopping, but not synapse formation. Nat Immunol 2004; 5: 531-538.
  • 25 Huang Y, Arora P, McCulloch CA, Vogel WF. The collagen receptor DDR1 regulates cell spreading and motility by associating with myosin IIA. J Cell Sci 2009; 122: 1637-1646.
  • 26 Even-Ram S, Doyle AD, Conti MA, Adelstein RS, Yamada KM. Myosin IIA regulates cell motility and actomyosin-microtubule crosstalk. Nat Cell Biol 2007; 9: 299-309.
  • 27 Sandquist JC, Swenson KI, Demali KA, Burridge K, Means AR. Rho kinase differentially regulates phophorilation of nonmuscle myosin isoforms A and B during cell rounding and migration. J Biol Chem 2006; 281: 35873-35883.
  • 28 Wang A, Ma X, Conti MA, Liu C, Kawamoto S, Adelstein RS. Nonmuscle myosin II isoform and domain specificity during early mouse development. Proc Natl Acad Sci USA 2010; 107: 14645-14650.
  • 29 Kolega J. The role of myosin II motor activity in distributing myosin asymmetrically and coupling protrusive activity to cell translocation. Mol Biol Cell 2006; 17: 4435-4445.
  • 30 Tenni R, Sonaggere M, Viola M. et al. Self-aggregation of fibrillar collagens I and II involves lysine side chains. Micron 2006; 37: 640-647.
  • 31 Greenberg SM, Rosenthal DS, Greeley TA, Tantravahi R, Handin RI. Characterization of a new megakaryocytic cell line: the Dami cell. Blood 1988; 72: 1968-1977.
  • 32 Zutter MM, Painter AA, Staatz WD, Tsung YL. Regulation of alpha 2 integrin gene expression in cells with megakaryocytic features: a common theme of three necessary elements. Blood 1995; 86: 3006-3014.
  • 33 Wang JF, Liu ZY, Groopman JE. The alpha-chemokine receptor CXCR4 is expressed on the megakaryocytic lineage from progenitor to platelets and modulates migration and adhesion. Blood 1998; 92: 756-764.
  • 34 Shi Q, Wilcox DA, Morateck PA. et al. Targeting platelet GPIbalpha transgene expression to human megakaryocytes and forming a complete complex with endogenous GPIbbeta and GPIX. J Thromb Haemost 2004; 2: 1989-1997.
  • 35 Lev PR, Goette NP, Glembotsky AC. et al. Production of functional platelet-like particles by the megakaryoblastic DAMI cell line provides a model for platelet biogenesis. Platelets 2011; 22: 26-36.
  • 36 Pippucci T, Savoia A, Perrotta S. et al. Mutations in the 5' UTR of ANKRD26, the Ankirin Repeat Domain 26 Gene, Cause an Autosomal-Dominant Form of Inherited Thrombocytopenia, THC2. Am J Hum Genet 2011; 88: 115-120.
  • 37 MacLeod RA, Dirks WG, Reid YA. et al. Identity of original and late passage Dami megakaryocytes with HEL erythroleukemia cells shown by combined cytogenetics and DNA fingerprinting. Leukemia 1997; 11: 2032-2038.
  • 38 Panza E, Marini M, Pecci A. et al. Transfection of the mutant MYH9 cDNA reproduces the most typical cellular phenotype of MYH9-related disease in different cell lines. Pathogenetics 2008; 1: 5.
  • 39 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.
  • 40 Larson MK, Watson SP. Regulation of proplatelet formation and platelet release by integrin alpha IIb beta3. Blood 2006; 108: 1509-1514.
  • 41 Maupin P, Phillips CL, Adelstein RS, Pollard TD. Differential localization of myosin-II isozymes in human cultured cells and blood cells. J Cell Sci 1994; 107: 3077-3090.
  • 42 Straight AF, Cheung A, Limouze J. et al. Dissecting temporal and spatial control of cytokinesis with a myosin II Inhibitor. Science 2003; 299: 1743-1747.
  • 43 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.
  • 44 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.
  • 45 Morin NA, Oakes PW, Hyun YM. et al. Nonmuscle myosin heavy chain IIA mediates integrin LFA-1 de-adhesion during T lymphocyte migration. J Exp Med 2008; 205: 195-205.
  • 46 Franke JD, Dong F, Rickoll WL. et al. Rod mutations associated with MYH9-related disorders disrupt nonmuscle myosin-IIA assembly. Blood 2005; 105: 161-169.