Thromb Haemost 1999; 82(02): 481-485
DOI: 10.1055/s-0037-1615868
Research Article
Schattauer GmbH

Insights and Questions Arising from Studies of a Mouse Model of Glanzmann Thrombasthenia

Richard O. Hynes
1   Howard Hughes Medical Institute, Center for Cancer Research and Department of Biology, Massachusettes Institute of Technology, Cambridge, MA, USA
,
Kairbaan M. Hodivala-Dilke
1   Howard Hughes Medical Institute, Center for Cancer Research and Department of Biology, Massachusettes Institute of Technology, Cambridge, MA, USA
› Author Affiliations
Further Information

Publication History

Publication Date:
09 December 2017 (online)

Introduction

Studies of patients suffering from Glanzmann thrombasthenia (GT) have provided many insights into the role of platelet adhesion in hemostasis and thrombosis,1-3 but they also have raised some unanswered questions. This genetically inherited bleeding disorder arises from mutations in the genes for either subunit of the major platelet integrin, αIIbβ3, or glycoprotein IIb/IIIa (CD41/CD61). These mutations lead to reduced or absent expression of this integrin on platelet surfaces or, in a few cases, to normal levels of expression of functionally defective forms of αIIbβ3.1-6 Normally, αIIbβ3 functions as one of the major adhesion receptors on platelets. It also serves as a receptor for fibrinogen during platelet aggregation and as a receptor for von Willebrand factor (vWF) during platelet adhesion to a damaged vessel wall.7,8 In addition, αIIbβ3 can also act as a fibronectin receptor9,10 or a thrombospondin receptor,11 though the exact importance of these interactions in platelet adhesion and/or aggregation remains unclear.

GT patients lacking αIIbβ3, or who express reduced levels or defective forms, have a number of characteristic defects in platelet function. These include defective platelet aggregation in response to a variety of agonists, failure to retract a fibrin clot, and reduced or absent intracellular fibrinogen.1-4 Patients show prolonged bleeding times and spontaneous mucocutaneous and gastrointestinal hemorrhage. Occasional bleeding crises can usually be managed by platelet transfusions, so the mortality is relatively low. Both male and female patients are fertile, and pregnancies can be carried to term, although postpartum hemorrhage and menorrhagia can occur.

The features of GT described above are common among patients with mutations in the αIIβ gene or the β3 gene. At one level, that is to be expected, since absence of either subunit destroys the functional heterodimeric integrin, and, indeed, compromises the export of the partner subunit to the surface. As a result, the partner subunit is degraded intracellularly.1-4 However, a complication arises in that the β3 subunit can also partner with another subunit, αv. The αvβ3 receptor is normally present at low levels (50 to 100 copies per platelet compared with 40,000 to 80,000 copies of αIIbβ3 per platelet), and its levels reportedly do not change on GT platelets lacking αIIb. Naturally, it is absent from β3-null platelets, but no defects attributable to the additional absence of αvβ3 have been described.

Given the low levels of αvβ3 on platelets, one might not expect a serious hemostatic consequence associated with its loss. However, αvβ3 is also expressed by many other cell types, in contrast with αIIbβ3, which is platelet-specific. Most notably, αvβ3 is expressed by endothelial cells during angiogenesis and has been implicated functionally in angiogenesis.12,13 It is also apparently an integrin of major importance on osteoclasts, where it is believed to play a role in bone remodeling.14,15 The αvβ3 receptor is also expressed by neutrophils, monocytes, macrophages, some T cells, melanomas, glioblastomas, and neural crest cells, among others.16-20 Given this widespread expression and the data implicating αvβ3 in important developmental and physiological processes, one would have predicted that mutations in the β3 gene would have much more severe consequences than the platelet-specific defects associated with mutations in the αIIb gene. Nonetheless, the clinical data on GT patients have failed to reveal clear distinctions between those defective in αIIb and those defective in β3.1-4 At least some of the β3 mutations are nulls, so it is unclear whether such GT patients might exhibit additional defects, for example in angiogenesis, wound repair, or bone remodeling. A few studies have been performed involving limited numbers of GT patients, but there are obvious limitations to the extent of experimentation that can be performed on these patients. Simply put, there are few GT patients, and ethical considerations severely limit possible interventions.

 
  • References

  • 1 George JN, Caen JP, Nurden AT. Glanzmann’s thrombasthenia: the spectrum of clinical disease. Blood 1990; 75: 1383-1395.
  • 2 Bray PF. Inherited diseases of platelet glycoproteins: considerations for rapid molecular characterization. Thromb Haemost 1994; 72: 492-502.
  • 3 Coller BS. Hereditary qualitative platelet disorders. In: Beutler E, Lichtman MA, Coller BS. 5th Ed. William’s Hematology. NY: McGraw Hill; 1995
  • 4 Kato A. The biologic and clinical spectrum of Glanzmann’s Thrombasthenia: implications of integrin αIIbβ3 for its pathogenesis. Crit Rev Oncol Hematol 1997; 26: 1-23.
  • 5 Loftus JC, O’Toole T., Plow EF, Glass A, Frelinger III AL, Ginsberg MH. A β3 integrin mutation abolishes ligand binding and alters divalent cation-dependent conformation. Science 1990; 249: 915-918.
  • 6 Bajt ML, Ginsberg MG, Frelinger III AL, Berndt MC, Loftus JC. A spontaneous mutation of integrin αIIbβ3 (platelet glycoprotein IIb-IIIa) helps define a ligand binding site. 1992 267. 3789 3794.
  • 7 Savage B, Saldivar E, Ruggeri ZM. Initiation of platelet adhesion by arrest onto fibrinogen or translocation on von Willebrand factor. Cell 1996; 84: 289-297.
  • 8 Savage B, Almus-Jacobs F, Ruggeri ZM. Specific synergy of multiple substrate-receptor interactions in platelet thrombus formation under flow. Cell 1998; 94: 657-666.
  • 9 Ginsberg M, Pierschbacher MD, Ruoslahti E, Marguerie G, Plow E. Inhibition of fibronectin binding to platelets by proteolytic fragments and synthetic peptides, which support fibroblast adhesion. J Biol Chem 1985; 260: 3931-3936.
  • 10 Gardner JM, Hynes RO. Interaction of fibronectin with its receptor on platelets. Cell 1985; 42: 439-448.
  • 11 Lawler J, Weinstein R, Hynes R O. (1988). Cell attachment to thrombospondin: the role of ARG-GLY-ASP, calcium, and integrin receptors. J Cell Biol 1988; 107: 2351-61.
  • 12 Varner JA, Brooks PC, Cheresh DA. The integrin alpha V beta 3: angiogenesis and apoptosis. Cell Adhes Commun 1995; 3: 367-374.
  • 13 Strömblad S, Cheresh DA. (1996). Cell adhesion and angiogenesis. Trends in Cell Biol 1996; 6: 462-468.
  • 14 Engleman VW, Nickols GA, Ross FP, Horton MA, Griggs DW, Settle SL, Ruminski PG, Teitelbaum SL. A peptidomimetic antagonist of the alpha(v)beta3 integrin inhibits bone resorption in vitro and prevents osteoporosis in vivo . J Clin Invest 1997; 99: 2284-2292.
  • 15 Crippes BA, Engleman VW, Settle SL, Delarco J, Ornberg RL, Helfrich MH, Horton MA, Nickols GA. (1996) Antibody to β3 integrin inhibits osteoclast-mediated bone resorption in the thyroparathyroidectomized rat. Endocrinology 1996; 137: 918-924.
  • 16 Byzova TV, Rabbani R, D’Souza SE, Plow EF. Role of integrin αvβ3 in vascular biology. Thromb. Haemost 1998; 80: 726-734.
  • 17 Felding-Habermann B, Cheresh DA. Vitronectin and its receptors. Curr Opin Cell Biol 1993; 5: 864-8.
  • 18 Albelda SM, Mette SA, Elder DE, Stewart R, Damjanovich L, Herlyn M, Buck CA. Integrin distribution in malignant melanoma: association of the beta 3 subunit with tumor progression. Cancer Res 1990; 20: 6757-6764
  • 19 Delannet M, Martin F, Bossy B, Cheresh DA, Reichardt LF, Duband JL. Specific roles of the αvβ1, αvβ3, and αvβ5 integrins in avian neural crest cell adhesion and migration on vitronectin. Development 1994; 120: 2687-2702.
  • 20 Bader BL, Rayburn H, Crowley D, Hynes RO. Extensive vasculogenesis, angiogenesis and organogenesis precede lethality in mice lacking all αv integrins. Cell 1998; 95: 507-519
  • 21 Hodivala-Dilke KM, McHugh K, Tsakiris DA, Rayburn H, Crowley D, Ullman-Culleré M, Ross FP, Coller BS, Teitelbaum S, Hynes RO. (1999). β3-integrin-deficient mice: a model for Glanzmann thrombasthenia showing placental defects and reduced survival. J Clin Invest 1999; 103: 229-238.
  • 22 Zhou Y, Damsky CH, Fisher SJ. Preeclampsia is associated with failure of human cytotrophoblasts to mimic a vascular adhesion phenotype. One cause of defective endovascular invasion in this syndrome. J Clin Invest 1997; 99: 2152-64.
  • 23 McHugh KP, Hodivala-Dilke K, Cheng S, Zheng MH, Avioli LV, Ross FP, Hynes RO, Teitelbaum SL. Osteoclasts and osteoblasts in the β3 integrin knockout mouse are dysfunctional. [SOURCE]. 1998
  • 24 Noden DM. Embryonic origins and assembly of blood vessels. Am Rev Respir Dis 1989; 140: 1097-1103.
  • 25 Risau W, Flamme I. Vasculogenesis. Annu Rev Cell Dev Biol. 1995; 11: 73-91.
  • 26 Risau W. Mechanisms of angiogenesis. Nature 1997; 386: 671-674.
  • 27 Folkman J, D’Amore PA. Blood vessel formation: what is its molecular basis?. Cell 1996; 87: 1153-1155.
  • 28 Hanahan D. Signaling vascular morphogenesis and maintenance. Science 1997; 277: 48-50.
  • 29 Beck Jr L, D’Amore PA. Vascular development: cellular and molecular regulation. Faseb J 1997; 11: 365-373.
  • 30 Korpelainen EI, Alitalo K. Signaling angiogenesis and lymphangiogenesis. Curr Opin Cell Biol 1998; 10: 159-164.
  • 31 Yancopoulos GD, Klagsbrun M, Folkman J. Vasculogenesis, angiogenesis, and growth factors: ephrins enter the fray at the border. Cell 1998; 93: 661-664.
  • 32 Patan S., Haenni B, Burri P.H.. Implementation of intussusceptive microvascular growth in the chicken chorioallantoic membrane (CAM): 1. Pillar formation by folding of the capillary wall. Microvasc Res 1996; a; 51: 80-98.
  • 33 Patan S, Haenni B, Burri PH. Implementation of intussusceptive microvascular growth in the chicken chorioallantoic membrane (CAM). 2. Pillar formation by capillary fusion. Microvasc Res 1997; 53: 33-52.
  • 34 Brooks PC, Stromblad S, Klemke R, Visscher D, Sarkar FH, Cheresh DA. Antiintegrin alpha v beta 3 blocks human breast cancer growth and angiogenesis in human skin. J Clin Invest 1995; 96: 1815-1822.
  • 35 Brooks PC, Clark RA, Cheresh DA. (1994a). Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 1994; a; 264: 569-571.
  • 36 Brooks PC, Montgomery AM, Rosenfeld M, Reisfeld RA, Hu T, Klier G, Cheresh DA. Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 1994; b; 79: 1157-1164.
  • 37 Brooks PC, Stromblad S, Klemke R, Visscher D, Sarkar FH., Cheresh DA. Antiintegrin alpha v beta 3 blocks human breast cancer growth and angiogenesis in human skin. J Clin Invest 1995; 96: 1815-1822.
  • 38 Brooks PC, Silletti S, von Schalscha TL, Friedlander M, Cheresh DA. Disruption of angiogenesis by PEX, a noncatalytic metalloproteinase fragment with integrin binding activity. Cell 1998; 92: 391-400.
  • 39 Friedlander M, Brooks PC, Shaffer RW, Kincaid CM, Varner JA, Cheresh DA. Definition of two angiogenic pathways by distinct alpha v integrins. Science 1995; 270: 1500-1502.
  • 40 Friedlander M, Theesfeld CL, Sugita M, Fruttiger M, Thomas MA, Chang S, Cheresh DA. (1996). Involvement of integrins alpha v beta 3 and alpha v beta 5 in ocular neovascular diseases. Proc Natl Acad Sci U S A 1996; 93: 9764-9.
  • 41 Hammes HP, Brownlee M, Jonczyk A, Sutter A, Preissner KT. Subcutaneous injection of a cyclic peptide antagonist of vitronectin receptor-type integrins inhibits retinal neovascularization. Nat Med 1996; 2: 529-33.
  • 42 Diaz-Gonzalez F, Forsyth J, Steiner B, Ginsberg MH. Transdominant inhibition of integrin function. Mol Biol Cell 1996; 7: 1939-1951.
  • 43 Yang JT, Rayburn H, Hynes RO. Embryonic mesodermal defects in alpha 5 integrin-deficient mice. Development 1993; 119: 1093-1105.
  • 44 George EL, Georges-Labouesse EN, Patel-King RS, Rayburn H, Hynes RO. Defects in mesoderm, neural tube and vascular development in mouse embryos lacking fibronectin. Development 1993; 119: 1079-91.
  • 45 George EL, Baldwin HS, Hynes RO. Fibronectins are essential for heart and blood vessel morphogenesis but are dispensable for initial specification of precursor cells. Blood 1997; 90: 3073-3081.
  • 46 Senger DR, Claffey KP, Benes JE, Perruzzi CA, Sergiou AP, Detmar M. Angiogenesis promoted by vascular endothelial growth factor: regulation through alpha1beta1 and alpha2beta1 integrins. Proc Natl Acad Sci U S A 1997; 94: 13612-13617.
  • 47 Chin L, Pomerantz J, Polsky D, Jacobson M, Cohen C, Cordon-Cardo C, Horner II JW, DePinho RA. Cooperative effects of INK4a and ras in melanoma susceptibility in vivo . Genes and Dev 1997; 11: 2822-2834.
  • 48 Bergers G, Hanahan D, Coussens LM. Angiogenesis and apoptosis are cellular parameters of neoplastic progression in transgenic mouse models of tumorigenesis. Int J Dev Biol 1998; 42: 995-1002.
  • 49 Wilcox DA, Olsen JC, Ishizawa L, Bray PF, French DL, Steeber DA, Bell WR, Griffith M, White III GC. 1999 Megakaryocyte-targeted synthesis of the integrin-β3-subunit results in the phenotypic correction of Glanzmann’s thrombasthenia. (submitted).
  • 50 Wilcox DA, Hodivala-Dilke KM, Hynes RO, White II GC. Of mice and men: detection of a functional murine αIIb-human β3 heterodimer complex on the surface of megakaryocytes derived from retrovirus-transduced bone marrow cells from β3-knockout mice. Blood 1998; 92: 2885a.
  • 51 Coburn J, Leong JM, Erban JK. (1993). Integrin αIIbβ3 mediates binding of the Lyme disease agent Borrelia burgdorferi to human platelets. Proc Natl Acad Sci U S A 1993; 90: 7059-7063.
  • 52 Coburn J, Barthold SW, Leong JM. Diverse Lyme disease spirochetes bind integrin αIIbβ3 on human platelets. Infect Immun. 1994; 62: 5559-5567.
  • 53 Coburn J, Magoun L, Bodary SC, Leong JM. Integrins αvβ3 and α5βq mediate attachment of Lyme disease spirochetes to human cells. Infect Immun 1998; 66: 1946-1952.
  • 54 Wickham TJ, Mathias P, Cheresh DA, Nemerow GR. Integrins αvβ3 and αvβ5 promote adenovirus internalization but not virus attachment. Cell 1993; 73: 309-319.
  • 55 Hebbel RP. Adhesive interactions of sickle erythrocytes with endothelium. J Clin Invest 1997; 99: 2561-2564.
  • 56 Panda D, Kundu GC, Lee BI, Peri A, Fohl D, Chackalaparampil I, Mukherjee BB, Li XD, Mukherjee DC, Seides S, Rosenberg J, Stark K, Mjkherjee AB. Potential roles of osteopontin and αvβ3 integrin in the development of coronary artery restenosis after angioplasty. Proc Natl Acad Sci U S A 1997; 94: 9308-9313.
  • 57 Weerasinghe D, McHugh KP, Ross FP, Brown EJ, Gisler RH, Imhof BA. A role for the αvβ3 integrin in the transmigration of monocytes. J Cell Biol 1998; 142: 595-607.
  • 58 Varner JA, Cheresh DA. Integrins and cancer. Curr Opin Cell Biol 1996; 8: 724-30.
  • 59 Giancotti FG, Mainiero F. Integrin-mediated adhesion and signaling in tumorigenesis. Biochim Biophys Acta 1994; 1198: 47-64.