Thromb Haemost 2016; 115(02): 250-256
DOI: 10.1160/th15-07-0570
Review Article
Schattauer GmbH

Translational approaches to functional platelet production ex vivo

Alessandra Balduini
1   Department of Molecular Medicine, University of Pavia, Pavia, Italy
2   Biotechnology Research Laboratories, IRCCS San Matteo Foundation, Pavia, Italy
3   Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
,
Christian A. Di Buduo
1   Department of Molecular Medicine, University of Pavia, Pavia, Italy
2   Biotechnology Research Laboratories, IRCCS San Matteo Foundation, Pavia, Italy
,
David L. Kaplan
3   Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
› Author Affiliations
Financial support: This paper was supported by Cariplo Foundation (2010–0807 and and 2013–0717), Italian Ministry of Health (grant RF-2009–1550218) and US National Institutes of Health (grant EB016041–01). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Further Information

Publication History

Received: 16 July 2015

Accepted after minor revision: 11 August 2015

Publication Date:
21 November 2017 (online)

Summary

Platelets, which are released by megakaryocytes, play key roles in haemostasis, angiogenesis, immunity, tissue regeneration and wound healing. The scarcity of clinical cures for life threatening platelet diseases is in a large part due to limited insight into the mechanisms that control the developmental process of megakaryocytes and the mechanisms that govern the production of platelets within the bone marrow. To overcome these limitations, functional human tissue models have been developed and studied to extrapolate ex vivo outcomes for new insight on bone marrow functions in vivo. There are many challenges that these models must overcome, from faithfully mimicking the physiological composition and functions of bone marrow, to the collection of the platelets generated and validation of their viability and function for human use. The overall goal is to identify innovative instruments to study mechanisms of platelet release, diseases related to platelet production and new therapeutic targets starting from human progenitor cells.

 
  • References

  • 1 Michel SG, Madariaga ML, Villani V. et al. Current progress in xenotransplantation and organ bioengineering. Int J Surg 2015; 13: 239-244.
  • 2 Benam KH, Dauth S, Hassell B. et al. Engineered in vitro disease models. Annu Rev Pathol 2015; 10: 195-262.
  • 3 Esch EW, Bahinski A, Huh D. Organs-on-chips at the frontiers of drug discovery. Nat Rev Drug Discov 2015; 14: 248-260.
  • 4 Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature 2014; 505: 327-334.
  • 5 Geddis AE, Kaushansky K. Immunology. The root of platelet production. Science 2007; 317: 1689-1691.
  • 6 Owens M, Holme S, Heaton A. et al. Post-transfusion recovery of function of 5-day stored platelet concentrates. Br J Haematol 1992; 80: 539-544.
  • 7 Cardigan R, Williamson LM. The quality of platelets after storage for 7 days. Transfus Med 2003; 13: 173-187.
  • 8 Anitua E, Andia I, Ardanza B. et al. Autologous platelets as a source of proteins for healing and tissue regeneration. Thromb Haemost 2004; 91: 4-15.
  • 9 Eppley BL, Woodell JE, Higgins J. Platelet quantification and growth factor analysis from platelet-rich plasma: implications for wound healing. Plast Re-constr Surg 2004; 114: 1502-1508.
  • 10 Anitua E, Sánchez M, Nurden AT. et al. New insights into and novel applications for platelet-rich fibrin therapies. Trends Biotechnol 2006; 24: 227-234.
  • 11 Andia I, Maffulli N. Platelet-rich plasma for managing pain and inflammation in osteoarthritis. Nat Rev Rheumatol 2013; 09: 721-730.
  • 12 Williamson LM, Devine DV. Challenges in the management of the blood supply. Lancet 2013; 381: 1866-1875.
  • 13 Estcourt LJ. Why has demand for platelet components increased? A review. Transfus Med 2014; 24: 260-268.
  • 14 Deutsch VR, Tomer A. Advances in megakaryocytopoiesis and thrombopoiesis: from bench to bedside. Br J Haematol 2013; 161: 778-793.
  • 15 Basciano PA, Bussel JB. Thrombopoietin-receptor agonists. Curr Opin Haema-tol 2012; 19: 392-398.
  • 16 Oshima Y, Yuji K, Tanimoto T. et al. Association between acute myelogenous leukemia and thrombopoietin receptor agonists in patients with immune thrombocytopenia. Intern Med 2013; 52: 2193-2201.
  • 17 Erickson-Miller CL, Delorme E, Tian SS. et al. Preclinical activity of eltrombo-pag (SB-497115), an oral, nonpeptide thrombopoietin receptor agonist. Stem Cells 2009; 27: 424-430.
  • 18 Ellis SL, Grassinger J, Jones A. et al. The relationship between bone, haemo-poietic stem cells, and vasculature. Blood 2011; 118: 1516-1524.
  • 19 Kaushansky K. The molecular mechanisms that control thrombopoiesis. J Clin Invest 2005; 115: 3339-3347.
  • 20 Pang L, Weiss MJ, Poncz M. Megakaryocyte biology and related disorders. J Clin Invest 2005; 115: 3332-3338.
  • 21 Hamada T, Möhle R, Hesselgesser J. et al. Transendothelial migration of megakaryocytes in response to stromal cell-derived factor 1 (SDF-1) enhances platelet formation. J Exp Med 1998; 188: 539-548.
  • 22 Italiano JE, Shivdasani RA. Megakaryocytes and beyond: the birth of platelets. J Thromb Haemost 2003; 01: 1174-1182.
  • 23 Avecilla ST, Hattori K, Heissig B. et al. Chaemokine-mediated interaction of haematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med 2004; 10: 64-71.
  • 24 Junt T, Schulze H, Chen Z. et al. Dynamic visualisation of thrombopoiesis within bone marrow. Science 2007; 317: 1767-1770.
  • 25 Nishimura S, Nagasaki M, Kunishima S. et al. IL-1a induces thrombopoiesis through megakaryocyte rupture in response to acute platelet needs. J Cell Biol 2015; 209: 453-466.
  • 26 Charras G, Sahai E. Physical influences of the extracellular environment on cell migration. Nat Rev Mol Cell Biol 2014; 15: 813-824.
  • 27 Malara A, Gruppi C, Pallotta I. et al. Extracellular matrix structure and nano-mechanics determine megakaryocyte function. Blood 2011; 118: 4449-4453.
  • 28 Di Buduo CA, Wray LS, Tozzi L. et al. Programmable 3D silk bone marrow niche for platelet generation ex vivo and modeling of megakaryopoiesis pathologies. Blood 2015; 125: 2254-2264.
  • 29 Kim DH, Provenzano PP, Smith CL. et al. Matrix nanotopography as a regulator of cell function. J Cell Biol 2012; 197: 351-360.
  • 30 Discher DE, Janmey P, Wang YL. Tissue cells feel and respond to the stiffness of their substrate. Science 2005; 310: 1139-1143.
  • 31 Shin JW, Swift J, Spinler KR. et al. Myosin-II inhibition and soft 2D matrix maximize multinucleation and cellular projections typical of platelet-producing megakaryocytes. Proc Natl Acad Sci USA 2011; 108: 11458-11463.
  • 32 Tangelder GJ, Slaaf DW, Arts T. et al. Wall shear rate in arterioles in vivo: least estimates from platelet velocity profiles. Am J Physiol 1988; 254: H1059-1064.
  • 33 Mazo IB, von Andrian UH. Adhesion and homing of blood-borne cells in bone marrow microvessels. J Leukoc Biol 1999; 66: 25-32.
  • 34 Zhang L, Orban M, Lorenz M. et al. A novel role of sphingosine 1-phosphate receptor S1pr1 in mouse thrombopoiesis. J Exp Med 2012; 209: 2165-2181.
  • 35 Ruggeri ZM. Platelet adhesion under flow. Microcirculation 2009; 16: 58-83.
  • 36 Neeves KB, Onasoga AA, Wufsus AR. The use of microfluidics in haemostasis: clinical diagnostics and biomimetic models of vascular injury. Curr Opin Hae-matol 2013; 20: 417-423.
  • 37 Dunois-Lardé C, Capron C, Fichelson S. et al. Exposure of human megakaryo-cytes to high shear rates accelerates platelet production. Blood 2009; 114: 1875-1883.
  • 38 Jiang J, Woulfe DS, Papoutsakis ET. Shear enhances thrombopoiesis and formation of microparticles that induce megakaryocytic differentiation of stem cells. Blood 2014; 124: 2094-2103.
  • 39 Torisawa YS, Spina CS, Mammoto T. et al. Bone marrow-on-a-chip replicates haematopoietic niche physiology in vitro. Nat Methods 2014; 11: 663-669.
  • 40 Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol 2014; 32: 760-772.
  • 41 Sullenbarger B, Bahng JH, Gruner R. et al. Prolonged continuous in vitro human platelet production using three-dimensional scaffolds. Exp Haematol 2009; 37: 101-110.
  • 42 Lasky LC, Sullenbarger B. Manipulation of oxygenation and flow-induced shear stress can increase the in vitro yield of platelets from cord blood. Tissue Eng Part C Methods 2011; 17: 1081-1088.
  • 43 Nakagawa Y, Nakamura S, Nakajima M. et al. Two differential flows in a bio-reactor promoted platelet generation from human pluripotent stem cell-derived megakaryocytes. Exp Haematol 2013; 41: 742-748.
  • 44 Schlinker AC, Radwanski K, Wegener C. et al. Separation of in-vitro-derived megakaryocytes and platelets using spinning-membrane filtration. Biotechnol Bioeng 2015; 112: 788-800.
  • 45 Thon JN, Mazutis L, Wu S. et al. Platelet bioreactor-on-a-chip. Blood 2014; 124: 1857-1867.
  • 46 Feng Q, Shabrani N, Thon JN. et al. Scalable generation of universal platelets from human induced pluripotent stem cells. Stem Cell Reports 2014; 03: 817-831.
  • 47 Omenetto FG, Kaplan DL. New opportunities for an ancient material. Science 2010; 329: 528-531.
  • 48 Pallotta I, Lovett M, Kaplan DL. et al. Three-dimensional system for the in vitro study of megakaryocytes and functional platelet production using silk-based vascular tubes. Tissue Eng Part C Methods 2011; 17: 1223-1232.
  • 49 Malara A, Currao M, Gruppi C. et al. Megakaryocytes contribute to the bone marrow-matrix environment by expressing fibronectin, type IV collagen, and laminin. Stem Cells 2014; 32: 926-937.
  • 50 Avraham H, Cowley S, Chi SY. et al. Characterisation of adhesive interactions between human endothelial cells and megakaryocytes. J Clin Invest 1993; 91: 2378-2384.
  • 51 Thiele W, Krishnan J, Rothley M. et al. VEGFR-3 is expressed on megakaryocyte precursors in the murine bone marrow and plays a regulatory role in megaka-ryopoiesis. Blood 2012; 120: 1899-1907.
  • 52 Pitchford SC, Lodie T, Rankin SM. VEGFR1 stimulates a CXCR4-dependent translocation of megakaryocytes to the vascular niche, enhancing platelet production in mice. Blood 2012; 120: 2787-2795.
  • 53 Choi ES, Nichol JL, Hokom MM. et al. Platelets generated in vitro from propla-telet-displaying human megakaryocytes are functional. Blood 1995; 85: 402-413.
  • 54 Hitchcock IS, Kaushansky K. Thrombopoietin from beginning to end. Br J Haematol 2014; 165: 259-268.
  • 55 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.
  • 56 Balduini A, Malara A, Balduini CL. et al. Megakaryocytes derived from patients with the classical form of Bernard-Soulier syndrome show no ability to extend proplatelets in vitro. Platelets 2011; 22: 308-311.
  • 57 Lambert MP, Sullivan SK, Fuentes R. et al. Challenges and promises for the development of donor-independent platelet transfusions. Blood 2013; 121: 3319-3324.
  • 58 Avanzi MP, Mitchell WB. Ex vivo production of platelets from stem cells. Br J Haematol 2014; 165: 237-247.
  • 59 Balduini A, Badalucco S, Pugliano MT. et al. In vitro megakaryocyte differentiation and proplatelet formation in Ph-negative classical myeloproliferative neoplasms: distinct patterns in the different clinical phenotypes. PLoS One 2011; 06: e21015.
  • 60 Currao M, Balduini CL, Balduini A. High doses of romiplostim induce proliferation and reduce proplatelet formation by human megakaryocytes. PLoS One 2013; 08: e54723.
  • 61 Di Buduo CA, Moccia F, Battiston M. et al. The importance of calcium in the regulation of megakaryocyte function. Haematologica 2014; 99: 769-778.
  • 62 Bluteau D, Balduini A, Balayn N. et al. Thrombocytopenia-associated mutations in the ANKRD26 regulatory region induce MAPK hyperactivation. J Clin Invest 2014; 124: 580-591.
  • 63 Espasandin YR, Glembotsky AC, Grodzielski M. et al. Anagrelide platelet-lowering effect is due to inhibition of both megakaryocyte maturation and pro-platelet formation: insight into potential mechanisms. J Thromb Haemost 2015; 13: 631-642.
  • 64 Shao Y, Sang J, Fu J. On human pluripotent stem cell control: The rise of 3D bioengineering and mechanobiology. Biomaterials 2015; 52: 26-43.
  • 65 Gaur M, Kamata T, Wang S. et al. Megakaryocytes derived from human embryonic stem cells: a genetically tractable system to study megakaryocytopoiesis and integrin function. J Thromb Haemost 2006; 04: 436-442.
  • 66 Takayama N, Nishikii H, Usui J. et al. Generation of functional platelets from human embryonic stem cells in vitro via ES-sacs, VEGF-promoted structures that concentrate haematopoietic progenitors. Blood 2008; 111: 5298-5306.
  • 67 Lu SJ, Li F, Yin H. et al. Platelets generated from human embryonic stem cells are functional in vitro and in the microcirculation of living mice. Cell Res 2011; 21: 530-545.
  • 68 Pick M, Azzola L, Osborne E. et al. Generation of megakaryocytic progenitors from human embryonic stem cells in a feeder- and serum-free medium. PLoS One 2013; 08: e55530.
  • 69 Takayama N, Nishimura S, Nakamura S. et al. Transient activation of c-MYC expression is critical for efficient platelet generation from human induced pluripo-tent stem cells. J Exp Med 2010; 207: 2817-2830.
  • 70 Nakamura S, Takayama N, Hirata S. et al. Expandable megakaryocyte cell lines enable clinically applicable generation of platelets from human induced pluripo-tent stem cells. Cell Stem Cell 2014; 14: 535-548.
  • 71 Schiffer CA. Diagnosis and management of refractoriness to platelet transfusion. Blood Rev 2001; 15: 175-180.
  • 72 Riolobos L, Hirata RK, Turtle CJ. et al. HLA engineering of human pluripotent stem cells. Mol Ther 2013; 21: 1232-1241.
  • 73 Wang Y, Hayes V, Jarocha D. et al. Comparative analysis of human ex vivo-gen-erated platelets vs megakaryocyte-generated platelets in mice: a cautionary tale. Blood 2015; 125: 3627-3636.