699
Views
5
CrossRef citations to date
0
Altmetric
Short review

Hematopoietic stromal cells and megakaryocyte development

, &
Pages 67-72 | Published online: 12 Nov 2013

Abstract

The hematopoietic microenvironment, and in particular the hematopoietic stromal cell element, are intimately involved in megakaryocyte development. The process of megakaryocytopoiesis occurs within a complex bone marrow microenvironment where adhesive interactions, chemokines, as well as cytokines play a pivotal role. Here we review the effect of stromal cells and cytokines on megakaryocytopoiesis with the aim of exploring new therapeutic strategies for platelet recovery after hematopoietic stem cell transplantation (HSCT).

Introduction

Prolonged thrombocytopenia is a serious complication of high‐dose chemotherapy in the context of autologous or allogeneic transplantation of hematopoietic stem cells (HSCs).Citation1,Citation2 Platelet transfusions are required to support platelet recovery in such patients. In recent years, experiments utilizing ex vivo expansion of megakaryocytes (MKs) from bone marrow (BM), and peripheral blood and cord blood cells have been developed in order to facilitate the platelet recovery.Citation3 However, megakaryocyte development is governed by a complex network of hematopoietic microenvironment, including the hematopoietic stromal cells, cell adhesion molecular, and cytokines. Damage to this microenvironment is one of the primary causes of hematopoietic stem cell transplantation (HSCT) failure; however, stromal cells could be used to restore this microenvironment.Citation4,Citation5 We review the effect of hematopoietic microenvironment on megakaryocytopoiesis, in order to analyse the mechanisms of platelet therapy. We postulate that promoting the development of MKs with stromal cells may be of research value and clinical significance.

Hematopoietic Microenvironment

The hematopoietic microenvironment consists of stromal cells, extracellular matrixes, and cytokines. It is believed that the secretion of various cytokines and growth factors by hematopoietic microenvironment plays an important role in megakaryocytopoiesisCitation6 and in the enhancement of HSCs engraftment in stem cell transplantation.Citation7,Citation8 Megakaryocyte development is coordinated and controlled by combinations of cytokines and mediators presented within specialized bone marrow ‘niches’.Citation9 Two microenvironmental niches are well described: the vascular niche and the osteoblastic niche. The megakaryocytes thrive at vascular niches, whereas the endosteal niche is specifically important in maintaining the stem cell reserve in the quiescent state. Normally, megakaryocytic cytoplasmic processes penetrate the endothelium and release platelets as a shower (thrombopoiesis).Citation10,Citation11

Human bone marrow‐derived stromal cells (hBMSCs)

There is a long‐standing controversy about the origins of stromal cells. On the one side, it is thought that stromal cells are homologous to hematopoietic cells. In the early stage of hematogenesis, ‘common stem cells’ differentiate into stromal cells, forming the hematopoietic microenvironment, before finally differentiating into hematopoietic cells.Citation12Citation15 Another group of researchers argues that human fetal bone marrow contains separate populations of hematopoietic and stromal progenitors, and therefore do not originate from hematopoietic cells.Citation16 Bone marrow stromal cells are derived from mesenchymal stem cells (MSCs), including fibroblasts, adipocytes, endothelial cells, and osteoblasts.Citation17,Citation18 It is noteworthy that MSCs, as the precursor cells of hBMSCs, were were proposed a term ‘multipotent mesenchymal stromal cells’ (with the acronym ‘MSCs’) by the International Society for Cellular Therapy.Citation19,Citation20

Stromal cells are firstly and generally found in bone marrow. Majumdar et al.Citation21 reported that primary and passage 1 cultures of hBMSCs contain a heterogeneous population of hematopoietic and stromal cells, After 14 days, primary cultures of hBMSCs show evidence of hematopoiesis, as indicated by formation of cobblestone area formation. Conventional wisdom has held that stromal cells in Dexter cultures comprise a mixture of macrophages, hematopoietic cells, adipocytes, osteoblasts, fibroblasts, muscle cells, and endothelial cells. However, there is evidence proved that the hBMSCs in Dexter cultures actually represent a single cell type (multi‐differentiated mesenchymal progenitor cell).Citation22 Stromal cells are the progeny of mesenchymal cells following a differentiation pathway of vascular smooth muscle cells.Citation23 Stromal cells from all lines, express mesenchymal markers (vimentin, laminin‐beta1, fibronectin, and osteopontin). Phenotypically, ex vivo expanded stromal cells express a number of non‐specific markers, including Stro‐1,Citation24 CD10,Citation25 SH2, SH3, SH4,Citation26 CD90,Citation27 CD105 (endoglin),Citation28 and CD106.Citation29 hBMSCs are devoid of hematopoietic and endothelial markers, such as CD11b, CD14, CD31, and CD45.Citation30

Significant clinical expectations have been associated with three functional aspects of hBMSCs: tissue repair, immune modulation, as well as hematopoietic support.Citation31 hBMSCs show similar cytokine and growth factor expression as MSCs.Citation21 hBMSCs are capable of supporting hematopoiesis by demonstrating the expression of several hematopoietic growth factors and extracellular matrix receptors including thrombopoietin (TPO), interleukins, granulocyte colony‐stimulating factor (G‐CSF), stem cell factor (SCF), vascular cell adhesion molecule‐1 (VCAM‐1), intercellular cell adhesion molecule‐1 (ICAM‐1), activated leukocyte cell adhesion molecule, stromal cell‐derived factor‐1 (SDF‐1), and fibroblast growth factor (FGF), all of which act directly on hematopoietic cells via the paracrine pathway, or combine with extracellular matrix to regulate hematogenesis.Citation32

Human umbilical cord blood‐derived stromal cells (hUCBDSCs)

Human umbilical cord blood is abundant, collected easily, and has a low probability of pathophoresis.Citation33 Therefore, human umbilical cord blood is often used as an alternative and attractive source for cellular or gene therapy.

hUCBDSCs are isolated and identified from cord blood. Following prolonged culture, cells are mainly composed of macrophage‐like, fibroblast‐like, and small‐sphere‐like cells.Citation34 By immunocytochemistry stain, the hUCB‐derived stromal cells are positive for CD106, CD29, CD44, CD45, CD50, CD68, and HLA‐ I, and negative for CD34, HLA‐ II, and T‐cell co‐stimulatory molecule CD80, CD86, CD40 and CD40L.Citation34Citation36 hUCB‐derived stromal cells can excrete hematopoietic factors, such as TPO, SDF‐1, GM‐CSF, and SCF. These factors may be closely linked with megakaryocyte development.Citation37,Citation38 Moreover, hUCBDSCs are superior to hBMSCs in promoting CFU‐Meg formation and the recovery of platelets in nude mice after transplant. This suggests that hUCBDSCs may play a unique role in megakaryocytopoiesis.Citation38

Cell adhesion molecules (CAMs)

CAMs play a key role in cell‐cell interactions between hematopoietic stromal cells and MK progenitor cells in bone marrow. CAMs include extracellular matrix molecules, integrins, selectins, cadherin, and immunoglobulin superfamily, such as VCAM‐1, very late antigen (VLA‐4 and ‐5), platelet endothelial cell adhesion molecule‐1(PECAM‐1), and ICAM‐1.

Integrins are reported to be involved in the adhesion of megakaryocytes to fibronectin and fibrinogen, which indicates that integrins have a role in MK maturation and platelet production.Citation39 The VLA‐4 is a receptor for VCAM‐1. VCAM‐1 is expressed by cultured BM stromal cells.Citation40,Citation41 Activation of VLA‐4 and ‐5 can induce the adhesion of megakaryocytic leukemia‐derived cell line Mo7e to fibronectin and VCAM‐1.Citation42 Previous studies have shown that VCAM‐1/VLA‐4 adhesion molecules support the attachment of megakaryocytes to bone marrow endothelial cells, as well as bone marrow fibroblasts.Citation43Citation45 It was found that megakaryocytes in PECAM‐1−/− mice cannot migrate along with the SDF‐1 concentration gradient.Citation46 Subsequent studies reported that PECAM‐1 on the megakaryocytes can chemotactically cause megakaryocytes to migrate by changing the distribution of its own CXCR4.Citation46,Citation47 There is report which suggests that the occurrence of megakaryocytic emperipoiesis is partly dependent on adhesion molecules via LFA‐1/ICAM‐1.Citation48 On the other hand, high levels of ICAM‐1 may be one of the factors initiating the events ultimately leading to clonal thrombocytosis, a pathological unregulated MKs development and platelet production.Citation49 Selectins (CD62L and CD62P) can mediate megakaryocyte–fibroblast interactions in human bone marrow.Citation50 Taken together, CAMs mainly mediate MKs adhesion to hematopoietic microenvironment, so as to regulate the development of MKs.

Cytokines

The regulation of megakaryocyte development appears to be controlled not only by the interactions between stromal cells and MKs, or the cell adhesion molecules and MKs, but also by soluble cytokines and chemokines. During the megakaryocytopoiesis, the nascent stage of MK progenitor cells is regulated primarily by TPO and to a lesser degree by other cytokines, such as IL‐1, IL‐3, and platelet‐derived growth factor (PDGF). Later differentiation stages are regulated primarily again by TPO and probably by IL‐6 and IL‐11.Citation51Citation54 In addition, SDF‐1 may play its role in the promotion of megakaryocytopoiesis ().Citation55,Citation56 Important cell factors include:

Table 1. Cytokines in megakaryocytopoiesis

1.

TPO: BM stromal cells are known to synthesize TPO.Citation27 TPO is the primary physiological growth factor for the MK lineage, which can stimulate the proliferation and maturation of MK progenitor cells. It stimulates MKs to increase in cell size and ploidy, and to form proplatelet processes that then fragment into single platelets.Citation57Citation59 Furthermore, when purified MKs were co‐cultured with stromal cells, most of the MKs adhered to the stromal cells and remained unchanged, while free MKs induced proplatelet formation. Thus, the interaction of MKs with stromal cells may suppress platelet formation.Citation60 Nagahisa et al.Citation61 believed that hBMSCs produce TPO and stimulate MK growth and maturation, but suppress the formation of lengthy beaded cytoplasmic processes;

2.

SDF‐1: SDF‐1 is a chemotactic factor produced by stromal cells. SDF‐1 supports megakaryocytopoiesis and homing of HSCs to the BM during fetal development.Citation62 SDF‐1, together with FGF‐4, supports platelet production in TPO−/− or mpl−/− mice through interactions of MK progenitors with the BM vascular niche.Citation59,Citation63 The activity of SDF‐1 is important for the movement of megakaryocyte progenitors from the proliferative ‘osteoblastic niche’ to the ‘vascular niche’ for platelet formation.Citation63,Citation64 Additionally, platelet production is enhanced during transendothelial migration of CXCR4+ MK in response to SDF‐1.Citation65 Perez et al.Citation55 have found that SDF‐1 and its analogue may be of clinical value in stimulating platelet recovery after chemo/radiation treatment as well as in stem cell mobilization;

3.

IL: the stromal cells can express mRNAs of IL‐1 through IL‐7 in bone marrow and fetal liver stromal cells.Citation66 IL‐1beta, IL‐3, and IL‐6, together with PDGF, are reported to increase the megakaryocytic progenitors (CD61+CD41+ cells and CFU‐MK).Citation67Citation69 IL‐6 and IL‐7 both derive from stromal cells and exert important effects in terms of MKs proliferation and differentiation.Citation70Citation72 Fujiki et al.Citation73 demonstrated that human IL‐9 can potentiate human megakaryocytopoiesis in the presence of erythropoietin (EPO) and/or SCF. IL‐11 is isolated from a marrow stromal fibroblast cell line, and this cytokine plays a key role in the regulation of human megakaryocytopoiesis in the BM. In the presence of rhIL‐3, rhIL‐11 stimulated an increase in the number, size, and ploidy value of the megakaryocyte colonies by non‐adherent, T cell‐depleted marrow mononuclear cells;Citation74,Citation75

4.

SCF: it is suggested that a close cell‐to‐cell interaction between MKs and stromal cells is important for an optimal stimulation of megakaryocytopoiesis through the SCF/c‐kit receptor system.Citation76 SCF is a ligand of the protein tyrosine kinase receptor c‐kit, which is found in two forms: transmembrane SCF (tm‐SCF) and soluble SCF (s‐SCF). Research in vitro indicates that tm‐SCF can induce CD34+ cell proliferation over time, but soluble SCF cannot.Citation77 SCF can increase the number of megakaryocytes per colony in the presence of IL‐3, GM‐CSF, or IL‐6. SCF also stimulates the proliferation of specific megakaryocytic cell lines (CMK and M‐07e).Citation78 SCF is recognized as a hematopoietic growth factor, acting on nascent hematopoietic stem cells or progenitor cells;

5.

FL: just like the SCF, FL is also produced by BM stromal cells. It is a ligand of protein tyrosine kinase receptors, and includes two types: soluble and combined. The soluble form is known to be active. FL synergistically augments the ability of IL‐3 and c‐kit ligand, alone or in association, to promote long‐term megakaryocytopoiesis.Citation79 Flt3/Flk‐2 ligand in combination with TPO increases the numbers of megakaryocyte progenitor cells in serum‐free cultures, and decreases apoptosis in megakaryocyte development;Citation80,Citation81

6.

FGF: expression of FGFs can be found in various tissue and cells, including BM stromal cells, smooth muscle cells, myocardial cells, etc. FGF as a potent mitogen, especially FGF‐2Citation82 and FGF‐4,Citation83,Citation84 has been reported to improve the performance of human long‐term hematopoietic cultures. Basic FGF can enhance the adhesion of BM megakaryocytes to marrow stromal fibrobasts and induce cytokine secretion (IL‐6, IL‐1beta, and GM‐CSF).Citation43 Furthermore, it is suggested that FGF is capable of stimulating megakaryocytopoiesis and the effect of FGF is mediated through an interaction with FGF receptor types 1 and 2 located on the megakaryocytic lineage.Citation43,Citation85

Additionally, a number of transcription factors have been implicated in MK differentiation. GATA‐1, FOG‐1, and NF‐E2 are essential regulators of megakaryocytopoiesis. Fli‐1 likely also plays an important role in the process.Citation54,Citation86 In vitro, some chemotactic factors can inhibit megakaryocytopoiesis, such as those in CXC categories (PF4, betaTG, NAP‐2, and IL‐8) and in CC categories (MIP‐1alpha, MIP‐1beta, and IP10).Citation87 Transforming growth factor‐beta1 and interferons, produced by stromal cells, can also inhibit megakaryocyte colony formationCitation88,Citation89 ().

Table 2. Transcriptional regulation of megakaryocytopoiesis

As mentioned above, stromal cells produce essential hematopoietic growth factors, express adhesion molecules and extracellular matrix proteins that are known to play a role in hematopoietic stem cell mobilization and homing, support the growth of hematopoietic stem and progenitor cells, and dramatically enhance megakaryocyte and platelet formation in vitro.Citation7,Citation90 This evidence suggests that co‐transplantation of stromal cells could facilitate HSC engraftment in patients. Indeed, several studies have already reported that co‐transplantation of stromal cells can enhance the success of HSCT,Citation91 prevent graft‐versus‐host disease,Citation92 and promote myelopoiesis and megakaryocytopoiesis.Citation7

Conclusions and Perspectives

In conclusion, the accumulation of knowledge about molecular mechanisms regulating megakaryocytopoiesis and platelet production provides a strong theoretical basis for the therapy of thrombocytopenic states. The hematopoietic microenvironment, in particular the hematopoietic stromal cells in it, is regarded as the ‘soil’ of HSCs, and shed a light on the physician. For example, ex vivo expansion of MKs through co‐culturing with the stromal cells or through an optimal cytokine combination and in vivo transfusion of hematopoietic stromal cells along with HSCT, might be used as supplemental approaches to conventional grafts to alleviate post‐transplant and chemotherapy‐induced thrombocytopenia.

This study was supported by grants from the National Natural Science Foundation of China (nos. 30800496 and 30971109), and the Chongqing Natural Science Foundation (nos. 2008BB5293 and CSTC2009BA5011).

References

  • First LR, Smith BR, Lipton J, Nathan DG, Parkman R, Rappeport JM. Isolated thrombocytopenia after allogeneic bone marrow transplantation: existence of transient and chronic thrombocytopenic syndromes. Blood 1985;65:368–74.
  • Kessinger A, Armitage JO, Landmark JD, Smith DM, Weisenburger DD. Autologous peripheral hematopoietic stem cell transplantation restores hematopoietic function following marrow ablative therapy. Blood 1988:71:723–7.
  • Maurer AM, Liu Y, Caen JP, Han ZC. Ex vivo expansion of megakaryocytic cells. Int J Hematol 2000;71:203–10.
  • Gibson LF, Fortney J, Landreth KS, Piktel D, Ericson SG, Lynch JP. Disruption of bone marrow stromal cell function by etoposide. Biol Blood Marrow Transplant 1997;3:122–32.
  • Hiwase SD, Dyson PG, To LB, Lewis ID. Cotransplantation of placental mesenchymal stromal cells enhances single and double cord blood engraftment in nonobese diabetic/severe combined immune deficient mice. Stem Cells 2009;27:2293–300.
  • Dorshkind K. Regulation of hemopoiesis by bone marrow stromal cells and their products. Annu Rev Immunol 1990;8:111–37.
  • Angelopoulou M, Novelli E, Grove JE, Rinder HM, Civin C, Cheng L, et al.. Cotransplantation of human mesenchymal stem cells enhances human myelopoiesis and megakaryocytopoiesis in NOD/SCID mice. Exp Hematol 2003;31:413–20.
  • Erices AA, Allers CI, Conget PA, Rojas CV, Minguell JJ. Human cord blood‐derived mesenchymal stem cells home and survive in the marrow of immunodeficient mice after systemic infusion. Cell Transplant 2003;12:555–61.
  • Gordge MP. Megakaryocyte apoptosis: sorting out the signals. Br J Pharmacol 2005;145:271–3.
  • Ayala F, Dewar R, Kieran M, Kalluri R. Contribution of bone microenvironment to leukemogenesis and leukemia progression. Leukemia 2009;23:2233–41.
  • Yin T, Li L. The stem cell niches in bone. J Clin Invest 2006;116:1195–201.
  • Islam A. Haemopoietic stem cell: a new concept. Leuk Res 1985;9:1415–32.
  • Huang S, Terstappen LW. Formation of haematopoietic microenvironment and haematopoietic stem cells from single human bone marrow stem cells. Nature 1992;360:745–9.
  • Singer JW, Keating A, Cuttner J, Gown AM, Jacobson R, Killen PD, et al.. Evidence for a stem cell common to hematopoiesis and its in vitro microenvironment: studies of patients with clonal hematopoietic neoplasia. Leuk Res 1984;8:535–45.
  • Simmons PJ, Torok‐Storb B. CD34 expression by stromal precursors in normal human adult bone marrow. Blood 1991;78:2848–53.
  • Waller EK, Olweus J, Lund‐Johansen F, Huang S, Nguyen M, Guo GR, et al.. The “common stem cell” hypothesis reevaluated: human fetal bone marrow contains separate populations of hematopoietic and stromal progenitors. Blood 1995;85:2422–35.
  • Simmons P, Gronthos S, Zannettino A. The development of stromal cells. In: Hematopoiesis: a developmental approach. New York: Oxford University Press; 2001. p. 718–26.
  • Short B, Brouard N, Occhiodoro‐Scott T, Ramakrishnan A, Simmons PJ. Mesenchymal stem cells. Arch Med Res 2003;34:565–71.
  • Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper‐Cortenbach I, Marini FC, et al.. Clarification of the nomenclature for MSC: the International Society for Cellular Therapy position statement. Cytotherapy 2005;7:393–5.
  • Dominici M, Le Blanc K, Mueller I, Slaper‐Cortenbach I, Marini F, Krause D, et al.. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315–7.
  • Majumdar MK, Thiede MA, Mosca JD, Moorman M, Gerson SL. Phenotypic and functional comparison of cultures of marrow‐derived mesenchymal stem cells (MSCs) and stromal cells. J Cell Physiol 1998;176:57–66.
  • Seshi B, Kumar S, Sellers D. Human bone marrow stromal cell: coexpression of markers specific for multiple mesenchymal cell lineages. Blood Cells Mol Dis 2000;26:234–46.
  • Dennis JE, Charbord P. Origin and differentiation of human and murine stroma. Stem Cells 2002;20:205–14.
  • Simmons PJ, Torok‐Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO‐1. Blood 1991;78:55–62.
  • Salles G, Chen CY, Reinherz EL, Shipp MA. CD10/NEP is expressed on Thy‐1low B220+ murine B‐cell progenitors and functions to regulate stromal cell‐dependent lymphopoiesis. Blood 1992;80:2021–9.
  • Haynesworth SE, Baber MA, Caplan AI. Cell surface antigens on human marrow‐derived mesenchymal cells are detected by monoclonal antibodies. Bone 1992;13:69–80.
  • Guerriero A, Worford L, Holland HK, Guo GR, Sheehan K, Waller EK. Thrombopoietin is synthesized by bone marrow stromal cells. Blood 1997;90:3444–55.
  • Rokhlin O, Cohen M, Kubagawa H, Letarte M, Cooper M. Differential expression of endoglin on fetal and adult hematopoietic cells in human bone marrow. J Immunol 1995;154:4456.
  • Simmons PJ, Masinovsky B, Longenecker BM, Berenson R, Torok‐Storb B, Gallatin WM. Vascular cell adhesion molecule‐1 expressed by bone marrow stromal cells mediates the binding of hematopoietic progenitor cells. Blood 1992;80:388–95.
  • Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al.. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–7.
  • Tolar J, Hippen KL, Blazar BR. Immune regulatory cells in umbilical cord blood: T regulatory cells and mesenchymal stromal cells. Br J Haematol 2009;147:200–6.
  • Kohler T, Plettig R, Wetzstein W, Schaffer B, Ordemann R, Nagels HO, et al.. Defining optimum conditions for the ex vivo expansion of human umbilical cord blood cells. Influences of progenitor enrichment, interference with feeder layers, early‐acting cytokines and agitation of culture vessels. Stem Cells 1999;17:19–24.
  • Barker JN, Wagner JE. Umbilical cord blood transplantation: current practice and future innovations. Crit Rev Oncol Hematol 2003;48:35–43.
  • Gao L, Chen X, Zhang X, Liu Y, Kong P, Peng X, et al.. Human umbilical cord blood‐derived stromal cell, a new resource of feeder layer to expand human umbilical cord blood CD34+ cells in vitro. Blood Cells Mol Dis 2006;36:322–8.
  • Hao L, Zhang C, Chen XH, Zou ZM, Zhang X, Kong PY, et al.. Human umbilical cord blood‐derived stromal cells suppress xenogeneic immune cell response in vitro. Croat Med J 2009;50:351–60.
  • Oh W, Kim DS, Yang YS, Lee JK. Immunological properties of umbilical cord blood‐derived mesenchymal stromal cells. Cell Immunol 2008;251:116–23.
  • Feng Y, Chen X, Gao L. Hypothesis: human umbilical cord blood‐derived stromal cells promote megakaryocytopoiesis through the influence of SDF‐1 and PECAM‐1. Cell Biochem Biophys 2010;58:25–30.
  • Gao L, Chen XH, Feng YM, Zhang X, Yu SC, Zhang C, et al.. Human umbilical cord blood‐derived stromal cells: multifaceted regulators of megakaryocytopoiesis. Cell Cycle 2010;9:1342–53.
  • Schick PK, Wojenski CM, He X, Walker J, Marcinkiewicz C, Niewiarowski S. Integrins involved in the adhesion of megakaryocytes to fibronectin and fibrinogen. Blood 1998;92:2650–6.
  • Miyake K, Medina K, Ishihara K, Kimoto M, Auerbach R, Kincade PW. A VCAM‐like adhesion molecule on murine bone marrow stromal cells mediates binding of lymphocyte precursors in culture. J Cell Biol 1991;114:557–65.
  • Juneja HS, Schmalsteig FC, Lee S, Chen J. Vascular cell adhesion molecule‐1 and VLA‐4 are obligatory adhesion proteins in the heterotypic adherence between human leukemia/lymphoma cells and marrow stromal cells. Exp Hematol 1993;21:444–50.
  • Cui L, Ramsfjell V, Borge OJ, Veiby OP, Lok S, Jacobsen SE. Thrombopoietin promotes adhesion of primitive human hemopoietic cells to fibronectin and vascular cell adhesion molecule‐1: role of activation of very late antigen (VLA)‐4 and VLA‐5. J Immunol 1997;159:1961–9.
  • Avraham H, Banu N, Scadden DT, Abraham J, Groopman JE. Modulation of megakaryocytopoiesis by human basic fibroblast growth factor. Blood 1994;83:2126–32.
  • Avraham H, Cowley S, Chi SY, Jiang S, Groopman JE. Characterization of adhesive interactions between human endothelial cells and megakaryocytes. J Clin Invest 1993;91:2378–84.
  • Avraham H, Scadden DT, Chi S, Broudy VC, Zsebo KM, Groopman JE. Interaction of human bone marrow fibroblasts with megakaryocytes: role of the c‐kit ligand. Blood 1992;80:1679–84.
  • Dhanjal TS, Pendaries C, Ross EA, Larson MK, Protty MB, Buckley CD, et al.. A novel role for PECAM‐1 in megakaryocytokinesis and recovery of platelet counts in thrombocytopenic mice. Blood 2007;109:4237–44.
  • Wu Y, Welte T, Michaud M, Madri JA. PECAM‐1: a multifaceted regulator of megakaryocytopoiesis. Blood 2007;110:851–9.
  • Tanaka M, Aze Y, Fujita T. Adhesion molecule LFA‐1/ICAM‐1 influences on LPS‐induced megakaryocytic emperipolesis in the rat bone marrow. Vet Pathol 1997;34:463–6.
  • Cobankara V, Oran B, Ozatli D, Haznedaroglu IC, Kosar A, Buyukasik Y, et al.. Cytokines, endothelium, and adhesive molecules in pathologic thrombopoiesis. Clin Appl Thromb Hemost 2001;7:126–30.
  • Wickenhauser C, Schmitz B, Baldus SE, Henze F, Farahmand P, Frimpong S, et al.. Selectins (CD62L, CD62P) and megakaryocytic glycoproteins (CD41a, CD42b) mediate megakaryocyte–fibroblast interactions in human bone marrow. Leuk Res 2000;24:1013–21.
  • Kaushansky K. Thrombopoietin. N Engl J Med 1998;339:746–54.
  • Yang M, Chesterman CN, Chong BH. Recombinant PDGF enhances megakaryocytopoiesis in vitro. Br J Haematol 1995;91:285–9.
  • Yang M, Li K, Chui CM, Yuen PM, Chan PK, Chuen CK, et al.. Expression of interleukin (IL) 1 type I and type II receptors in megakaryocytic cells and enhancing effects of IL‐1beta on megakaryocytopoiesis and NF‐E2 expression. Br J Haematol 2000;111:371–80.
  • Yang M, Li K. The role of cytokines and transcription factors in megakaryocytopoiesis. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2002;10:580–5.
  • Perez LE, Alpdogan O, Shieh JH, Wong D, Merzouk A, Salari H, et al.. Increased plasma levels of stromal‐derived factor‐1 (SDF‐1/CXCL12) enhance human thrombopoiesis and mobilize human colony‐forming cells (CFC) in NOD/SCID mice. Exp Hematol 2004;32:300–7.
  • Hodohara K, Fujii N, Yamamoto N, Kaushansky K. Stromal cell‐derived factor‐1 (SDF‐1) acts together with thrombopoietin to enhance the development of megakaryocytic progenitor cells (CFU‐MK). Blood 2000;95:769–75.
  • Kaushansky K. The molecular mechanisms that control thrombopoiesis. J Clin Invest 2005;115:3339–47.
  • Kaushansky K. Thrombopoietin and the hematopoietic stem cell. Ann NY Acad Sci 2005;1044:139–41.
  • Deutsch V, Tomer A. Megakaryocyte development and platelet production. Br J Haematol 2006;134:453–66.
  • Nagata Y, Nagahisa H, Nagasawa T, Todokoro K. Regulation of megakaryocytopoiesis by thrombopoietin and stromal cells. Leukemia 1997;11(Suppl. 3):435–8.
  • Nagahisa H, Nagata Y, Ohnuki T, Osada M, Nagasawa T, Abe T, et al.. Bone marrow stromal cells produce thrombopoietin and stimulate megakaryocyte growth and maturation but suppress proplatelet formation. Blood 1996;87:1309–16.
  • 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;92:756–64.
  • Avecilla ST, Hattori K, Heissig B, Tejada R, Liao F, Shido K, et al.. Chemokine‐mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med 2004;10:64–71.
  • Pang L, Weiss MJ, Poncz M. Megakaryocyte biology and related disorders. J Clin Invest 2005;115:3332–8.
  • Lane WJ, Dias S, Hattori K, Heissig B, Choy M, Rabbany SY, et al.. Stromal‐derived factor 1‐induced megakaryocyte migration and platelet production is dependent on matrix metalloproteinases. Blood 2000;96:4152–9.
  • Gutierrez‐Ramos JC, Olsson C, Palacios R. Interleukin (IL1 to IL7) gene expression in fetal liver and bone marrow stromal clones: cytokine‐mediated positive and negative regulation. Exp Hematol 1992;20:986–90.
  • Su R, Li K, Yang M, Zhang X, Tsang K, Fok T, et al.. Platelet‐derived growth factor enhances ex vivo expansion of megakaryocytic progenitors from human cord blood. Bone Marrow Transplant 2001;27:1075.
  • Su R, Zhang X, Li K, Yang M, Li C, Fok T, et al.. Platelet‐derived growth factor promotes ex vivo expansion of CD34+ cells from human cord blood and enhances long‐term culture‐initiating cells, non‐obese diabetic/severe combined immunodeficient repopulating cells and formation of adherent cells. Br J Haematol 2002;117:735–46.
  • Bruno E, Miller ME, Hoffman R. Interacting cytokines regulate in vitro human megakaryocytopoiesis. Blood 1989;73:671–7.
  • Fry T, Mackall C. The many faces of IL‐7: from lymphopoiesis to peripheral T cell maintenance. J Immnol 2005;174:6571–6.
  • Gupta P, Blazar BR, Gupta K, Verfaillie CM. Human CD34+ bone marrow cells regulate stromal production of interleukin‐6 and granulocyte colony‐stimulating factor and increase the colony‐stimulating activity of stroma. Blood 1998;91:3724–33.
  • Baatout S: Interleukin‐6 and megakaryocytopoiesis: an update. Ann Hematol 1996;73:157–62.
  • Fujiki H, Kimura T, Minamiguchi H, Harada S, Wang JF, Nakao M, et al.. Role of human interleukin‐9 as a megakaryocyte potentiator in culture. Exp Hematol 2002;30:1373–80.
  • Teramura M, Kobayashi S, Hoshino S, Oshimi K, Mizoguchi H. Interleukin‐11 enhances human megakaryocytopoiesis in vitro. Blood 1992;79:327.
  • Du X, Williams D. Interleukin‐11: review of molecular, cell biology, and clinical use. Blood 1997;89:3897.
  • Avraham H, Vannier E, Cowley S, Jiang SX, Chi S, Dinarello CA, et al.. Effects of the stem cell factor, c‐kit ligand, on human megakaryocytic cells. Blood 1992;79:365–71.
  • Friel J, Itoh K, Bergholz U, Jucker M, Stocking C, Harrison P, et al.. Hierarchy of stroma‐derived factors in supporting growth of stroma‐dependent hemopoietic cells: membrane‐bound SCF is sufficient to confer stroma competence to epithelial cells. Growth Factors 2002;20:35–51.
  • Erickson‐Miller CL, Ji H, Murphy MJ. Megakaryocytopoiesis and platelet production: does stem cell factor play a role? Stem Cells 1993;11(Suppl. 2):163–9.
  • Piacibello W, Garetto L, Sanavio F, Severino A, Fubini L, Stacchini A, et al.. The effects of human FLT3 ligand on in vitro human megakaryocytopoiesis. Exp Hematol 1996;24:340–6.
  • Sigurjonsson OE, Gudmundsson KO, Haraldsdottir V, Rafnar T, Agnarsson BA, Gudmundsson S. Flt3/Flk‐2 ligand in combination with thrombopoietin decreases apoptosis in megakaryocyte development. Stem Cells Dev 2004;13:183–91.
  • Sigurjonsson OE, Gudmundsson KO, Haraldsdottir V, Rafnar T, Gudmundsson S. Flt3/Flk‐2‐ligand in synergy with thrombopoietin delays megakaryocyte development and increases the numbers of megakaryocyte progenitor cells in serum‐free cultures initiated with CD34+ cells. J Hematother Stem Cell Res 2002;11:389–400.
  • Allouche M, Bikfalvi A. The role of fibroblast growth factor‐2 (FGF‐2) in hematopoiesis. Prog Growth Factor Res 1995;6:35–48.
  • Quito F, Beh J, Bashayan O, Basilico C, Basch R. Effects of fibroblast growth factor‐4 (k‐FGF) on long‐term cultures of human bone marrow cells. Blood 1996;87:1282.
  • Liu CH, Hwang SM. Cytokine interactions in mesenchymal stem cells from cord blood. Cytokine 2005;32:270–9.
  • Bikfalvi A, Han ZC, Fuhrmann G. Interaction of fibroblast growth factor (FGF) with megakaryocytopoiesis and demonstration of FGF receptor expression in megakaryocytes and megakaryocytic‐like cells. Blood 1992;80:1905–13.
  • Shivdasani RA. Molecular and transcriptional regulation of megakaryocyte differentiation. Stem Cells 2001;19:397–407.
  • Gewirtz AM, Zhang J, Ratajczak J, Ratajczak M, Park KS, Li C, et al.. Chemokine regulation of human megakaryocytopoiesis. Blood 1995;86:2559–67.
  • Scandura JM, Boccuni P, Massague J, Nimer SD. Transforming growth factor beta‐induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up‐regulation. Proc Natl Acad Sci USA 2004;101:15231–6.
  • Verma A, Deb DK, Sassano A, Uddin S, Varga J, Wickrema A, et al.. Activation of the p38 mitogen‐activated protein kinase mediates the suppressive effects of type I interferons and transforming growth factor‐beta on normal hematopoiesis. J Biol Chem 2002;277:7726–35.
  • Cheng L, Qasba P, Vanguri P, Thiede MA. Human mesenchymal stem cells support megakaryocyte and pro‐platelet formation from CD34+ hematopoietic progenitor cells. J Cell Physiol 2000;184:58–69.
  • Almeida‐Porada G, Porada CD, Tran N, Zanjani ED. Cotransplantation of human stromal cell progenitors into preimmune fetal sheep results in early appearance of human donor cells in circulation and boosts cell levels in bone marrow at later time points after transplantation. Blood 2000;95:3620–7.
  • Chung NG, Jeong DC, Park SJ, Choi BO, Cho B, Kim HK, et al.. Cotransplantation of marrow stromal cells may prevent lethal graft‐versus‐host disease in major histocompatibility complex mismatched murine hematopoietic stem cell transplantation. Int J Hematol 2004;80:370–6.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.