Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 May 5.
Published in final edited form as: Curr Opin Hematol. 2008 Jul;15(4):307–311. doi: 10.1097/MOH.0b013e3283007db5

Cytokines regulating hematopoietic stem cell function

Cheng C Zhang a, Harvey F Lodish b,c
PMCID: PMC2677548  NIHMSID: NIHMS105584  PMID: 18536567

Abstract

Purpose of review

Regulation of the multiple fates of hematopoietic stem cells – including quiescence, self-renewal, differentiation, apoptosis, and mobilization from the niche – requires the cooperative actions of several cytokines and other hormones that bind to receptors on these cells. In this review we discuss recent advances in the identification of novel hematopoietic stem cell supportive cytokines and the mechanisms by which they control hematopoietic stem cell fate decisions.

Recent findings

Several extrinsic factors that stimulate ex-vivo expansion of hematopoietic stem cells were recently identified by a number of experimental approaches, including forward genetic screening and transcriptional profiling of supportive stromal cells. Recent experiments in which multiple cytokine signaling pathways are activated or suppressed in hematopoietic stem cells reveal the complexity of signal transduction and cell-fate choice in hematopoietic stem cells in vivo and in vitro.

Summary

The study of genetically modified mice and improvements in the in-vitro hematopoietic stem cell culture system will be powerful tools to elucidate the functions of cytokines that regulate hematopoietic stem cell function. These will further reveal the complex nature of the mechanisms by which extrinsic factors regulate signal transduction and cell-fate decisions of hematopoietic stem cells.

Keywords: cytokines, ex-vivo expansion, growth factors, hematopoietic stem cells, signal transduction

Introduction

Cytokines are secreted proteins that regulate many aspects of hematopoiesis – immune responses and inflammation in particular. Many hematopoietic cytokines were identified and purified on the basis of their abilities to support in-vitro formation of hematopoietic colonies from progenitors; the functions of many of these cytokines and their receptors were confirmed and extended through studies of genetically modified mice. Subsequently, many cytokines were shown to bind directly to receptors on hematopoietic stem cells (HSCs) and regulate many HSC functions, including quiescence, self-renewal, differentiation, apoptosis, and mobility. HSCs reside in ‘stem cell niches’ in the bone marrow and in other tissues; stromal cells are thought to synthesize many HSC cytokines but, in general, we know little about which cells in a niche make specific cytokines. Many cell culture experiments have shown that HSCs respond to multiple cytokines and that the fate of an HSC – self renewal, apoptosis, mobilization from the niche, formation of differentiated progeny cells – depends on multiple cytokines, adhesion proteins, and other signals produced by stromal cells and likely other cells in the body. The study of HSC cytokines will facilitate the development of new strategies for HSC-based cell and gene therapies. Despite a great deal of progress in the past decade, identification and functional study of HSC cytokines are still in a primitive stage and many secreted or cell surface proteins that affect HSC function remain to be discovered.

Identification of hematopoietic stem cell cytokines

To isolate HSC cytokines, sensitive assays for HSC functions are critical. For many years, due to the rarity of HSCs, the lack of an efficient culture system for them, and the difficulty in measuring their activities, direct identification of cytokines that target HSCs was virtually impossible. Some HSC cytokines, including Wnts and bone morphogenic proteins (BMPs), were initially identified from studies of invertebrate development. Some, such as thrombopoietin (TPO), were isolated based on functions of cells other than HSCs.

Two spatially and likely functionally distinct bone marrow microenvironments, or HSC niches, have been proposed (reviewed in [1,2]). Recent work shows that the endosteal HSC niche contains osteoblasts as the main supportive cell type (reviewed in [2,3]). In the vascular niche, many HSCs in the bone marrow and spleen are associated with the sinusoidal endothelium [4].

About a decade ago stromal cell lines were established from in-vivo fetal or adult hematopoietic organs, including aorta-gonado-mesonephros (AGM), fetal liver, yolk sac, and bone marrow. Moore et al. [5] established numerous fetal liver stromal cell lines and used them to isolate secreted proteins that support HSCs. Recently, this group isolated HSC supportive stromal cell lines from the AGM region [6]. The Williams lab isolated yolk sac stromal cell lines that support HSC and progenitor activities [7]. We identified primary mouse fetal liver CD3+Ter119 cells as a novel HSC supportive population, and by transcriptional profiling we identified insulin-like growth factor 2 (IGF-2) and a group of angiopoietin-like proteins (Angptls) as HSC growth factors [810]. We also showed that IGF binding protein 2 (IGFBP2), a protein secreted by HEK 293T cells, supports HSC expansion (Huynh HD et al., unpublished data), and we anticipate that cancer cells will be a rich source of other HSC-stimulating proteins.

Discussion of individual factors

Below we discuss very recent results for several cytokines that affect HSC fates.

Stem cell factor

Stem cell factor (SCF), also known as steel factor, is a cytokine expressed by a number of cell types. SCF functions by binding to c-Kit, a tyrosine kinase receptor expressed on all HSCs. Expression of a defective c-Kit leads to a decrease in repopulating HSCs [11]. Although SCF may not be essential for the generation of HSCs [11], numerous studies [12] have shown that it prevents HSC apoptosis. Almost all cytokine combinations used to date for culturing HSCs include SCF. SCF potentiates the greater ability of fetal liver HSCs than adult HSCs to undergo symmetric self-renewal in culture [13•]; this activity likely needs the cooperation of other factors. The membrane-bound form of SCF is also an adhesive molecule for HSCs to the bone marrow environment [14], and an increased number of osteoclasts was associated with HSC mobilization. Receptor activator of nuclear factor (NF)-κB (RANK) ligand and cathepsin K mediate the cleavage of membrane-bound SCF; this decreases the abundance of SCF and, therefore, increases HSC mobilization [15]. The involvement of SCF in survival, mobility, and possibly self-renewal of HSCs in culture and in the HSC niche likely reflects the complex relationship of different cell fates of HSCs.

Thrombopoietin

TPO is the primary cytokine that regulates megakaryocyte and platelet development. TPO and its receptor Mpl also exert profound effects on primitive hematopoietic cells. All HSCs express Mpl; TPO−/− or Mpl−/− mice have a decreased number of repopulating HSCs [16]. In-vitro culture studies [17] also indicate a role of TPO in promoting the survival of repopulating HSCs. Through study of AGM and fetal liver Mpl−/− HSCs, Petit-Cocault et al. [18•] showed that TPO contributes to both generation and expansion of HSCs during definitive hematopoiesis. An intracellular adaptor, Lnk, induces a negative signaling pathway downstream of TPO in HSCs [19,20]. A recent study [21] on mice that express Mpl lacking the C-terminal 60 amino acids revealed a pivotal role of an unknown signal emanating from the membrane proximal region of the Mpl receptor or from JAK2 that is critical for maintenance of HSC activity.

Notch ligands

In addition to their major roles in lymphopoiesis, the Notch ligands Delta and Jagged are involved in the generation, antidifferentiation, and expansion of HSCs (reviewed in [22]). Importantly, Calvi et al. [23] and Duncan et al. [24] demonstrated that the Notch signaling pathway plays a role in the osteoblast bone marrow HSCs niche. Notch ligands have positive effects on ex-vivo expansion of HSCs: activated Notch is able to immortalize primitive mouse hematopoietic progenitors and Notch ligands support HSC expansion in culture (reviewed in [22]). Recently, by culturing human cord blood cells in serum-free medium supplemented with SCF, TPO, Flt3L, IL-3, IL-6/sIL-6R, and Delta 1, Suzuki et al. [25] reported an approximate six-fold increase in SCID-repopulating cell (SRC) number. It is noteworthy that there exists a dose effect for Notch ligands in HSC culture. Whereas a low amount of Delta 1 supports human cord blood SRC expansion, high amounts of the cytokine induce apoptosis (reviewed in [22]). This emphasizes the complicated relationship among the different fates of HSCs. As conditional knockouts of Notch1 and Jagged1 have normal in-vivo HSC activities [26], there likely is functional redundancy of different Notch isoforms and their ligands.

Wnts

Wnt signaling is involved in HSC generation and expansion (reviewed in [27•]). The Wnt pathway supports mouse HSC expansion and has most recently been shown to be important for self-renewal of stem cells of B-cell antigen receptor (BCR)-ABL-induced chronic myelogenous leukemia [28]. Nemeth et al. [29•] showed that the noncanonical Wnt5a supports HSC repopulation by inhibiting canonical Wnt signaling in HSCs and maintains HSCs in a quiescent state. Wnt also promotes generation of hematopoietic cells from human embryonic stem cells [30•]. In gene-altered mice, prolonged activation of Wnt/β-catenin signaling results in an increase of phenotypical HSCs but failure of hematopoietic functions, including blocks in HSC differentiation and loss of repopulating activity [31,32]. Deletion of both β-catenins and γ-catenins in mice does not change HSC repopulation activity [33,34]. Nevertheless, Wnt signaling is still maintained in these double-deficient HSCs [34]. Most recently, Zhao et al. [28] showed that β-catenin-deficient HSCs have defects in long-term growth and maintenance.

Bone morphogenic proteins and transforming growth factor-β

Transforming growth factor (TGF)-β potently inhibits HSC activity in vitro (reviewed in [3,35••]). However, a TGF-β signaling deficiency in vivo does not affect proliferation of HSCs. BMPs, members of the TGF-β superfamily, play important roles in HSC specification during development. A negative role of BMP signaling in maintenance of mouse HSCs was shown by its control of the size of the HSC endosteal niche [3]. BMP4 supports HSC expansion in culture and partially mediates the effects of Sonic hedgehog on cultured human HSCs [36]. Recently, the Karlsson lab characterized the expression of TGF-β superfamily ligands, receptors, and Smads in mouse HSCs; primary HSCs and the Lhx2-HPC cell line express most of the proteins required to transmit signals from several TGF-β family ligands [37]. In addition, Pimanda et al. [38•] demonstrated the integration of BMP4/Smad pathway and Scl and Runx1 activity in HSC development. Importantly, Trowbridge et al. [39] showed that a glycogen synthase kinase 3 (GSK-3) inhibitor that can modulate Wnt, Hedgehog, and Notch pathways enhances HSC repopulation.

Fibroblast growth factors

All long-term repopulating bone marrow HSCs express a fibroblast growth factor (FGF) receptor [40]; both FGF-1 and FGF-2 support HSC expansion when unfractionated mouse bone marrow cells are cultured in serum-free medium [40,41]. Crcareva et al. [42] confirmed that FGF-1 stimulates ex-vivo expansion of HSCs and showed that the expanded cells were efficiently transduced by retrovirus vectors. Conditional derivatives of FGF receptor-1 have also been used to support short-term HSC expansion and long-term HSC survival in culture [43]. However, the role of the FGF pathway in regulating adult HSCs or embryonic hematopoietic development is controversial as Schiedlmeier et al. [44•] showed that the treatment of purified mouse HSCs that ectopically express HoxB4 with the fibroblast growth factor receptor (FGFR) inhibitor SU5402 enhanced HSC repopulating activity. Similar results were obtained using primitive hematopoietic colonies derived from embryonic stem cells [44•]. These inconsistent results were obtained from different starting cell populations and under different culture conditions, suggesting that the crosstalk of FGF signaling with other pathways is complex.

Angiopoietin-1

The angiopoietin (Ang) family of growth factors is composed of four members that bind to the Tie-2 tyrosine kinase receptor; Ang growth factors are important modulators of angiogenesis. Members of the angiopoietin family of proteins contain an N-terminal coiled–coil domain that mediates homo-oligomerization and a C-terminal fibrinogen-like domain that binds Tie-2. The Tie-2/Ang1 signaling pathway plays a critical role in the maintenance of HSCs in a quiescent state in the bone marrow niche [45]. This pathway also supports SRC activity of human bone marrow CD34 cells in culture [46].

Insulin-like growth factor 2

Recently, we identified a novel cell population that supports HSC expansion: CD3+Ter119− cells isolated from embryonic day 15 (E15) mouse fetal livers [8]. DNA array experiments showed that, among other proteins, IGF-2 is specifically expressed in these cells but not in several other cell types that do not support HSC expansion in culture. We showed that all fetal liver and bone marrow HSCs express receptors for IGF-2. IGF-2 stimulates ex-vivo expansion of both fetal liver and bone marrow HSCs [8]. The inclusion of IGF-2 with SCF, TPO, and FGF-1 supports an eight-fold increase of highly enriched HSCs in culture [8]. Whether IGF-2 acts on self-renewal, apoptosis, differentiation, or homing of HSCs is unclear. Interestingly, IGF-2 was found to bind and stimulate self-renewal of human embryonic stem cells [47•].

Angiopoietin-like proteins

Angptls are a family of seven secreted glycoproteins that share sequence homology with the angiopoietins [48]. Similar to the angiopoietins, each Angptl contains an N-terminal coiled-coil domain and a C-terminal fibrinogen-like domain. However, unlike angiopoietins, Angptls do not bind to Tie-2 or Tie-1 and their receptors are unknown. This suggests that Angptls may have functions different from the angiopoietins. Limited studies [48] on Angptls have shown that several members play a role in regulating angiogenesis, metabolism, and tumorgenesis. Angptl7 was suggested to be a target of the Wnt/β-catenin signaling pathway [49]. However, most of the physiological activities of the Angptls remain unknown. Recently we identified Angptl2 and Angptl3 as growth factors that stimulate ex-vivo expansion of bone marrow HSCs. We further showed that several other analogues, including Angptl5, Angptl7, and Mfap4, also support ex-vivo expansion of HSCs. These studies have enabled us to optimize our serum-free culture system such that we now obtain up to 30-fold expansion of long-term-reconstituting mouse HSCs in culture [10]. Angptl5 also stimulates ex-vivo expansion of human cord blood SRCs [50•]. The mechanism by which Angptls regulate HSCs expansion is under investigation.

Other factors

The IL-10 receptor is expressed on mouse Lin -Sca-1+c-Kit+ (LSK) and side population (SP) cells and IL-10 stimulated a three to four-fold increase in the frequency of repopulating HSCs in culture [51]. mKirre is a membrane protein identified in the stem cell supportive OP9 stromal cells. The cleaved extracellular domain acted as a factor that supports HSC expansion in vitro [52]. A matricellular protein ‘nephroblastoma overexpressed’ (also called NOV or CCN3) was identified from CD34+ human cord blood cells; recombinant Nov protein enhances SRC activity and its knockdown abrogates SRC function. This suggests that Nov is a regulator of human hematopoietic stem or progenitor cells [53••].

Conclusion

One can make two general observations from studies on known HSC cytokines. One is that there are differences in the roles of some cytokines in embryos and in adults. This is not surprising considering the differences between fetal and adult HSCs and the dissimilarities in their microenvironments. Another is that most, if not all, cytokines affecting HSCs exhibit pleiotropy and redundancy. Pleiotropic actions appear to be the result of their different activities in conjunction with other cytokines. Their redundancy may be explained by convergence of different intracellular signaling pathways to ‘master regulators’ for different stem cell fates, self-renewal for example. The study of genetically modified mice and improvements in the in-vitro HSC culture system will be powerful tools to elucidate the functions of cytokines that regulate HSC function. Through studying how the different cell fates of HSCs are regulated by extrinsic factors, we will gain exciting insights into the molecular mechanisms by which the environment controls the fates of HSCs, as well as the pathophysiological origins of many disorders. The knowledge we obtain from these studies promises to result in better development of hematopoietic cell and gene therapies.

Acknowledgments

We regret that we have been unable to cite many relevant primary references due to space limitations. Support to C. C. Z. is from a National Institutes of Health (NIH) grant K01 CA 120099-01 and the Michael. L. Rosenberg Endowed Scholar Fund from University of Texas Southwestern Medical Center. Support to H. F. L. is from NIH grant R01 DK 067356.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest

•• of outstanding interest Additional references related to this topic can also be found in the Current World Literature section in this issue (p. 425).

  • 1.Wilson A, Trumpp A. Bone-marrow haematopoietic-stem-cell niches. Nat Rev Immunol. 2006;6:93–106. doi: 10.1038/nri1779. [DOI] [PubMed] [Google Scholar]
  • 2.Adams GB, Scadden DT. The hematopoietic stem cell in its place. Nat Immunol. 2006;7:333–337. doi: 10.1038/ni1331. [DOI] [PubMed] [Google Scholar]
  • 3.Ross J, Li L. Recent advances in understanding extrinsic control of hematopoietic stem cell fate. Curr Opin Hematol. 2006;13:237–242. doi: 10.1097/01.moh.0000231420.92782.8f. [DOI] [PubMed] [Google Scholar]
  • 4.Kiel MJ, Yilmaz OH, Iwashita T, et al. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121:1109–1121. doi: 10.1016/j.cell.2005.05.026. [DOI] [PubMed] [Google Scholar]
  • 5.Moore KA, Ema H, Lemischka IR. In vitro maintenance of highly purified, transplantable hematopoietic stem cells. Blood. 1997;89:4337–4347. [PubMed] [Google Scholar]
  • 6.Weisel KC, Gao Y, Shieh JH, Moore MA. Stromal cell lines from the aorta-gonado-mesonephros region are potent supporters of murine and human hematopoiesis. Exp Hematol. 2006;34:1505–1516. doi: 10.1016/j.exphem.2006.06.013. [DOI] [PubMed] [Google Scholar]
  • 7.Yoder MC, King B, Hiatt K, Williams DA. Murine embryonic yolk sac cells promote in vitro proliferation of bone marrow high proliferative potential colony-forming cells. Blood. 1995;86:1322–1330. [PubMed] [Google Scholar]
  • 8.Zhang CC, Lodish HF. Insulin-like growth factor 2 expressed in a novel fetal liver cell population is a growth factor for hematopoietic stem cells. Blood. 2004;103:2513–2521. doi: 10.1182/blood-2003-08-2955. [DOI] [PubMed] [Google Scholar]
  • 9.Zhang CC, Lodish HF. Murine hematopoietic stem cells change their surface phenotype during ex vivo expansion. Blood. 2005;105:4314–4320. doi: 10.1182/blood-2004-11-4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zhang CC, Kaba M, Ge G, et al. Angiopoietin-like proteins stimulate ex vivo expansion of hematopoietic stem cells. Nat Med. 2006;12:240–245. doi: 10.1038/nm1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ikuta K, Weissman IL. Evidence that hematopoietic stem cells express mouse c-kit but do not depend on steel factor for their generation. Proc Natl Acad Sci USA. 1992;89:1502–1506. doi: 10.1073/pnas.89.4.1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hassan HT, Zander A. Stem cell factor as a survival and growth factor in human normal and malignant hematopoiesis. Acta Haematol. 1996;95:257–262. doi: 10.1159/000203893. [DOI] [PubMed] [Google Scholar]
  • 13•.Bowie MB, Kent DG, Copley MR, Eaves CJ. Steel factor responsiveness regulates the high self-renewal phenotype of fetal hematopoietic stem cells. Blood. 2007;109:5043–5048. doi: 10.1182/blood-2006-08-037770. The paper provides evidence that the SCF/c-Kit signaling pathway accounts for the greater symmetric self-renewal capacity of fetal HSCs than their adult counterparts. [DOI] [PubMed] [Google Scholar]
  • 14.Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002;109:625–637. doi: 10.1016/s0092-8674(02)00754-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med. 2006;12:657–664. doi: 10.1038/nm1417. [DOI] [PubMed] [Google Scholar]
  • 16.Solar GP, Kerr WG, Zeigler FC, et al. Role of c-mpl in early hematopoiesis. Blood. 1998;92:4–10. [PubMed] [Google Scholar]
  • 17.Matsunaga T, Kato T, Miyazaki H, Ogawa M. Thrombopoietin promotes the survival of murine hematopoietic long-term reconstituting cells: comparison with the effects of FLT3/FLK-2 ligand and interleukin-6. Blood. 1998;92:452–461. [PubMed] [Google Scholar]
  • 18•.Petit-Cocault L, Volle-Challier C, Fleury M, et al. Dual role of Mpl receptor during the establishment of definitive hematopoiesis. Development. 2007;134:3031–3040. doi: 10.1242/dev.001818. The study of HSCs in Mpl−/− AGM and fetal liver shows that Mpl is important for the generation and expansion of HSCs in definitive hematopoiesis. [DOI] [PubMed] [Google Scholar]
  • 19.Buza-Vidas N, Antonchuk J, Qian H, et al. Cytokines regulate postnatal hematopoietic stem cell expansion: opposing roles of thrombopoietin and LNK. Genes Dev. 2006;20:2018–2023. doi: 10.1101/gad.385606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Seita J, Ema H, Ooehara J, et al. Lnk negatively regulates self-renewal of hematopoietic stem cells by modifying thrombopoietin-mediated signal transduction. Proc Natl Acad Sci USA. 2007;104:2349–2354. doi: 10.1073/pnas.0606238104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tong W, Ibarra YM, Lodish HF. Signals emanating from the membrane proximal region of the thrombopoietin receptor (mpl) support hematopoietic stem cell self-renewal. Exp Hematol. 2007;35:1447–1455. doi: 10.1016/j.exphem.2007.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chiba S. Notch signaling in stem cell systems. Stem Cells. 2006;24:2437–2447. doi: 10.1634/stemcells.2005-0661. [DOI] [PubMed] [Google Scholar]
  • 23.Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841–846. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
  • 24.Duncan AW, Rattis FM, DiMascio LN, et al. Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nat Immunol. 2005;6:314–322. doi: 10.1038/ni1164. [DOI] [PubMed] [Google Scholar]
  • 25.Suzuki T, Yokoyama Y, Kumano K, et al. Highly efficient ex vivo expansion of human hematopoietic stem cells using Delta1-Fc chimeric protein. Stem Cells. 2006;24:2456–2465. doi: 10.1634/stemcells.2006-0258. [DOI] [PubMed] [Google Scholar]
  • 26.Mancini SJ, Mantei N, Dumortier A, et al. Jagged1-dependent Notch signaling is dispensable for hematopoietic stem cell self-renewal and differentiation. Blood. 2005;105:2340–2342. doi: 10.1182/blood-2004-08-3207. [DOI] [PubMed] [Google Scholar]
  • 27•.Nemeth MJ, Bodine DM. Regulation of hematopoiesis and the hematopoietic stem cell niche by Wnt signaling pathways. Cell Res. 2007;17:746–758. doi: 10.1038/cr.2007.69. A very nice review of the roles of Wnt signaling in HSCs and HSC niches. [DOI] [PubMed] [Google Scholar]
  • 28.Zhao C, Blum J, Chen A, et al. Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 2007;12:528–541. doi: 10.1016/j.ccr.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29•.Nemeth MJ, Topol L, Anderson SM, et al. Wnt5a inhibits canonical Wnt signaling in hematopoietic stem cells and enhances repopulation. Proc Natl Acad Sci USA. 2007;104:15436–15441. doi: 10.1073/pnas.0704747104. This study of the role of the noncanonical Wnt5a on HSCs in culture showed that Wnt5a inhibits the Wnt3a-mediated canonical Wnt pathway and results in quiescence of HSCs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30•.Woll PS, Morris JK, Painschab MS, et al. Wnt signaling promotes hematoendothelial cell development from human embryonic stem cells. Blood. 2008;111:122–131. doi: 10.1182/blood-2007-04-084186. The authors studied the role of the canonical Wnt pathways in hematoendothelial differentiation of human embryonic stem cells in vitro. Inhibition of Wnt signaling decreases, and overexpression of canonical Wnt1 increases the hamatoendothelial differentiation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Scheller M, Huelsken J, Rosenbauer F, et al. Hematopoietic stem cell and multilineage defects generated by constitutive beta-catenin activation. Nat Immunol. 2006;7:1037–1047. doi: 10.1038/ni1387. [DOI] [PubMed] [Google Scholar]
  • 32.Kirstetter P, Anderson K, Porse BT, et al. Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block. Nat Immunol. 2006;7:1048–1056. doi: 10.1038/ni1381. [DOI] [PubMed] [Google Scholar]
  • 33.Koch U, Wilson A, Cobas M, et al. Simultaneous loss of beta- and gamma-catenin does not perturb hematopoiesis or lymphopoiesis. Blood. 2008;111:160–164. doi: 10.1182/blood-2007-07-099754. [DOI] [PubMed] [Google Scholar]
  • 34.Jeannet G, Scheller M, Scarpellino L, et al. Long-term, multilineage hematopoiesis occurs in the combined absence of beta-catenin and gamma-catenin. Blood. 2008;111:142–149. doi: 10.1182/blood-2007-07-102558. [DOI] [PubMed] [Google Scholar]
  • 35••.Blank U, Karlsson G, Karlsson S. Signaling pathways governing stem cell fate. Blood. 2008;111:492–503. doi: 10.1182/blood-2007-07-075168. An excellent review of signaling pathways important for HSC regulation. [DOI] [PubMed] [Google Scholar]
  • 36.Bhardwaj G, Murdoch B, Wu D, et al. Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation. Nat Immunol. 2001;2:172–180. doi: 10.1038/84282. [DOI] [PubMed] [Google Scholar]
  • 37.Utsugisawa T, Moody JL, Aspling M, et al. A road map toward defining the role of Smad signaling in hematopoietic stem cells. Stem Cells. 2006;24:1128–1136. doi: 10.1634/stemcells.2005-0263. [DOI] [PubMed] [Google Scholar]
  • 38•.Pimanda JE, Donaldson IJ, de Bruijn MF, et al. The SCL transcriptional network and BMP signaling pathway interact to regulate RUNX1 activity. Proc Natl Acad Sci USA. 2007;104:840–845. doi: 10.1073/pnas.0607196104. This paper showed that Smads can regulate both Scl and Runx1 promoters, and therefore integrates three key determinants of HSC development: Scl, Runx1, and Smad signaling. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Trowbridge JJ, Xenocostas A, Moon RT, Bhatia M. Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation. Nat Med. 2006;12:89–98. doi: 10.1038/nm1339. [DOI] [PubMed] [Google Scholar]
  • 40.de Haan G, Weersing E, Dontje B, et al. In vitro generation of long-term repopulating hematopoietic stem cells by fibroblast growth factor-1. Dev Cell. 2003;4:241–251. doi: 10.1016/s1534-5807(03)00018-2. [DOI] [PubMed] [Google Scholar]
  • 41.Yeoh JS, van Os R, Weersing E, et al. Fibroblast growth factor-1 and 2 preserve long-term repopulating ability of hematopoietic stem cells in serum-free cultures. Stem Cells. 2006;24:1564–1572. doi: 10.1634/stemcells.2005-0439. [DOI] [PubMed] [Google Scholar]
  • 42.Crcareva A, Saito T, Kunisato A, et al. Hematopoietic stem cells expanded by fibroblast growth factor-1 are excellent targets for retrovirus-mediated gene delivery. Exp Hematol. 2005;33:1459–1469. doi: 10.1016/j.exphem.2005.09.001. [DOI] [PubMed] [Google Scholar]
  • 43.Weinreich MA, Lintmaer I, Wang L, et al. Growth factor receptors as regulators of hematopoiesis. Blood. 2006;108:3713–3721. doi: 10.1182/blood-2006-01-012278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44•.Schiedlmeier B, Santos AC, Ribeiro A, et al. HOXB4’s road map to stem cell expansion. Proc Natl Acad Sci USA. 2007;104:16952–16957. doi: 10.1073/pnas.0703082104. The authors examined the gene expression profiling of adult HSCs and embryonic stem-derived hematopoietic cells that express inducible HoxB4 and demonstrated that HoxB4 coordinates the HSC response to extrinsic signals for self-renewal. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Arai F, Hirao A, Ohmura M, et al. Tie2/Angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118:149–161. doi: 10.1016/j.cell.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 46.Nakamura Y, Yahata T, Muguruma Y, et al. Angiopoietin-1 supports induction of hematopoietic activity in human CD34 (−) bone marrow cells. Exp Hematol. 2007;35:1872–1883. doi: 10.1016/j.exphem.2007.08.007. [DOI] [PubMed] [Google Scholar]
  • 47•.Bendall SC, Stewart MH, Menendez P, et al. IGF and FGF cooperatively establish the regulatory stem cell niche of pluripotent human cells in vitro. Nature. 2007;448:1015–1021. doi: 10.1038/nature06027. The authors demonstrated the important roles of IGF-2/IGF-IR and FGF signaling in maintenance of the survival and pluripotency of human embryonic stem cells. [DOI] [PubMed] [Google Scholar]
  • 48.Morisada T, Kubota Y, Urano T, et al. Angiopoietins and angiopoietinlike proteins in angiogenesis. Endothelium. 2006;13:71–79. doi: 10.1080/10623320600697989. [DOI] [PubMed] [Google Scholar]
  • 49.Katoh Y, Katoh M. Comparative integromics on Angiopoietin family members. Int J Mol Med. 2006;17:1145–1149. [PubMed] [Google Scholar]
  • 50•.Zhang CC, Kaba M, Iizuka S, et al. Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation. Blood. 2008;111:3415–3423. doi: 10.1182/blood-2007-11-122119. This study reported a serum-free culture medium containing angiopoietin-like protein 5 and IGFBP2 that supports a 14–20-fold ex-vivo expansion of human cord blood SCID repopulating cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kang YJ, Yang SJ, Park G, et al. A novel function of interleukin-10 promoting self-renewal of hematopoietic stem cells. Stem Cells. 2007;25:1814–1822. doi: 10.1634/stemcells.2007-0002. [DOI] [PubMed] [Google Scholar]
  • 52.Ueno H, Sakita-Ishikawa M, Morikawa Y, et al. A stromal cell-derived membrane protein that supports hematopoietic stem cells. Nat Immunol. 2003;4:457–463. doi: 10.1038/ni916. [DOI] [PubMed] [Google Scholar]
  • 53••.Gupta R, Hong D, Iborra F, et al. NOV (CCN3) functions as a regulator of human hematopoietic stem or progenitor cells. Science. 2007;316:590–593. doi: 10.1126/science.1136031. This study identified NOV/CCN3 from CD34+ human cord blood cells and showed it enhances SRC activity, demonstrating that Nov is a regulator of human hematopoietic stem or progenitor cells. [DOI] [PubMed] [Google Scholar]

RESOURCES