Gene Therapy (2002) 9, 606–612  2002 Nature Publishing Group All rights reserved 0969-7128/02 $25.00 www.nature.com/gt REVIEW Cell fate determination from stem cells

AJ Wagers1, JL Christensen1 and IL Weissman1,2 1Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; and 2Department of Developmental , Stanford University School of Medicine, Stanford, CA, USA

In the adult, tissue-specific stem cells are thought to be that influence fate, taking as a primary example responsible for the replacement of differentiated cells within the cell fate determination of hematopoietic stem cells. continuously regenerating tissues, such as the liver, skin, Gene Therapy (2002) 9, 606–612. DOI: 10.1038/sj/gt/3301717 and blood system. In this review, we will consider the factors

Keywords: stem cells; progenitors; hematopoiesis; self-renewal; differentiation; migration

Introduction to hematopoietic stem and and characterized in the fetal liver (FL).9–11 FL HSC differ progenitor cells from adult HSC both phenotypically and functionally. Unlike adult LT-HSC, FL HSC express the cell surface The study of stem cells within the hematopoietic system markers Mac-1 and AA4.1, are cycling rapidly, and give began nearly 40 years ago, with the demonstration by Till a more robust and rapid reconstitution of irradiated and McCulloch that a single precursor cell exists in the hosts.9–15 While adult HSC readout in CFU-S assays at (BM) of adult animals that is capable of day 12, FL HSC form visible colonies at day 8.16 Finally, both extensive self-renewal and multi-lineage differen- while the pool is composed mostly of tiatiation.1–3 This finding gave rise to the hypothesis that, short-term and multipotent progenitors,6 most stem cells during hematopoiesis, a small population of multipotent, in FL are thought to be LT-HSC.15 self-renewing, clonogenic stem cells divides to generate The oligopotent progenitors downstream of FL HSC progeny cells, each progressively more restricted in also differ slightly from their adult counterparts (Figure developmental potential, until ultimately mature effector 1). Unlike adult CMP, FL CMP, but not GMP or MEP, cells are formed. Careful regulation of cell fate decisions give rise to B cells (but never T cells) both in vivo and in in the stem cell pool profoundly influences the pro- vitro.16 Likewise, while adult CLP have no macrophage duction of mature blood cells, and is required both to activity in vivo or in vitro, FL CLP form macrophage (but maintain homeostasis and to enable rapid and robust never granulocyte, erythrocyte, or megakaryocyte) colon- responses to physiologic stresses, including blood loss, ies in methylcellulose and stromal cell cultures.17 In infection and injury. addition to these differences in the lineage relationships Hematopoietic stem cells (HSC) are rare cells within of fetal progenitors, several blood cells that cannot be the BM or fetal liver which can both self-renew and dif- generated by adult HSC, including B-1a B cells and V␥3+ ferentiate to form all of the major blood cell lineages. The and V␥4+ T cells, arise during fetal hematopoiesis from HSC pool has been characterized phenotypically and is fetal HSC.18–20 separable into distinct subpopulations based on both The prospective isolation of HSC and progenitor cells phenotype and function (Figure 1).4,5 Long-term reconsti- in mice has provided key insights into the molecular and tuting HSC (LT-HSC) have the greatest self-renewal cellular regulators of hematopoiesis. Careful control of capacity and give rise to all hematopoietic lineages HSC developmental decisions is clearly essential for throughout life.4,6 The immediate progeny of LT-HSC are blood cell homeostasis. As discussed below, the develop- short-term (ST-) HSC, which also generate all hematopo- mental options of HSC include not only self-renewal and ietic lineages, but do so for only 8–10 weeks. Further differentiation, but also apoptosis and migration. The downstream are lineage-restricted oligopotent progenitor synthesis of signals from diverse hematopoietic regu- cells, including both lymphoid (common lymphoid pro- lators ultimately determines the fate of dividing HSC. genitors, CLP) and myeloid (common myeloid progeni- tors, CMP; granulocyte–monocyte progenitors, GMP; and Self-renewal versus differentiation in the megakaryocyte–erythrocyte progenitors, MEP) restricted precursors.7,8 control of HSC fate Multipotent, clonogenic HSC also have been identified Most studies of HSC cell fate determination focus on the choice between stem cell self-renewal, in which the daughter(s) of a dividing HSC retains the properties of Correspondence: IL Weissman, Department of Pathology, Beckman B257, the parent cell, and differentiation, in which the daughter Stanford University School of Medicine, Stanford, CA 94305, USA cell(s) becomes restricted in its potential and diminished The first two authors contributed equally to this work in its capacity for self-renewal. Both stochastic and Cell fate determination from stem cells AJ Wagers et al 607

Figure 1 Hematopoietic cell ontogeny. This illustration describes the development of mature blood cells through the sequential restriction of the cell fate potential of oligopotent progenitor cells derived from multipotent HSC. The stem cell pool is comprised of both LT- and ST-HSC. LT-HSC have a significantly greater potential for self-renewal than ST-HSC. Oligopotent progenitor cells do not self-renew appreciably. Dashed lines indicate developmen- tal pathways that operate only during fetal hematopoiesis (see text for details). instructive models have been proposed to describe this genitor cells has demonstrated that HSC constitutively choice, and while both models invoke a key role for cyto- express low levels of many lineage-specific cytokine kines, different functions are assigned to these factors. receptors and transcription factors.7,24,28 Among progeni- Under the stochastic model, HSC randomly commit to tors, lineage commitment is accompanied by loss of either self-renew or differentiate. Cytokines present in the expression of genes associated with unrelated lineages,7 BM milieu do not direct this choice per se, but do allow indicating that hematopoietic maturation may be survival and proliferation of the cells that ultimately mediated by sequential restriction of gene expression. develop into mature lineages. Under the instructive Promiscuous gene expression by HSC may provide a model, the particular cytokines to which HSC are framework for stochastic fluctuations in expression of exposed directly determine the outcome of each mitosis, signaling or transcriptional complexes, which ultimately whether self-renewing or differentiating. are amplified or repressed to cement lineage choice.26 While several studies support an instructive role for Specific molecular mediators that influence HSC self- cytokines in the maturation of terminally differentiated renewal versus differentiation decisions have only cells from progenitor and precursor cells,21–24 the initial recently been identified. Increased HSC self-renewal divisions of HSC are likely to involve stochastic fate appears to be associated with HSC-intrinsic over- choices.25 In fact, it has been postulated that stochastic expression of HoxB4,29,30 and induction of the Wnt (Reya determination of HSC cell fate may be important for HSC et al, submitted), Notch,31,32 and Sonic hedgehog (Shh)33 function, in that this scheme allows for the maintenance signaling pathways. Extrinsic signals that promote HSC of production of all blood cell lineages even in the face self-renewal have been more difficult to identify, as of substantial demand for one particular lineage.26 Sup- shown by the notorious lack of effective means of porting the stochastic model for HSC differentiation, in expanding HSC in vitro, while maintaining their self- vitro culture of Bcl-2 transgenic HSC with steel factor renewing properties. Currently published literature34–38 (SLF) alone induces these cells to proliferate, but does not points to several cytokines, including SLF, Flt3L, throm- preserve their primitive status.27 This induction of HSC bopoietin, IL-3, and IL-6, as working individually or syn- differentiation in the absence of any overt differentiating ergistically to permit one to two self-renewing divisions signals, suggests that lineage choice need not be initiated in liquid culture. However, long-term maintenance of by particular cytokine signals. Similarly, lineage-restric- HSC function clearly requires more complex systems and ted progenitors can develop from HSC at normal fre- may rely on as yet uncharacterized factors provided by quency, even in the absence of lineage-specific cytokines. stromal support cells (see below). For example, although maturation of lymphocytes from A central issue in HSC fate choices is the consideration CLP requires IL-7, and IL-7 induces CLP proliferation in of symmetry within stem cell divisions. Are the individ- vitro, CLP develop at normal frequency in mice deficient ual mitoses of HSC obligately symmetric (always produc- ␥ in expression of the common signaling receptor ( c)of ing two daughter cells with identical fates), obligately the IL-7 receptor.8 asymmetric (always producing two daughters with dis- RT-PCR analysis of rigorously purified stem and pro- tinct fates), or variably symmetric and asymmetric?

Gene Therapy Cell fate determination from stem cells AJ Wagers et al 608 Because in the steady state the total number of BM HSC and a unique cloned marrow stromal line (ATF024), remains constant,4 half of all HSC divisions, at the popu- derived from murine FL, can maintain murine HSC lation level, must be self-renewing. However, HSC must activity in culture for weeks.42,43 On the other hand, while also have the capacity to expand their numbers, by freshly purified marrow myofibroblasts facilitate hemato- increasing the frequency of self-renewal, in response to poietic cell proliferation in vitro, marrow endothelial cells hematopoietic stress. Thus, HSC divisions cannot be obli- may inhibit hematopoiesis.51 gately asymmetric, as this would prohibit HSC expan- Whether direct contact of HSC with stromal cells is sion. Experiments in vitro have provided evidence for required for their effects on HSC is somewhat contro- both symmetric and asymmetric divisions of cultured versial. ATF024 cells fail to support long-term mainte- HSC,34,39 suggesting that the outcomes of HSC mitoses nance of primitive human progenitor cells in non-contact are variable, but the relevance of these findings to HSC cultures,42 but can maintain SCID-repopulating activity divisions in vivo remains to be determined. of umbilical cord blood CD34+ cells in transwell assays.52 Some stromal cell cultures support hematopoiesis in non- contact conditions, but inhibit it in direct co-cultures.53 Programmed cell death as a determinant of The differences in outcome of these in vitro cultures may HSC fate relate to differences in the source or type of HSC and/or stromal elements studied, or may depend on other differ- The hematopoietic compartment must produce large ences, such as the presence or absence of cytokines, numbers of cells daily (~2.4 x 108 in the mouse40), and serum, or other factors, in these culture systems. must further scale up this effort following injury or hem- The emergence during development of distinct blood atopoietic stress. Programmed cell death (apoptosis) of cell lineages, dependent on particular stromal microen- HSC has recently been shown to play an important role vironments, supports the importance of stromal cell in regulating HSC number. Transgenic mice with interactions to HSC fate choices in vivo. For example, the enforced overexpression of the proto-oncogene Bcl-2 + + + first TCR thymocytes are V␥3 and V␥4 .20 These T cells exhibit increased steady-state HSC frequency and arise only from FL HSC and absolutely require the fetal increased competitive repopulation potential.41 When thymic environment for their generation.19 The involve- transplanted into irradiated wild-type recipients, Bcl-2 ment of stromal cells in HSC function in vivo is further transgenic HSC give increasing hematopoietic readout evident from studies of naturally occurring mutations of over time, and in vitro culture of Bcl-2 transgenic HSC the Steel (Sl) locus in mice. BM transplantation has dem- with SF alone is sufficient to prevent HSC death.27 These onstrated that defective hematopoiesis in these mice data demonstrate that regular turnover of BM HSC results from a specificdeficiency (loss or impairment of through programmed cell death is an important regulator SLF expression) within the BM microenvironment.54,55 of HSC cell fate and hematopoietic homeostasis. The HSC from Sl mutant mice reconstitute the ablated BM of inability of Bcl-2 transgenic HSC to be removed from BM wild-type animals, while wild-type HSC fail to reconsti- niches by apoptosis may underlie the competitive advan- tute hematopoiesis in transplanted Sl mice and BM stro- tage of these HSC in wild-type transplant recipients. mal cells from these mice fail to support hematopoiesis in vitro.54–58 Finally, in recent studies, co-transplantation Adhesion and migration as regulators of of stromal elements with either adult or fetal BM HSC HSC fate significantly enhanced engraftment of preimmune fetal sheep,59 providing further evidence for support of HSC In the BM, hematopoietic cell development is thought to function by marrow stromal cells. occur in various specialized microenvironments, called Extensive and complex interactions within the BM ‘niches’. At present, support for the niche hypothesis microenvironment clearly influence the cell fate decisions remains indirect, and these specialized areas of BM have of HSC; however, these interactions do not necessarily never been described at the ultrastructural level. Evi- imply that HSC remain permanently lodged in specific dence for the existence of HSC-supporting niches derives BM niches. On the contrary, recent data suggests signifi- from several lines of investigation, including the lack of cant dynamism among HSC niches, and supports the in vitro conditions that can fully support HSC self- notion that migration through the bloodstream to distinct renewal, the infrequency of isolation of hematopoiesis BM sites or to other peripheral tissues is an additional supporting stromal cell lines,42,43 and the low level of BM developmental alternative for HSC (Figure 2). The impor- engraftment that can be achieved following intravenous tance of HSC migration is clearly evident during fetal injection of HSC, unless those HSC are given in extraordi- hematopoiesis. HSC arise first in the embryonic yolk narily high numbers or unless the BM is ablated by sac60–65 and aorta/gonad/mesonephros (AGM) region,66–69 irradiation or cytotoxic drugs.44,45 The hypothetical func- and are thought to migrate to and seed the fetal tion of the HSC niche is to provide HSC survival and liver.63,70,71 FL HSC subsequently engraft the newly self-renewal factors, either through direct contact with developing fetal BM. In the adult, the ability of hemato- HSC or through secreted factors. HSC niches are likely poietic cells to use the blood as a transit mechanism for formed from a subset of bone marrow stromal cells, migration from one organ to another was demonstrated which may play a further role in ‘translating’ external more than 40 years ago,72–74 but the realization that such signals to influence HSC developmental decisions.46 migration in the bloodstream is a property not only of BM stromal cells can play either supportive or inhibi- mature blood cells, but also of hematopoietic stem and tory roles in the maintenance of primitive hematopoietic progenitor cells, occurred more recently. HSC and pro- progenitors. Co-culture of human CD34+CD38Ϫ/lo human genitor cells are constantly present at low frequency in progenitor cells with marrow-derived stromal cells the blood of normal animals,75,76 and these blood-borne appears to promote maintenance of the primitive state,47–50 HSC can, at least, re-engraft BM to functionally contrib-

Gene Therapy Cell fate determination from stem cells AJ Wagers et al 609

Figure 2 Transit of HSC and progenitor cells through the bloodstream. BM-derived HSC and progenitor cells rapidly and constitutively enter the bloodstream and may traffic to distinct hematopoietic and non-hematopoietic tissues, as well as back to BM. Thus, HSC are constantly present in most, if not all, tissues of the body, and may represent a source of pluripotent cells that participate in the repair of damaged or degenerating tissues. These cells also may be a source of contaminating hematopoietic activity in ‘transdifferentiation’ assays. ute to on-going hematopoiesis.76,110 In genetically marked cell transdifferentiation, some caution is warranted in parabiotic mice, which share a common blood circulation, considering these findings. First, it is important to significant cross-engraftment of BM HSC is observed in remember that BM contains several stem cell popu- the absence of BM ablation following as little as 2–3 lations, including mesenchymal and hematopoietic stem weeks of parabiosis (A. Wagers, unpublished cells, and that mesenchymal stem cells are defined by observation). This data suggests that constitutive, low- their potential to give rise to muscle, cartilage, bone, fat level HSC mobilization occurs in normal, unconditioned and stroma.85 Furthermore, because circulating tissue- animals. Surprisingly, the normal flux of HSC and pro- specific stem cells, including HSC, have been described genitor cells in the bloodstream of normal animals for several lineages, such blood-borne stem cells rep- appears to be quite high, as predicted by their extremely resent significant potential sources of contamination in short residence time in the blood, such that the effective transdifferentiation assays.75,76,105–107,110 Finally, some pool of circulating cells may include more than 30 000 claims of transdifferentiation involve cells grown in cul- stem and progenitor cells.110 While the full effects of ture, using high concentrations of growth factors, which constitutive migration on HSC developmental decisions may impart a more primitive potential to the cultured remain to be determined, the possibility that HSC may cells.103,104 For example, fetal murine primordial germ find themselves in unique microenvironments in tissues cells grown in basic fibroblast growth factor (bFGF) and outside of the BM raises the interesting possibility that leukemia inhibitory factor (LIF) take on embryonic stem itinerant HSC or their progeny may contribute to the (ES) cell qualities.108 Thus, to demonstrate true transdif- regeneration of non-hematopoietic, as well as hematopo- ferentiation, it is necessary that the stem cells in question ietic cells (see below). Furthermore, this finding provides are prospectively isolated and characterized, able to self- a physiological framework for understanding the renew and differentiate into all relevant cell types at the efficiency of bone marrow transplantation and the exist- clonal level, and that the transdifferentiated cells gener- ence of induced HSC mobilization, which occurs follow- ated by these stem cells are demonstrated to be fully ing myeloablation and/or increases in circulating functional in the target tissue. cytokine/chemokine levels,77 as these transplanted or Despite the caveats discussed above, good evidence for mobilized HSC likely exploit similar mechanisms of a greater than expected plasticity of adult stem cells is migration as those employed by constitutively circulating emerging, and this research has exciting implications for HSC in normal animals. potential clinical application to the repair of damaged or degenerating tissues. Highly purified HSC have been shown able to differentiate into colonies of hepatocytes Plasticity of stem cell fate Ϫ Ϫ when introduced into fumarylacetoacetate hydrolase / Recently much attention has been paid to stem cells in the (FAHϪ/Ϫ) mice.109 HSC-derived hepatocytes restored adult organism, and the possibility that stem cells may liver function in transplanted FAHϪ/Ϫ mice, and all hep- transdifferentiate into cell types of unrelated tissues. atic engrafted recipients also showed hematopoietic Stem cells or cells with stem cell-like qualities have been reconstitution. Whether hepatic regeneration is a normal isolated or described in several tissues, including neural role of HSC, or whether this function is induced by the (CNS78–80 and PNS81), pancreatic,82 epidermal,83 mes- mutant background of the recipient mice, remains to be enchymal,84-87 hepatic,88,89 bone,86 muscle,90 and endo- determined, but these findings certainly demand further thelial91,92 tissues. In recent years, published reports have inquiry into the possibility of HSC mediated therapy for indicated that cells found in BM are capable of giving liver degenerative disorders. rise to endothelial precursors,93 brain microglia and mac- 94–96 97–99 98,100 roglia, skeletal muscle, cardiac muscle, hep- Concluding remarks atic cells,101,102 and mesenchymal progeny.86,87 Other groups have reported that cultured stem cells of neural103 Hematopoiesis is regulated by the developmental or muscle104 origin can give rise in vivo to hematopoietic decisions of HSC, which include self-renewal, differen- cells. While these results may constitute evidence of stem tiation, death, and migration. Much has been learned

Gene Therapy Cell fate determination from stem cells AJ Wagers et al 610 about the molecular and cellular regulators that influence 20 Havran WL, Allison JP. Developmentally ordered appearance HSC fate decisions, and recent data suggest that these of thymocytes expressing different T-cell antigen receptors. cells may be significantly more dynamic than previously Nature 1988; 335: 443–445. appreciated. Exciting new studies have called into ques- 21 Metcalf D, Burgess AW. Clonal analysis of com- tion long-standing beliefs regarding the tissue-restriction mitment of granulocyte or macrophage production. J Cell Phy- siol 1982; 111: 275–283. of HSC, and of adult tissue-specific stem cells in general, 22 Kaushansky K et al. Thrombopoietin, the Mp1 ligand, is essen- suggesting the possibility that these stem cells may some- tial for full megakaryocyte development. Proc Natl Acad Sci day be useful targets for therapeutic strategies aimed at USA 1995; 92: 3234–3238. tissue regeneration and repair. 23 Kondo M et al. Bcl-2 rescues T lymphopoiesis, but not B or NK cell development, in common gamma chain-deficient mice. References Immunity 1997; 7: 155–162. 24 Kondo M et al. Cell-fate conversion of lymphoid-committed 1 Wu A, Till J, Siminovitch L, McCulloch E. A cytological study progenitors by instructive actions of cytokines. Nature 2000; of the capacity for differentiation of normal hemopoietic col- 407: 383–386. ony-forming cells. J Cell Physiol 1967; 69: 177–184. 25 Ogawa M. Differentiation and proliferation of hematopoietic 2 Wu A, Till J, Siminovitch L, McCulloch E. Cytological evidence stem cells. Blood 1993; 81: 2844–2853. for a relationship between normal hematopoietic colony-for- 26 Enver T, Heyworth CM, Dexter TM. Do stem cells play dice? ming cells and cells of the lymphoid system. J Exp Med 1968; Blood 1998; 92: 348–351. 127: 455–467. 27 Domen J, Weissman IL. Hematopoietic stem cells need two sig- 3 Till J, McCulloch E. A direct measurement of the radiation sen- nals to prevent apoptosis; BCL-2 can provide one of these, sitivity of normal mouse bone marrow cells. Radiat Res 1961; Kitl/c-Kit signaling the other. J Exp Med 2000; 192: 1707–1718. 14: 1419–1430. 28 Nakauchi H, Takano H, Ema H, Osawa M. Further characteriz- 4 Morrison SJ, Weissman IL. The long-term repopulating subset ation of CD34-low/negative mouse hematopoietic stem cells. of hematopoietic stem cells is deterministic and isolatable by Ann NY Acad Sci 1999; (872) 57–66. phenotype. Immunity 1994; 1: 661–673. 29 Thorsteinsdottir U, Sauvageau G, Humphries RK. Enhanced in 5 Spangrude GJ, Heimfeld S, Weissman IL. Purification and vivo regenerative potential of HOXB4-transduced hematopo- characterization of mouse hematopoietic stem cells. Science ietic stem cells with regulation of their pool size. Blood 1999; 1988; 241:58–62. 94: 2605–2612. 6 Morrison SJ et al. Identification of a lineage of multipotent hem- 30 Sauvageau G et al. Overexpression of HOXB4 in hematopoietic atopoietic progenitors. Development 1997; 124: 1929–1939. cells causes the selective expansion of more primitive popu- 7 Akashi K, Traver D, Miyamoto T, Weissman IL. A clonogenic lations in vitro and in vivo. Genes Dev 1995; 9: 1753–1765. common myeloid progenitor that gives rise to all myeloid lin- 31 Karanu FN et al. The notch ligand jagged-1 represents a novel eages. Nature 2000; 404: 193–197. growth factor of human hematopoietic stem cells. J Exp Med 8 Kondo M, Weissman IL, Akashi K. Identification of clonogenic 2000; 192: 1365–1372. common lymphoid progenitors in mouse bone marrow. Cell 32 Varnum-Finney B et al. Pluripotent, cytokine-dependent, hema- 1997; 91: 661–672. topoietic stem cells are immortalized by constitutive Notch1 9 Morrison SJ, Hemmati HD, Wandycz AM, Weissman IL. The signaling. Nat Med 2000; 6: 1278–1281. purification and characterization of fetal liver hematopoietic 33 Bhardwaj G et al. Sonic hedgehog induces the proliferation of stem cells. Proc Natl Acad Sci USA 1995; 92: 10302–10306. primitive human hematopoietic cells via BMP regulation. Nat 10 Jordan CT. Cellular and developmental properties of fetal hem- Immunol 2001; 2: 172–180. atopoietic stem cells. Cell 1990; 61: 953–963. 11 Jordan CT et al. Long-term repopulating abilities of enriched 34 Goff JP, Shields DS, Greenberger JS. Influence of cytokines on the growth kinetics and immunophenotype of daughter cells fetal liver stem cells measured by competitive repopulation. + + Exp Hematol 1995; 23: 1011–1015. resulting from the first division of single CD34( )Thy-1( )lin- 12 Holyoake TL, Nicolini FE, Eaves CJ. Functional differences cells. Blood 1998; 92: 4098–4107. between transplantable human hematopoietic stem cells from 35 Ema H, Takano H, Sudo K, Nakauchi H. In vitro self-renewal fetal liver, cord blood, and adult marrow. Exp Hematol 1999; division of hematopoietic stem cells. J Exp Med 2000; 192: 27: 1418–1427. 1281–1288. 13 Rebel VI, Miller CL, Eaves CJ, Lansdorp PM. The repopulation 36 Yagi M et al. Sustained ex vivo expansion of hematopoietic stem potential of fetal liver hematopoietic stem cells in mice exceeds cells mediated by thrombopoietin. Proc Natl Acad Sci USA 1999; that of their liver adult bone marrow counterparts. Blood 1996; 96: 8126–8131. 87: 3500–3507. 37 Bryder D, Jacobsen SE. Interleukin-3 supports expansion of 14 Pawliuk R, Eaves C, Humphries RK. Evidence of both long-term multilineage repopulating activity after multiple ontogeny and transplant dose-regulated expansion of hemato- stem cell divisions in vitro. Blood 2000; 96: 1748–1755. poietic stem cells in vivo. Blood 1996; 88: 2852–2858. 38 Luskey BD, Rosenblatt M, Zsebo K, Williams DA. Stem cell 15 Harrison DE et al. Relative to adult marrow, fetal liver repopu- factor, interleukin-3, and interleukin-6 promote retroviral- lates nearly five times more effectively long-term than short- mediated gene transfer into murine hematopoietic stem cells. term. Exp Hematol 1997; 25: 293–297. Blood 1992; 80: 396–402. 16 Traver D et al. Fetal liver myelopoiesis occurs through distinct, 39 Huang S et al. Symmetry of initial cell divisions among primi- prospectively isolatable progenitor subsets. Blood 2001; 98: tive hematopoietic progenitors is independent of ontogenic age 627–635. and regulatory molecules. Blood 1999; 94: 2595–2604. 17 Mebius RE et al. The fetal liver counterpart of adult common 40 Cheshier SH, Morrison SJ, Liao X, Weissman IL. In vivo pro- lymphoid progenitors gives rise to all lymphoid lineages, liferation and cell cycle kinetics of long-term self-renewing cd45(+)cd4(+)cd3(-) cells, as well as macrophages. J Immunol hematopoietic stem cells. Proc Natl Acad Sci USA 1999; 96: 2001; 166: 6593–6601. 3120–3125. 18 Hayakawa K, Hardy RR, Parks DR, Herzenberg LA. The ‘Ly- 41 Domen J, Cheshier SH, Weissman IL. The role of apoptosis in 1B’ cell subpopulation in normal immunodefective, and auto- the regulation of hematopoietic stem cells: overexpression of immune mice. J Exp Med 1983; 157: 202–218. Bcl-2 increases both their number and repopulation potential. 19 Ikuta K et al. A developmental switch in thymic lymphocyte J Exp Med 2000; 191: 253–264. maturation potential occurs at the level of hematopoietic stem 42 Thiemann FT et al. The murine stromal cell line AFT024 acts cells. Cell 1990; 62: 863–874. specifically on human CD34+CD38- progenitors to maintain

Gene Therapy Cell fate determination from stem cells AJ Wagers et al 611 primitive function and immunophenotype in vitro. Exp Hematol 64 Moore MA, Metcalf D. Ontogeny of the haemopoietic system: 1998; 26: 612–619. yolk sac origin of in vivo and in vitro colony forming cells in 43 Moore KA, Ema H, Lemischka IR. In vitro maintenance of the developing mouse . Br J Haematol 1970; 18: 279–296. highly purified, transplantable hematopoietic stem cells. Blood 65 Huang H, Auerbach R. Identification and characterization of 1997; 89: 4337–4347. hematopoietic stem cells from the yolk sac of the early mouse 44 Harrison DE. Competitive repopulation in unirradiated normal embryo. Proc Natl Acad Sci USA 1993; 90: 10110–10114. recipients. Blood 1993; 81: 2473–2474. 66 Garcia-Porrero JA, Godin IE, Dieterlen-Lievre F. Potential 45 Takada A, Takada Y, Ambrus JL. Proliferation of donor spleen intraembryonic hemogenic sites at pre-liver stages in the and bone-marrow cells in the spleens and bone marrows of mouse. Anat Embryol (Berl) 1995; 192: 425–435. unirradiated and irradiated adult mice. Proc Soc Exp Biol Med 67 Medvinsky A, Dzierzak E. Definitive hematopoiesis is auton- 1971; 136: 222–226. omously initiated by the AGM region. Cell 1996; 86: 897–906. 46 Muller-Sieburg CE, Deryugina E. The stromal cells’ guide to 68 Muller AM et al. Development of the stem cell universe. Stem Cells 1995; 13: 477–486. activity in the mouse embryo. Immunity 1994; 1: 291–301. 47 Koller MR, Palsson MA, Manchel I, Palsson BO. Long-term cul- 69 Sanchez MJ, Holmes A, Miles C, Dzierzak E. Characterization ture-initiating cell expansion is dependent on frequent medium of the first definitive hematopoietic stem cells in the AGM and exchange combined with stromal and other accessory cell liver of the mouse embryo. Immunity 1996; 5: 513–525. effects. Blood 1995; 86: 1784–1793. 70 Johnson GR, Moore MA. Role of stem cell migration in 48 Gan OI, Murdoch B, Larochelle A, Dick JE. Differential mainte- initiation of mouse foetal liver haemopoiesis. Nature 1975; 258: nance of primitive human SCID-repopulating cells, clonogenic 726–728. progenitors, and long-term culture-initiating cells after incu- 71 Houssaint E. Differentiation of the mouse hepatic primordium. bation on human bone marrow stromal cells. Blood 1997; 90: II. Extrinsic origin of the haemopoietic cell line. Cell Differ 1981; 641–650. 10: 243–252. 49 Breems DA et al. Stroma-contact prevents loss of hematopoietic 72 Gowans J, Knight E. The route of recirculation of lymphocytes + stem cell quality during ex vivo expansion of CD34 mobilized in the rat. Phil Trans R Soc Lond B 1964; 159: 257–282. peripheral blood stem cells. Blood 1998; 91: 111–117. 73 Gowans J. Life-span, recirculation, and transformation of lym- 50 Bennaceur-Griscelli A et al. Stromal cells retard the differen- phocytes. Inter Rev Exp Path 1966; 5:1–24. + tiation of CD34( )CD38(low/neg) human primitive progeni- 74 Weissman IL. Thymus cell migration. J Exp Med 1967; 126: tors exposed to cytokines independent of their mitotic history. 291–304. Blood 2001; 97: 435–441. 75 Morrison SJ, Wright DE, Weissman IL. Cyclophosphamide/ 51 Sitnicka E, Wang QR, Tsai S, Wolf NS. Support versus inhi- granulocyte colony-stimulating factor induces hematopoietic bition of hematopoiesis by two characterized stromal cell types. stem cells to proliferate prior to mobilization. Proc Natl Acad Stem Cells 1995; 13: 655–665. Sci USA 1997; 94: 1908–1913. 52 Lewis ID et al. Umbilical cord blood cells capable of engrafting 76 Fleming WH et al. Steel factor influences the distribution and in primary, secondary, and tertiary xenogeneic hosts are pre- activity of murine hematopoietic stem cells in vivo. Proc Natl served after ex vivo culture in a noncontact system. Blood 2001; Acad Sci USA 1993; 90: 3760–3764. 97: 3441–3449. 77 Papayannopoulou T. Hematopoietic stem/progenitor cell 53 Verfaillie CM, Catanzaro P. Direct contact with stroma inhibits mobilization. A continuing quest for etiologic mechanisms. proliferation of human long-term culture initiating cells. Leuke- Ann NY Acad Sci 1999; 872: 187–197. mia 1996; 10: 498–504. 78 Gage FH et al. Survival and differentiation of adult neuronal 54 McCulloch E et al. The cellular basis of the genetically deter- progenitor cells transplanted to the adult brain. Proc Natl Acad mined hemopoietic defect in anemic mice of genotype Sl-Sld. Sci USA 1995; 92: 11879–11883. Blood 1965; 26: 399–410. 79 Ray J, Peterson DA, Schinstine M, Gage FH. Proliferation, dif- 55 Bernstein A et al. The murine W/c-kit and Steel loci and the ferentiation, and long-term culture of primary hippocampal control of hematopoiesis. Semin Hematol 1991; 28: 138–142. neurons. Proc Natl Acad Sci USA 1993; 90: 3602–3606. 56 Fried W et al. Studies on the defective haematopoietic microen- vironment of Sl/Sl d mice. Br J Haematol 1973; 24: 643–650. 80 Palmer TD, Ray J, Gage FH. FGF-2-responsive neuronal pro- 57 Dexter TM, Moore MA. In vitro duplication and ‘cure’ of genitors reside in proliferative and quiescent regions of the haemopoietic defects in genetically anaemic mice. Nature 1977; adult rodent brain. Mol Cell Neurosci 1995; 6: 474–486. 269: 412–414. 81 Morrison SJ, White PM, Zock C, Anderson DJ. Prospective 58 Barker JE. Early transplantation to a normal microenvironment identification, isolation by flow cytometry, and in vivo self- prevents the development of Steel hematopoietic stem cell renewal of multipotent mammalian neural crest stem cells. Cell defects. Exp Hematol 1997; 25: 542–547. 1999; 96: 737–749. 59 Almeida-Porada G, Flake AW, Glimp HA, Zanjani ED. 82 Ramiya VK et al. Reversal of insulin-dependent diabetes using Cotransplantation of stroma results in enhancement of islets generated in vitro from pancreatic stem cells. Nat Med engraftment and early expression of donor hematopoietic stem 2000; 6: 278–282. cells in utero. Exp Hematol 1999; 27: 1569–1575. 83 Jones PH, Watt FM. Separation of human epidermal stem cells 60 Wong PM, Chung SW, Chui DH, Eaves CJ. Properties of the from transit amplifying cells on the basis of differences in inte- earliest clonogenic hemopoietic precursors to appear in the grin function and expression. Cell 1993; 73: 713–724. developing murine yolk sac. Proc Natl Acad Sci USA 1986; 83: 84 Pittenger MF et al. Multilineage potential of adult human mes- 3851–3854. enchymal stem cells. Science 1999; 284: 143–147. 61 Yoder MC et al. Characterization of definitive lymphohemato- 85 Pittenger MF, Mosca JD, McIntosh KR. Human mesenchymal poietic stem cells in the day 9 murine yolk sac. Immunity 1997; stem cells: progenitor cells for cartilage, bone, fat and stroma. 7: 335–344. Curr Top Microbiol Immunol 2000; 251:3–11. 62 Weissman I, Papaioannou V, Gardner R. Fetal hematopoietic 86 Pereira RF. Cultured adherent cells from marrow can serve as origins of the adult hematolymphoid system. In: Clarkson B, long-lasting precursor cells for bone, cartilage, and lung in Mark P, Till J (eds). Differentiation of Normal and Neoplastic Hem- irradiated mice. Proc Natl Acad Sci USA 1995; 92: 4857–4861. atopoietic Cells. Cold Spring Harbor Laboratory Press: New 87 Lazarus HM et al. Ex vivo expansion and subsequent infusion York, 1978, pp 33–47. of human bone marrow-derived stromal progenitor cells 63 Toles JF et al. Hemopoietic stem cells in murine embryonic yolk (mesenchymal progenitor cells): implications for therapeutic sac and peripheral blood. Proc Natl Acad Sci USA 1989; 86: use. Bone Marrow Transplant 1995; 16: 557–564. 7456–7459. 88 Overturf K et al. Serial transplantation reveals the stem-cell-

Gene Therapy Cell fate determination from stem cells AJ Wagers et al 612 like regenerative potential of adult mouse hepatocytes. Am J 99 Gussoni E et al. Dystrophin expression in the mdx mouse Pathol 1997; 151: 1273–1280. restored by stem cell transplantation. Nature 1999; 401: 390–394. 89 Theise ND, et al.Badve S, Saxena R, Henegariu O, Sell S, Craw- 100 Orlic D. Transplanted adult bone marrow cells repair myocar- ford JM, Krause DS. Derivation of hepatocytes from bone mar- dial infarcts in mice. Ann NY Acad Sci 2001; 938: 221–229. row cells in mice after radiation-induced myeloablation. Hepa- 101 Alison MR et al. Hepatocytes from non-hepatic adult stem cells. tology 2000; 31: 235–240. Nature 2000; 406: 257. 90 Pavlath GK, et al.Thaloor D, Rando TA, Cheong M, English 102 Petersen BE et al. Bone marrow as a potential source of hepatic AW, Zheng B. Heterogeneity among muscle precursor cells in oval cells. Science 1999; 284: 1168–1170. adult skeletal muscles with differing regenerative capacities. 103 Bjornson CR et al. Turning brain into blood: a hematopoietic Dev Dyn 1998; 212: 495–508. fate adopted by adult neural stem cells in vivo. Science 1999; 91 Asahara T et al. Isolation of putative progenitor endothelial 283: 534–537. cells for angiogenesis. Science 1997; 275: 964–967. 104 Jackson KA, Mi T, Goodell MA. Hematopoietic potential of 92 Takahashi T et al. Ischemia- and cytokine-induced mobilization stem cells isolated from murine skeletal muscle. Proc Natl Acad of bone marrow-derived endothelial progenitor cells for neo- Sci USA 1999; 96: 14482–14486. vascularization. Nat Med 1999; 5: 434–438. 105 Peichev M et al. Expression of VEGFR-2 and AC133 by circulat- 93 Asahara T et al. Bone marrow origin of endothelial progenitor ing human CD34(+) cells identifies a population of functional cells responsible for postnatal vasculogenesis in physiological endothelial precursors. Blood 2000; 95: 952–958. and pathological neovascularization. Circ Res 1999; 85: 221–228. 106 Shi Q et al. Evidence for circulating bone marrow-derived 94 Mezey E et al. Turning blood into brain: cells bearing neuronal endothelial cells. Blood 1998; 92: 362–367. antigens generated in vivo from bone marrow. Science 2000; 290: 107 Kuznetsov SA et al. Circulating skeletal stem cells. J Cell Biol 1779–1782. 2001; 153: 1133–1140. 95 Brazelton TR, Rossi FM, Keshet GI, Blau HM. From marrow to 108 Matsui Y, Zsebo K, Hogan BL. Derivation of pluripotential brain: expression of neuronal phenotypes in adult mice. Science embryonic stem cells from murine primordial germ cells in cul- 2000; 290: 1775–1779. ture. Cell 1992; 70: 841–847. 96 Eglitis MA, Mezey E. Hematopoietic cells differentiate into 109 Lagasse E et al. Purified hematopoietic stem cells can differen- both microglia and macroglia in the brains of adult mice. Proc tiate into hepatocytes in vivo. Nat Med 2000; 6: 1229–1234. Natl Acad Sci USA 1997; 94: 4080–4085. 97 Ferrari G et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science 1998; 279: 1528–1530. Reference added in proof 98 Bittner RE et al. Recruitment of bone-marrow-derived cells by skeletal and cardiac muscle in adult dystrophic mdx mice. Anat 110 Wright DE et al. Physiological migration of hematopoietic stem Embryol (Berl) 1999; 199: 391–396. and progenitor cells. Science 2001; 294: 1933.

Gene Therapy