Blood cell progenitors: Insights into the ... - Wiley Online Library

33 downloads 0 Views 102KB Size Report
Nearly 200 billion red blood cells, 10 billion white blood cells, and 400 billion platelets are produced daily through- out our lifetime. In addition to the requirement ...
THE ANATOMICAL RECORD PART A 276A:66 –74 (2004)

Blood Cell Progenitors: Insights into the Properties of Stem Cells MERVIN C. YODER* Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana

ABSTRACT Hematopoiesis is a dynamic process in which eight lineages of mature blood cells are derived from a common stem cell. Great progress has been made in identifying the functionally disparate progenitors that emerge from the stem cell and in elucidating the molecules required for their growth and survival. Further work will be required to understand the molecular mechanisms that regulate commitment of stem and progenitor cells to each stage of progenitor cell development and ultimately into the mature blood cells. Anat Rec Part A 276A:66 –74, 2004. © 2004 Wiley-Liss, Inc.

Key words: hematopoiesis; stem cell; progenitor

The homeostatic production of circulating blood cells is one of the most dynamic processes in the human body. Nearly 200 billion red blood cells, 10 billion white blood cells, and 400 billion platelets are produced daily throughout our lifetime. In addition to the requirement for high cell production, the concentration of individual blood cell lineages must be maintained within narrow limits in the peripheral blood and tissues. Superimposed on this dynamic system is the requirement of the blood-forming tissues to produce particular cell types in response to specific demands. For example, microbial breach of the skin and mucosal barriers must be met with an increase in the number and antimicrobial properties of granulocytes to protect the host, acute blood loss requires an increase in oxygen-carrying erythroid cells to sustain normal host organ function, and so on. When the tightly regulated production of blood cells fails, the host may develop severe disease as a result of too few (marrow aplasia and anemia) or too many (leukemia) blood cells. Hematopoiesis is the process of blood cell production. The stem cell theory of hematopoiesis purports that all hematopoietic elements are ultimately derived from a transplantable multilineage long-term repopulating hematopoietic stem cell. This chapter will review some of the experimental evidence, primarily documented in the murine system, that supports this theory and provide an overview of the in vitro and in vivo assays that discriminate self-renewing hematopoietic stem cells from multipotent and lineage committed progenitor and mature cells. The intent of this overview is to provide a basic understanding of the hematopoietic hierarchy. Hopefully, the experimentally defined paradigm for the hierarchy of hematopoietic stem and progenitor cell compartments will prove useful in understanding the biology of stem cells in other organs, including the cardiovascular system. ©

2004 WILEY-LISS, INC.

ORIGINS OF BLOOD CELL INVESTIGATION Circulating erythrocytes were the first blood cells identified with the aid of the microscope in the late 1600s. The colorless white blood cells, however, remained obscured from direct observation until the late 1800s. With further improvements in microscopes and the use of a variety of cellular stains, a new era in hematology was born at the turn of the 20th century where changes in blood cell number, morphology, or location were now correlated with specific human diseases. Further progress in studying hematopoiesis was largely limited to conjecture and opinion until the mid-20th century when hematologists began to examine the hematopoietic consequences of animal exposure to ionizing radiation (Wintrobe, 1980).

METHODS OF IDENTIFYING HEMATOPOIETIC STEM AND PROGENITOR CELLS Whole-body irradiation of animals was noted to depress hematopoiesis and cause life-threatening deficiencies in all circulating blood cells (pancytopenia) in exposed mice

Grant sponsor: National Institutes of Health Grant; Grant number: HL63169. *Correspondence to: Mervin C. Yoder, Professor of Pediatrics and Biochemistry and Molecular Biology, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut St., R4-419, Indianapolis, IN 46202. Fax: (317) 274-8679. E-mail: [email protected] Received 10 January 2003; Accepted 14 February 2003 DOI 10.1002/ar.a.10133 Published online in Wiley InterScience (www.interscience.wiley.com).

HEMATOPOIETIC STEM AND PROGENITOR CELLS

67

Fig. 1. The stem cell theory of hematopoiesis predicts that all the circulating cells arise from a single stem cell. The stem cell may divide and one daughter cell retains all the properties of the parent (self-renewal) while the second daughter cell commits to becoming a progenitor cell. The committed cell proliferates extensively to provide distinct stages of progenitor cells from the CFU-S to mixed lineage colony-forming cells (CFU-Mix) and finally committed progenitor colony-forming cells of each lineage (CFC-C). The committed progenitor cells create all of the mature cells that circulate in the blood.

(Jacobson et al., 1949). Shielding the spleen of the experimental animal from the radiation beam resulted in recovery from an otherwise lethal dose of x-ray exposure (Jacobson et al., 1949). The splenic protection of the irradiated subject was thought to be conferred by a humoral factor since removal of the spleen 1– 6 hr after the lethal dose of irradiation was demonstrated to be sufficient for the protection (Jacobson et al., 1951a). However, transplantation of intact spleens into the abdomen of the lethally irradiated hosts also provided radioprotection (Jacobson et al., 1951b). In 1956, Ford et al. (1956) provided definitive evidence that hematopoietic cells and not plasma or subcellular molecules conferred radiation protection. Using donor cells from a strain of mice carrying a balanced (but morphologically identifiable) chromosome translocation, blood cells of a lethally irradiated mouse were demonstrated to be effectively replaced by donor hematopoietic cells after transplantation. Soon after, Till and McCulloch (1961) provided evidence that single multipotent hematopoietic progenitor cells (cells giving rise to more than one lineage of blood cells) could be identified in vivo by injecting donor marrow cells into a lethally irradiated recipient animal and examining the recipient spleen for hematopoietic colonies 8 –12 days later. Each colony of hematopoietic cells in the spleen was demonstrated to arise from a single precursor cell, the colonyforming unit in spleen cell (CFU-S) (Becker et al., 1963). Use of this assay provided the first compelling evidence that hematopoietic cells were clonally derived. While this method advanced the study of hematopoietic stem cell biology, little was known in the early 1960s of the mechanisms that caused and/or permitted stem cells to differentiate into the mature cells. It was clear to experimental hematologists in the mid1960s that new methods would be required to isolate the clonal precursors of the mature blood cells and to determine the factors that permitted their proliferation, survival, and differentiation. Pluznik and Sachs (1965) and Bradley and Metcalf (1966) reported that murine hematopoietic cells could be cultured in vitro and that addition of soluble fluid from several murine organs (i.e., urine or pregnant uterine extract) resulted in the in vitro forma-

tion of myeloid colonies. This biologically active tissue fluid was called colony-stimulating activity (CSA). Furthermore, these investigators reported that each myeloid colony developing in vitro arose from a single precursor cell called the colony-forming unit in culture (CFU-C) (Fig. 1). Further work revealed that myeloid colonies were comprised of granulocytes (CFU-G), macrophages (CFU-M), or both (CFU-GM) and that the type of colony formed was related in part to the type of CSA added. Later, using anemic mouse serum as a source of red blood cell CSA, Axelrad et al. (1974) succeeded in demonstrating that red blood cell colonies were clonally derived in vitro and called the most primitive red cell precursors the erythroid burstforming unit (BFU-E) and the most committed erythroid CFC were called colony-forming unit erythroid cells (CFUE). Multipotent progenitor cells were also identified and called CFU-Mix or CFU-GEMM to reflect the colony composition of granulocytes, erythroid cells, mast cells, megakaryocytes, and/or macrophages (Fig. 1). Plating of hematopoietic cells in special double-layer agar cultures with multiple recombinant cytokines permits the identification of hematopoietic progenitors that are highly proliferative (highly proliferative potential colony-forming cells (HPP-CFC)) (Kriegler et al., 1994). HPPCFC colonies contain ⬎50,000 cells that are visible in the culture dishes without need for magnification. HPP-CFC can be plucked from the agar medium, dispersed into a single-cell suspension, and replated in standard CFC assays with emergence of CFU-Mix, CFU-GM, and some BFU-E (McNiece et al., 1990). Thus, the HPP-CFC is considered to be a more primitive cell than the multipotent hematopoietic progenitor (CFU-GEMM). In fact, HPP-CFC are the most primitive hematopoietic progenitor cells that can be cultured in vitro without the presence of hematopoietic stromal cells in co-culture. Nonetheless, HPP-CFC are heterogenous progenitors. Several investigators have fully characterized the murine HPP-CFC population and have allocated HPP-CFC into differentiation classes (Kriegler et al., 1994). The most primitive HPPCFC require four or more growth factors to form colonies, while more committed HPP-CFC will emerge in culture in the presence of only three or as few as two growth factors.

68

YODER

The most committed HPP-CFC display a more limited replating potential and are somewhat smaller in diameter with fewer cells comprising the colony. In the late 1970s, Allen and Dexter (1984) reported on techniques to culture murine bone marrow for prolonged periods in vitro. These cultures were dependent upon the development of complex monolayers of nonhematopoietic stromal cells adherent to the culture plates. Further analysis revealed that the stromal elements were composed of endothelial cells, fibroblasts, macrophages, mesenchymal cells, and adipocytes (Penn et al., 1993). In time, procedures arose that permitted the establishment of primary stromal cell layers that were subsequently treated with DNA cross-linking agents to prevent further cell division but allow cell survival. Isolated populations of hematopoietic stem and progenitor cells could then be added to the mitotically quiescent stromal layers and analysis of hematopoietic cell proliferation and differentiation performed. Such study revealed that the most primitive hematopoietic cells (those giving rise to hematopoietic progenitors for up to 35 days and longer) resided beneath the stromal monolayers and were phase contrast dark (colony areaforming cells (CAFC)), whereas the mature blood cells produced via progenitor cell differentiation were released into the tissue culture medium as nonadherent cells and were phase contrast bright (Ploemacher et al., 1989). Using these morphologic characteristics some investigators determined that the number of day 35 CAFC was found to correlate with the number of long-term repopulating hematopoietic stem cells in vivo (Ploemacher et al., 1989). A modification of this assay called for the co-culture of hematopoietic cells with mitotically inactive primary stromal cells (or some stromal cell lines) and, after varying times in culture, isolation of the cells in co-culture and plating of the cells in standard CFC assays. Those hematopoietic progenitors co-cultured for prolonged periods with retention of the ability to give rise to CFC were called long-term culture-initiating cells (LTC-IC) (Sutherland et al., 1989). Using limiting dilution analysis, the number of such murine or human LTC-IC that give rise to multipotent and committed progenitors for 3– 4 weeks in vitro can be calculated (Ponchio et al., 1995; Verfaillie and Miller, 1995; Leemhuis et al., 1996) and are considered to be the most primitive hematopoietic precursor detectable via in vitro assays. The above CFC assays identify progenitor cells of the myeloerythroid series but not B or T lymphocytes. B lymphocytes require the presence of bone marrow or fetal liver stromal cells to promote proliferation and maturation in vitro (Muller-Sieburg et al., 1986; Whitlock and Witte, 1987). Addition of certain growth factors to the stromal cell cultures improves B cell maturation in vitro (Pietrangeli et al., 1988). T cell clones can be identified in the thymus in irradiated recipient mice following direct injection (Ezine et al., 1984). T lymphocyte progenitor cells can also be identified using fetal thymic organ cultures (Eren et al., 1987). Combining these lymphocyte progenitor assays with the myeloerythroid CFC assays permits identification of essentially all clonal hematopoietic progenitors. The frequency of human and murine hematopoietic stem cells has been estimated to be 1/104–105 bone marrow cells (Harrison et al., 1989; Sutherland et al., 1989; Harrison, 1993; Szilvassy and Hoffman, 1995; Bhatia et al., 1998; Abkowitz et al., 2002). The rarity of this population presented considerable impediments to isolation

TABLE 1. Hematopoietic stem cell phenotype* Cell surface phenotypic markers on adult murine bone marrow stem cells Positive selection

Negative selection

CD11a CD24 CD31 CD34a CD38 CD41a CD43 CD49da CD49e CD90a CD105 CD117 AA4.1a Sca-1 MDR Hoechst 33342a Rhodamine 123a

CD4 CD11b CD16 CD27 CD32 CD45R CD62L CD127 Gr-1 TER119

*Cells expressing the “positive” markers are enriched for hematopoietic stem cell repopulating ability while those expressing the “negative” markers are devoid of stem cell activity. Stem cell isolation protocols generally employ both positive and negative selection strategies to obtain highly enriched cells. Abbreviations: Stem cell antigen-1 (Sca-1), multidrug resistence gene (MDR), granulocyte-1 (Gr-1), and terminal erythroid 119 antigen (TER119). A more detailed description of the monoclonal antibodies used and stem cell selection strategies has recently been published (Srour and Yoder, 2003). a The most potent stem cells display low levels of these antigens and limited nuclear or cytoplasmic accumulation of these vital stains (Hoechst 33342 and Rhodamine 123, respectively).

until recently. Several significant advances that have led to hematopoietic stem cell identification include development of bone marrow transplantation techniques, production of monoclonal antibodies to stem cell surface antigens, and availability of high-speed cell-sorting techniques to enrich for the rare stem cells. Murine bone marrow transplantation experiments have provided the necessary evidence to reliably identify the murine hematopoietic stem cell. In fact, murine hematopoietic stem cells are now defined as cells that self-renew in vivo and also proliferate and differentiate into all lineages of circulating peripheral blood cells for more than 4 months after transplantation into recipient animals (Orlic and Bodine, 1994). Murine hematopoietic stem and progenitor cells can be enriched (negative selection) using flow cytometric cell sorting (Table 1) by selecting bone marrow cells that fail to express cell surface antigens (lin–) typically displayed by mature B and T lymphocytes, neutrophils, macrophages, natural killer cells, and red blood cells (Muller-Sieburg et al., 1986). Three commonly used phenotypic markers expressed by stem cells (positive selection) include stem cell antigen-1 (Sca-1), c-kit (CD117), and Thy-1 (CD90) (Morrison et al., 1995). Sca-1 is a cell surface molecule that is required for normal stem cell self-renewal and progenitor cell proliferation and lineage maturation (Ito et al., 2003). C-kit is a cell surface receptor tyrosine kinase that is

HEMATOPOIETIC STEM AND PROGENITOR CELLS

Fig. 2. Adult bone marrow hematopoietic hierarchy. The long-term repopulating hematopoietic stem cell (LT-HSC) may self-renew or produce transplantable short-term repopulating hematopoietic progenitors (ST-HPC). These progenitors differentiate into the CLP or CMP. The CMP differentiates into GMP and MEP, which further commit to unilineage committed progenitors prior to initiating full maturation into a circulating blood cell.

necessary for hematopoietic stem cell survival and in utero embryo survival (Reith and Bernstein, 1991). Thy-1 is a cell surface molecule expressed by stem and lymphoid cells (Basch and Berman, 1982). CD34 is a cell surface sialomucin expressed by hematopoietic and endothelial cells (Krause et al., 1996). Stem cells in the murine fetal liver highly express CD34 and the C1q complement receptor (AA4.1) (Tavassoli, 1994). These antigens (CD34 and AA4.1) are expressed on proliferating bone marrow stem cells but are downregulated and not detectable on the surface of quiescent marrow stem cells (Szilvassy and Cory, 1993; Sato et al., 2000). Other antigens that have been utilized to enrich (positive or negative) murine bone marrow stem cells include AC133, CD31, CD38, CD43, CD105, and leukocyte function antigen-1 (CD11a). Stem cells may also be enriched from other marrow cells by the fact that quiescent stem cells bind the least amount of a DNA-binding dye (Hoechst 33342) and appear as a side population distinguishable (by cell sorting) from other hematopoietic cells. Retention of low amounts of another vital dye, Rhodamine 123, is also a feature of hematopoietic stem cells. Use of several different combinations of these cell surface markers has permitted enrichment of stem cells to homogeneity as evidenced by long-term repopulation of all blood lineages in recipient mice from a single transplanted cell with a phenotype, Sca-1⫹cKit⫹CD34–lin–. The hematopoietic system is comprised of a series of functionally and phenotypically distinguishable progenitor cell compartments (Fig. 1). Bone marrow and fetal liver cells isolated using the above strategies can be further fractionated into several distinct progenitor cell populations. C-kit⫹, Thy-1lo, Sca-1⫹lin– cells possess longterm repopulating ability, but cells that are c-kit⫹Thy1loSca-1⫹ and Mac-1lo (a granulocyte adhesion molecule) possess only short-term repopulating ability (Morrison and Weissman, 1994). While the long-term repopulating cells can differentiate into the short-term repopulating cells, the opposite is not true (Fig. 2) (Morrison et al., 1997). Downstream of the short-term repopulating pro-

69

genitors are the common lymphoid (CLP) and common myeloid progenitors (CMP) (Fig. 2) (Kondo et al., 1997; Akashi et al., 2000). These progenitors differ in that the CLP express the interleukin-7 receptor (IL-7R) but not the granulocyte-macrophage colony-stimulating factor receptor (GM-CSFR) while the CMP are GM-CSFR⫹IL-7R–. The CMP can be further differentiated into committed granulocyte-macrophage progenitors (GMP) or megakaryocyte-erythroid progenitors (MEP) (Fig. 2). GMP express the macrophage colony-stimulating factor receptor (M-CSFR) but not the erythropoietin receptor (Epo-R), while the MEP are MCSFR–Epo-R⫹ (Akashi et al., 2000). Recent data demonstrate that lethally irradiated mice can be rescued from radiation-induced pancytopenia by infusion of MEP alone (Nakorn et al., 2002). While the recipient animals are not long-term repopulated by the MEP, these committed progenitors differentiate into platelets and red blood cells that support the animals until surviving (radiation-resistant) endogenous hematopoietic stem cells repopulate all of the peripheral blood cell lineages. Human hematopoietic cells have also been isolated using monoclonal antibodies and flow cytometric cell sorting (reviewed in Payne and Crooks (2002) and Verfaillie (2002)). Human stem cells have been isolated from bone marrow, umbilical cord blood, fetal liver, and mobilized peripheral blood as cells expressing CD34, Thy-1, AC133, and c-kit but not CD38 or mature blood cell lineage markers (lin–). While CFC assays and LTC-IC assays have proven useful in identifying the functional properties of flow cytometric sorted human cells, in vivo testing of selected cell populations in human subjects has not been possible for ethical reasons. Nonetheless, CD34⫹ selected hematopoietic cells have been proven to support patients long term after bone marrow transplantation (Verfaillie, 2002). In an attempt to develop an in vivo system for detection of human hematopoietic stem cells, several groups have developed xenotransplantation models. Enriched populations of human hematopoietic stem cells from human bone marrow, mobilized peripheral blood, cord blood, or fetal liver have been observed to engraft in the preimmune sheep fetus (⬍60 days of gestation) with long-term evidence of multilineage peripheral human blood cell chimerism in the transplanted animals (Zanjani et al., 1992; Srour et al., 1993; Civin et al., 1996). Human cells present in the marrow of transplanted sheep fetuses can engraft in secondarily transplanted preimmune fetuses in vivo suggesting the presence of self-renewing stem cell populations. Nonobese diabetic (NOD) mice bred with severe combined immunodeficient (SCID) mice result in NOD/SCID mice that accept human hematopoietic grafts (Schultz et al., 1995). This NOD/SCID model has permitted calculation of the frequency of human repopulating cells present in a donor sample (Dick et al., 1997). Similarly, SCID mice can be implanted with fragments of human fetal thymus and bone that will survive (Heike et al., 1995). Sublethal irradiation of these mice will impair hematopoiesis in the implanted human tissue and permit engraftment of intravenously administered human cells following transplantation (Namikawa et al., 1990). Other immunodeficient mice models that permit human hematopoietic cell engraftment have also been reported; however, it remains unclear whether any of these assays can accurately identify the same hematopoietic stem cells that repopulate the blood system of transplanted human patients (Dao et al., 1999).

70

YODER

STEM/PROGENITOR CELL DIFFERENTIATION: STOCHASTIC OR INDUCTIVE The above methods have permitted subfraction of the hematopoietic system into a hierarchy of functionally and phenotypically distinct stem and progenitor cell compartments. However, little is known of the mechanisms that permit differentiation of the stem cells into the committed progenitor cell pools and further development into mature blood cells. Controversy remains whether stem/progenitor cell commitment and differentiation is the result of a stochastic (purely random process intrinsic to the cell) or an instructive (signals outside the cells dictate or determine cell fate) process (Enver et al., 1998). Growth factors have been largely implicated as modulators of hematopoietic progenitor cell fate in support of the instructive model of lineage commitment. Experiments using hematopoietic cell lines or primary hematopoietic progenitor cells have provided results that the presence of stem cell factor (SCF) or granulocyte colonystimulating factor (G-CSF) promotes neutrophil differentiation while M-SCF stimulates macrophage differentiation from GM-CFC in vitro (Metcalf, 1998). When individual GM-CFC are isolated as single cells in vitro, allowed to divide, and the paired daughter cells are then separated and exposed to single growth factors, GM-CSF has been demonstrated to promote neutrophil differentiation, but M-CSF promotes macrophage differentiation (Metcalf and Burgess, 1982). These results would support an instructive model of lineage commitment whereby the presence of a particular growth factor dictates the fate of progenitor cell differentiation. However, alternative interpretations of these data are possible. Provision of bipotent or multipotent progenitors with a single growth factor in vitro and deriving a single lineage of differentiated cells could be interpreted (as above) that the growth factor dictated the fate of the progenitors. The same result could also be interpreted as the growth factor simply promoted the growth and survival of a subpopulation of the progenitors that had already committed to that particular lineage. Evidence that growth factors may play a limited instructive role for progenitors has been provided in analyzing several knockout mouse models. Mice that fail to express GM-CSF are viable but die as young adult mice due to pulmonary failure as a result of surfactant protein accumulation due to faulty macrophage handling of the protein. These mutant mice have apparently normal numbers of GM-CFC and neutrophils despite the absence of GM-CSF (Stanley et al., 1994). Similarly, GCSF null mice display severely reduced numbers of circulating neutrophils, but GM-CFC are present at near normal frequencies and some neutrophils persist (Lieschke et al., 1994). These results suggest that the absence of GMCSF and G-CSF stimulation of the progenitors has no apparent effect on GM-CFC commitment to mature cell lineage differentiation. One must be cautious, however, since compensatory pathways are often invoked when one molecular pathway is interrupted in knockout mice. An alternative approach to addressing the role of growth factors in determining cell fate can be achieved by misexpressing a growth factor receptor for one lineage into another lineage using gene therapy. Transducing primary erythroid cells with a retrovirus encoding the MCSFR has demonstrated that addition of M-CSF to the

transduced cells promotes erythroid CFC formation but does not cause a shift in differentiation to macrophages or monocytes (McArthur et al., 1994). Similarly, providing multipotent progenitors with constitutively active Epo-R and M-CSFR does not result in a preferential production of erythroid or macrophage progeny, respectively, but does promote the proliferation of the progenitor cells in vitro (Pharr et al., 1994). As noted above, transplantation of the CLP into lethally irradiated hosts demonstrates the restricted differentiation potential of these cells into T and B lymphocytes, natural killer cells, and antigen-presenting dendritic cells. However, introduction of the GM-CSFR into the CLP causes these cells to differentiate into granulocytes and macrophages with diminished lymphoid commitment (Kondo et al., 2000). Culturing CLP with IL-7 for several days in vitro causes these progenitors to become resistant to the signaling effects of an introduced GM-CSFR, and no differentiation into myeloid cells occurs. Most of the IL-7stimulated CLP commit to B lymphoid development. Isolation of primary pro-B cells and introduction of the GMCSFR reveal that these progenitors are resistant to the GM-CSFR transduced signals (Kondo et al., 2000). Early pro-T cells from the thymus can be reprogrammed for a myeloid outcome via introduction of the GM-CSFR and GM-CSF stimulation. These results suggest that during normal lymphocyte lineage specification, a phase of differentiation exists that only results in a lymphoid type outcome; however, the genome remains sufficiently plastic to permit rescue of another blood cell phenotype given certain conditions (Weissman et al., 2001). Creation of transgenic animals carrying modified growth factor receptors has also been utilized to examine the role of growth factor signaling in lineage commitment. One example of such work was the creation of a transgenic mouse expressing a mutant growth factor receptor in which the cytoplasmic domain of the c-mpl receptor (receptor for thrombopoietin) was replaced with the cytoplasmic domain of the G-CSFR (Stoffel et al., 1999). The anticipated result was that transgenic mice homozygous for the mutant receptor would demonstrate deficiencies in platelet production similar to those of mice homozygous deficient for the c-mpl receptor. However, platelet production was relatively unaffected, suggesting that the mutant receptor was sufficient to mediate the intracellular signaling required for megakaryocyte proliferation and differentiation. These results support a stochastic lineage commitment process. The effects of a particular growth factor on the differentiation of a progenitor population depend on the relative expression of the relevant receptor for that growth factor on the progenitor cells. May and Enver (2001) have argued that the overall pattern of growth factor receptors expressed by a population of progenitor cells is likely a consequence rather than the determining cause of progenitor cell commitment. This conclusion begs the question of how the pattern of growth factor receptor expression is dictated in the various progenitor cell populations. Transcription factors have been postulated to play a role in the process of cellular commitment to lineage differentiation. The SCL/tal-1 transcription factor is a basic helixloop-helix protein that is expressed primarily in hematopoietic, endothelial, and central nervous cells (Shivdasanl et al., 1995; Robb and Begley, 1997). Knockout of this gene in mice results in embryonic lethality with a complete

HEMATOPOIETIC STEM AND PROGENITOR CELLS

absence of hematopoietic cells (Shivdasanl et al., 1995; Robb et al., 1996). This deficiency most likely occurs at the level of mesoderm cell commitment to the hematopoietic lineage and is upstream of the hematopoietic stem cell. PU.1 is a protein expressed exclusively in hematopoietic cells (Fisher and Scott, 1998). Disruption of this molecule blocks both lymphoid and myeloid cell development (McKercher et al., 1996). Fetal liver hematopoietic stem cells isolated from homozygous null PU.1 mice failed to reconstitute lymphoid or myeloid lineages in lethally irradiated mice, although erythroid cells were derived from the mutant donor cells. Production of chimeric embryos via injection of blastocysts with PU.1 null embryonic stem (ES) cells resulted in viable mice that lacked evidence of PU.1 null ES cell contribution to B or T cells, monocytes, or neutrophils (Scott et al., 1997). GM-CSFR and G-CSFR are expressed normally in the absence of PU.1, but MCSFR is not expressed. PU.1 null progenitor cells are not responsive to GM-CSF, G-CSF, or M-CSF. Transduction of PU.1 null progenitor cells with the M-CSFR overcomes the block in M-CSF-induced cell proliferation but does not restore full myeloid cell differentiation to these progenitor cells (Olson et al., 1995; DeKoter et al., 1998). Further studies concluded that PU.1 controls cellular differentiation by regulating distinct proliferation and differentiation pathways. The role of transcription factors in controlling lineage specification can be addressed using other genetic approaches. Heyworth et al. (2002) recently transduced primary murine GM-CFC with a retroviral construct encoding a transcription factor (GATA-1) in an inducible form. They reported the progenitors differentiated into neutrophils and monocyte/macrophages in the presence of interleukin-3 (IL-3) or IL-3 plus Epo. In the presence of the retroviral vector alone or the inducible GATA-1 construct, transduced GM-CFC stimulated with the growth factors also differentiated into granulocytes only. However, induction of GATA-1 expression in the GM-CFC resulted in a dramatic appearance of erythroblasts, eosinophils, and basophils. This lineage switch was a high-frequency event occurring in ⬎95% of individual clones of transduced GMCFC. The induction of erythroid cells, eosinophils, and basophilic neutrophils occurred at the primary expense of monocyte differentiation in these experiments. These results suggest that a single transcription factor can reprogram the fate of committed GM-CFC. How might the process of lineage specification be identified at the molecular level in multipotent cells? Recent evidence suggests that transcription of a number of lineage-affiliated effector genes, growth factor receptors, and transcription factors can be concomitantly identified in multipotent cells cultured in vitro (Hu et al., 1997). Similar evidence of simultaneous transcription of multiple lineage affector genes in single sorted stem and progenitor cells from mice and humans suggests that the low-level heterogenous expression of these molecules may provide potential starting points for lineage specification (Cheng et al., 1996; Delassus et al., 1999). For example, accessibility of gene transcripts for multiple lineage commitment pathways may lead to interactive or competing interactions leading to an upregulation (or downregulation of an alternative pathway) of appropriate molecules as a dominant commitment pathway emerges. In the future, gene expression profiling of stem and progenitor cells at different stages of normal or experimentally induced differen-

71

tiation may permit elucidation of specific molecules associated with each state of commitment to lineage specification.

DEVELOPMENTAL HEMATOPOIESIS The primary sites of hematopoiesis change during mammalian development. Blood cells are first detectable in the mouse on embryonic day 7 (E7.0) and on the 18th day postconception in the human (Palis and Yoder, 2001). In both species, blood cells appear first in the yolk sac. The first murine hematopoietic cells are predominantly erythroid cells with few macrophages (Palis et al., 1999). Limited numbers of megakaryocytes are also produced within 12 hr of the first erythroid cells in the yolk sac. The first erythroid progenitors are called primitive erythroid progenitors (EryP) since these cells produce primitive erythroblasts. The primitive erythroblasts are large nucleated erythrocytes that contain fetal and adult hemoglobins. These cells are formed exclusively in the yolk sac from E7.0 –E8.5 (Palis et al., 1999). The earliest macrophages are also unique in their cell surface protein expression and phagocytic function and are often referred to as primitive macrophages (Lichanska and Hume, 2000). Recently, a unique population of primitive megakaryocytes has also been identified. These cells demonstrate accelerated maturation of platelet formation compared to megakaryocytes produced later in development. In sum, the primitive erythroblasts, macrophages, and megakaryocytes comprise the primitive hematopoietic phase of blood cell development. The first definitive hematopoietic progenitor cells are also produced in the murine yolk sac on E8.25 (Palis et al., 2001). Of interest, these definitive progenitor cells do not normally differentiate into mature blood cells in the yolk sac, but rather enter the bloodstream at the time the liver is beginning to accumulate hematopoietic progenitors. Thus, yolk sac-derived definitive progenitor cells likely seed the liver where they subsequently mature into circulating granulocytes and red blood cells. Are the primitive and definitive hematopoietic progenitors derived from a common stem cell? The most evidence of such a relationship has been determined from experiments using murine ES cell differentiation in vitro (Kennedy et al., 1997). ES cells expressing flk-1 can give rise to primitive and definitive hematopoietic progenitor cells. This question has not been answered in murine embryos, but the controversy has been reviewed extensively (Cumano and Godin, 2001; Yoder and Palis, 2001). Current evidence suggests that primitive hematopoiesis may be derived via mechanisms distinct from definitive hematopoiesis. Available data suggest that stem cells derived from the aorta-gonad-mesonephros (AGM) region may be the first to seed the fetal liver (de Bruijn et al., 2002; North et al., 2002). Recent studies suggest that wherever the stem cells originate from, these cells are expanded locally in the AGM and within the yolk sac circulation concomitant with proliferation of the stem cells in the early fetal liver to account for the dramatic increase in stem cell numbers during the first 48 hr of liver hematopoiesis (Kumaravelu et al., 2002). The liver continues as the primary hematopoietic site until late in gestation when stem cells reenter the circulation and seed the marrow and spleen. The spleen serves as an active hematopoietic site throughout the life of the mouse but is principally a lymphoid organ in humans. While stem cells can be identified

72

YODER

in the adult murine liver, recent data suggest that a certain fraction of marrow-derived stem cells are constantly circulating throughout the body and thus may contaminate all perfused organs (Wright et al., 2001). In general, the murine liver loses the ability to support hematopoiesis within a few weeks of postnatal life but can regain such function in the face of extreme hematopoietic stress.

STEM CELL PLASTICITY Stem cells residing in adult tissues have typically been thought to be committed to the production of progeny specific for that particular tissue. Thus, hematopoietic stem cells give rise to blood cells, not liver cells, neurons, or muscle cells. Or do they? Numerous publications over the past 3– 4 years have provided some evidence for more plasticity of stem cell commitment and differentiation than previously considered possible. Bone marrow cells, including isolated populations of hematopoietic stem cells, have been reported to differentiate into muscle, neurons, liver, vascular, lung, intestine, and kidney tissue (Gussoni et al., 1999; Jackson et al., 1999; Brazelton et al., 2000; Lagasse et al., 2000; Krause et al., 2001; Grompe, 2002). These results have created significant controversy, particularly with regard to the criteria used to demonstrate that a specific stem cell directly differentiated into an unexpected lineage. Recent publications point out that some of the results of stem cell plasticity studies may be explained due to cell fusion events or occur at such low frequency that the progeny of the stem cells would not contribute to tissue function (Iscove, 2001; Weissman et al., 2001; Wagers et al., 2002; Ying et al., 2002). Several investigators have suggested some basic principles that should be addressed in future stem cell studies (Anderson et al., 2001; Weissman et al., 2001; Graf, 2002; Lemischka, 2002). First, the stem cell population should be localized to a specific cell phenotype that can be isolated and therefore comparable from one study to the next. The contribution of the stem cell to a particular organ should be clonal. That is, one should prove that the differentiated tissues derived from a donor source arose from single stem cells. The tissue derived from the stem cell should be a significant population of cells that demonstrate functions consistent with the organ of residence. Following these simple principles should clarify the question of whether an adult tissue stem cell can be stimulated to commit to differentiation to a lineage of cells of a different tissue upon transplantation.

LITERATURE CITED Abkowitz JL, Catlin S, McCallie M, Guttorp P. 2002. Evidence that the number of hematopoietic stem cells per animals is conserved in mammals. Blood 100:2665–2667. Akashi K, Traver D, Miyamoto T, Weissman I. 2000. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404:193–196. Allen TD, Dexter TM. 1984. The essential cells of the hemopoietic microenvironment. Exp Hematol 12:517–521. Anderson D, Gage F, Weissman I. 2001. Can stem cells cross lineage boundaries? Nat Med 7:393–395. Axelrad A, McLeod D, Shreeve M, Heath D. 1974. Properties of cells that produce erythrocytic colonies in vitro. In: Robinson W, editor. Hemopoiesis in culture, vol. 74-205. Washington: National Institutes of Health. p 226 –237.

Basch R, Berman J. 1982. Thy-1 determinants are present on many murine hematopoietic cells other than T cells. Eur J Immunol 12:359 –364. Becker A, McCulloch E, Till J. 1963. Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature 197:452– 455. Bhatia M, Bonnet D, Murdoch B, Gan O, Dick J. 1998. A newly discovered class of human hematopoietic cells with SCID-repopulating activity. Nat Med 4:1038 –1045. Bradley T, Metcalf D. 1966. The growth of mouse bone marrow cells in vitro. Aust J Exp Biol Med Sci 44:287–300. Brazelton T, Rossi F, Kanet G, Blau H. 2000. From marrow to brain: expression of neuronal phenotypes in adult mice. Science 290:1775– 1779. Cheng T, Shen H, Giokas D, Gere J, Tenen D, Scadden D. 1996. Temporal mapping of gene expression levels during the differentiation of individual primary hematopoietic cells. Proc Natl Acad Sci USA 93:13158 –13163. Civin C, Porada G, Lee M-P, Olweus J, Terstappen L, Zanjani E. 1996. Sustained, retransplantable, multilineage engraftment of highly purified adult human bone marrow stem cells in vivo. Blood 88: 4102– 4109. Cumano A, Godin I. 2001. Pluripotent hematopoietic stem cell development during embryogenesis. Curr Opin Immunol 13:166 –171. Dao MA, Tsark E, Nolta JA. 1999. Animal xenograft models for evaluation of gene transfer into human hematopoietic stem cells. Curr Opin Mol Ther 1:553–557. de Bruijn M, Ma X, Robin C, Ottersbach K, Sanchez M-J, Dzierzak E. 2002. Hematopoietic stem cells localize to the endothelial cell layer in the midgestation mouse aorta. Immunity 16:673– 683. DeKoter R, Walsh J, Singh H. 1998. PU.1 regulates both cytokinedependent proliferation and differentiation of granulocyte/macrophage progenitors. EMBO J 17:4456 – 4468. Delassus S, Titley I, Enver T. 1999. Functional and molecular analysis of hematopoietic progenitors derived from the aorta-gonadmesonephros region of the mouse embryo. Blood 94:1495–1503. Dick J, Bhatia M, Gan O, Kapp U, Wang J. 1997. Assay of human cells by repopulation of NOD/SCID mice. Stem Cells 15:199 –203. Enver T, Heyworth C, Dexter T. 1998. Do stem cells play dice? Blood 92:348 –351. Eren R, Auerbach R, Globerson A. 1987. T cell ontogeny: extrathymic and intrathymic development of embryonic lymphohematopoietic stem cells. Immunol Res 6:279 –283. Ezine S, Weissman I, Rouse R. 1984. Bone marrow cells give rise to distinct cell clones within the thymus. Nature 309:629 – 631. Fisher R, Scott E. 1998. Role of PU.1 in hematopoiesis. Stem Cells 16:25–37. Ford C, Hamerton J, Barnes W, Loutit J. 1956. Cytological identification of radiation-chimaeras. Nature 177:452– 454. Graf T. 2002. Differentiation plasticity of hematopoietic cells. Blood 99:3089 –3101. Grompe M. 2002. Transition of stem cells to therapeutically functional tissue-specific cells. Ann NY Acad Sci 961:305–306. Gussoni E, Soneoka Y, Strickland C, Buzney E, Khan MK, Flint A, Kunkel L, Mulligan R. 1999. Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 401:390 –394. Harrison D, Astle C, Stone M. 1989. Numbers and functions of transplantable primitive immunohematopoietic stem cells. J Immunol 142:3833–3840. Harrison D. 1993. Competitive repopulation in unirradiated normal recipients. Blood 81:2473–2474. Heike Y, Ohira T, Takahashi M, Saijo N. 1995. Long-term human hematopoiesis in SCID-hu mice bearing transplanted fragments of adult bone and bone marrow cells. Blood 86:524 –530. Heyworth C, Pearson S, May G, Enver, T. 2002. Transcription factormediated lineage switching reveals plasticity in primary committed progenitor cells. EMBO J 21:3770 –3781. Hu M, Drause D, Greaves M, Sharkis S, Dexter M, Heyworth C, Enver T. 1997. Multilineage gene expression precedes commitment in the hemopoietic system. Gene Dev 11:774 –785. Iscove N. 2001. Is plasticity here to stay? Blood 98:1999.

HEMATOPOIETIC STEM AND PROGENITOR CELLS Ito C, Bernstein A, Dick J, Stanford WL. 2003. Hematopoietic stem cell and progenitor defects in Sca-1/Ly-6A-null mice. Blood 101: 517–523. Jackson K, Mi T, Goodell MA. 1999. Hematopoietic potential of stem cells isolated from murine skeletal muscle. Proc Natl Acad Sci USA 96:14482–14485. Jacobson L, Marks E, Robson M, Gaston E, Zirkle R. 1949. The effect of spleen protection on mortality following X-irradiation. J Lab Clin Med 34:1538 –1542. Jacobson L, Simmons E, Marks E, Eldredge J. 1951a. Recovery from radiation injury. Science 113:510 –511. Jacobson L, Simmons E, Marks E, Gaston E, Robson M, Eldredge J. 1951b. Further studies on recovery from radiation injury. J Lab Clin Med 37:683– 697. Kennedy M, Firpo M, Choi K, Wall C, Robertson S, Kabrun N, Keller G. 1997. A common precursor for primitive erythropoiesis and definitive haematopoiesis. Nature 386:488 – 493. Kondo M, Weissman I, Akashi K. 1997. Identification of clonogenic common lymphoid progenitor in mouse bone marrow. Cell 91:661– 672. Kondo M, Scherer D, Miyamoto K, King AG, Akashi K, Sugamura K, Weissman I. 2000. Cell-fate conversion of lymphoid-committed progenitors by instructive actions of cytokines. Nature 407:383–386. Krause D, Fackler M, Civin C, May W. 1996. CD34: Structure, biology, and clinical utility. Blood 87:1–13. Krause DS, Theise N, Collecter M, Henegariu O, Hwang S, Gardner R, Neutzel S, Sharkis S. 2001. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 105:369 –377. Kriegler AB, Verschoor SM, Bernardo D, Bertoncello I. 1994. The relationship between different high proliferative potential colonyforming cells in mouse bone marrow. Exp Hematol 22:432– 440. Kumaravelu P, Hook L, Morrison A, Ure J, Zhal S, Zuayev S, Ansell J, Medvinsky A. 2002. Quantitative developmental anatomy of definitive haematopoeitic stem cells/long-term repopulating units (HSC/RUs): role of the aorta-gonad-mesonephros (AGM) region and the yolk sac in colonisation of the mouse embryonic liver. Development 129:4891– 4899. Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse M, Osborne L, Wang X-S, Finegold M, Weissman I, Grompe M. 2000. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med 6:1229 –1234. Leemhuis T, Yoder MC, Grigsby S, Aguero B, Eder P, Srour E. 1996. Isolation of primitive human bone marrow hematopoietic progenitor cells using Hoechst 33342 and Rhodamine 123. Exp Hematol 24:1215–1224. Lemischka I. 2002. A few thoughts about the plasticity of stem cells. Exp Hematol 30:848 – 852. Lichanska AM, Hume DA. 2000. Origins and functions of phagocytes in the embryo. Exp Hematol 28:601– 611. Lieschke G, Grail D, Hodgson G, Metcalf D, Stanley E, Cheers C, Fowler K, Basu S, Zhan Y, Dunn A. 1994. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood 84:1737–1746. May G, Enver T. 2001. The lineage commitment and self-renewal of blood stem cells. In: Zon L, editor. Hematopoiesis. A developmental approach. Oxford: Oxford University Press. p 61– 81. McArthur G, Rohrschneider L, Johnson G. 1994. Induced expression of c-fms in normal hematopoietic cells shows evidence for both conservation and lineage restriction of signal transduction in response to macrophage colony-stimulating factor. Blood 83:972–981. McKercher SR, Torbett BE, Anderson KL, Henkel G, Vestal DJ, Baribault H, Klemsz M, Feeney A, Wu G, Paige C, Maki RA. 1996. Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J 15:5647–5658. McNiece I, Bertoncello I, Kriegler A, Quesenberry P. 1990. Colonyforming cells with high proliferative potential (HPP-CFC). Int J Cell Cloning 8:146 –160. Metcalf D, Burgess A. 1982. Clonal analysis of progenitor cell commitment to granulocyte or macrophage production. J Cell Physiol 111:275–283.

73

Metcalf D. 1998. Lineage commitment and maturation in hematopoietic cells: the case for extrinsic regulation. Blood 92:345–352. Morrison S, Weissman I. 1994. The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype. Immunity 1:661– 673. Morrison S, Uchida N, Weissman I. 1995. The biology of hematopoietic stem cells. Annu Rev Cell Dev Biol 11:35–71. Morrison S, Wandycz A, Hemmati H, Wright D, Weissman I. 1997. Identification of a lineage of multipotent hematopoietic progenitors. Development 124:1929 –1939. Muller-Sieburg C, Whitlock C, Weissman I. 1986. Isolation of two early B lymphocyte progenitors from mouse marrow: a committed pre-pre-B cell and a clongenic thy-1 hematopietic stem cell. Cell 44:653– 662. Nakorn T, Traver D, Weissman I, Akashi K. 2002. Myeloerythroidrestricted progenitors are sufficient to confer radioprotection and provide the majority of day 8 CFU-S. J Clin Invest 109:1579 –1585. Namikawa R, Weilbaecher K, Kaneshima H, Yee E, McCune J. 1990. Long-term human hematopoiesis in the SCID-hu mouse. J Exp Med 172:1055–1063. North T, de Bruijn M, Stacy T, Talebian L, Lind E, Robin C, Binder M, Dzierak E, Speck N. 2002. Runx1 expression marks long-term repopulating hematopoietic stem cells in the midgestation mouse embryo. Immunity 16:661– 672. Olson M, Scott EW, Hack A, Su G, Tenen D, Singh H, Simon MC. 1995. PU.1 Is not essential for early myeloid gene expression but is required for terminal myeloid differentiation. Immunity 3:703–714. Orlic D, Bodine D. 1994. What defines a pluripotent hematopoietic stem cell (PHSC): will the real PHSC please stand up! Blood 84: 3991–3994. Palis J, Robertson S, Kennedy M, Wall C, Keller G. 1999. Development of erythroid and myeloid progenitors in the yolk sac and embryo proper of the mouse. Development 126:5073–5081. Palis J, Yoder M. 2001. Yolk sac hematopoiesi—the first blood cells of mouse and man. Exp Hematol 29:927–936. Palis J, Chan R, Koniski A, Patel R, Starr M, Yoder M. 2001. Spatial and temporal emergence of high proliferative potential hematopoietic precursors during murine embryogenesis. Proc Natl Acad Sci USA 98:4528 – 4533. Payne K, Crooks GM. 2002. Human hematopoietic lineage commitment. Immunol Rev 187:48 – 64. Penn PE, Jiang D-Z, Fei R-G, Sitnicka E, Wolf NS. 1993. Dissecting the hematopoietic microenvironment. IX. Further characterization of murine bone marrow stromal cells. Blood 81:1205–1213. Pharr P, Ogawa M, Hofbauer A, Longmore G. 1994. Expression of an activated erythropoietin or a colony-stimulating factor 1 receptor by pluripotent progenitors enhances colony formation but does not induce differentiation. Proc Natl Acad Sci USA 91:7482–7486. Pietrangeli C, Hayashi S, Kincade P. 1988. Stromal cell lines which support lymphocyte growth: characterization, sensitivity to radiation and responsiveness to growth factors. Eur J Immunol 18:863– 872. Ploemacher R, vanderSluijs J, Voerman J, Brons N. 1989. An in vitro limiting-dilution assay of long-term repopulating hematopoietic stem cells in the mouse. Blood 74:2755–2763. Pluznik D, Sachs L. 1965. The cloning of normal “mast” cells in tissue culture. J Cell Comp Physiol 66:319 –327. Ponchio L, Conneally E, Eaves C. 1995. Quantitation of the quiescent fraction of long-term culture-initiating cells in normal human blood and marrow and the kinetics of their growth factor-stimulated entry into S-phase in vitro. Blood 86:3314 –3321. Reith A, Bernstein A. 1991. Molecular basis of mouse developmental mutants. Genes Dev 5:1115–1123. Robb L, Elwood N, Elefanty A, Kontgen F, Li R, Barnett L, Begley C. 1996. The scl gene product is required for the generation of all hematopoietic lineages in the adult mouse. EMBO J 15:4123– 4129. Robb L, Begley C. 1997. The SCL/TAL1 gene: roles in normal and malignant haematopoiesis. Bioessays 19:607– 613. Sato T, Laver J, Ogawa M. 2000. Reversible expression of CD34 by murine hematopoietic stem cells. Blood 94:2548 –2554. Schultz L, Schweitzer P, Christianson S, Gott B, Schweitzer I, Tennent B, McKenna S, Mobraten L, Rajan T, Greiner D, Leiter EH.

74

YODER

1995. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol 154:180 –191. Scott EW, Fisher CR, Olson M, Kehrli E, Simon MC, Singh H. 1997. PU.1 functions in a cell-autonomous manner to control the differentiation of multipotential lymphoid-myeloid progenitors. Immunity 6:437– 447. Shivdasanl R, Mayer E, Orkin S. 1995. Absence of blood formation in mice lacking the T-cell leukaemia oncoprotein tal-1/SCL. Nature 373:432– 434. Srour E, Zanjani E, Cornetta K, Traycoff C, Flake A, Hedrick M, Brandt J, Leemhuis T, Hoffman R. 1993. Persistence of human multilineage, self-renewing lymphohematopoietic stem cells in chimeric sheep. Blood 82:3333–3342. Srour EF, Yoder MC. Flow cytometric analysis of hematopoietic development. In: Baron MH, editor. Developmental hematopoiesis. Totowa, NJ: Human Press, in press. Stanley E, Lieschke G, Grail D, Metcalf D, Hodgson G, Gall J, Maher D, Cebon J, Sinickas V, Dunn A. 1994. Granulocyte/macrophage colony-stimulating factor-deficient mice show no major perturbation of hematopoiesis but develop a characteristic pulmonary pathology. Proc Natl Acad Sci USA 91:5592–5596. Stoffel R, Ziegler S, Ghilardi N, Ledermann B, DeSauvage F, Skoda R. 1999. Permissive role of thrombopoietin and granulocyte colonystimulating factor receptors in hematopoietic cell fate decisions in vivo. Proc Natl Acad Sci USA 96:698 –702. Sutherland H, Eaves C, Eaves A. 1989. Characterization and partial purification of human marrow cells capable of initiating long-term hematopoiesis in vitro. Blood 74:1563–1570. Szilvassy S, Cory S. 1993. Phenotypic and functional characterization of competitive long-term repopulating hematopoietic stem cells enriched from 5-fluorouracil-treated murine marrow. Blood 81:2310 – 2320. Szilvassy S, Hoffman R. 1995. Enriched hematopoietic stem cells: basic biology and clinical utility. Biol Blood Marrow Transplant 1:3–17.

Tavassoli M. 1994. Embryonic origin of hematopoietc stem cells. Exp Hematol 22:7. Till J, McCulloch E. 1961. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res 14:213– 222. Verfaillie C, Miller J. 1995. A novel single-cell proliferation assay shows that long-term culture-initiating cell (LTC-IC) maintenance over time results from the extensive proliferation of a small fraction of LTC-IC. Blood 86:2137–2145. Verfaillie C. 2002. Hematopoietic stem cells for transplantation. Nat Immunol 3:314 –317. Wagers A, Sherwood R, Christensen J, Weissman I. 2002. Little evidence for developmental plasticity of adult hematpoietic stem cells. Science 297:2256 –2260. Weissman I, Anderson D, Gage F. 2001. Stem and progenitor cells: origins, phenotypes, lineage commitments, and transdifferentiations. Annu Rev Cell Dev Biol 17:387– 403. Whitlock C, Witte O. 1987. Long-term culture of murine bone marrow precursors of B lymphocytes. Methods Enzymol 150:275–286. Wintrobe M. 1980. Milestones on the path of progress. In: Wintrobe M, editor. Blood, pure and eloquent. New York: McGraw-Hill Book Company. p 1–31. Wright D, Wagers A, Gulati A, Johnson F, Weissman I. 2001. Physiological migration of hematopoietic stem and progenitor cells. Science 294:1933–1936. Ying Q, Nichols J, Evans E, Smith A. 2002. Changing potency by spontaneous fusion. Nature 416:485– 487. Yoder M, Palis J. 2001. Ventral (yolk sac) hematopoiesis in the mouse. In: Zon L, editor. Hematopoiesis. London: Oxford University Press. p 180 –191. Zanjani E, Pallavicini M, Ascensao J, Flake A, Langlois R, Reitsma M, MacKintosh F, Stutes D, Harrison M, Tavassoli M. 1992. Engraftment and long-term expression of human fetal hematopoietic stem cells in sheep following transplantion in utero. J Clin Invest 89: 1178 –1188.