haematopoietic stem cells

11
Review Article Haematopoietic stem cells { Dominique Bonnet* Cancer Research UK, London Research Institute, London, UK * Correspondence to: D. Bonnet, PhD, Haematopoietic Stem Cell Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln’s Inn Fields, London, WC2A 3PX, UK. E-mail: dominique.bonnet@ cancer.org.uk Abstract Considerable efforts have been made in recent years in determining the composition of the cell types that constitute the human haematopoietic stem cell (HSC) compartment. These studies have emphasized the heterogeneity of the human HSC in terms of proliferative and self-renewal capacities. Recent studies have indicated that CD34 is not the universal marker of all human HSCs. New markers for purifying HSCs have been described. A number of genes that regulate the formation, self-renewal, or differentiation of HSCs has been identified. The elucidation of the molecular phenotype of the HSC has just begun. Finally, an unexpected degree of developmental or differentiation plasticity of HSC has emerged. This review summarizes all the recent advances made in the human HSC field and examines the impacts that these discoveries may have both clinically and in understanding the organization of the human haematopoietic system. Copyright # 2002 John Wiley & Sons, Ltd. Keywords: haematopoietic stem cell (HSC); xenotransplantation model; NOD/SCID; fetal sheep; regulation; plasticity; transdifferentiation Introduction Stem cell populations from a variety of tissues offer great promise for tissue regeneration, cell-based trans- plantation therapies, and the eventual development of clinically effective gene therapy protocols [1–3]. The best characterized stem cells are those responsible for haematopoiesis. All the experimental strategies and conceptual paradigms that are applicable to stem cells in general were defined first in this system [4]. The hall- mark properties of haematopoietic stem cells (HSCs) are as follows: HSCs have the ability to balance self- renewal against differentiation cell fate decisions; they are multipotent, a single stem cell producing at least eight to ten distinct lineages of mature cells; they have an extensive proliferative capacity that yields a large number of mature progeny; HSCs are rare, with a frequency of 1 in 10 000 to 100 000 total blood cells; they are slowly cycling in a steady-state adult haema- topoietic system. Thus, determining the composition and relationship of the cell types that constitute the human stem cell compartment may help both to identify the cellular and molecular factors that govern normal and leukaemic stem cell development and to advance clinical applications of transplantation, gene therapy, stem cell expansion, and tumour cell purging. Furthermore, recent studies have provided compelling evidence that the adult stem cells may have a pre- viously unsuspected degree of developmental or differ- entiation plasticity (see the review in this issue by Poulsom et al. and reviews in refs [5–11]). These recent findings have shattered the existing dogma that only embryonic stem cells are capable of giving more than one tissue. In this review, we present a brief summary of the most recent findings in the field of human HSCs, discussing the assays used so far, the cellular and mole- cular phenotypes of HSCs, and the regulatory mechan- isms involved. We will finish with a short overview on recent results on HSC plasticity and the potential cli- nical implications of this new property. Characterization of HSCs Surrogate in vitro assays A number of in vitro assays have been described that assess primitive human progenitors. These include long- term culture-initiating cell (LTC-IC) assays, which detect primitive cells capable of giving rise to colony- forming cells (CFCs) after 5 weeks of culture on com- petent feeder layers [12,13]; cobblestone area-forming cell (CAFC) assays [14,15]; and extended-LTC-IC (E- LTC-IC) assays [16]. The E-LTC-IC defines a small subpopulation of LTC-IC, which has more extensive proliferative capacity and can be maintained for up to 10 weeks in culture. These assays enumerate primitive myeloid progenitors, but not cells with multi-lineage differentiation or self-renewal potential. More recently, several groups have developed cultures that allow the differentiation of single human Lin x CD34 + cells into cells with myeloid, natural killer (NK), B-lymphoid, dendritic, and/or T-lymphoid phenotypes, showing that a single cell can differentiate in vitro into multiple lineages [17–19]. However, none of these assays is able to generate secondary primitive progenitors that again {Note: Cancer Research UK, London Research Institute comprises the Lincoln’s Inn Fields and Clare Hall Laboratories of the former Imperial Cancer Research Fund following the merger of the ICRF with the Cancer Research Campaign in February 2002. Journal of Pathology J Pathol 2002; 197: 430–440. Published online 30 May 2002 in Wiley InterScience (www.interscience.wiley.com). DOI: 10.1002 / path.1153 Copyright # 2002 John Wiley & Sons, Ltd.

Upload: dominique-bonnet

Post on 06-Jul-2016

213 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Haematopoietic stem cells

Review Article

Haematopoietic stem cells{

Dominique Bonnet*Cancer Research UK, London Research Institute, London, UK

*Correspondence to:D. Bonnet, PhD, HaematopoieticStem Cell Laboratory, CancerResearch UK, London ResearchInstitute, 44 Lincoln’s Inn Fields,London, WC2A 3PX, UK.E-mail: [email protected]

Abstract

Considerable efforts have been made in recent years in determining the composition of the cell

types that constitute the human haematopoietic stem cell (HSC) compartment. These studies have

emphasized the heterogeneity of the human HSC in terms of proliferative and self-renewal

capacities. Recent studies have indicated that CD34 is not the universal marker of all human

HSCs. New markers for purifying HSCs have been described. A number of genes that regulate

the formation, self-renewal, or differentiation of HSCs has been identified. The elucidation of the

molecular phenotype of the HSC has just begun. Finally, an unexpected degree of developmental

or differentiation plasticity of HSC has emerged. This review summarizes all the recent advances

made in the human HSC field and examines the impacts that these discoveries may have both

clinically and in understanding the organization of the human haematopoietic system. Copyright

# 2002 John Wiley & Sons, Ltd.

Keywords: haematopoietic stem cell (HSC); xenotransplantation model; NOD/SCID; fetal

sheep; regulation; plasticity; transdifferentiation

Introduction

Stem cell populations from a variety of tissues offergreat promise for tissue regeneration, cell-based trans-plantation therapies, and the eventual development ofclinically effective gene therapy protocols [1–3]. Thebest characterized stem cells are those responsible forhaematopoiesis. All the experimental strategies andconceptual paradigms that are applicable to stem cellsin general were defined first in this system [4]. The hall-mark properties of haematopoietic stem cells (HSCs)are as follows: HSCs have the ability to balance self-renewal against differentiation cell fate decisions; theyare multipotent, a single stem cell producing at leasteight to ten distinct lineages of mature cells; they havean extensive proliferative capacity that yields a largenumber of mature progeny; HSCs are rare, with afrequency of 1 in 10 000 to 100 000 total blood cells;they are slowly cycling in a steady-state adult haema-topoietic system. Thus, determining the compositionand relationship of the cell types that constitute thehuman stem cell compartment may help both toidentify the cellular and molecular factors that governnormal and leukaemic stem cell development and toadvance clinical applications of transplantation, genetherapy, stem cell expansion, and tumour cell purging.Furthermore, recent studies have provided compellingevidence that the adult stem cells may have a pre-viously unsuspected degree of developmental or differ-entiation plasticity (see the review in this issue byPoulsom et al. and reviews in refs [5–11]). These recent

findings have shattered the existing dogma that onlyembryonic stem cells are capable of giving more thanone tissue.

In this review, we present a brief summary of themost recent findings in the field of human HSCs,discussing the assays used so far, the cellular and mole-cular phenotypes of HSCs, and the regulatory mechan-isms involved. We will finish with a short overview onrecent results on HSC plasticity and the potential cli-nical implications of this new property.

Characterization of HSCs

Surrogate in vitro assays

A number of in vitro assays have been described thatassess primitive human progenitors. These include long-term culture-initiating cell (LTC-IC) assays, whichdetect primitive cells capable of giving rise to colony-forming cells (CFCs) after 5 weeks of culture on com-petent feeder layers [12,13]; cobblestone area-formingcell (CAFC) assays [14,15]; and extended-LTC-IC (E-LTC-IC) assays [16]. The E-LTC-IC defines a smallsubpopulation of LTC-IC, which has more extensiveproliferative capacity and can be maintained for up to10 weeks in culture. These assays enumerate primitivemyeloid progenitors, but not cells with multi-lineagedifferentiation or self-renewal potential. More recently,several groups have developed cultures that allow thedifferentiation of single human LinxCD34+ cells intocells with myeloid, natural killer (NK), B-lymphoid,dendritic, and/or T-lymphoid phenotypes, showing thata single cell can differentiate in vitro into multiplelineages [17–19]. However, none of these assays is ableto generate secondary primitive progenitors that again

{Note: Cancer Research UK, London Research Institute comprisesthe Lincoln’s Inn Fields and Clare Hall Laboratories of the formerImperial Cancer Research Fund following the merger of the ICRFwith the Cancer Research Campaign in February 2002.

Journal of PathologyJ Pathol 2002; 197: 430–440.Published online 30 May 2002 in Wiley InterScience (www.interscience.wiley.com). DOI: 10.1002 /path.1153

Copyright # 2002 John Wiley & Sons, Ltd.

Page 2: Haematopoietic stem cells

have multi-lineage differentiation potential (i.e. self-renewal potential). Using a stroma-based culture sys-tem supplemented with early-acting cytokines, Verfaillie’sgroup defined a very primitive human progenitor, themyeloid–lymphoid initiating cells (ML-ICs), capable ofgenerating multiple secondary progenitors that havethe ability to reinitiate long-term multi-lineage haema-topoiesis [20].

In vivo assays

A conclusive way to assay stem cells is based on theircapacity to repopulate the entire hematopoietic systemin conditioned recipients after transplantation [21]. Inmice, the phenotype and function of haematopoieticstem cells (HSCs) have been characterized using com-petitive in vivo repopulation assays [22–25]. As suchrepopulation assays cannot be performed in humans,surrogate in vivo and in vitro assays are used to evalu-ate human HSCs. In an attempt to develop in vivoanimal models for human haematopoiesis, severalgroups have transplanted human cells in xenogeneictransplant recipients, such as fetal sheep [26,27] orimmune-deficient mice [28,29].

Non-obeses diabetic–severe combined immunodeficient

(NOD/SCID) mouse model

The engraftment of normal human haematopoieticcells in immunodeficient mice provides an assay thatmeasures the repopulating capacity of human stemcells. Dick’s group has shown that intravenous injec-tion of human bone marrow (BM) [30] or cord blood[31] into severe combined immunodeficient (SCID)mice resulted in the engraftment of primitive cells thatproliferated and differentiated in the murine BM, pro-ducing large numbers of LTC-ICs, CFCs, immatureCD34+Thy.1+, CD34+CD38x cells, and mature mye-loid, erythroid and lymphoid cells. The primitive cellsthat initiated the graft were operationally defined asSCID repopulating cells (SRCs). Kinetic experimentsshowed that only 0.1% of the injected CFCs and LTC-ICs were detectable in the murine BM 2 days post-transplant and that there was a large expansion ofthese cells, as well as primitive CD34+ cells, over thenext 4 weeks, implying their production from a moreprimitive cell [32]. Since this pioneering work, a num-ber of research groups have confirmed these results[33–35].

Most of the earlier studies used SCID mice, whichwere not ideal, as they still possessed significant antigen-non-specific immunity. As a result, high cell doses wererequired to overcome any residual host resistance,ruling out the development of quantitative assays andany purification strategies. A new mouse strain, createdby crossing the SCID gene onto the non-obese diabetic(NOD) background, proved to be a better recipient.Compared with SCID mice, this new strain of mice –NOD/Lt-Sz-Scid/Scid (NOD/SCID) – appears to havelower NK and complement activities, and a defect inmacrophages [36]. A lower number of cells (ten to

20-fold less) were necessary to engraft NOD/SCIDmice, when compared with SCID mice. Overall, thesemice showed high levels of engraftment for normal andleukaemic human transplants and, more importantly,enabled engraftment with lower cell doses, renderingpurification strategies possible [29,37,38]. The onlylimitation of these NOD/SCID mice is their inabilityto support human T-cell development. On the con-trary, beige-nude-SCID (Bnx) mice allow T-cell devel-opment, but not B-cell differentiation [39,40]. Othermouse strains have also become available recently: b2-microglobulin knockout/NOD/SCID, Rag1 knockout/NLD, and the nude/NOD/SCID mice [41–43]. Thecapabilities of each of these new strains still need toexplored.

The fetal sheep HSC assay

This assay, based on the permissive environment of theearly gestational age fetus, aims at the development ofa large animal model of human haematopoiesis insheep. The preimmune sheep fetus assay allows thelong-term engraftment and multi-lineage expression ofhuman HSCs in the absence of irradiation or other mye-loablative therapies [44–46], possibly due to reducedNK cells and the preimmune status in early sheepgestation [47]. An essential feature of this model isthat human HSCs primarily engraft host marrow andpersist for long periods into post-natal life [44–46],showing multi-lineage expression and biological respon-siveness to human cytokines [45]. The multi-lineageexpression included T- and B-lymphoid cells [48].Furthermore, this model is relatively specific of thehuman HSC pool. Indeed, while both CD34+CD38+

and CD38x subpopulations engraft the sheep, onlyprimary recipients engrafted with CD34+CD38x cellsexhibited long-term persistence of human cells, whereasCD34+CD38+ cells persist for a short period of timeonly and were unable to engraft into a secondaryrecipient. Although not ideal, the human/sheep xeno-graft model is comparable to the NOD/SCID assay; inparticular, the sheep model does not require myelo-ablation, while it allows prolonged follow-up studiesafter birth. However, widespread utilization of thismodel is hindered by its high costs.

Isolation and purification of HSCs

Haematopoietic stem cells have been enriched using avariety of techniques, including density centrifugation,activation and/or cell-cycle status, and surface antigenexpression, but no unique characteristics have beenfound to identify these elusive cells specifically. Animportant point in the isolation of HSCs is the one-to-one correspondence between physically purified cellsand their potential ability to function as stem cells.

Cell surface markers

Systematic functional analysis of haematopoietic cellsexpressing a particular cell surface antigen or other

Haematopoietic stem cells 431

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 3: Haematopoietic stem cells

markers has led to the identification of rare popula-tions highly enriched for stem cell (SRC and/or LTC-IC) activity. HSCs do not express many of the surfaceantigens (‘lineage markers’) that are characteristic ofterminally differentiating haematopoietic cells. Thus,the removal of such lineage-positive cells leaves asuspension of predominantly immature cells.

CD34 as the universal marker of HSCs

CD34 was discovered originally as the result of astrategy to develop antibodies that recognize smallsubsets of human marrow cells, but not mature bloodand lymphoid cells [49]. The discovery of the sialomu-cin CD34 as a haematopoietic cell surface antigen hastransformed and accelerated studies on human haema-topoietic development. Cell surface expression of theCD34 antigen has rapidly become the distinguishingfeature used as the basis for the enumeration, isolation,and manipulation of human stem cells, because CD34is down-regulated as cells differentiate into moreabundant mature cells [50,51]. Despite this extensiveuse, the normal function of the CD34 molecule inhaematopoiesis has remained enigmatic. Studies havedescribed its potential role in cell adhesion and in thehoming process [52]. Human and murine CD34 homo-logues are highly conserved in their protein codingregions [53,54]. The cytoplasmic domains of the humanand mouse proteins share 90% amino acid identity; thetransmembrane and C-terminal regions of the extra-cellular domains are also well conserved, with 73–82%amino acid identity. The N-terminal portions of theextracellular domains are the least conserved regions ofthe molecule (45% amino acid identity). The expressionpattern of CD34 is also conserved between human andmouse. Thus, in addition to being expressed selectivelyon stem cells and early progenitors during human[49,55,56] and murine [53,54] haematopoiesis, bothmouse and human CD34 are expressed outside thehaematopoietic system on vascular endothelial cells[57,58] and some fibroblasts [53,59]. This distributionsuggests a function outside haematopoiesis. Transplantstudies in several species, including baboons and mice,have shown that long-term marrow repopulation canbe provided by CD34+ selected cells. Thus, all relevantclinical and experimental protocols are designed forCD34+ cells enriched by a variety of selection methods.However, several recent studies have suggested thatthere may be human and murine stem cells that do notexpress CD34.

Other stem cell markers

CD133 represents the human homologue of prominin5 transmembrane glycoproteins (PROML 1) [60–63].Several studies have shown the presence of CD133 cellsthat co-express CD34, c-kit, and other cell surfacemarkers [64,65]. Taken together, these studies clearlyindicate that CD133 represents a significant cell surfacemarker for the identification of human HSCs, but itremains unclear whether the use of this marker

provides any distinct advantage over CD34 expression.Further details on the expression of CD133 expressionin human stem cells can be found in the recent reviewby Bhatia [66].

Another recent marker allowing the isolation ofhuman HSCs is the vascular growth factor receptor 2(KDR) [67]. The KDR+ cell fraction, essentially Linx,is largely present in populations enriched for HSCs,namely CD34+CD38x, CD90+, and CD117low cells.It has been reported that CD34+KDR+ is highlyenriched in putative HSCs (SRC and E-LTC-IC orCAFC). Conversely, haematopoietic progenitors withno self-renewal activity are restricted to and highlyenriched in the CD34+KDRx cell fraction. Severalother markers have proven useful in further dividingthe population into more functionally homogeneouspopulations, e.g. CD90, CD117, and CD38 [68,69].Further details on the procedures of isolation of theseHSCs can be found in the review article by Thomaset al. [70].

Side population

In 1996, Goodell et al. reported a new method ofobtaining enriched populations of HSCs from adultmouse bone marrow [71]. This procedure exploits theability of HSCs to efflux the fluorescent dye, which,like the activity of P-glycoprotein (encoded by theMDR gene), is verapamil-sensitive [71]. The Hoechst33342 low cells thus isolated were called side popu-lation (SP) cells and were found to have the sameLin xScal+CD34x phenotype independently identifiedin adult murine HSCs [72]. SP cells have since beenidentified in adult bone marrow from several speciesincluding humans [73]. To date, a description of thefunctional activities of human SP cells in normal indi-viduals has been limited to an in vitro study of cordblood [74] and more recently to an in vivo study ofhuman fetal liver [75]. In this latter study, it was demon-strated that SP cell are present in the secondxtrimesterhuman fetal liver. These cells include all transplantableHSC activity detectable in NOD/SCID mice and alsoother more differentiated haematopoietic cell types [75].More recently, Sorrentino’s group established the linkbetween Bcrp1/ABCG2 expression and the SP pheno-type [76].

Heterogeneity of the human HSCcompartment

Initially, it was assumed that in humans, only cellsexpressing CD34 would display HSC activity, as thefrequency of CD34+ cells is now commonly used toanticipate the adequacy of clinical haematopoietic celltransplants. Recently, however, several groups includ-ing ours have provided evidence of various types ofhuman HSCs that do not express detectable levels ofCD34. Xenograft repopulation assays using fetal sheepand immunodeficient mice have been crucial for theidentification of human CD34x stem cells, as little or

432 D. Bonnet

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 4: Haematopoietic stem cells

no clonogenic cell (CFC) or LTC-IC activity wasobserved within the human LinxCD34x cell popula-tion. Using the sheep xenograft model, Zanjani et al.[77] showed that LinxCD34x cells contained stem cellscapable of long-term repopulation and multi-lineage

differentiation in vivo. Moreover, these cells were alsoable to repopulate secondary recipients, attesting to theextensive self-renewal potential of the engrafting cells.The fact that large numbers of CD34+ cells werefound in repopulated sheep suggests that the stem cellswithin the LinxCD34+ cell fraction are more primi-tive than CD34+ cells. Even though the possibility ofslight contamination by CD34+ cells could not be

ruled out, this seems unlikely based on limiting dilu-tion analysis.

Using the NOD/SCID model, the presence of anovel human haematopoietic repopulating cell that isdevoid of lineage-specific markers and of the CD34antigen has also been reported [78]. This population isnot only distinct from LinxCD34+ cells in the absenceof CD34, but also in the lack of HLA-DR and Thy. 1(CD90) markers. In addition to phenotypic differences,

several additional lines of evidence functionallydistinguish this novel stem cell population. WhileLinxCD34x cells have limited haematopoietic activityin the CFC and LTC-IC assays, LinxCD34+CD38x

cells are highly clonogenic in these assays. Further-more, the in vitro response to growth factor stimulationof LinxCD34x cells is clearly distinct from that ofLinxCD34+CD38x cells [6]. LinxCD34xCD38x cells

were unable to proliferate or increase their clonogeniccapacity under culture conditions known to induceproliferate or increase their clonogenic capacity underculture conditions known to induce proliferative anddifferentiative responses in LinxCD34+CD38x cells.Similar to the sheep model, the development ofCD34+ cells, as well as the more differentiated pro-

geny in vivo, suggests that CD34x cells might be moreprimitive than the CD34+ stem cells. Thus, it can beconcluded that the CD34x SRC found within theLinxCD34xCD38x cell fraction represents a novelrepopulating cell within the human haematopoietichierarchy. The fact that the LinxCD34x cell fractionfrom mice and humans contains repopulating cells

indicates an evolutionary conservation of this novelstem cell population [74,77–79]. The identification ofCD34x SRCs within the LinxCD34x subfractionestablishes that the human HSC compartment is morecomplex than previously recognized. However, it is notknown whether CD34x stem cells are important clini-cally. Our current knowledge on the nature of theprecise relationship between CD34x and CD34+ stemcells is illustrated in Figure 1 and has been reviewedrecently [80].

Regulation of the HSC

Over the years, several models have been advancedproposing that haematopoietic lineage determination isdriven extrinsically (through growth factors, stroma orother external influences) [3,81–84], intrinsically (asdescribed in stochastic models) [85,86], or both [87,88].Within the haematopoietic microenvironment, earlyprogenitors are maintained in specific compartmenta-lized niches, where they interact with other cell typesand components of the extracellular matrix [89,90].The microenvironment has been reported to influencesurvival, proliferation, and differentiation [91,92]. Morerecently, it has been suggested that the primary func-tion of these extrinsic signals, including growth factors,is to support the survival and development of committed

Figure 1. Human haematopoietic stem cell hierarchy

Figure 2. Evolving view of adult haematopoietic stem cellcapacity

Haematopoietic stem cells 433

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 5: Haematopoietic stem cells

cells, whereas lineage commitment can be attributedto cell intrinsic mechanisms [85,86,88,93]. Overall, itappears that the regulation of haematopoiesis is theresult of multiple processes involving cell–cell andcell–extracellular matrix interactions, the action ofspecific growth factors and other cytokines, as well asintrinsic modulators of haematopoietic development.

Role of transcription factors in the regulation ofHSC fate

Ultimately, all signal transduction pathways convergeat the level of gene expression where positive and nega-tive modulators of transcription interact and delineatethe pattern of genes expressed by the cell and itsoverall haematopoietic response. As such, transcriptionfactors represent the nodal point of the control ofhaematopoiesis [88,94,95]. It is the alternative expres-sion of specific combinations of transcription factorsthat determines the survival, proliferation, commit-ment, and differentiation responses of haematopoieticprogenitors to such signals, whether they arise fromextrinsic or intrinsic regulatory factors. Increasingevidence suggests that different families of transcrip-tion factors regulate the developmental programme ofstem cells [95] and when their expression is disrupted,leukaemic proliferation is initiated [96]. The mostimportant specific transcription factors identified todate that act at the earliest stage of blood formationare members of the GATA and SCL/tal-1 gene families[95]. GATA-1/2 are the earliest haematopoietic specificfactors to be expressed during mesodermal inductionand in amphibians, expression can be used to defineregions of the ventral mesoderm that give rise tohaematopoietic progenitors. Mice in which the SCL/tal-1 gene was disrupted do not generate definitivehaematopoietic cells, suggesting that SCL/tal-1 func-tions very early in haematopoietic development at thestage of specification of ventral mesoderm to a bloodcell fate [95,97]. Disruption of the GATA-1 and LMO-2/rbtn-2 genes also severely impairs early erythroidlineage development. The fact that the most frequenttargets of chromosomal translocations in acute leukae-mias are genes that encode transcription factorsemphasizes the critical role of these master regulatorymolecules in the control of blood development [96,98].Translocations that inappropriately activate transcrip-tion factor genes in ALL and AML show remarkablespecificity for haematopoietic cells blocked in definedstages of differentiation [98]. This property suggeststhat the different oncoproteins produced by chromo-somal translocations interfere specifically with trans-criptional networks that normally function in concertwith growth factors and their receptors to regulatehaematopoiesis.

Finally, a number of myeloid transcription factorshave been identified through their involvement inleukaemias, either as a result of abnormal expression,such as PU.1 and WT-1 [99,100], or through theirinvolvement at the site of a consistent chromosomal

translocation (examples include AML1, CBFb, andSCL/Tal.1) [96,98]. The technique of gene disruptionhas been used to confirm the role of these specificfactors in myeloid development [95]. Several transcrip-tion factors that are expressed in haematopoietic cellsand play a role in leukaemogenesis (chromosomal trans-locations) contain a homeo-box DNA binding region.HOX gene family members encode DNA-bindingtranscription factors characterized by a conserved60-amino acid homeo-domain which is homologousto the Drosophila homeo-box proteins and also plays acrucial role in mammalian embryonic axis formation[101]. Human HOX genes are organized on differentchromosomes in four major clusters, A, B, C, and D,each of which consists of nine to 12 tandem genes[102]. While their role in embryonic axis formationhas been well studied, the role that they play in regula-ting haematopoiesis and leukaemogenesis is less clear,although several recent studies point to a major role[103,104]. Furthermore, further detailed analysis of Hoxgene expression in functionally distinct subpopulationsof CD34+ cells has shown that genes, primarilylocated at the 3k end of the clusters (e.g. HOX B3 andB4), are preferentially expressed in the subpopulationcontaining the most primitive haematopoietic cells[104]. Overexpression of Hox B4 has been shown tofavour self-renewal over differentiation [105,106].

Some initial clues to the regulatory relationshipsbetween transcription factors are coming. For example,three recent studies over the past year have reportedthe direct physical interactions and cross-antagonismsof PU.1 and GATA-1 [107–109]. Additional examplesof cross-antagonisms between lineage-specific trans-cription factors, or their co-factors, are beginning toemerge and reveal novel mechanisms (see the review byCantor and Orkin [110]). Thus, there appears to be adynamic balance of forces that ultimately determinethe phenotype of a cell.

Role of mesodermal inducing factors in theregulation of HSC fate

As indicated above, amongst the approximately 50different chromosomal translocations that have beencloned, the majority involve transcription factors.However, there are numerous other molecules forwhich there is reasonable information to warrant con-sideration for roles in stem cell regulation. One of theseis the collection of mammalian Notch molecules, theirligands Delta-like, Jagged family, the fringe family ofsignalling modifiers, as well as the variety of down-stream regulators [111]. Notch can affect cell fate byregulating transcription directly via association withnuclear factors and can thus affect the growth anddifferentiation of HSCs. Incubation of human HSCswith soluble Notch ligands, Jagged 1, has recentlybeen shown to drive in vitro HSC self-renewal. Wheninjected into NOD/SCID mice, Jagged 1-treated HSCswere capable of reconstituting lymphoid and myeloiddifferentiation [112]. Furthermore, the constitutive

434 D. Bonnet

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 6: Haematopoietic stem cells

expression of Notch 1C using a retroviral vector is capa-

ble of generating a clonal population of immortalized

HSCs [113].Members of the TGF-b superfamily and those that

act through the TGF-b pathway also appear to be very

prominent [114,115]. TGF-b itself is a potent inhibitor

of murine stem cells and human LTC-ICs and appears

to be a major regulator that keeps stem cells in a

quiescent state [116]. Interestingly, treatment of human

LTC-ICs with anti-TGF-b can induce them to enter

the cell cycle [117]. Recently, it has been demonstrated

that bone morphogenetic protein (BMP-2 and BMP-4)

and activin A are potent ventralizing factors and

inducers of haematopoietic tissue, and that BMP-4

and GATA-2 can function in two adjacent germ layers,

mesoderm and ectoderm, to participate in blood cell

formation during embryogenesis [118]. We reported

that BMPs are capable of regulating the proliferation

and differentiation of highly purified primitive human

haematopoietic cells. Populations of LinxCD34+-

CD38x cells isolated from human haematopoietic

tissue were shown to express the BMP type I receptors

ALK-3 and ALK-6, and their downstream transducers

SMAD 1, 4 and 5. Soluble BMP-2, BMP-4, and

BMP-7 induced dose-dependent responses in human

LinxCD34+CD38x cord blood cells, as determined by

changes in proliferation, clonogenicity, cell surface

phenotype, and multi-lineage repopulation capacity in

NOD/SCID mice (SCID-repopulating cells; SRCs).

Similar to TGF-b, treatment of purified cells with

BMP-2 or BMP-7 at high concentrations inhibited

proliferation, yet maintained primitive CD34+CD38x

phenotype and SRC activity. In contrast, low concen-

trations of BMP-4 induced the proliferation and

differentiation of LinxCD34+CD38x cells, whereas

at higher concentrations BMP-4 extended the period in

which ex vivo cultured cells maintained repopulating

function. This study illustrates a novel role for the

BMP pathway and suggests that this family of mor-

phogens continues to play an important role in human

blood stem cells beyond haematopoietic tissue specifi-

cation [119].Other factors that play a role in mesodermal tissue

include basic fibroblast growth factor (bFGF), which

has been shown by Allouche to be involved in the

proliferation and differentiation of numerous cell types

including those in the haematopoietic lineage [120].

bFGF is expressed mostly in tissues of mesoderm and

neuroectoderm origin, and plays an important role in

mesoderm induction, together with TGF-b. bFGF is

expressed and produced by bone marrow stromal cells,

as well as by cells from several mature peripheral blood

lineages. FGF-receptors (FGF-Rs) are expressed on

nearly every cell of haematopoietic origin tested so far

and bFGF can regulate haematopoiesis, by acting on

stromal cells, early and committed haematopoietic

progenitors, and mature blood cells. It synergizes with

haematopoietic cytokines, or antagonizes the negative

regulatory effects of TGF-b.

The genetic programme of the HSC

As mentioned above, several molecules have beenshown to play a role in several aspects of haemato-poetic development, but the elucidation of the mole-cular phenotype of the HSC has just begun [121,122].Indeed, key aspects of the stem cell regulation arelikely to be emergent properties of interacting path-ways and networks, the elucidation of which requiresan extensive description of the genetic programmeavailable to the stem cell. Recently, using a highly sen-sitive single-cell RT-PCR approach, or a quantitativereal-time PCR method, it has been demonstrated thatHSCs co-expressed several lineage-restricted gene sets[123,124]. These data indicate the complexity of geneexpression from which distinct patterns must emergeduring the lineage commitment process by selective up-regulation of the relevant gene cohorts, accompaniedby the inactivation of others. To take into account thisnew concept of regulation, a model for the molecularnature of the uncommitted stem cell ‘state’ has recentlybeen postulated [125]. This model proposes that a HSCpossesses a molecular ‘ground state’ composed of lowlevels of transcripts normally associated with thefunction of various mature cell lineages. According tothis model, a commitment process would involve theselection and amplification of an appropriate subset ofthe available transcriptional programme and possiblerepression of the remainder. Interestingly, the featuresof the ‘ground state’ model are consistent with theclinical description of gene expression promiscuity incertain mixed leukaemias. The features of the groundstate model fit well with the stochastic model of stemcell commitment.

Engraftment of HSC

In a transplantation context, a stem cell is definedretroactively as a biological activity that can give riseto substantial measurable numbers of mature cells.Similarly, the presence of a single clonotypic marker indonor-derived cells of different lineages defines multi-potentiality. Thus, following transplantation into therecipient, HSCs, in order to engraft and reconstitutethe bone marrow functions, must home to and lodge inthe specialized niches of the BM microenvironment. Atpresent, only partial understanding of the cellular andmolecular mechanisms governing homing exists. It isbelieved that an intricate process involving interac-tions between adhesion molecules and their counter-receptors expressed on HSCs and endothelial cellsdirects the cells. In many aspects, the homing of HSCsduring transplantation mimics the natural movementof these cells during ontogeny. The direct involvementof particular adhesion molecules in homing has beenelucidated [126,127]. Recently, Zanjani et al. demon-strated that VLA-4 played a central role in the homingand engraftment of transplanted human cells to theBM of sheep fetuses [128]. Another ligand receptor

Haematopoietic stem cells 435

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 7: Haematopoietic stem cells

pairing, SDF-1–CXCR4, has also been implicated inselectively directing the homing of HSCs to the BM.The engraftment of human cells in NOD/SCID micewas prevented by treatment with antibodies againstCXCR4. Furthermore, the expression of CXCR4 onCD34+ human HSCs has suggested a role for thesechemoattractants in the homing process [129]. How-ever, Rosu-Myles et al. recently demonstrated thatCXCR4 expression on human HSCs was not requiredfor effective stem cell repopulation function [130].Redundancy between different chemoattractant mole-cules may be responsible for this discrepancy (for moredetails see the reviews in refs 131 and 132), Whilemore is known about homing, less is understood aboutHSC niches [133].

Recently, the spatial organization of subpopulationsof haematopoietic cells following syngeneic transplan-tation in mice has been investigated. The studydemonstrated that the spatial distribution of trans-planted cells is not a random process; candidate stemcells exhibited selective lodgement in the endostealregion of the bone [134].

HSC plasticity

The notion of adult stem cell plasticity has alreadybeen discussed in this issue by Poulsom et al. and canbe found in different recent review articles [5–11].Thus, we will restrict our comments here on the recentstudies involving HSCs, including some of our recentwork on HSC plasticity using the NOD/SCID model.

Several striking observations have been reportedwhich are beginning to raise questions about the tradi-tional view of HSC biology (illustrated in Figure 2).For example, some observations have challenged thedogma that HSCs are committed solely to the haema-topoietic lineage [135–142]. In particular, reports haveindicated that the bone marrow of adult rodents con-tains cells with the capacity to give rise to hepatocytes,muscle tissue, or even neurons. Lagasse et al. estab-lished that some haematopoietic stem cells present inadult bone marrow co-purified with stem cells thatgave rise to hepatocytes, supporting the new conceptthat somatic adult stem cells can change cell fate [143].However, these reports were based on adult rodentstem cell populations, which may differ from humanstem cells in their capacity to give rise to multipletissues. Nevertheless, the studies by Theise et al. andAlison et al. based on patients who received bonemarrow transplants, indicate that some human adultstem cells present in bone marrow also have the abilityto give rise to hepatocytes [144,145]. These two studiesdid not distinguish whether HSCs, mesenchymal stemcells or hepatocyte stem cells circulating in the bonemarrow were responsible for the observed liver engraft-ment.

We reported recently the potential of a highlypurified population present in adult bone marrow andumbilical cord blood (human C1qRp

+ stem cells) to

differentiate in vivo into hepatocytes (Danet et al.,in preparation). These data provide the first directdemonstration that a highly purified and phenotypi-cally defined human adult stem cell population canrepopulate the bone marrow and differentiate in vivointo functional hepatocytes, using the NOD/SCIDmouse model. Not only HSCs, but also mesenchymalstem cells (MSCs) present within the adult humanmarrow possess remarkable plasticity. Kopen et al.showed that cells that gave rise to neurons and gliawere derived from cultures of adherent bone marrowstroma, suggesting that they included MSCs [146].More recently, Verfaillie’s group identified mesodermalprogenitor cells (MPCs), which have the capacity todifferentiate into osteoclasts, chondrocytes, adipocytes,skeletal myoblasts, and endothelial cells [147,148]. Inthe light of the recent identification of a single mousebone marrow-derived stem cell with multi-organ andmulti-lineage engraftment [149], additional experimentswill be required to determine the full developmentalcapacity of marrow-derived stem cells.

Conclusion

Despite the lack of definitive proof of plasticity inmany of the present studies, the use of HSCs for thepotential treatment of human diseases such as liverdiseases and muscular dystrophy represents an excitingnew therapeutic strategy. HSCs represent a safe andaccessible source of stem cells that can be geneticallymanipulated and may thus prove to be an ideal vehiclefor delivering therapeutic genes to other organs. TheNOD/SCID xenotransplant model will play an impor-tant role in evaluating this potential.

A more detailed and systematic analysis of differentsomatic stem cell types based on their gene expressionprofiles and functional properties is needed for a betterunderstanding of the nature of stem cells and theirdevelopmental plasticity.

The study of HSCs has become extremely exciting,with new insights into HSC biology being reported ona weekly basis. HSCs may be more plastic than pre-viously appreciated and assumptions about theirphenotype may be under revision. We are entering avery exciting area of investigation where dogma willevolve rapidly and new techniques will open up newexperimental approaches.

References

1. Fuchs E, Sefre J. Stem cells: a new lease of life. Cell 2000; 100:

143–155.

2. Weissman I. Stem cells; units of development, units of

regeneration, and units in evolution. Cell 2000; 100: 157–168.

3. Watt F, Hogan B. Out of Eden: stem cells and their niches.

Science 2000; 287: 1427–1430.

4. Morrison SJ, Shah NM, Anderson DJ. Regulatory mechanisms

in stem cell biology. Cell 1997; 88: 287–298.

5. Lemischka I. The power of stem cells reconsidered? Proc Natl

Acad Sci U S A 1999; 96: 14193–14195.

436 D. Bonnet

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 8: Haematopoietic stem cells

6. Orkin SH. Stem cell alchemy. Nature Med 2000; 6: 1212–1213.

7. Blau HM, Brazelton TR, Weimann JM. The evolving concept

of stem cell: entity or function? Cell 2001; 105: 829–841.

8. Morrison SJ. Stem cell potential: can anything make anything?

Curr Biol 2001; 11: R7–R9.

9. Anderson DJ, Gage FH, Weissman IL. Can stern cells cross

boundaries? Nature Med 2001; 7: 393–395.

10. Lagasse E, Shizuru JA, Uchida N, Tsukamoto A, Weissman IL.

Toward regenerative medicine. Immunity 2001; 14: 425–436.

11. Wulf GG, Jackson KA, Goodell MA. Somatic stem cell plas-

ticity: current evidence and emerging concepts. Exp Hematol

2001; 29: 1361–1370.

12. Sutherland HJ, Lansdorp PM, Henkelman DH, et al. Func-

tional characterization of individual human hematopoietic stem

cells cultured at limiting dilution on supportive marrow stromal

layers. Proc Natl Acad Sci U S A 1990; 87: 3584–3588.

13. Sutherland HJ, Eaves CJ, Eaves AC. Characterization and

partial purification of human marrow cells capable of initiating

long-term hematopoiesis in vitro. Blood 1989; 74: 1563–1570.

14. Ploemacher RE, van der Sluijs JP, Voerman JS, Brons NH. An

in vitro limiting dilution assay of long-term repopulating hema-

topoietic stem cells in the mouse. Blood 1989; 74: 2755–2764.

15. Breems DA, Blokland EAW, Neben S, Ploemacher RE.

Frequency analysis of human primitive haematopoietic stem

cell subsets using a cobblestone area forming cell assay.

Leukemia 1994; 8: 1095–1102.

16. Hao QL, Thiemann FT, Petersen D, et al. Extended long term

culture reveals a highly quiescent and primitive human hema-

topoietic progenitor population. Blood 1996; 88: 3306–3310.

17. Hao QL, Smogorzewska EM, Barsky LW, Crooks GM. In vitro

identification of single CD34+/CD38xcells with both lymphoid

and myeloid potential. Blood 1998; 91: 4145–4152.

18. Miller JS, McCullar V, Punzel M, et al. Single adult human

CD34+/Linx/CD38x progenitors give rise to NK cells, B-

lineage cells, dendritic cells and myeloid cells. Blood 1999; 93:

96–105.

19. Baum CM, Weissman IL, Tsukamoto AS, et al. Isolation of a

candidate human hematopoietic stem cell population. Proc Natl

Acad Sci U S A 1992; 89: 2804–2810.

20. Punzel M, Wissink JS, Miller KA, et al. The myeloid-lymphoid

initiating cell (ML-IC) assay assesses the fate of multipotent

human progenitors in vitro. Blood 1999; 93: 3750–3756.

21. Phillips R. Hematopoietic stem cells: concepts, assays, and

controversies. Semin Immunol 1991; 3: 337–347.

22. Hodgson GS, Bradley TR. Properties of hematopoietic stem

cells surviving 5-fluorouracil treatment. Nature 1979; 281:

381–384.

23. Spangrude GJ, Heimfield S, Weissman IL. Purification and

characterization of murine hematopoietic stem cells. Science

1988; 241: 58–63.

24. Spangrude GJ, Johnson JR. Resting and activated subsets of

mouse multipotent hematopoietic stem cells. Proc Natl Acad Sci

U S A 1990; 87: 7433–7440.

25. Morrison SJ, Weissman IL. The long-term repopulating subset

of hematopoietic stem cells is deterministic and isolatable by

phenotype. Immunity 1994; 1: 661–673.

26. Srour EF, Zanjani ED, Cornetta K, et al. Persistence of human

multilineage, self-renewing lymphohematopoietic stem cells in

chimeric sheep. Blood 1993; 82: 3333–3339.

27. Zanjani ED, Almeida-Porada G, Ascensao JL, et al. Trans-

plantation of hematopoietic stem cells in utero. Stem Cells 1997;

15 (Suppl 1): 79–84.

28. McCune JM. Development and applications of the SCID-hu

mouse model. Semin Immunol 1996; 8: 187–196.

29. Torbett BE, Picchio G, Mosier DE. Hu-PBL-SCID mice: a

model for human immune function, AIDS, and lymphomagen-

esis. Immunol Rev 1991; 124: 139–164.

30. Dick JE. Normal and leukemic human stem cells assayed in

SCID mice. Semin Immunol 196 ; 8: 197–206.

31. Lapidot T, Pflumio F, Doedens M, et al. Cytokine stimulation

of multilineage hematopoiesis from immature human cells

engrafted in SCID mice. Science 1992; 255: 1137–1141.

32. Vormoor J, Lapidot T, Pflumio F, et al. Immature human cord

blood progenitors engraft and proliferate to high levels in

immune-deficient SCID mice. Blood 1994; 83: 2489–2497.

33. Cashman JD, Lapidot T, Wang JCY, et al. Kinetic evidence of

the regeneration of multi-lineage hematopoiesis from primitive

cells in normal human bone marrow transplanted into

immunodeficient mice. Blood 1997; 89: 4307–4316.

34. Pflumio F, Izac B, Katz A, et al. Phenotype and function of

human hematopoietic cells engrafting immune-deficient

CB17–severe combined immunodeficiency mice and nonobese

diabetic–severe combined immunodeficiency mice after trans-

plantation of human cord blood mononuclear cells. Blood 1996;

88: 3731–3740.

35. Hogan CJ, Shpall EJ, McNutty O, et al. Engraftment and

development of human CD34(+)-enriched cells from umbilical

cord blood in NOD/LtSz-scid/scid mice. Blood 1997; 90: 85–96.

36. Rottman GA, Shultz LD, Civin CI. Mature human hemato-

poietic cells in donor bone marrow complicate interpretation of

stem/progenitor cell assays in xenogeneic hematopoietic chi-

meras. Exp Hematol 1998; 26: 332–344.

37. Shultz LD, Schweitzer PA, Christianson SW, et al. Multiple

defects in innate and adaptive immunological function in NOD/

LtSz-scid mice. J Immunol 1995; 154: 180–191.

38. Gan O, Murdoch B, Larochelle A, et al. Differential main-

tenance of primitive human SCID-repopulating cells, clono-

genic progenitors, and long-term culture marrow stromal cells.

Blood 1997; 2: 641–650.

39. Dao MA, Nolta JA. Use of the bnx/hu xenograft model of

human hematopoiesis to optimize methods for retroviral-

mediated stem cell transduction. Int J Mol Med 1998; 1:

257–264.

40. Nolta JA, Dao MA, Wells D, et al. Transduction of pluripotent

human hematopoietic stem cells demonstrated by clonal

analysis after engraftment in immune-deficient mice. Proc Natl

Acad Sci U S A 1996; 93: 2414–2420.

41. Kollet O, Peled A, Byk T, et al. b2 microglobulin-deficient

(B2mmull) NOD/SCID mice are excellent recipients for study-

ing human stem cell function. Blood 2000; 95: 3102–3105.

42. Shultz LD, Lang PA, Christianson SH, et al. NOD/LtSz-

Rag1null mice: an immunodeficient and radioresistant model

for engraftment of human hematolymphoid cells, HIV infec-

tion, and adoptive transfer of NOD mouse diabetogenic T cells.

J Immunol 2000; 164: 2496–2507.

43. Arevalo JMG, Ertl DC, Dao MA, et al. A new immunodefi-

cient mouse strain: the nude NOD/SCID for human hemato-

poietic cell xenotransplantation. Blood 1999; 94 (Suppl 1: 129a.

44. Zanjani ED, Almeida-Porada G, Falke AW. The human/sheep

xenograft model: a large animal model of human hematopoi-

esis. Int J Hematol 1996; 63: 179–192.

45. Zanjani ED, Pallavicini MG, Ascensao JL, et al. Engraftment

and long term expression of human fetal hematopoietic stem

cells in sheep following transplantation in utero. J Clin Invest

1992; 89: 1178–1188.

46. Srour EF, Zanjani ED, Brandt JE, et al. Sustained human

hematopoiesis in sheep transplanted in utero during early gesta-

tion with fractionated adult human bone marrow cells. Blood

1992; 79: 1404–1412.

47. Morris B, Miyasaka M (eds). Immunology of the sheep. Roche:

Basel, 1985.

48. Civin CI, Almeida-Porada G, Lee MJ, et al. Sustained,

retransplantable, multilineage engraftment of highly purified

adult human bone marrow stem cells in vivo. Blood 1996; 88:

4102–4109.

49. Civin CI, Strauss LC, Browall C, et al. Antigenic analysis of

hematopoiesis III. A hematopoietic progenitor cell surface

antigen defined by a monoclonal antibody against KG-1a

cells. J Immunol 1985; 133: 157–164.

50. Andrews RE, Singer JW, Bernstein ID. Precursors of colony-

forming cells in humans can be distinguished from colony-

forming cells by expression of Cd33 and CD34 antigen and light

scatter. J Exp Med 1989; 169: 1721–1731.

51. Krause DS, Fackler MJ, Civin CI, et al. CD34: structure,

Haematopoietic stem cells 437

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 9: Haematopoietic stem cells

biology, and clinical utility [see comments]. Blood 1996; 87:

1–13.

52. Healy L, May G, Gale K, et al. The stem cell antigen CD34

functions as a regulator of hematopoietic cell adhesion. Proc

Natl Acad Sci U S A 1995; 92: 12240–12244.

53. Brown J, Greaves MF, Molgaard HV. The gene encoding the

stem cell antigen, CD34, is conserved in mouse and expressed in

hematopoietic cell lines, brain, and embryonic fibroblasts. Int

Immunol 1991; 3: 75–84.

54. Simmons DL, Satterwaite AB, Tenen DG, Seed BJ. Molecular

cloning of a cDNA encoding CD34, a sialomucin of human

hematopoietic stem cells. J Immunol 1992; 148: 267–271.

55. Berenson RJ, Bensinger WI, Hill RS, et al. Engraftment after

infusion of CD34+ marrow cells in patients with breast cancer

or neuroblastoma. Blood 1991; 77: 1717–1722.

56. Siena S, Bregni M, Brando B, et al. Flow cytometry for clinical

estimation of circulating hematopoietic progenitors for auto-

logous transplantation in cancer patients. Blood 1991; 77:

400–409.

57. Fina L, Molgaard HV, Robertson D, et al. Expression of the

CD34 gene in vascular endothelial cells. Blood 1990; 75:

2417–2426.

58. Baumhueter S, Dybdal N, Kyle C, et al. global expression of

murine CD34, a sialomucin-like endothelial ligand for L-

selectin. Blood 1994; 84: 2554–2565.

59. Greaves MF, Brown J, Molgaard HV, et al. molecular features

of CD34: a hematopoietic progenitor cell-associated molecule.

Leukemia 1992; 6 (Suppl 1): 31–36.

60. Corbeil D, Roper K, Weigmann A, Huttner WB. AC133

hematopoietic stem cell antigen: human homologue of mouse

kidney prominin or distinct member of a novel protein family?

Blood 1998; 91: 2625–2626.

61. Miraglia S, Godfrey W, Yin AH, et al. A novel five-

transmembrane hematopoietic stem cell antigen: isolation,

characterization, and molecular cloning. Blood 1997; 90:

5013–5021.

62. Miraglia S, Godfrey W, Buck D. A response to AC133

hematopoietic stem cell antigen: human homologue of mouse

kidney prominin or distinct member of a novel protein family?

Blood 1998; 91: 4390–4391.

63. Yin AH, Miraglia S, Zanjani EM, et al. AC133, a novel marker

for human hematopoietic stem and progenitor cells. Blood 1997;

90: 5002–5012.

64. Wuchter C. Impact of CD133 (AC133) and CD90 expression

analysis for acute leukemia immunophenotyping. Haematolo-

gica 2001; 86: 154–161.

65. Majka M, Ratajack J, Machalinski B, et al. Expression and

function of AC133, a putative cell surface marker of primitive

human haematopoietic cells. Folia Histochem Cytobiol 2000; 38:

53–63.

66. Bhatia M. AC133 expression in human stem cells. Leukemia

2001; 15: 1685–1688.

67. Ziegler BL, Valtieri M, Almeida-Porada G, et al. KDR

receptor: a key marker defining hematopoietic stem cells.

Science 1999; 285: 553–558.

68. Craig W, Kay R, Cutler RB, Lansdorp PM. Expression of

Thy-1 on human hematopoietic progenitor cells. J Exp Med

1993; 177: 1331–1342.

69. Bhatia M, Wang JCY, Kapp U, et al. Pruification of primitive

human hematopoietic cells capable of repopulating immune-

deficient mice. Proc Natl Acad Sci U S A 1997; 94: 5320–5325.

70. Thomas TE, Miller CL, Eaves CJ. Purification of hematopoietic

stem cells for further biological study. Methods 1999; 17:

202–218.

71. Goodell MA, Brose K, Paradis G, et al. Isolation and

functional properties of murine hematopoietic stem cells that

are replicating in vivo. Exp Med 1996; 183: 1797–1806.

72. Yusa K, Tsuruo T. Reversal mechanism of multidrug resistance

by verapamil: direct binding of verapamil to P-glycoprotein on

specific sites and transport of verapamil outward across the

plasma membrane of K562/ADM cells. Cancer Res 1989; 49:

5002–5006.

73. Miller CL, Eaves CJ. Expansion in vitro of adult murine

hematopoietic stem cells with transplantable lympho-myeloid

reconstitution. Proc Natl Acad Sci U S A 1997; 94: 13648–

13653.

74. Goodell MA, Rosenzweig M, Kim H, et al. Dye efflux studies

suggest that hematopoietic stem cells expressing low or

undetectable levels of CD34 antigen exist in multiple species.

Nature Med 1997; 3: 1337–1345.

75. Uchida N, Fujisaki T, Eaves AC, Eaves CJ. Transplantable

hematopoietic stem cells in human fetal liver have a CD34+

side population (SP) phenotype. J Clin Invest 2001; 108:

1071–1077.

76. Zhou S, Schuetz JD, Bunting KD, et al. The ABC transporter

Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is

a molecular determinant of the side-population phenotype.

Nature Med 2001; 7: 1028–1034.

77. Zanjani ED, Almeida-Porada G, Livingston AG, et al. Human

bone marrow CD34x cells engraft in vivo and undergo multi-

lineage expression that includes giving rise to CD34+ cells. Exp

Hematol 1998; 26: 353–360.

78. Bhatia M, Bonnet D, Murdoch B, et al. Identification of a

newly discovered class of human hematopoietic cells with

SCID-repopulating activity. Nature Med 1998; 4: 1038–1044.

79. Osawa M, Hanada K, Hamada H, et al. Long-term lymphohe-

matopoietic reconstitution by a single CD34-low/negative

hematopoietic stem cell. Science 1996; 273: 242–245.

80. Bonnet D. Normal and leukemic CD34-negative human

hematopoietic stem cells. Rev Clin Exp Hematol 2001; 5: 42–61.

81. Roberts R, Gallagher J, Spooncer EE, et al. Heparan sulfate

bound growth factors: a mechanism for stromal cell mediated

hematopoiesis. Nature 1988; 332: 376–378.

82. Metcalf D. Hemopoietic regulators: redundancy or subtely?

Blood 1993; 82: 3515–3523.

83. Van Zant G, Goldwasser E. Competition between erythro-

poietin and colony-stimulating factor for target cells in mouse

marrow. Blood 1979; 53: 946–965.

84. Trentin JJ. Influence of hematopoietic organ stroma (hemato-

poietic inductive microenvironment) on stem cell differentia-

tion. In Regulation of Hematopoieisis, Gordon AS (ed).

Appleton-Century-Crofts: New York, 1970: 161–185.

85. Ogawa M. The stochastic model revisited. Int J Hematol 1999;

69: 2–5.

86. Ogawa M. Differentiation and proliferation of hematopoietic

stem cells. Blood 1993; 81: 2844–2853.

87. Just U, Friel J, Heberlein C, et al. Upregulation of lineage

specific receptors and ligands in multipotential progenitor cells

is part of an endogenous program of differentiation. Growth

Factors 1993; 9: 291–300.

88. Tenen DG, Hromas R, Licht JD, et al. Transcription factors,

normal myeloid development, and leukemia. Blood 1997; 90:

489–519.

89. Taichman RS, Emerson SG. The role of osteoblasts in the

hematopoietic microenvironment. Stem Cells 1998; 16: 7–15.

90. Potonik AJ, Brakebusch C, Fassler R. Fetal and adult

hematopoietic stem cells require beta 1 integrin function for

colonizing fetal liver, spleen and bone marrow. Immunity 2000;

12: 653–663.

91. Rafii S, Mohle R, Shapiro F, et al. Regulation of hematopoiei-

sis by microvascular endothelium. Leuk Lymphoma 1997; 27:

357–386.

92. Torok-Storb B, Iwata M, Graf L, et al. Dissecting the marrow

microenvironment. Ann N Y Acad Sci 1999; 872: 164–170.

93. Cross MA, Enver T. The lineage commitment of hematopoietic

progenitor. Curr Opin Genet Dev 1997; 7: 609–613.

94. Orkin SH. Transcription factors and hematopoietic develop-

ment. J Biol Chem 1995; 270: 4955–4958.

95. Shivdasani RA, Orkin SH. The transcription control of

hematopoiesis. Blood 1996; 87: 4025–4029.

96. Rabbitts TH. Chromosomal translocations in human cancer.

Nature 1994; 372: 143–145.

97. Robb L, Lyons I, Li R, et al. Absence of yolk sac hematopoiesis

438 D. Bonnet

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 10: Haematopoietic stem cells

from mice with a targeted disruption of the scl gene. Proc Natl

Acad Sci U S A 1995; 92: 7075–7079.

98. Look TA. Oncogenic transcription factors in human acute

leukemias. Science 1996; 278: 1059–1064.

99. Moreau-Gachelin L, Tavitian A, Tambourin P, et al. Spi-1 is a

putative oncogene in virally induced murine erythroleukemias.

Nature 1988; 331: 277–279.

100. Miwa H, Beran M, Saunders GF. Expression of the Wilm’s

tumor (WT) gene in human leukemia. Leukemia 1992; 6:

405–408.

101. Bacchetta R, de Waal Malefgk R, Yssel H, et al. Host-reactive

CD4+ and CD8+ T cell clones isolated from a human chimera

produce IL-5, IL-2, IFN-gamma and granulocyte/macrophage-

colony-stimulating factor by not IL-4. J Immunol 1990; 144:

902–908.

102. Scott MP. Vertebrate homeobox gene nomenclature. Cell 1992;

71: 551–553.

103. Lawrence HJ, Sauvageau G, Humphries RK, Largman C.

Stage- and lineage-specific expression of the HOXA10 homeo-

box gene in normal and leukemic hematopoietic cells. Exp

Hematol 1995; 23: 1160–1166.

104. Sauvageau G, Lansdorp PM, Eaves CJ, et al. Differential

expression of homeobox genes in functionally distinct CD34+subpopulations of human bone marrow cells. Proc Natl Acad

Sci U S A 1994; 91: 12223–12227.

105. Sauvageau G, Thorsteinsdottir U, Eaves CJ, et al. Over-

expression of HOXB4 in hematopoietic cells causes the selective

expansion of more primitive populations in vitro and in vivo.

Genes Dev 1995; 9: 1753–1765.

106. Thorsteindottir U, Sauvageau G, Humphries RK. Enhanced

in vivo regenerative potential of Hox B4-transduced hemato-

poietic stem cells with regulation of their pool size. Blood 1999;

94: 2605–2612.

107. Rekhtman N, Radparvar F, Evans T, et al. Direct interaction

of hematopoietic transcription factors PU.1 and GATA.1

functional antagonism in erythroid cells. Genes Dev 1999; 13:

1398–1411.

108. Nerlov C, Querfurth E, Kulessa H, et al. GATA-1 interacts

with the myeloid PU-1 transcription factor and represses PU-1

dependent transcription. Blood 2000; 95: 2543–2551.

109. Zhang P, Zhang X, Iwama A, et al. PU.1 inhibits GATA-1

function and erythroid differentiation by blocking GATA-1

DNA binding. Blood 2000; 96: 2641–2648.

110. Cantor AB, Orkin SH. Hematopoietic development: a balanc-

ing act. Curr Opin Genet Dev 2001; 11: 513–519.

111. Artavanis-Tsakonas S, Matsuno K, Fortini M. Notch signal-

ling. Science 1995; 268: 225–232.

112. Karanu FN, Murdoch B, Gallacher L, et al. The Notch ligand

jagged-1 represents a novel growth factor of human hemato-

poietic stem cells. J Exp Med 2000; 192: 1365–1372.

113. Varnum-Finney B, Xu L, Brashem-Stein C, et al. Pluripotent,

cytokine-dependent hematopoietic stem cells are immortalized

by constitutive Notch 1 signalling. Nature Med 2000; 6: 1278–

1281.

114. Attisano L, Wrana JL, Lopez-Casillas F, Massague J. TGF-

beta receptors and actions. Biochim Biophys Acta 1994; 1222:

71–80.

115. Massague J. TGFbeta signaling: receptors, transducers, and

Mad proteins. Cell 1996; 85: 947–950.

116. Eaves AC, Eaves CJ. Maintenance and proliferation control of

primitive hemopoietic progenitors in long-term cultures of

human marrow cells. Blood Cells 1988; 14: 355–368.

117. Hatzfeld J, Li ML, Brown EL, et al. Release of early human

hematopoietic progenitors from quiescence by antisense trans-

forming growth factor beta 1 or Rb oligonucleotides. J Exp

Med 1991; 174: 925–929.

118. Maeno M, Mead PE, Kelley C, et al. The role of BMP-4 and

GATA-2 in the induction and differentiation of hematopoietic

mesoderm in Xenopus laevis. Blood 1996; 88: 1965–1972.

119. Bhatia M, Bonnet D, Wu D, et al. Bone morphogenetic

proteins regulate the developmental program of human hema-

topoietic stem cells. J Exp Med 1999; 189: 1139–1147.

120. Allouche M. Basic fibroblast growth factor and hematopoiesis.

Leukemia 1995; 9: 937–942.

121. Phillips RL, Ernst RE, Brunk B, et al. The genetic program of

hematopoietic stem cells. Science 2000; 288: 1635–1640.

122. Zhou G, Chen J, Lee S, Clark T, Rowley J, Wang SM. The

pattern of gene expression in human CD34+ Stem/progenitor

cells. Proc Natl Acad Sci U S A 2001; 98: 13966–13971.

123. Hu M, Krause D, Greaves M, et al. Multilineage gene

expression levels during the differentiation of individual pri-

mary hematopoietic cells. Genes Dev 1997; 11: 774–785.

124. Cheng T, Shen H, Giokas D, Gere J, Tene DG, Scadden DT.

Temporal mapping of gene expression levels during the differ-

entiation of individual primary hematopoietic cells. Proc Natl

Acad Sci U S A 1996; 93: 13158–13163.

125. Enver T, Greaves M. Loops, Lineage and leukaemia. Cell 1998;

94: 9–12.

126. Papayannopoulos T, Nakamoto B. Peripheralization of hema-

topoietic progenitors in primates treated with anti-VLA4 inte-

grin. Proc Natl Acad Sci U S A 1993; 90: 9374–9378.

127. Prosper F, Stroncek D, McCarthy JB, Verfaillie CM. Mobiliza-

tion and homing of peripheral blood is related to reversible

downregulation of alpha4-betal integrin and function. J Clin

Invest 1998; 101: 2456–2467.

128. Zanjani ED, Flake AW, Almeida-Porada G, Tran N, Papayan-

nopoulou T. Homing of human cells in the fetal sheep model:

modulation by antibodies activating or inhibition very late

activation antigen 4 dependent function. Blood 1999; 94:

2515–2522.

129. Peled A, Petit I, Kollet O, et al. Dependence of human stem cell

engraftment and repopulation of NOD/SCID mice on CXCR4.

Science 1999; 283: 845–848.

130. Rosu-Myles, Gallacher L, Murdoch B, et al. The human

hematopoietic stem cell compartment is heterogeneous for

CXCR4 expression. Proc Natl Acad Sci U S A 2000; 97:

14626–14631.

131. Srour EF, Jetmore A, Wolber FM, et al. Homing, cell cycle

kinetics and fate of transplanted hematopoietic stem cells.

Leukemia 2001; 15: 1681–1684.

132. Whetton AD, Graham GJ. Homing mobilization in the stem

cell niche. Trends Cell Biol 1999; 9: 233–238.

133. Nilsson SK, Dooner MS, Tianks CY, Weier HU, Quesenberry

PJ. Potential and distribution of transplanted hematopoietic

stem cells in a non-ablated mouse model. Blood 1997; 89:

4013–4020.

134. Nilsson SK, Johnston HM, Coverdale JA. Spatial localization

of transplanted hematopoietic stem cells: interferences for the

localization of stem cell niches. Blood 2001; 97: 2293–2299.

135. Ferrari G, Cusella-DeAngelis G, Coletta M, et al. Muscle

regeneration of bone marrow derived myogenic progenitors.

Science 1998; 279: 1528–1530.

136. Gussoni E, Soneaoka Y, Stickland C, et al. Dystrophin

expression in the mdx mouse restored by stem cell transplanta-

tion. Nature 1999; 401: 390–394.

137. Petersen B, Bowen W, Pattiene L, et al. Bone marrow as a poten-

tial source of hepatic oval cells. Science 1999; 284: 1168–1170.

138. Theise ND, Badve S, Saxena R, et al. Derivation of hepatocyte

from bone marrow cells in mice after radiation-induced

myeloablation. Hepatocyte 2000; 31: 235–240.

139. Woodbury D, Schwarz EJ, Prockop D, Black IR. Adult rat and

marrow stromal cells differentiate into neurons. J Neurosci Res

2000; 61: 364–370.

140. Jackson K, Mi T, Goodell M. Hematopoietic potential of stem

cells isolated from murine skeletal muscle. Proc Natl Acad Sci

U S A 1999; 96: 14482–14486.

141. Bjornson CR, Rietze RL, Reynolds BA, et al. Turning brain

into blood: a hematopoietic fate adopted by adult neural stem

cells in vivo. Science 1999; 283: 534–538.

142. Eglitis MA, Mezey E. Hematopoietic cells differentiate into

both microglia and macroglia in the brains of adult mice. Proc

Natl Acad Sci U S A 1997; 94: 4080–4085.

143. Lagasse E, Connors H, Al-Dhalimy M, et al. Purified hema-

Haematopoietic stem cells 439

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.

Page 11: Haematopoietic stem cells

topoietic stem cells can differentiate to hepatocytes in vivo.

Nature Med 2001; 6: 1229–1234.

144. Theise ND, Nimmakayalu M, Gardner R, et al. Liver from

marrow in humans. Hepatocyte 2000; 32: 11–16.

145. Alison MR, Poulsom R, Jeffery R, et al. Hepatocytes from non-

hepatic adult stem cells. Nature 2000; 406.

146. Kopen GC, Prockop DJ, Phinney DG. Marrow stromal cells

migrate throughout forebrain and cerebellum, aand they differ-

entiate into astrocytes after injection into neonatal mouse

brains. Proc Natl Acad Sci U S A 2000; 96: 10711–10716.

147. Reyes M, Lund T, Lenvik T, Agular D, Koodie L, Verfaillie C.

Purification and ex vivo expansion of postnatal human marrow

mesodermal progenitor cells. Blood 2001; 98: 2615–2625.

148. Reyes M, Dudek A, Jahagindar B, Koodie L, Marker PH,

Verfaillie CM. Origin of endothelial progenitors in human

postnatal bone marrow. J Clin Invest 2002; 109: 337–346.

149. Krause DS, Theise ND, Collector MI, et al. Multi-Organ,

multi-lineage engraftment by a single bone marrow-derived

stem cell. Cell 2001; 105: 369–377.

440 D. Bonnet

Copyright # 2002 John Wiley & Sons, Ltd. J Pathol 2002; 197: 430–440.