Assay systems for hematopoietic stem and progenitor cells

11
Assay Systems for Hematopoietic Stem and Progenitor Cells THOMAS A. BOCK Department of Hematology and Oncology, University of Tiibingen, Tiibingen, Germany Key Words. In vitro assays . In vivo assays . Hemntopoietic stem cells Henzatopoietic progenitor cells Long-tern7 culture ' Immunodeficient mice Fetal sheep ABSTRACT Progress in our understanding of the hematopoietic system as well as novel cellular and molecular biology techniques are increasing- ly promoting the ex vivo manipulation and ther- apeutic use of hematopoietic stem and progenitor cells. For both, development of stem cell therapies and basic hematopoietic research, test systems for hematopoietic stem cells are required to mon- itor the intrinsic and ex vivo-induced properties of these cells. In vitro assays for primitive hematopoietic cells (colony-forming units-blast, cobblestone area-forming cells, long-term culture-initiating cells [LTC-IC]) have been established which demonstrate the proliferative and differentiation capacities of these populations. The potentials of these assays have been recently enhanced by the extended LTC and the switch LTC modifca- tions. Although some hematopoietic cells charac- terized in vitro have the multipotential and proliferative properties of pluripotent hematopoi- etic stem cells (PHSC), their capacity to long-term repopulate hematopoiesis in vivo, a hallmark of PHSC, has not been established. Without this confirmation, populations defined in vitro should not be considered the equivalent of PHSC. In animals, the properties of primitive hematopoietic cells can be systematically ana- lyzed by multiple in vivo assays. Therefore, var- ious strategies have been pursued to develop an animal model for human hematopoiesis. In fetal sheep and immunodeficient mice, the functions of human PHSC are reproduced, and long-term multilineage repopulation capacity and exten- sive proliferative potential have been demon- strated for xenografted human cells. Thus, both models can be considered stem cell assays and may significantly enhance the study of early hematopoiesis and the development of therapeu- tic strategies. Stem Cells 1997:15(supp1 1):185-195 Recent studies of early hematopoiesis have expanded our knowledge of the phenotypic proper- ties, biology and regulation of hematopoietic stem cells. Phenotypic characterization of the most primitive pluripotent hematopoietic stem cells (PHSC), however, is still not discriminative enough to purify these cells to homogeneity. One reason for this is that surface markers on early hematopoi- etic stem cells have not yet been completely identified. Also, antigen expression, as well as other properties, are not specific for a singular hematopoietic cell type. There is considerable overlap in phenotype for cell morphology and surface markers between different cell types. Therefore, only a combination of multiple markers and characteristics could unequivocally define a PHSC, if at all. Furthermore, different stem cell phenotypes probably exist that are related to different functional states of these cells. As a consequence, even a pure population of stem cells may contain different phenotypes. Hematopoietic Stem Cells. STEM CELLS 1997;15(suppl 1):185-195 0 1997 AlphaMed Press. All rights reserved.

Transcript of Assay systems for hematopoietic stem and progenitor cells

Assay Systems for Hematopoietic Stem and Progenitor Cells

THOMAS A. BOCK

Department of Hematology and Oncology, University of Tiibingen, Tiibingen, Germany

Key Words. In vitro assays . I n vivo assays . Hemntopoietic stem cells Henzatopoietic progenitor cells Long-tern7 culture ' Immunodeficient mice Fetal sheep

ABSTRACT Progress in our understanding of the

hematopoietic system as well as novel cellular and molecular biology techniques are increasing- ly promoting the ex vivo manipulation and ther- apeutic use of hematopoietic stem and progenitor cells. For both, development of stem cell therapies and basic hematopoietic research, test systems for hematopoietic stem cells are required to mon- itor the intrinsic and ex vivo-induced properties of these cells.

In vitro assays for primitive hematopoietic cells (colony-forming units-blast, cobblestone area-forming cells, long-term culture-initiating cells [LTC-IC]) have been established which demonstrate the proliferative and differentiation capacities of these populations. The potentials of these assays have been recently enhanced by the extended LTC and the switch LTC modifca- tions. Although some hematopoietic cells charac- terized in vitro have the multipotential and

proliferative properties of pluripotent hematopoi- etic stem cells (PHSC), their capacity to long-term repopulate hematopoiesis in vivo, a hallmark of PHSC, has not been established. Without this confirmation, populations defined in vitro should not be considered the equivalent of PHSC.

In animals, the properties of primitive hematopoietic cells can be systematically ana- lyzed by multiple in vivo assays. Therefore, var- ious strategies have been pursued to develop an animal model for human hematopoiesis. In fetal sheep and immunodeficient mice, the functions of human PHSC are reproduced, and long-term multilineage repopulation capacity and exten- sive proliferative potential have been demon- strated for xenografted human cells. Thus, both models can be considered stem cell assays and may significantly enhance the study of early hematopoiesis and the development of therapeu- tic strategies. Stem Cells 1997:15(supp1 1):185-195

Recent studies of early hematopoiesis have expanded our knowledge of the phenotypic proper- ties, biology and regulation of hematopoietic stem cells. Phenotypic characterization of the most primitive pluripotent hematopoietic stem cells (PHSC), however, is still not discriminative enough to purify these cells to homogeneity. One reason for this is that surface markers on early hematopoi- etic stem cells have not yet been completely identified. Also, antigen expression, as well as other properties, are not specific for a singular hematopoietic cell type. There is considerable overlap in phenotype for cell morphology and surface markers between different cell types. Therefore, only a combination of multiple markers and characteristics could unequivocally define a PHSC, if at all. Furthermore, different stem cell phenotypes probably exist that are related to different functional states of these cells. As a consequence, even a pure population of stem cells may contain different phenotypes.

Hematopoietic Stem Cells. STEM CELLS 1997;15(suppl 1):185-195 0 1997 AlphaMed Press. All rights reserved.

186 Stem Cell Assays

Because of these current limitations in identifying a PHSC by its phenotype alone, functional in vivo and in vitro assays have been developed to indirectly demonstrate the presence of primitive hematopoiet- ic cells in a heterogenous test population by determining the biologic functions of these cells. PHSC have been defined as cells with the biological properties of self-renewal and long-term repopulation of all lym- phohematopoietic lineages in a transplant recipient. Therefore, each assay attempts to define conditions under which early hematopoietic cells can exhibit those biological capacities. If those have been suffi- ciently demonstrated, then the elusive cell exhibiting these capacities can be considered the equivalent of a PHSC. Because of the indirect nature of these approaches, demonstration of presence of a specific cell type does not imply, however, that this cell can be identified as an individual cell of the input population, unless the test population contains only one single cell.

Multilineage differentiation is only indicative of PHSC when originating from one single ancestral cell as opposed to several single lineage-committed progenitors. Confirmation of clonal hematopoiesis either requires single cell studies or analyses of genetic markers within different lineages [l , 21. These analyses are elaborate and have not always been included in studies of stem cells. Instead, it has been assumed that committed progenitors are only short-lived because of their limited proliferative potential and because they die in differentiation. Thus, the presence of stem cells has been deduced from the demonstration of ongoing growth after several weeks.

It has been widely debated whether self-renewal, defined as the potential of giving rise to identical daugh- ter cells, actually exists. Restrictions in serial transfer potential and telomer shortening during differentiation and ontogeny of hematopoietic cells both suggest that differences in proliferative potential within stem cell compartments exist [3,4]. However, if all daughter cells must differ in some respect from their parents, then the stem cell compartment would be extremely heterogeneous and hierarchical. The experimental demonstra- tion of extensive proliferative potential includes the in vitro replating potential or the in vivo serial transfer capacity of hematopoietic cells, but this experimental evidence is not sufficient to settle this issue.

According to the above definition of a PHSC, lifelong monitoring of multilineage hematopoietic repopulation in a transplanted animal or person constitutes the golden standard for PHSC assays. Consequently, results from alternative assays obviously must comply with in vivo repopulation studies. However, demonstration of complete and stable hematopoietic reconstitution requires a period of months or years. To facilitate the study of early hematopoietic cells, many strategies have been developed for assaying these cells in vivo and in vitro over shorter time periods. However, none of these assays fully satisfies the criteria for PHSC analysis [5 , 61. Rather, it appears that the cells studied in these assays do not achieve the same degree of self-renewal and range of differentiation as they do in vivo.

Other obstacles include the technical and quantitative limitations of stem and progenitor cell assays. The poorer our knowledge of PHSCs, the larger and more heterogeneous the input population must be chosen and the more unwanted nonstem cells are contained. High numbers of contaminating cells, however, may obscure the effects of the stem cells. Also, PHSC could potentially contaminate the selected test cells, read out in the assay and cause a false positive result.

On the other hand, the greater our knowledge a priori, the earlier hematopoietic cells can be enriched. As a consequence, very few cells are contained in the test samples and techniques may not be sensitive enough to assay such a small number of cells. There is a high risk of losing these cells during processing and transfer resulting in false negative results. This particularly concerns in vivo assays, which have rela- tively low seeding efficiencies for transplanted cells (-20%). In addition, highly enriched test cells are deprived of accessory cells which may be necessary for their proper functions. Therefore, accessory cells or their signals must be complemented.

In summary, in order to assess the validity of a PHSC assay, it must be tested whether the assay para- meters provide sufficient evidence for clonal multilineage differentiation, extensive proliferative potential and repopulation ability. In addition, technical issues have to be addressed due to the multiplicity of existing procedures and methods which are presented below.

Bock 187

DIRECT CLONOGENIC ASSAYS Clonogenic assays are the most direct quantitative means of measuring hematopoietic cells by colony

formation in vitro. Hematopoietic colonies are clones of cells produced by a single progenitor cell. Hence, the number of colony-forming progenitors is directly related to the number of colonies. By replating the cells into different clonogenic systems, information regarding self-renewal and the differentiation capacities of the colony-forming cell (CFC) is generated.

Blast Colony-Forming Cell Assay (CFC-Blast) Ogawa has described a clonogenic assay for CFC-blast which detects a 5-fluorouracil (5-FU)-resistant

progenitor with secondary recloning potential and the capacity to generate single lineage and multilineage- committed progeny [7-121. The assay is defined for steady-state conditions and is based on the premise that the earliest hematopoietic cells are in Go. Bone marrow cells are plated in the presence of fetal calf serum, without complementary cytokines, under the assumptions that early cells are kept in Go and actively cycling progenitors are not supported. Colonies produced by CFC-blast consist of 50-500 immature hematopoietic cells. Their growth is delayed relative to the formation of granulocyte-macrophage colonies and erythroid bursts. However, the appeareance of colonies could be hastened by adding cytokines. Colonies produced by CFC-blast contained new CFC-blast which were detected by replating cells from the colonies under condi- tions that are identical to the primary culture. This reproduction capacity allows expansion of CFC-blast to relatively high numbers, but impairs reliable quantification of these cells. Alternative conditions for lineage commitment and differentiation can be used in the secondary culture in order to determine the functional spectrum of CFC-blast.

High Proliferative Potential-Colony-Forming Cells (HPP-CFC) HPP-CFC are characterized by their ability to generate very large colonies containing greater than

50,000 cells at 10-12 days of culture [13, 141. The frequency of HPP-CFC in normal marrow was report- ed at about 2 per lo5 mononuclear cells. HPP-CFC are resistant to 5-FU toxicity. They are made up of several subpopulations, suggesting a hierarchical organization within this progenitor cell compartment [ 14, 151. A portion of replated HPP-CFC-derived colonies contained further HPP-CFC, suggesting some self-renewal potential of HPP-CFC [ 161. The most primitive subsets have been described as being close- ly related with long-term reconstituting cells [13, 171. Murine, as well as human HPP-CFC, produced colonies which contained progenitors of granulocytes, erythrocytes and megakaryocytes [ 181.

Cobblestone Area-Forming Cell (CAFC) Assay When murine marrow is added to preformed stromal layers, colonies of adherent hematopoietic cells

(cobblestone areas) can he observed [ 191. Following up this observation in the human system, Gordon et al. [20] described a human cell type that adhered to stroma and produced colonies of undifferentiated blast cells. Replating of these colonies showed limited self-renewal and the presence of CFU-granulo- cyte-erythroid-macrophage-megakaryocyte (GEMM), CFU-GM and BFU-E, suggesting that CAFC are ancestral to CFU-GEMM [2 11. In the murine system, cells generating early-appearing cobblestone areas (days 7-10) appeared to coenrich with CFU-spleen and committed progenitors. On the other hand, cells forming late-appearing cobblestone areas (day 28) coenriched with cells providing short-term hematopoietic repopulation [22,23]. Although CAFC determined at weeks 5-8 correlated closely with in vivo repopulating ability [23], there is not sufficient evidence to support that CAFC represent PHSC [24].

In order to simplify the initial culture setup and eliminate inhomogeneities in the stromal feeder layer, fresh marrow-derived stromal layers were substituted with clonal cell lines. A murine bone mar- row stromal cell line GB1/6 [25] and a preadipose cell line FBMD-l [26] were both found to support the growth of overlaid fresh marrow with the same efficiency as fresh stromal layers. For quantification of CAFC, Ploemacher described a long-term culture (LTC) system using microtiter wells. Cells are overlaid

188 Stem Cell Assays

at a limiting dilution on previously established irradiated stromal layers. The percentage of wells with at least one hematopoietic cobblestone area is then determined over a period of approximately 40 days, and CAFC frequencies can be calculated using Poisson statistics [23].

Frequency of CAFC in steady-state human marrow was determined at about 45 (week 5) and 15 (week 8) per lo5 Ficolled cells [26]. Murine CAFC frequency declined by over one log between days 7 and 28 with values of 2-40 per lo5 cells at day 28 [23, 251. Murine CAFC have been described as wheat germ agglutinin (WGA)d'm [27], and the phenotype of human CAFC was reported as CD34', HLA-DR-, Rh123dU" [26], 1% and Thy-1' [28].

SECONDARY CLONOGENIC ASSAYS This group includes assays for progenitor cells that do not themselves form colonies in semisolid

media but instead can produce clonogenic progeny. Test populations are cultured for several weeks and then analyzed for the number of clonogenic cells present.

LTC Assays Stromal cell-associated hematopoiesis has been extensively studied by Dexter and others as a means for

reproducing the in vivo environment for stem and progenitor cells [29-3 11. Several groups have established various LTC assays which detect an LTC-initiating cell (LTC-IC) [32-381. After five to eight weeks, prima- ry stroma-supported cultures are scored for replatable colony-forming cells (CFC) that are released into the culture supernatant and assayed in semisolid culture systems. LTC can be set up either by inoculating tissue culture flasks with bone marrow, or alternatively, in two stages by growing the stromal layer to confluence and then adding hematopoietic cell suspensions. As in the CAFC assay, stromal cell lines have been employed to replace primary human marrow adherent feeder layers [39-411.

Since the LTC system is a qualitative method, frequency analysis of progenitor subsets is not feasi- ble unless carried out as a limiting dilution approach. Eaves et al. [38] demonstrated a linear relationship between the number of cells used to seed a preformed stroma and the numbers of CFU-GM released at five weeks. However, individual LTC-IC produced variable numbers (1-30) of clones, and the number of LTC-IC continously declined with only a fraction persisting after five weeks [42]. Therefore, the absolute numbers of LTC-IC in different hematopoietic tissues cannot be compared unless the rate of decline is known for the test sample and corrected accordingly. Furthermore, long-term repopulating ability was lost in LTC of murine bone marrow 1431. Frequency of LTC-IC at weeks 5 and 8 was deter- mined to be about 40 and 15, respectively, per 10' Percolled bone marrow cells, and represented about one percent of the CD34' cell population [36, 401. In peripheral blood, LTC-IC concentration was two logs lower and amounted to six cells per ml [44].

LTC-IC and CAFC share a series of functional and phenotypic properties. A considerable portion of both were resistant to cytotoxic drugs [45]. Both were preferentially, but not exclusively, contained in the CD34' Rhl 23dU" fraction of Ficolled bone marrow cells, and had a Thy-1' and HLA-DR'"" phenotype [40, 46-48]. LTC-IC were distributed between the CD38' and CD38- fractions and appeared to express low levels of c-kit [48, 491. Another study suggested that the c-kit' fraction contains the most primitive progenitor cells in human marrow [SO].

Crooks and colleagues extended the period of standard long-term stromal cocultures (supplemented with interleukin 3 [IL-31, IL-6 and stem cell factor [SCF]) to over 60 days [49, 511. They observed two peaks of CFC production, the first during days 35-60 and the second during days 60-100. Extended (E)LTC-IC were exclusively contained in the CD34'CD38- fraction and arose almost exclusively from late proliferating cells which formed visible clones after day 30. In contrast, standard LTC-IC arose from early proliferating cells between days 14-28 and were distributed between the CD38' and CD38- frac- tions. Clones that appeared after 60 days from single late proliferating cells contained tenfold more cells than from early proliferating cells. Whereas retroviral gene marking of LTC-IC showed efficient gene

Bock 189

marking, ELTC-IC were not marked with vector. These studies suggest that ELTC-IC are a primitive progenitor population functionally distinct from LTC-IC.

Recently, a different modification of the LTC assay has been described by the Vancouver group [52,53]. It allows the identification and quantitation of a subset of murine LTC-IC (designated as LTC-ICML) with the capacity to generate both myeloid and B lymphoid progeny in vitro. Test cells are cultured at limiting dilu- tions on irradiated mouse marrow feeder layers for an initial four weeks under conditions supporting myelopoiesis and then for an additional week under conditions permissive for B lymphopoiesis. Clonogenic pre-B progenitors detected in such postswitch LTC appeared to be derived from uncommitted cells present in the original cell suspension. However, clonal myelolymphoid hematopoiesis was not analyzed. LTC-ICML were 5-F'U-resistant and coenriched 500-fold with LTC-IC in the SCA-l', lin-, WGA' fraction [52]. Their frequency was determined at approximately 1 per 5 x lo5 normal adult mouse bone marrow cells, which rep- resented a 15-30-fold lower level than standard LTC-IC or cells with long-term in vivo myelolymphoid repop- ulation potential. Both standard LTC-IC and LTC-ICML were significantly better maintained in vitro than repopulating cells [53], suggesting differences in detectability or the developmental stages of these cells types.

Delta (A)-Assay A group of assay systems has become known as A-assays because they measure the production of

multiple clonogenic progeny (base of the A) by a single progenitor (apex of the A symbol). Delta assays permit analyses of recruitment, proliferation potential and lineage potentials of early hematopoietic cells. Bone marrow depleted of committed progenitors and enriched for primitive hematopoietic cells is grown in a seven-day suspension culture in the presence of various cytokine combinations. The effect of these cytokines on early progenitor cells is measured in a secondary clonogenic assay for committed progenitors [54, 551. Other variants of A assays utilize the properties of early hematopoietic cells to adhere to stroma [24] or plastic [56,57]. Cells producing CFU-GM in the latter assay modification have been described as 5-FU-resistant and as expressing CD34 and Thy-1 [56,58]. The cells detected by the different various A assays are heterogeneous but probably related and overlap with the LTC-IC [59].

IN VIVO XENOTRANSPLANTATION ASSAYS In vitro assays have greatly expanded our knowledge of the phenotype of primitive human progenitor cells

and their lineage relationships. Although some hematopoietic cells characterized in vitro have the multipoten- tial and proliferative properties of PHSC, their capacity for long-term hematopoietic repopulation has not yet been established. Until this capacity has been confirmed, the populations defined by in vitro assays cannot be considered the equivalent of long-term repopulating PHSC. In animals, the properties of the most primitive hematopoietic cells can be systematically analyzed by multiple in vivo assays [ S , 60-631 which are not feasi- ble in humans for ethical reasons. Consequently, various strategies have been pursued to develop an animal model for human hematopoiesis. Ideally, this model would provide the critical microenvironment for the engraftment of human hematopoietic cells without mediating rejection or permitting graft-versus-host-disease. Growth and differentiation of human cells would be supported and reflected by a physiological distribution of primitive and differentiated human cells in the host.

In the past few years, novel chimeric animal models have been developed that may significantly enhance our ability to analyze the functions of human PHSC in vivo, but outside of the human body. These models include xenotransplantation of human hematopoietic cells in immunodeficient mouse strains and preimmune fetal sheep. Recent results from these chimeric systems suggest that these animals provide a permissive environment for the study of human stem cells in vivo.

Fetal Sheep In sheep, long-term engraftment of human hematopoietic cells that have been injected into the peri-

toneal cavity of preimmune fetuses has been achieved [64-661. In utero transplanted cells from human

190 Stem Cell Assays

fetal liver, adult bone marrow and adult peripheral blood reconstituted myelolymphoid hematopoiesis, including T lymphocytes. Human cells could be monitored in all hematopoietic tissues and the peripher- al blood for at least several months and up to several years [65-671. Engraftment was successful with as few as several hundred progenitor-enriched human bone marrow cells, and was restricted to the c-kirl"" as opposed to the c-kit' or c-kit- fractions [68]. Isolated CD45' human cells from the marrow of adult sheep, previously injected in utero with human fetal liver cells, could be transferred and engrafted to sec- ondary sheep recipients [66]. However, the level of engraftment of human cells was still low in the pri- mary sheep (3%-4%), and only two of six secondary recipients supported continued growth of human cells. Since myeloablative regimens are not feasible in pregnant sheep because of teratogenic hazards, xenografted donor cells are in competition with the endogenous pool of stem cells. Early engraftment of a certain percentage of donor cells occurs at a low proportion and stability cannot be fully achieved in the course of gestation. However, the engraftment level could be enhanced by ex vivo incubation of fetal liver cells with hematopoietic growth factors [67].

In order to establish a short-term assay for primitive hematopoietic cells, Zunjuni and coworkers transplanted human cells into the ovine fetus, sacrificed the fetal recipient two months later and trans- ferred the recovered human CD45' cells into secondary recipients. Engraftment capacity in the secondary recipient (determined two months after secondary transfer) strictly correlated with the long-term multi- lineage hematopoietic reconstitution capacity (one to two years) of selected test populations in primary sheep (Zanjuni, personal communication). If confirmed, this modification may allow the identification of populations containing human long-term repopulating cells in a four-month assay.

Immunodeficient Mouse Models Xenotransplantation approaches would be facilitated by the development of simple transfer proce-

dures and models that are more economical than sheep. The identification of genetically immunodefi- cient mouse strains has provided a small animal model with a permissive microenvironment for the engraftment of human hematopoietic cells [69-7 I]. The most commonly used mouse strains include beige, nude and X-linked immunodeficient (bnx) mice, severe combined immunodeficient (SCID) mice and recently, nonobese diabetic (NOD-)KID mice.

Bnx mice were constructed by combining three recessive mutations which synergistically affect different cellular immune activities. The reduced immunity of the parental nude mouse (affecting T cell differentiation) was further reduced by the effects of the beige (cytotoxic T lymphocyte and natural killer [NK] cells) and X-linked immunodeficiency (lymphokine-activated killer cells and certain B cell responses) mutations [72-741. SCID mice are severely deficient in B and T lymphocytes, since lymphocyte differentiation is arrested as a result of a dysfunctional VDJ recombination of immunoglobulin and T cell receptor genes [70, 75, 761. However, residual immunity in both SCID and bnx mice, due to NK cells and macrophages, can mediate rejec- tion of xenografted human cells [77]. To generate a SCID strain with low NK cell activity, the scid mutation was recently backcrossed onto the NOD/Lt strain background, which is characterized by a deficiency in NK cell activity, by functional defects of both antigen presenting cells and macrophages, and by reduced lytic com- plement activity [71]. NOD/Lt mice display a high incidence of autoimmune T cell-mediated diabetes melli- tus [78] which is prevented in NODLtSz-scidscid (NOD-SCID) mice due to the scid-related absence of mature T cells [7 1,791,

Engraftment of human hematopoietic cells has been achieved in all three mouse strains, and engraftment levels appeared highest in NOD-SCID mice [80-831. This may reflect the extensive immunodeficiency of this strain, but may also result from the particular support of human hematopoietic cells by NOD-SCID stre ma. Multiple strategies have been pursued to provide complementary support for proliferation and differen- tiation of xenografted cells. In the SCID-hu model, human fetal thymus or fetal bone fragments have been cotransplanted with human fetal liver cells in order to provide a human microenvironment [84-871. Alternatively, recombinant human (rHu)SCF and PIXY 321, a fusion protein of HuGM-CSF and HuIL-3,

Bock 191

have been injected into SCID mice [88, 891. Selection of these cytokines was based on the rationale that SCF, GM-CSF and IL-3 act highly species-specific and promoted growth and differentiation of bone mar- row progenitor cells in vitro [go]. In another approach, expression of the HuIL-3, GM-CSF and SCF trans- genes enhanced the engraftment of umbilical cord blood or adult bone marrow cells in SCID mice [82]. Finally, human hematopoietic cells were cotransplanted into bnx mice with primary human bone marrow stromal cells engineered to produce HuIL-3 [SO].

Immunodeficient mice have been permissive to the engraftment of human hematopoietic cells from var- ious sources including fetal liver, cord blood, adult bone marrow or adult peripheral blood. Xenografted cells differentiated into the myeloid, erythroid and B lymphoid lineages [83, 871. T lymphocytes were also recon- stituted when human fetal thymus was cotransplanted with fetal donor cells [84, 911. Hematopoiesis was clonal as demonstrated by retroviral marking and persisted long-term for at least several months [2, 80, 82, 871. Donor cells recovered from human fetal bone grafts of SCID-hu mice eight weeks postinjection had the capacity to reconstitute myelolymphoid hematopoiesis in bone grafts of secondary SCID-hu recipients [87].

Using cell purification, retroviral gene marking and limiting dilution approaches, Dick and coworkers established that the cells capable of engrafting NOD-SCID mice are biologically distinct from LTC-IC and ancestral to these progenitors [83]. The frequency of the SCID-repopulating cell (SRC) was determined at about 1 per lo6 human cord blood and 1 per 3 x 10' bone marrow cells. SRC were exclusively CD34'CD38-. The attempts to retrovirally transduce these cells have been very inef- ficient [ 831.

SUMMARY The ultimate proof of PHSC requires demonstration of long-term multilineage hematopoietic repopula-

tion in a transplant recipient. Alternatively, short-term assays may be very useful and enhance the study of early hematopoiesis. This is particularly true for the human system. However, validity and specificity of alter- native assays must be confirmed in vivo. This has either not been performed for most in vitro assay systems or results have not been satisfying [S, 921.

As a recent improvement, the LTC-ICML extends the range of differentiation that hematopoietic cells have achieved in other culture systems. Furthermore, the extended LTC system appears to detect a cell more primitive than standard in vitro-defined progenitors. The properties of the (E)LTC-IC cell, as far as they are known, are shared with the presumed PHSC phenotype.

The development of xenotransplantation strategies using immunodeficient mice or fetal sheep has result- ed in attractive models for human hematopoietic cells. Since long-term lymphomyeloid in vivo repopulation capacity, as well as extensive proliferative potential, have been demonstrated for the cells engrafting immuno- deficient mice and preimmune fetal sheep, these xenograft models both appear to be promising assays for PHSC. However, additional progress could be helpful in establishing these systems as a better quantitative assay for xenorecipient repopulating-cells (XRC). Since quantitation often involves Poisson analysis of mul- tiple limiting dilution setups, chimeric mice may be more useful than sheep. The coenrichment of cells exhibiting secondary transfer potential with long-term XRC supports the prospect that short-term in vivo assays for human hematopoietic stem cells are feasible.

However, until purification of PHSC to homogeneity is achieved and a specific PHSC phenotype is established, proof of PHSC requires the demonstration of long-term hematopoietic repopulation of all lineages in a transplant recipient. Only cells providing such evidence should be considered PHSC. Concern exists that differences in functional state or proliferative capacity of individual PHSC may result in extreme phenotypic heterogeneity and multiple stem cell subsets, and may challenge our ability to purify PHSC to homogeneity. However, only with a phenotypically homogeneous input population of PHSC can studies using different cell culture and animal models be considered truly comparative. Additionally, these studies may be affected by the limited assay sensitivity to minimal numbers of PHSC in a presumably homogeneous subpopulation of stem cells. In view of these many obstacles in studying

192 Stem Cell Assays

the most primitive hematopoietic cell, the isolation of the PHSC truly does earn the designation of the holy grad of hematology.

ACKNOWLEDGMENT

longstanding support as well as that of Dr. Lothar Kunz and Dr. Arthur W. Nienhuis. I am indebted to Dr. David M. Bodine and Dr. Donald Orlic, and gratefully acknowledge their

REFERENCES 1

2

3

4

5

6

7

8

9

10

11

Jordan CT, Lemischka IR. Clonal and systemic analysis of long-term hematopoiesis in the mouse. Genes Dev 1990;4:220-232.

N o h JA, Dao MA, Wells S et al. Transduction of pluripotent human hematopoietic stem cells demon- strated by clonal analysis after engraftment in immune-deficient mice. Proc Natl Acad Sci USA 1996;93:2414-24 19.

Harrison DE, Astle CM, DeLaittre JA. Loss of proliferative capacity in immunohemopoietic stem cells caused by serial transplantation rather than aging. J Exp Med 1978;147:1526-1531.

Lansdorp PM. Developmental changes in the func- tion of hematopoietic stem cells. Exp Hematol 1995;23: 187- I9 1.

Orlic D, Bodine DM. What defines a pluripotent hematopoietic stem cell (PHSC): will the real PHSC please stand up! Blood 1994;84:3991-3994.

Morrison SJ, Uchida N, Weissman IL. The biolo- gy of hematopoietic stem cells. Annu Rev Cell Biol 1995;11:35-71.

Nakahata T, Ogawa M. Identification in culture of a class of hemopoietic colony-forming units with extensive capability to self-renew and generate multi- potential hemopoietic colonies. Proc Natl Acad Sci

Koike K, Ihle JN, Ogawa M. Declining sensitivity to interleukin 3 of murine multipotential hemopoi- etic progenitors during their development. Appli- cation to a culture system that favors blast cell colony formation. J Clin Invest 1986;77:894-899.

Leary AG, Ogawa M. Blast cell colony assay for umbilical cord blood and adult bone marrow progenitors. Blood 1987;69:953-956.

Brandt E, Baird N, Lu L e t al. Characterization of a human hematopoietic progenitor cell capable of forming blast cell containing colonies in vitro. J Clin Invest 1988;82:1017-1027.

Leary AG, Ikebuchi K, Hirai Y et al. Synergism between interleukin-6 and interleuhn-3 in sup- porting proliferation of human hematopoietic stem cells: comparison with interleukin- 1 alpha. Blood 1988;71: 1759- 1763.

USA 1982;79:3843-3847.

12

13

14

15

16

17

18

19

20

21

Ogawa M. Effects of hematopoietic growth factors on stem cells in vitro. Hematol Oncol Clin North Am 1989;3:453-464.

Bradley TR, Hodgson GS. Detection of primitive macrophage progenitor cells in the mouse bone marrow. Blood 1979;54:1446-1450.

McNiece IK, Stewart DM, Deacon DS et al. Detection of a human CFC with high proliferative potential. Blood 1989;74: 609-6 12.

Bertoncello I, Bradley T, Hodgson G et al. The resolution, enrichment, and organization of normal bone marrow high proliferative potential colony- forming cell subsets on the basis of rhodamine- 123 fluorescence. Exp Hematol 1991;19: 174-178.

Srour EF, Brandt JE, Bride11 RA et al. Long-term generation and expansion of human primitive hematopoietic progenitor cells in vitro. Blood 1993;8 11661.669.

Bradley TR, Hodgson GS, Bertoncello I. Character- istics of primitive macrophage progenitor cells with high proliferative potential: relationship to cells with marrow repopulating ability with 5-fluorouracil treated mouse bone marrow. In: Baum SJ, Ledney GD, van BeWcum DW, eds. Experimental Hematology Today. Berlin, New York: Springer International, 1982:285-292.

McNiece IK, Bertoncello I, Kriegler AB et al. Colony-forming cells with high proliferative poten- tial (HPP-CFC). Int J Cell Cloning 1990;8: 146.160.

Cohen GI, Canellos GP, Greenberger JS. In vitro quantitation of engraftment between purified popu- lations of bone marrow hemopoietic stem cells and stromal cells. In: Gale RP, Fox CF, eds. Biology of Bone Marrow Transplantation. New York: Academic Press, 1980:491-506.

Gordon MY, Hibbin JA, Kearney LU et al. Colony formation by primitive hemopoietic progenitor cells in cocultures of bone marrow cells and stromal cells. Br J Hematol 1985;60:129-136,

Gordon MY, Dowding CR, Riley GP et al. Characterization of stroma dependent blast colony- forming cells in human marrow. J Cell Physiol 1987;130:150-156.

Bock 193

22 Ploemacher R, van der Sluijs J, Voerman J et al. An in vitro limiting-dilution assay of long-term repopulating hematopoietic stem cells in the mouse.

23 Ploemacher R, van der Sluijs J, van Beurden C et al. Use of limiting-dilution type long-term marrow cul- tures in frequency analysis of marrow-repopulating and spleen colony-forming hematopoietic stem cells in the mouse. Blood 1991;10:2527-2533.

24 Dowding CD, Gordon MY. Physical, phenotypic and cytochemical characterization of stroma- adherent blast colony-forming cells. Leukemia

25 Neben S, Anklesaria P, Greenberger J et al. Quanti- tation of murine hematopoietic stem cells in vitro by limiting dilution analysis of cobblestone area forma- tion on a clonal stromal cell line. Exp Hematol

Blood 1989;74:2755-2763.

1992;6:347-35 I .

1993;21:438-443.

26 Breems DA, Blokland EA, Neben S et al. Frequency analysis of human primitive haematopoietic stem cell subsets using a cobblestone area forming cell assay. Leukemia 1994;8:1095-1104.

27 Ploemacher RE, Van der Loo JCM, Van Beurden CAJ et al. Wheat germ agglutinin affinity of murine hematopoietic stem cell subpopulations is an inverse function of their long-term repopulating ability in vitro and in vivo. Leukemia 1993;7:120-130.

28 Uchida N, Combs J, Chen S et al. Primitive human hematopoietic cells displaying differential efflux of the rhodamine 123 dye have distinct biological activities. Blood 1996;88: 1297-1305.

29 Dexter TM, Allen TD, Lajtha LG et al. In vitro analysis of self-renewal and commitment of hematopoietic stem cells. In: Clarkson B, Marks PA, Till JE, eds. Differentiation of Normal and Neoplastic Hemopoietic Cells. Cold Spring Harbor Conference on Cell Proliferation, Vol. 5 . Plainview, NY: Cold Spring Harbor Laboratory, 1977:63-80.

Cell Physiol 1982;l(suppl):87-94.

31 Dexter TM, Allen TD, Lajtha LG. Conditions con- trolling the proliferation of hemopoietic stem cells in vitro. J Cell Physiol 1977;91:335-344.

32 Dexter TM. Cell interactions in vitro. Clin Hematol

30 Dexter T. Stromal cell associated haemopoiesis. J

1979;8:453-468.

33 Gartner S, Kaplan HS. Long-term culture of human bone marrow cells. Proc Natl Acad Sci USA 1980;77:4756-4759.

34 Eaves AC, Cashman JD, Gaboury LA et a]. Unregulated proliferation of primitive chronic myeloid leukemia progenitors in the presence of normal marrow adherent cells. Proc Natl Acad Sci USA 1986;83:5306-5310.

35 Sutherland HJ, Eaves CJ, Eaves AC et al. Characterization and partial purification of human marrow cells capable of initiating long-term hematopoiesis in vitro. Blood 1989;74: 1563-1570.

36 Sutherland HJ, Lansdorp PM, Henkelman DH et al. Functional characterization of individual hematopoietic stem cells cultured at limiting dilu- tion on supportive marrow stromal layers. Proc Natl Acad Sci USA 1990;87:3584-3588.

37 Szilvassy SJ, Fraser CC, Eaves CJ et al. Retrovirus- mediated gene transfer to purified hemopoietic stem cells with long-term lymphomyelopoietic repopulating ability. Proc Natl Acad Sci USA 1989;86:8798-8802.

38 Eaves CJ, Sutherland HS, Udomsakdi C et al. The human hematopoietic stem cell in vitro and in vivo. Blood Cells 1992; 18:301-307.

39 Tsai S, Emerson SG, Sieff CA et al. Isolation of a human stromal cell strain secreting hematopoietic growth factors. J Cell Physiol 1986; 127: 137-145.

40 Sutherland HJ, Eaves CJ, Lansdorp PM et al. Differential regulation of primitive human hemato- poietic cells in long-term cultures maintained on genetically engineered murine stromal cells. Blood 1991;78:666-672.

41 Issaad C, Croisille L, Katz A et al. Murine stromal cell line allows the proliferation of very primitive human CD34”/CD38- progenitor cells in long-term cultures and semi-solid assays. Blood 1993;81:2916-2924.

42 Udomsakdi C, Eaves CJ, Swolin B et al. Rapid decline of chronic myeloid leukemia cells in long- term culture due to a defect at the leukemic stem cell. Proc Natl Acad Sci USA 1992;89:6 192-6 196.

43 Van der Sluijs JP, Van den Bos C, Baert MRM et al. Loss of long-term repopulating ability in long-term bone marrow culture. Leukemia 1993;7:725-732.

44 Udomsakdi C, Lansdorp PM, Hogge D et al. Characterization of primitive hematopoietic cells in normal human peripheral blood. Blood 1992;80:2513-2521.

45 Eaves CJ, Cashman JD, Zoumbos NC et al. Biological strategies for the selective manipulation of normal and leukemic stem cells. STEM CELLS 1993;l l(suppl 3): 109-1 21.

46 Udomsakdi C, Eaves CJ, Sutherland HJ et al. Separation of functionally distinct subpopulations of primitive human hematopoietic cells using Rhodamine-123. Exp Hematol 1991;19:338-342.

47 Chaudhary PM, Roninson IB. Expression and activity of p-glycoprotein, a multidrug efflux pump, in human hematopoietic stem cells. Cell 199 I ;66:85-94.

194

48 Craig W, Kay R, Cutler RL et al. Expression of Thy-I on human hematopoietic progenitor cells. J Exp Med 1993;177: 133 1-1342.

49 Crooks GM, Hao QL, Thiemann FT et al. Extended long-term culture-initiating cells (ELTC-IC) in cord blood are more quiescent and functionally more primitive than LTC-IC. Exp Hematol 1996;24: 1028a.

50 Bride11 RA, Broudy VC, Bruno E et al. Further

51

52

53

54

55

56

57

phenotypic characterization and isolation of human hernatopoietic progenitor cells using a monoclonal antibody to the c-kit receptor. Blood 1992;79:3 159-3167.

Hao QL, Shah AJ, Thiemann FT et al. A function- al comparison of CD34’CD38- cells in cord blood and bone marrow. Blood 1995;86:3745-3753.

Lemieux RE, Rebel VI, Lansdorp PM et al. Characterization and purification of a primitive hematooietic cell type in adult mouse marrow capable of lymphomyeloid differentiation in long-term marrow “switch” cultures. Blood 1995;86: 1339-1347.

Lemieux ME, Eaves CJ. Identification of proper- ties that can distinguish primitive populations of stromal-cell-responsive lympho-myeloid cells from cells that are stromal-cell-responsive but lymphoid-restricted and cells that have lympho- myeloid potential but are also capable of competi- tively repopulating myeloablated recipients. Blood 1996i88: 1639-1648.

Moore MAS. Clinical implications of positive and negative hematopoietic stem cell regulators. Blood 1991;78:1-19.

Shapiro F, Yao TJ, Raptis G et al. Optimization of conditions for ex vivo expansion of CD34’ cells from patients with stage IV breast cancer. Blood 1994;84:3567-3574.

Gordon MY, Riley GP, Greaves MF. Plastic-adher- ent progenitor cells in human bone marrow. Exp Hematol 1987;15:772-778,

Bearpark AD, Gordon MY. Adhesive properties distinguish sub-populations of haemopoietic stem cells with different spleen-forming and marrow repopulating capacities. Bone Marrow Transplant 1989;4:625-628.

58 Gordon MY, Clarke D, Healy LE. An in vitro model for the production of committed haemopoietic pro- genitor cells stimulated by exposure to single and combined recombinant growth factors. Bone Marrow Transplant 1989;4:353-358.

Blood Rev 1993;7:190-197. 59 Gordon MY. Human haemopoietic stem cell assays.

Stem Cell Assays

60 Brecher G, Neben S, Yee M et al. Plnripotent stem

61

62

63

64

65

66

67

68

cells with normal or reduced self-renewal survive lethal irradiation. Exp Hematol 1988; 16:627-630.

Harrison DE. Competitive repopulation: a new assay for long-term stem cell functional capacity. Blood 1980;55:77-81.

Odic D, Anderson S, Bodine DM. Biological properties of pluripotent hematopoietic stem cells enriched by elutriation and flow cytometry. Blood Cells 1994;20:107-I 17.

Bodine DM, Seidel NE, Orlic D. Bone marrow C O I L lected 14 days after in vivo administration ofgranu- locyte colony-stimulating factor and stem cell factor to mice has 10-fold more repopulating ability than untreated bone marrow. Blood 1996;88:89-97.

Flake AW, Harrison MR, Adzick NS et al. Transplantation of fetal hematopoietic stem cells in utero: the creation of hematopoietic chimeras. Science 1986;233:776-778.

Srour EF, Zanjani ED, Brandt JE et al. Sustained human hematopoiesis in sheep transplanted in utero during early gestation with fractionated adult human bone marrow cells. Blood 1992;79:1404-1412.

Zanjani ED, Flake AW, Rice H et al. Long-term repopulating ability of xenogeneic transplanted human fetal liver hematopoietic stem cells in sheep. J Clin Invest 1994;93:1051-1055.

Zanjani ED, Ascensao JL, Harrison MR et al. Ex vivo incubation with growth factors enhances the engraftrnent of fetal hematopoietic cells transplanted in sheep fetuses. Blood 1992;79:3045-3049.

Kawashima I, Zanjani ED, Almaida-Porada G et al. CD34’ human marrow cells that express low lev- els of kit protein are enriched for long-term mar- row-engrafting cells. Blood 1996;87:4136-4142.

69 Flanagan SP. ‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genet Res 1966;8:295-309.

70 Bosma GC, Custer RP, Bosma MJ. A severe com-

71

72

73

bined immunodeficiency mutation in the mouse. Nature 1983;301:527-530.

Shultz LD, Schweitzer PA, Christianson SW et al. Multiple defects in innate and adaptive immuno- logic function in NOD-LtSz-scid mice. J Immunol 1995;154:IXO-l9l.

Pantelouris EM. Observations on the immunobiology of the ‘nude’ mouse. Immunology 1971 ;20:247-252.

Scher I, Steinberg AD, Berning AK et al. X-linked B-lymphocyte immune defect in CBA/N mice. 11. Studies on the mechanisms underlying the immune defect. J Exp Med 1975;142:637-650.

Bock 195

74 Roder J, Duwe A. The beige mutation in the mouse selectively impairs natural killer cell function. Nature 1979;278:45 1-453.

75 M d y ~ BA, Blackwell TK, Fulop GM et al. The scid defect affects the final step of the immunoglobulin VDJ recombinase mechanism. Cell 1988;54:453-460.

76 Bosma MJ, Carroll AM. The SCID mouse mutant: definition, characterization, and potential uses. Annu Rev Immunol 1991;9:323-350.

77 Murphy WJ, Bennett M, Anver MR et al. Human-mouse lymphoid chimeras: host-vs.- graft and graft-vs.-host reactions. Eur J Immunol 1992;22: 1421 -1427.

78 Leiter E. The nonobese diabetic mouse: a model for analyzing the interplay between heredity and envi- ronment in development of autoimmune disease. ILAR News 1993;35:4-6.

79 Chnstianson SW, Shutz LD, Leiter EH. Adoptive transfer of diabetes into immunodeficient NOD- scidscid mice: relative contributions of CD4' and CD8' T lymphocytes from diabetic versus prediabet- ic NOD.NON-thy la donors. Diabetes 1993;42:44.

80 Nolta JA, Hanley MB, Kohn DB. Sustained human hematopoiesis in immunodeficient mice by cotrans- plantation of marrow stroma expressing human inter- leukin-3: analysis of gene transduction of long-lived progenitors. Blood 1994;83:3041-305 1.

81 Vormoor J, Lapidot T, Pflumio F et al. Immature human cord blood progenitors engraft and proliferate to high levels in severe combined immunodeficient mice. Blood 1994;83:2489-2497.

82 Bock TA, Orlic D, Dunbar CE et al. Improved engraftment of human hematopoietic cells in severe combined immunodeficient (SCID) mice carrying human cytokine transgenes. J Exp Med 1995; 18212037-2043.

83 Larochelle A, Lapidot T, Vormoor J et al. Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse

bone marrow: implications for gene therapy. Nat Med 1996;2: 1329-1337.

84 McCune JM, Namikawa R, Kaneshima H et al. The SCID-hu mouse: murine model for the analy- sis of human hematolymphoid differentiation and function. Science 1988;241: 1632-1639.

85 McCune JM. SCID mice as immune system models. Curr Opin Immunol 199 1 ;3:224-228.

86 Kyoizumi S, Baum CM, Kaneshima H et al. Implantation and maintenance of functional human bone marrow in SCID-hu mice. Blood 1992;78: 1704-17 1 1.

87 Chen BP, Galy A, Kyoizumi S et al. Engr a f tment of human hematopoietic precursor cells with sec- ondary transfer potential in SCID-hu mice. Blood 1994;84:2497-2505.

88 Kamel-Reid S, Dick JE. Engraftment of immune- deficient mice with human hematopoietic stem cells. Science 1988;242: 1706- 1709.

89 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.

90 Bernstein ID, Andrews RG, Zsebo KM. Recom- binant human stem cell factor enhances the forma- tion of colonies by CD34' and CD34'lin- cells. and the generation of colony-forming cell progeny from CD34'lin- cells cultured with interleukin-3, granulocyte colony-stimulating factor, or granulo- cyte-macrophage colony-stimulating factor. Blood 199 I ;77:23 16-232 1.

91 Peault B, Weissman IL, Baum C et a]. Lymphoid reconstitution of the human fetal thymus in SCID mice with CD34' precursor cells. J Exp Med I99 I;174: 1283-1286.

92 Harrison DE, Lerner CP, Spooncer E. Erythro- poietic repopulating ability of stem cells from long- term marrow cultures. Blood 1987;69:1021-1025.