Hematopoiesis has been considered hierarchical in nature, but recent data suggest that the system is not hierarchical and is, in fact, quite functionally plastic. Existing data indicate that engraftment and progenitor phenotypes vary inversely with cell cycle transit and that gene expression also varies widely. These observations suggest that there is no progenitor/stem cell hierarchy, but rather a reversible continuum. This may, in turn, be dependent on shifting chromatin and gene expression with cell cycle transit. If the phenotype of these primitive marrow cells changes from engraftable stem cell to progenitor and back to engraftable stem cell with cycle transit, then this suggests that the identity of the engraftable stem cell may be partially masked in nonsynchronized marrow cell populations. A general model indicates a marrow cell that can continually change its surface receptor expression and thus responds to external stimuli differently at different points in the cell cycle.

We present here a different view of early hematopoiesis. This is not the standard dogma of many investigators in the field, but is built upon solid work by many laboratories over many years. This is a perspective, and thus naturally reflects the biases of the authors. These, in turn, have a basis in many published studies from our laboratory and other laboratories and also from extensive unpublished, but abstracted, data from our laboratory. It is appropriate to consider this contribution as speculative in nature.

The term chiaroscuro refers to the treatment of light and shade in painting. Our current view of the changing phenotype of the marrow stem cell suggests a chiaroscuro nature to this picture.

In general, models of stem cell regulation have been hierarchical.1,2 A primitive stem cell, with great potential, gives rise to a proliferating progenitor pool, which in turn gives rise to recognizable differentiated cells. During this process, proliferative potential is lost, while specific differentiated features are acquired. Presumptively, but without definite proof, there is also self-renewal at the most primitive stem cell level, and this is also lost with differentiation. Many data exist to support such a hierarchical model. Marrow cells have been separated with short- and long-term repopulation potential3,4 and progenitors have been characterized as exclusively committed to the production of restricted progeny.5,6 In addition, the clear expansion of different progenitor types in cytokine-stimulated in vitro culture with a loss of long-term engraftment capacity speaks to the existence of a progenitor hierarchy, at least at the more differentiated progenitor levels.7,8 A model encompassing these features is shown in Figure 1.

Fig. 1.

Classic hierarchical model of hematopoiesis.

S represents stem cell; P, progenitor; and D, differentiated cell.

Fig. 1.

Classic hierarchical model of hematopoiesis.

S represents stem cell; P, progenitor; and D, differentiated cell.

Close modal

This model does not fit with all published data. For instance, the “daughter cell” or paired-progenitor experiments of Suda and colleagues9 indicate that a primitive progenitor cell can make totally different lineage choices during one cell cycle transit, such that, for example, one daughter cell forms an erythroid colony while the other daughter (or sister) cell forms a neutrophil-macrophage colony. Out of a total of 387 pairs evaluated, 68 pairs of colonies consisted of dissimilar combinations of cell lineages. Admittedly, these studies were carried out using spleen as a cell source with erythropoietin and an uncharacterized conditioned media as a source of stimulation. In addition, others have reported high degrees of congruence in the phenotype of individual colonies observed after replating single cells from colony starts.10 Despite these caveats, the elegant experiments of Suda and colleagues9would seem to weigh against an ordered hematopoietic stem cell hierarchy.

An intrinsic component of this hierarchical model is that the most primitive hematopoietic stem cell is a quiescent cell in G0 and a fount of potential without differentiated characteristics. It has generally been believed that primitive hematopoietic stem cells were dormant or quiescent and were thus protected from depletion or exhaustion.11,12 On the basis of a number of transplant studies, a clonal succession model has been proposed.13 This model proposes that the production of blood cells is maintained sequentially by one or just a few lymphohematopoietic stem cells, the residual stem cells remaining dormant. This model is consistent with reports evaluating the contributions of individually marked hematopoietic stem cells in transplant studies.14-16 This scenario formed the basis for the concept that hematopoietic stem cells are hierarchically ordered based on their relative quiescence.17,18 Other investigators have presented data indicating that hematopoietic stem cells are functioning concurrently, continuously cycling and contributing to blood cell production.19-21 This latter view suggests that hematopoietic stem cell quiescence is relative, the cells being relatively quiescent compared with more differentiated progenitor cells, but not dormant, often proceeding through cell cycle and undergoing cell division. Certainly, stem cells, as evaluated at any one point in time, are relatively quiescent. Our laboratory's standard stem cell separation is based on this relative quiescence, separating lineage-negative, rhodamine dull and Hoechst dull (LinRhodullHodull) cells,22-27 although other separations based on Sca-1 and c-kit epitopes include a small percentage of proliferating cells.28-30 In studies on Hoechst-stained side population marrow stem cells, Goodell et al31 determined that between 1% and 3% of these cells were in S/G2/M. Previous studies with cell cycle–specific killing drugs did not distinguish between cells that are truly dormant and cells that are either in prolonged cell cycle or intermittently entering and exiting cell cycle at a slow rate. In previous studies of hematopoietic cell cycling the cells analyzed were populations with functional characteristics of late progenitors.32-37 Bone marrow repopulating ability had been assessed with 7 days of bromodeoxyuridine (BrdU) labeling and a turnover time of more than 11.5 days was demonstrated.36 Labeling after 32 days of exposure to BrdU was assessed by one group.37 They showed a T½ of 21 days for colony-forming unit spleen (CFU-S) on day 14, but in this study long-term repopulating cells were not assessed.

Bradford and colleagues, studying long-term repopulating cells as represented by LinRhodullHodull–separated murine marrow cells and using in vivo exposure to BrdU, evaluated the proliferative history of these stem cells over longer periods of time in vivo.38 They studied mice continuously exposed to BrdU in their drinking water and then assessed the percentage of LinRhodullHodull marrow stem cells labeling over different time intervals of BrdU administration. BrdU is incorporated into DNA during DNA synthesis, and thus labeling with this agent is an accurate measure of whether a cell has transited S phase while progressing through the cell cycle. These researchers isolated LinRhodullHodull cells from these BrdU-exposed mice. They found that 60% (± 14%) of these primitive stem cells were labeled after 4 weeks and showed a time to 50% labeling (T½) of 19 days. Cheshier and colleagues, using a different mouse strain and a different stem cell separation, confirmed these data; they showed more rapid labeling of stem cells.30 We evaluated whether these data could be explained by DNA damage and repair, rather than proliferation, and after extensive studies came to the conclusion that the BrdU incorporation did, in fact, indicate proliferation of these primitive stem cells over time.39 We studied BALB/c mice and LinRhodullHodull cells in a fashion identical to that of Bradford et al38 and obtained virtually identical results. Thus, when the cycling status of stem cells is approached over a longer time frame, it appears that these relatively quiescent cells are either in a prolonged active cell cycle or, perhaps more likely, repeatedly entering and leaving the cell cycle. The latter possibility is supported by studies showing that transplanted stem cells rapidly enter cell cycle40,41 and that primitive marrow stem cells are easily induced into cell cycle on in vitro cytokine exposure.42 Abkowitz and colleagues,43 using stochastic modeling and computer simulation to study the replication, apoptosis, and differentiation of murine hematopoietic cells, estimated that murine hematopoietic stem cells replicated (on average) every 2.5 weeks. When this approach was used in cats, heterozygous for glucose-6-phosphate dehydrogenase, different estimates of hematopoietic stem cell replication rate (1 per 8.3 to 10 weeks) were derived. Thus this present work, along with the work of others, indicates that virtually all primitive marrow stem cells, while relatively quiescent, pass through the cell cycle over a varying period of time, depending on mouse strain and possibly the specifics of the stem cell separation.

In a similar vein, while these cells do not express terminal differentiation functions, they robustly express certain stem cell functions. They are highly motile, showing very rapid directed movement and rapid membrane deformation on cytokine exposure.44These cells also express a large number of stem cell–specific genes. Phillips et al45 have reported on 2119 nonredundant gene products from fetal liver hematopoietic stem cells, using a subtracted cDNA library to generate a micro array chip. They have identified several genes specific to fetal liver hematopoietic cells. In addition, when comparing fetal hematopoietic cells with adult hematopoietic cells (LinRhodull Hodullc-kit+Sca-1+), they found several genes that were coexpressed on fetal and adult stem cells as well as genes specific for either fetal or adult stem cells. Park et al46 have also reported on murine hematopoietic stem cells gene profiling. They used a 5000-cDNA array obtained by subtraction of cDNA from lineage-positive cell populations and studied both hematopoietic adult stem cells and multipotent progenitors (with minimal self-renewal capacity). Genes primarily expressed in stem cells were transcription factors, RNA binding proteins, chromatin modifiers, and protein kinases. In differential gene display studies using a gel-based method that uses the display of 3′ end fragments of cDNA generated by cutting with specific enzymes,47 we have identified a total of 637 differentially expressed genes in murine LinRhodullHodull cells compared with differentiated (Lin+) cells (S. Weissman et al, unpublished data, 2001). These genes include 411 unknowns and 19 different gene categories. These categories included transcription factors (22), protein synthesis regulators (11), surface proteins (11), mitochondrial sequences (10), RNA metabolism proteins (10), signaling pathway factors (9), and cytokines (8). In separate studies, we have also shown that LinRhodullHodull marrow cells express adhesion proteins48,49 and cytokine receptors50 on the cell surface. These data indicate a different picture of the primitive marrow stem cell than is usually envisioned—a functional cell in which a large number of stem cell–specific, as opposed to differentiated, functions are manifest (Figure 2).

Fig. 2.

Stem cell–specific functions.

This figure illustrates proteopodia membrane extensions and adherence to a mesenchymal stromal cell (M). Surface-based symbols illustrate cell surface–based receptors.

Fig. 2.

Stem cell–specific functions.

This figure illustrates proteopodia membrane extensions and adherence to a mesenchymal stromal cell (M). Surface-based symbols illustrate cell surface–based receptors.

Close modal

In addition, work by others has indicated that marrow-derived stem cells may express lineage-specific genes prior to commitment.51-53 These studies indicate that primitive hematopoietic stem cells express a wide variety of genes, including a number of transcription factors. Presumptively, transcription factor access to DNA is a major determinant of such gene expression.

Chromatin remodels during cell cycle progression. In previous studies chromatin modulation at the B-globin and lysozyme gene loci were evaluated54-57 and myeloid specificcis-regulatory elements showed a specific chromatin pattern at the lysozyme locus in myelomonocytic cells at different differentiation stages. This chromatin pattern was also found in multipotent hematopoietic progenitors, but it disappeared when cells differentiated into the erythroid lineage. Evaluation of the chromatin structure of multipotent hematopoietic cells indicated that the lineage-associated genes, globin, myeloperoxidase, immunoglobulin H (IgH), and CD3δ have accessible control regions before unilineage commitment.55-59 Other work indicates that chromatin remodeling factors may be recruited in one phase of the cell cycle for ultimate action in a later phase, which in turn results in changes in transcriptional programs. Consistent with the model of a fluctuating cell cycle–based stem cell phenotype and asymmetric division is the proposal of McConnell and Kaznowski60 that cell cycle passage could determine the fate of cells derived from stem cell division and renew stem cell multipotency. Studying laminar determination in the developing neocortex, these investigators transplanted embryonic neural progenitor cells into older host brains. The fate of the transplanted neurons correlated with the position of the progenitors in the cell cycle at the time of transplantation. Daughters of cells transplanted in S phase migrated to layer 2/3, while progenitors transplanted later in the cell cycle produced daughters that were committed to their normal deep-layer fate. These studies indicate that the cell fate of the immediate stem cell progeny is restricted by environmental signals shortly after S phase; passage through the next S phase then restores multipotency. These observations might be explained by alterations in chromatin structure during DNA replication. Such alterations could have marked effects on gene expression and therefore on cell fate.

This hypothesis is supported by studies in yeast61,62showing that chromatin-remodeling factors are recruited during M/G1 and chromatin is modeled in the next G1. Thus a reasonable sequence of events would be chromatin remodeling with cytokine-induced cell cycle transit leading to varying levels of DNA access to transcription factors, followed by alterations in patterns of gene expression.

In models of stem cell–cycle progression and division, there are several possibilities. The stem cell could have a symmetric division with the production of 2 identical stem cells, a symmetric division with the production of 2 differentiating cells, or an asymmetric division with the production of 1 stem cell and 1 differentiated cell. Most models assume that, on a population basis, stem cells would show asymmetric division; the other alternatives would lead to either hyperproliferation or stem cell exhaustion. These events might also be modulated by cellular apoptosis. The above-described stem cell phenotypic shifts with cell cycle transit and the concept of asymmetric divisions form the basis for the model shown in Figure3.

Fig. 3.

Fluctuating stem cell phenotype and genotype with cell cycle transit.

Chromatin modulation and resulting changes in access of transcription factors to different control regions. Shown is an asymmetric division in which a stem cell (S) produces a phenotypically similar stem cell and a cell (D) destined for terminal differentiation. Cylinders represent DNA chromatin coverage; boxes, active transcription factors.

Fig. 3.

Fluctuating stem cell phenotype and genotype with cell cycle transit.

Chromatin modulation and resulting changes in access of transcription factors to different control regions. Shown is an asymmetric division in which a stem cell (S) produces a phenotypically similar stem cell and a cell (D) destined for terminal differentiation. Cylinders represent DNA chromatin coverage; boxes, active transcription factors.

Close modal

We have been investigating the phenotype of marrow stem cells as they progress through the cell cycle under cytokine stimulation. Initial studies showed that cytokine stimulation in vitro with interleukin-3 (IL-3), interleukin-11 (IL-11), interleukin-6 (IL-6), and steel factor for 48 hours resulted in a loss of engraftment capacity at 3 to 24 weeks and that longer-term engraftment was most strongly affected.63-65 Early engraftment, at 1 to 3 weeks, was either modestly augmented or unaffected. Subsequent studies evaluated the engraftment capacity of BALB/c or C57BL/6J marrow cells cultured in IL-3, IL-6, IL-11, and steel factor in nonadherent Teflon bottles every 2 to 4 hours from 24 to 80 hours of culture.66 Separate studies had determined the cell cycle status of LinRhodullHodull marrow cells under the same cytokine and culture conditions.42These hematopoietic stem cells isolated on the basis of quiescence progressed in a highly synchronized fashion through the cell cycle, entering S phase at about 18 hours and completing the first population doubling at about 36 to 38 hours. These studies showed dramatic and reversible fluctuations in 8-week and 6-month engraftment capacity at 2- to 4-hour intervals, with a nadir and recovery appearing in the first cell cycle transit. Cell cycle–related shifts in the functional and surface phenotype of both murine and human primitive hematopoietic stem cells have been observed by a number of investigators. Fleming and colleagues showed functional heterogeneity associated with the cell cycle status of murine stem cells.67 They found that purified lineage-negative Sca-1+ and thy-1lowcells showed decreased engraftment in S/G2/M as compared with G1. Orschell-Traycoff et al68 showed that when engrafting phenotypes of Sca-1+Lin stem cells were Hoechst fractionated into G0 /G1 or S/G2/M, cells with long-term engraftment capacity were found in G0/G1. Gothot et al,69studying primitive human progenitor in bone marrow and peripheral blood, found multipotent progenitors in G0 or early G1, while lineage-restricted granulomonocytic progenitors were more abundant in late G1. These investigators used Hoechst 33342 and pyronin to isolate cells. The same group found that the repopulating capacity of human mobilized peripheral blood CD34+ cells in nonobese diabetic/severe combined immunodeficient mice (NOD/SCID ) was increased in G0 as opposed to G1.70 In a similar vein, Glimm and colleagues, studying human cord blood with lymphomyeloid repopulating activity in NOD/SCID mice, showed that transplantable stem cell activity was restricted to the G1fraction, even though colony forming cells (CFCs) and long-term culture initiating cells (LTC-ICs) were equally distributed between G0/G1 and S/G2/M.71G0 cells had (and were defined by) reduced K167 and cyclin D expression and low Hoechst expression. CD34+ cord blood cells stimulated with steel factor, thrombopoietin, and Flt-3 ligand showed functional differences between G0 and G1cell cycle phases, as reported by Summers et al.72 They showed a 1000-fold increase in granulocyte-macrophage colony forming cells (GM-CFCs) in G0 as compared with G1, a 250-fold increase in burst-forming unit-erythroid (BFU-e), and a 600-fold increase in CD34+ cells. Finally, in studies of murine hematopoietic stem cells, using 2 different stem cell separative approaches, long-term in vivo engraftment was shown to be better in G0 than in G1 and in G0/G1 than in S/G2/M.73,74 

Studies of adhesion protein cell-surface expression on LinRhodullHodull and LinSca-1+ hematopoietic stem cells with cell cycle transit showed fluctuations of α-4, α-5, α-6, β-1, L-selectin, and platelet-endothelial cell adhesion molecule-1 (PECAM-1) at different points in the cell cycle.48,49 A probable causal role for these fluctuations in the engraftment fluctuations was shown by homing studies in which it was found that CFDA-SE (5- (and -6)-carboxyfluorescein diacetate succinimidyl ester)–labeled LinSca+ stem cells cultured in IL-3, IL-6, IL-11, and steel factor showed a marked depression in homing at a time when engraftment and α-4 were also markedly depressed (48 hours of culture).75 A schematic summarizing the results with marrow cells cultured in IL-3, IL-6, IL-11, and steel factor is shown in Figure 4. These results indicated that engraftment was reversibly lost at late S/early G2 and recovered in the next G1.

Fig. 4.

Fluctuating engraftment phenotype with cytokine-induced cell cycle transit.

Based on studies of murine marrow stem cells stimulated with IL-3, IL-6, IL-11, and steel factor.31,32 Engraftment is lost in late S early G2 and recovers in the next G1. ▾, ●, and ⋄ represent adhesion molecules.

Fig. 4.

Fluctuating engraftment phenotype with cytokine-induced cell cycle transit.

Based on studies of murine marrow stem cells stimulated with IL-3, IL-6, IL-11, and steel factor.31,32 Engraftment is lost in late S early G2 and recovers in the next G1. ▾, ●, and ⋄ represent adhesion molecules.

Close modal

In addition, Yamaguchi et al,76 studying CD34+ human progenitors, showed different adhesive characteristics and very-late antigen 4 (VLA-4) expression in the G0/G1 and S/G2/M phases of the cell cycle. A wide variety of other genes, including cytokine receptors, were also modulated with cycle progression.50Other gene families that were either down-regulated or up-regulated in purified murine hematopoietic stem cells after 48 hours' culture in IL-3, IL-6, IL-11, and steel factor included genes involved in energy metabolism, cell cycle regulation, signaling pathways, transcription fate, cytoskeleton, apoptosis regulation, membrane trafficking, RNA metabolism, and chromatin.77 In addition, a total of 246 unknown genes were modulated. These studies indicate that a large variety of genes are modulated with cell cycle transit, presumably underlying the shifting stem cell phenotype seen with cell cycle transit (Figure 5). All together, these studies showed a reversible fluctuating engraftment phenotype associated with cell cycle phase position, which is associated with a loss of homing and a modulation of the stem cell genotype.

Fig. 5.

Cell cycle–related fluctuations in gene expression.

The primitive marrow stem cells express a large variety of genes, and that expression changes with cell cycle transit. S represents engraftable stem cell; D, differentiated cell.

Fig. 5.

Cell cycle–related fluctuations in gene expression.

The primitive marrow stem cells express a large variety of genes, and that expression changes with cell cycle transit. S represents engraftable stem cell; D, differentiated cell.

Close modal

Differentiation was also assessed with cell cycle transit, this time with different cytokines and with 2 different culture systems. Unseparated BALB/c or C57BL/6J murine marrow cells were cultured in Teflon bottles or in microgravity rotating wall vessels in the presence of thrombopoietin, FLT3, and steel factor, and progenitor and engraftable stem cell levels were determined at various times in culture corresponding to different points in stem cell–cycle transit.78 These studies showed that there were marked and reversible increases in progenitor cell (high-proliferative-potential colony-forming cell [HPP-CFC] or colony-forming unit culture [CFU-C]) numbers during the first cell cycle transit. Remarkably, these increases in progenitor cells were tightly linked to decreases in engraftment capacity (8-week competitive engraftment into lethally irradiated mice), and generally, when the progenitor numbers returned to baseline there was a reciprocal change in engraftment capacity. A total of 20 marked increases in HPP-CFC or CFU-C numbers were observed, and in all but one case this increase was linked to a significant decrease in engraftable stem cells. These progenitor/stem cell shifts were significant at P < .0001. With longer times in cytokine culture, the progenitors expanded, indicating that the immediate kinetics were not those that might be seen in the setting of progenitor exhaustion. In a smaller number of observations, when the number of progenitors was decreased, the number of engraftable stem cells was elevated or at baseline. In general, in these experiments, when engraftable stem cell levels decreased and then returned to baseline, they did not show increases. This is consistent with an asymmetric-division stem cell model. We have termed these phenomena “stem cell inversions.” Others have reported similar findings. Knaan-Shanzer79 has proposed that there is “no hierarchy within the primitive hemopoietic stem cell compartment.” This investigator, studying umbilical cord blood cells, showed that primitive CD34+CD33,38,71NOD/SCID repopulating cells were interconvertible with the less primitive CD34+CD33+,38+,71+cells in culture. These latter showed practically “no NOD/SCID repopulating activity.” The less primitive cells could give rise to the more primitive cells. Finally, Kirkland and Borokov80have developed a phase space model of hemopoiesis and stem cell proliferation and differentiation, which is used to describe the differentiative state of cells in 2 or more dimensions. In this model the stem cell population is viewed as a continuum, rather than being composed of discrete states.

Such a model includes the possibility that some daughter cells will have greater “stemness” than the parent cells in a renewing model. This is also consistent with our observations of stem/progenitor cell inversions. A schematic encompassing these observations is presented in Figure 6.

Fig. 6.

Stem cell/progenitor cell inversion.

As marrow stem cells progress through the cell cycle, there is an inverse correlation between progenitor and stem cells. When progenitors increase, engraftable stem cells decrease, and this is reversible.

Fig. 6.

Stem cell/progenitor cell inversion.

As marrow stem cells progress through the cell cycle, there is an inverse correlation between progenitor and stem cells. When progenitors increase, engraftable stem cells decrease, and this is reversible.

Close modal

These studies indicate that previous views of the stem cell hierarchy, at least at the more primitive stem/progenitor cell levels, may be untenable. Rather than a hierarchical transition from stem to progenitor cell, it appears that a fluctuating continuum exists, in which the phenotype of these primitive marrow cells shifts from one state to another and back. Presumptively, these phenotypic shifts are based on chromatin remodeling associated with cell cycle transit, which reversibly alters the surface phenotype of the stem cell, which then determines its response to environmental stimuli. This model would also be consistent with an asymmetric model of stem cell regulation in steady state hematopoiesis in which one daughter cell of a division returns to its base primitive (stem/progenitor) cell phenotype, while the other daughter cell commits to a differentiation pathway or an apoptotic death.

If the phenotype of the stem cell reversibly modulates with cell cycle transit, with a predominantly progenitor phenotype at one point in the cycle and a predominantly engraftable multipotent stem cell phenotype at another point in the cycle, then estimates of long-term engraftable stem cell incidence in a cell population are probably underestimates. In essence, the identity of the stem cell would be masked at certain points in the cycle, and in a nonsynchronized population of cells, at any one point in time, a number of true stem cells would not be detectable. This could be termed the “masked” or “stealth” stem cell concept.

The present model builds upon a large base of previously accomplished work.81 It suggests that cell cycle transit, a fundamental characteristic of primitive hematopoietic stem cells, is associated with a continually changing stem cell phenotype and that, at least at the more primitive stem/progenitor cell levels, a fluctuating continuum, rather than a stem cell/progenitor hierarchy, may exist. Thus, outcomes would be determined by the changing cell cycle surface phenotype of the stem cell, that is, its receptor expression, and the delivered environmental stimuli, that is, fluid-phase or membrane-based cytokines, adhesion protein, or other ligands.

This provides, ultimately, a very flexible system for hematopoietic regulation, in which multiple different outcomes could occur sequentially, dependent on cell cycle phase and specific microenvironment. One can speculate that such flexibility might also hold for the recently described transdifferentiation of marrow stem cells to nonhematopoietic cells in different tissues, although at present there are no data addressing this point. This theory is presented in model form in Figure 7.

Fig. 7.

The chiaroscuro stem cell model.

As early marrow stem cells move through cell cycle transit, chromatin coverage changes, resulting in activation of different transcription regions and thus different gene expression. This change underlies the reversibly shifting phenotype of stem cells. Here stem cells are shown altering their phenotype from hematopoietic engraftable stem cell (S) to hematopoietic progenitor 1 (P1) to hematopoietic progenitor 2 (P2) with different phenotypes (eg, proteopodia) and back to hematopoietic engraftable stem cell. Cylinders represent DNA chromatin coverage; boxes, active transcription factors.

Fig. 7.

The chiaroscuro stem cell model.

As early marrow stem cells move through cell cycle transit, chromatin coverage changes, resulting in activation of different transcription regions and thus different gene expression. This change underlies the reversibly shifting phenotype of stem cells. Here stem cells are shown altering their phenotype from hematopoietic engraftable stem cell (S) to hematopoietic progenitor 1 (P1) to hematopoietic progenitor 2 (P2) with different phenotypes (eg, proteopodia) and back to hematopoietic engraftable stem cell. Cylinders represent DNA chromatin coverage; boxes, active transcription factors.

Close modal

We wish to thank Dr Abby Maizel for his careful critique and helpful suggestions.

Prepublished online as Blood First Edition Paper, August 22, 2002; DOI 10.1182/blood-2002-04-1246.

Supported by grants PO1 DK50222-02, PO1 HL56920-04, and RO1 DK27424-19.

1
Quesenberry
PJ
Hematopoiesis and hematopoietic growth factors.
Cecil Textbook of Medicine.
21st ed.
Goldman
L
Bennett
JC
1999
834
840
WB Saunders
Philadelphia, PA
2
Quesenberry
PJ
Colvin
GA
Hematopoietic stem cells, progenitor cells and cytokines.
Williams Hematology.
6th ed.
Beutler
E
Lichtman
MA
Coller
BS
Kipps
TJ
Seligsohn
U
2001
153
174
McGraw-Hill
New York, NY
3
Guenechea
G
Gan
OI
Dorrell
C
Dick
JE
Distinct classes of human stem cells that differ in proliferative and self-renewal potential.
Nat Immunol.
2
2001
75
82
4
Harrison
DE
Astle
CM
Short- and long-term multilineage repopulating hematopoietic stem cells in late fetal and newborn mice: models for human umbilical cord blood.
Blood.
90
1997
174
181
5
Akashi
K
Traver
D
Miyamoto
T
Weissman
IL
A clonogenic common myeloid progenitor that gives rise to all myeloid lineages.
Nature.
404
2000
193
197
6
Borge
OJ
Adolfsson
J
Jacobsen
AM
Lymphoid-restricted development from multipotent candidate murine stem cells: distinct and complimentary functions of the c-kit and flt3-ligands.
Blood.
94
1999
3781
3790
7
McNiece
IK
Almeida-Porada
G
Shpall
EJ
Zanjani
E
Ex vivo expanded cord blood cells provide rapid engraftment in fetal sheep but lack long-term engrafting potential.
Exp Hematol.
30
2002
612
616
8
Wiesmann
A
Kim
M
Georgeias
A
et al
Modulation of hematopoietic stem/progenitor cell engraftment by transforming growth factor beta.
Exp Hematol.
28
2000
128
139
9
Suda
T
Suda
J
Ogawa
M
Disparate differentiation in mouse hemopoietic colonies derived from paired progenitors.
Proc Natl Acad Sci U S A.
81
1984
2520
2524
10
Brady
G
Iscove
NN
Construction of cDNA libraries from single cells.
Methods Enzymol.
225
1993
611
623
11
Hodgson
GS
Bradley
TR
Properties of haemopoietic stem cells surviving 5-fluorouracil treatment: evidence for a pre-CFU-S cell?
Nature.
281
1979
381
382
12
Lerner
C
Harrison
DE
5-Fluouracil spares hemopoietic stem cells responsible for long-term repopulation.
Exp Hematol.
18
1990
114
118
13
Kay HEM. How many cell generations? Lancet. 1965:418.
14
Mintz
B
Anthony
K
Litwin
S
Monoclonal derivation of mouse myeloid and lymphoid lineages from totipotent hematopoietic stem cells experimentally engrafted in fetal hosts.
Proc Natl Acad Sci U S A.
81
1984
7835
7839
15
Lemischka
IR
Raulet
DH
Mulligan
RC
Developmental potential and dynamic behavior of hematopoietic stem cells.
Cell.
45
1986
917
927
16
Capel
B
Hawley
RG
Mintz
B
Long- and short-lived murine haematopoietic stem cell clones individually identified with retroviral integration markers.
Blood.
75
1990
2267
2270
17
Rosendaal
M
Hodgson
GS
Bradley
TR
Organisation of haematopoietic stem cells: the generation-age hypothesis.
Cell Tissue Kinet.
12
1979
17
29
18
Hodgson
GS
Bradley
TR
Radley
JM
The organization of hemopoietic tissue as inferred from the effects of 5-fluorouracil.
Exp Hematol.
10
1982
26
35
19
Harrison
DE
Lerner
C
Hoppe
PC
Carlson
GA
Alling
D
Large numbers of primitive stem cells are active simultaneously in aggregated embryo chimeric mice.
Blood.
69
1987
773
777
20
Micklem
HS
Lennon
JE
Ansell
JD
Gray
RA
Numbers and dispersion of repopulating hematopoietic cell clones in radiation chimeras as functions of injected cell dose.
Exp Hematol.
15
1987
251
257
21
Harrison
DE
Astle
CM
Lerner
C
Number and continuous proliferative pattern of transplanted primitive immunohematopoietic stem cells.
Proc Natl Acad Sci U S A.
85
1988
822
826
22
Bertoncello
I
Bradley
TR
Hodgson
GS
The concentration and resolution of primitive hemopoietic cells from normal mouse bone marrow by negative selection using monoclonal antibodies and dynabead monodisperse magnetic microspheres.
Exp Hematol.
17
1989
171
176
23
Mulder
AH
Viser
JWM
Separation and functional analysis of bone marrow cells separated by rhodamine 123 fluorescence.
Exp Hematol.
15
1987
99
104
24
Wolf
NS
Kone
A
Priestley
GV
Bartelmez
SH
In vivo and in vitro characterization of long-term repopulating primitive hematopoietic cells isolated by sequential Hoechst-33342-rhodamine 123 FACS selection.
Exp Hematol.
21
1993
614
622
25
Neben
S
Redfearn
WJ
Parra
M
Brecher
G
Pallavicini
MG
Short- and long-term repopulation of lethally irradiated mice by bone marrow stem cells enriched on the basis of light scatter and Hoechst fluorescence.
Exp Hematol.
19
1991
958
967
26
Nilsson
SK
Dooner
MS
Tiarks
CY
Weier
H
Quesenberry
PJ
Potential and distribution of transplanted hematopoietic stem cells in a nonablated mouse model.
Blood.
89
1997
4013
4020
27
Ploemacher
RE
Brons
NHC
Cells with marrow and spleen repopulating ability and forming spleen colonies on days 16, 12 and 8 are sequentially ordered on the basis of increasing rhodamine 123 retention.
J Cell Physiol.
136
1988
531
536
28
Spangrude
GJ
Heimfeld
S
Weissman
IL
Purification and characterization of mouse hematopoietic stem cells.
Science.
241
1988
58
62
29
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S
Miura
Y
Suda
T
In vivo and in vitro stem cell function of c-kit and Sca-1-positive murine hematopoietic cells.
Blood.
80
1992
3044
3050
30
Cheshier
SH
Morrison
SJ
Liao
X
Weissman
IL
In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells.
Proc Natl Acad Sci U S A.
96
1999
3120
3125
31
Goodell
MA
Brose
K
Paradis
G
Conner
AS
Mulligan
RC
Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
J Exp Med.
183
1996
1797
1806
32
Becker
AJ
McCulloch
EA
Siminovitch
L
Till
JE
The effect of differing demands for blood cell production on DNA synthesis by hemopoietic colony-forming cells of mice.
Blood.
26
1965
296
33
Vassort
F
Winterholer
M
Frindel
E
Tubiana
M
Kinetic parameters of bone marrow stem cells using in vivo suicide by tritiated thymidine or by hydroxyurea.
Blood.
41
1973
789
796
34
Patt
HM
Maloney
MA
Lamela
RA
Hematopoietic stem cell proliferative behavior as revealed by bromodeoxyuridine labeling.
Exp Hematol.
8
1980
1075
1079
35
Hagan
MP
MacVittie
TJ
CFUs kinetics observed in vivo by bromodeoxyuridine and near-UV light treatment.
Exp Hematol.
9
1981
123
128
36
Hodgson
GS
Bradley
TR
In vivo kinetic status of hematopoietic stem and progenitor cells as inferred from labeling with bromodeoxyuridine.
Exp Hematol.
12
1984
683
687
37
Pietrzyk
ME
Priestley
GV
Wolf
NS
Normal cycling patterns of hematopoietic stem cell subpopulations: an assay using long-term in vivo BrdU infusion.
Blood.
66
1985
1460
1462
38
Bradford
GB
Williams
B
Rossi
R
Bertoncello
I
Quiesence, cycling, and turnover in the primitive hematopoietic stem cell compartment.
Exp Hematol.
25
1997
445
453
39
Pang
L
Reddy
PV
Quesenberry
PJ
Are bone marrow stem cells quiescent? [abstract].
Exp Hematol.
27
1999
106
40
Nilsson
SK
Dooner
MS
Quesenberry
PJ
Synchronized cell-cycle induction of engrafting long-term repopulating stem cells.
Blood.
90
1997
4646
4650
41
Hendrikx
PJ
Martens
ACM
Hagenbeek
A
Keij
JF
Visser
JWM
Homing of fluorescently labeled hemopoietic stem cells.
Exp Hematol.
24
1996
129
140
42
Reddy
GPV
Tiarks
CY
Pang
L
Quesenberry
PJ
Synchronization and cell cycle analysis of pluripotent hematopoietic progenitor stem cells.
Blood.
90
1997
2293
2299
43
Abkowitz
JL
Galinelli
D
Harrison
DE
Gutturp
P
In vivo kinetics of murine hematopoietic stem cells.
Blood.
96
2000
3399
3405
44
Frimberger
A
McAuliffe
CI
Werme
KA
et al
The fleet feet of haematopoietic stem cells: rapid motility, interaction and proteopodia.
Br J Haematol.
112
2001
644
654
45
Phillips
RL
Ernst
RE
Brunk
B
et al
The genetic program of hematopoietic stem cells.
Science.
288
2000
1635
1640
46
Park
IK
He
Y
Lin
F
et al
Differential gene expression profiling of adult murine hematopoietic stem cells.
Blood.
99
2002
488
498
47
Prashar
Y
Weissman
SM
Analysis of differential gene expression by display of 3′ end restriction fragments of cDNAs.
Proc Natl Acad Sci U S A.
93
1996
659
663
48
Becker
PS
Nilsson
SK
Zhifang
L
et al
Adhesion receptor expression by hematopoietic cell lines and murine progenitors: modulation by cytokines and cell cycle status.
Exp Hematol.
27
1999
533
541
49
Berrios
VM
Dooner
GJ
Nowakowski
G
et al
The molecular basis for the cytokine-induced defect in homing and engraftment of hematopoietic stem cells.
Exp Hematol.
29
2001
1326
1335
50
Reddy
GP
McAuliffe
CI
Pang
L
Quesenberry
PJ
Bertoncello
I
Cytokine receptor repertoire and cytokine responsiveness of Hodull/Rhdull stem cells with differing potentials for G1/S phase progression.
Exp Hematol.
30
2002
792
800
51
Tada
T
Tada
M
Toti-/pluripotential stem cells and epigenetic modifications.
Cell Struct Funct.
3
2001
149
160
52
Forsberg
EC
Downs
KM
Christensen
HM
Im
H
Nuzzi
PA
Bresnick
EH
Developmentally dynamic histone acetylation pattern of a tissue-specific chromatin domain.
Proc Natl Acad Sci U S A.
97
2000
14494
14499
53
Miyata
Y
Towatari
M
Maeda
T
Ozawa
Y
Saito
H
Histone acetylation induced by granulocyte colony-stimulating factor in a map kinase-dependent manner.
Biochem Biophys Res Commun.
283
2001
655
660
54
Bonifer
C
Long-distance chromatin mechanisms controlling tissue-specific gene locus activation.
Gene.
2238
1999
277
289
55
Maes
J
O'Neill
LP
Cavelier
P
Turner
BM
Rougeon
F
Goodhardt
M
Chromatin remodeling at the Ig loci prior to V(D)J recombination.
J Immunol.
167
2001
866
874
56
Jimenez
G
Griffiths
SD
Ford
AM
Greaves
MF
Enver
T
Activation of the beta-globin locus control region precedes commitment to the erythroid lineage.
Proc Natl Acad Sci U S A.
89
1992
10618
10622
57
Kontaraki
J
Chen
HH
Riggs
A
Bonifer
C
Chromatin fine structure profiles for a developmentally regulated gene: reorganization of the lysozyme locus before transactivator binding and gene expression.
Genes Dev.
14
2000
2106
2122
58
Hu
M
Krause
D
Greaves
M
et al
Multilineage gene expression precedes commitment in the hemopoietic system.
Genes Dev.
11
1997
774
785
59
Ford
AM
Bennett
CA
Healy
LE
Navarro
E
Spooncer
E
Greaves
MF
Immunoglobulin heavy-chain and CD3 delta-chain gene enhancers are DNAse I-hypersensitive in hemopoietic progenitor cells.
Proc Natl Acad Sci U S A.
89
1992
3424
3428
60
McConnell
S
Kaznowski
C
Cell cycle dependence of laminar determination in developing neocortex.
Science.
254
1991
282
285
61
Cosma
M
Tanaka
T
Nasmyth
K
Ordered recruitment of transcription and chromatin remodeling factors to a cell cycle and developmentally regulated promoter.
Cell.
97
1999
299
311
62
Krebs
JE
Kuo
MH
Allis
CD
Peterson
CL
Cell cycle related histone acetylation required for expression of the yeast H0 gene.
Genes Dev.
13
1999
1412
1421
63
Peters
SO
Kittler
EL
Ramshaw
HS
Quesenberry
PJ
Murine marrow cells expanded in culture with IL-3, IL-6, IL-11, and SCF acquire an engraftment defect in normal hosts.
Exp Hematol.
23
1995
461
469
64
Peters
SO
Kittler
ELW
Ramshaw
HS
Quesenberry
PJ
Ex vivo expansion of murine marrow cells with interleukin-3, interleukin-6, interleukin-11, and stem cell factor leads to impaired engraftment in irradiated hosts.
Blood.
87
1996
30
37
65
Peters
SO
Habibian
HK
Vergilis
K
Quesenberry
PJ
Effects of cytokines on stem cell engraftment depends on time of evaluation post-marrow infusion.
Int J Hematol.
70
1999
112
118
66
Habibian
HK
Peters
SO
Hsieh
CC
et al
The fluctuating phenotype of the lymphohematopoietic stem cell with cell cycle transit.
J Exp Med.
188
1998
393
398
67
Fleming
WH
Alpern
EJ
Uchida
N
Ikuta
K
Spangrude
GJ
Weissman
IL
Functional heterogeneity is associated with the cell cycle status of murine marrow cells.
J Cell Biol.
122
1993
897
902
68
Orschell-Traycoff
CM
Hiatl
K
Dagher
RN
Rice
S
Yoder
MC
Srour
EF
Homing and engraftment potential of Sca-1+ lin- cells fractionated on the basis of adhesion molecule expression and position in cell cycle.
Blood.
96
2000
1380
1387
69
Gothot
A
Pyatt
R
McMahel
J
Rice
S
Srour
EF
Functional heterogeneity of human CD34 (+) cells isolated in subcompartments of the G0/G1 phase of cell cycle.
Blood.
90
1997
4384
4393
70
Gothot
A
van der loo
JC
Clapp
DW
Srour
EF
Cell cycle-related changes in repopulating capacity of human mobilized peripheral blood CD34 (+) cells in non-obese diabetic/severe combined immune-deficient mice.
Blood.
92
1998
2641
2649
71
Glimm
H
Il-Hoan
O
Eaves
CJ
Human hematopoietic stem cells stimulated to proliferate in vitro lose engraftment potential during their S/G2/M transit and do not re-enter G0.
Blood.
96
2000
4185
4193
72
Summers
YJ
Heyworth
CM
deWynter
EA
Testa
NG
Cord blood G(0) CD34+ cells have a thousand-fold higher capacity for generating progenitors in vitro than G(1) CD34+ cells.
Stem Cells.
19
2001
505
513
73
Szilvassy
SJ
Meyerrose
TE
Grimes
B
Effects of cell cycle activation on the short-term engraftment properties of ex-vivo expanded murine hematopoietic cells.
Blood.
95
2000
2829
2837
74
Huttman
A
Liu
SL
Boyd
AW
Li
CL
Functional heterogeneity within rhodamine 123 (lo) Hoechst 33342 (lo/sp) primitive stem cells revealed by pyronin Y.
Exp Hematol.
29
2001
1109
1116
75
Cerny J, Dooner M, McAuliffe C, et al. Homing of purified murine lymphohematopoietic stem cells: a cytokine-induced defect. J Hematother Stem Cell Res. In press.
76
Yamaguchi
M
Ikebuchi
K
Hirayama
F
et al
Different adhesive characteristics and VLA-4 expression of CD34 (+) progenitors in G0/G1 versus S/G2/M phases of cell cycle.
Blood.
92
1998
842
848
77
Lambert
JF
Liu
M
Baskaran
NV
et al
Purified hematopoietic stem cells show a major shift of gene expression through cell cycle that coincides with an engraftment nadir [abstract].
Exp Hematol.
30
2002
118
78
Colvin
GA
Lambert
JF
McAuliffe
CI
Quesenberry
PJ
Stem/progenitor cell inversions: the chiaroscuro stem cell model of hematopoiesis [abstract].
Exp Hematol.
30
2002
128
79
Knaan-Shanzer S. No hierarchy within the primitive hemopoietic stem cell compartment. Abstract presented at: The New Stem Cells; September 18-19, 2000; Leiden, The Netherlands.
80
Kirkland
M
Borokov
K
A phase space model of hemopoiesis [abstract].
Exp Hematol.
30
2002
125
81
Quesenberry
PJ
Colvin
GA
Lambert
JF
et al
The new stem cell biology.
Trans Am Clin Climatol Assoc.
113
2002
182
207

Author notes

Peter J. Quesenberry, Center for Stem Cell Biology, Roger Williams Medical Center, 825 Chalkstone Ave, Providence, RI 02908-4735; e-mail: pquesenberry@rwmc.org.

Sign in via your Institution