Skip to main content

Rb and hematopoiesis: stem cells to anemia

Abstract

The retinoblastoma protein, Rb, was one of the first tumor suppressor genes identified as a result of the familial syndrome retinoblastoma. In the period since its identification and cloning a large number of studies have described its role in various cellular processes. The application of conditional somatic mutation with lineage and temporally controlled gene deletion strategies, thus circumventing the lethality associated with germ-line deletion of Rb, have allowed for a reanalysis of the in vivo role of Rb. In the hematopoietic system, such approaches have led to new insights into stem cell biology and the role of the microenvironment in regulating hematopoietic stem cell fate. They have also clarified the role that Rb plays during erythropoiesis and defined a novel mechanism linking mitochondrial function to terminal cell cycle withdrawal. These studies have shed light on the in vivo role of Rb in the regulation of hematopoiesis and also prompt further analysis of the role that Rb plays in both the regulation of hematopoietic stem cells and the terminal differentiation of their progeny.

Introduction

Decisions to enter the cell cycle are regulated by the G1-S phase restriction point [1]. One of the major molecular circuits involved in this restriction point is centered on the retinoblastoma protein (Rb) and is termed the "Rb pathway" [2]. Through a series of sequential inhibitory phosphorylation events by cyclin dependent kinases (Cdk), Rb family members are inactivated and release E2F proteins which drive the cell through the G1 phase into S phase where cell division will occur independent of extracellular signals [3, 4]. Mutations of Rb itself and other components of the "Rb pathway" occur universally in human cancer, where differentiation is perturbed, and cancer initiating cells are thought to reacquire the capacity to self-renew [5, 6]. Rb has also been linked to the cell cycle arrest that accompanies terminal differentiation. As such the study of the role of these pathways in self-renewal will improve our understanding not only of the normal regulation of self-renewal but will also be applicable to understanding the initiation and maintenance of cancer.

Hematopoiesis represents an attractive system in which to study the role of the cell cycle in the regulation of cell fate. Hematopoiesis is hierachically structured, defined cell populations can be prospectively isolated and manipulated using flow cytometry and rigorous analysis of stem cell and lineage restricted progenitor function can be performed using transplantation assays. Coupling of the advances in in vivo genetic manipulation techniques, such as Cre-lox technologies, with the study of hematopoiesis and HSCs has led to rapid advances in our understanding of the programs involved in the maintenance of HSC self-renewal and differentiation and in the identification of transcription factors critical for lineage choice and identity [7–13]. Recent studies have sought to define the role that the cell cycle control machinery plays in the regulation of HSC fate and in the differentiation of specific hematopoietic lineages such as neutrophils and erythroid cells [14–17]. These studies, particularly those focused on the retinoblastoma protein (Rb), have revealed new roles for the cell cycle in the regulation of hematopoietic stem cells and in the coupling of cell cycle arrest to cellular differentiation. This review will focus on the role of Rb in HSC biology and in the terminal differentiation of the erythroid lineage.

A historical perspective on the role of Rb in hematopoiesis

Beginning with the initial descriptions of the germline knockout mice for the Rb gene in 1992, a role for Rb in hematopoiesis, and in particular erythropoiesis, was suspected. Rb deficient embyros had a profound anemia, proposed to lead to embryonic lethality, along with marked neurodevelopmental defects [18–20]. However upon further examination of chimeric mice it was noted that Rb-null cells could contribute to hematopoiesis and generate erythroid cells [21, 22]. Hematopoietic contribution in the chimeric animals was largely normal. These observations raised the possibility that the anemia in the Rb-null embryos resulted from non-cell autonomous defects.

The nature of the influence of other cell types, hematopoietic or otherwise, to the described phenotypes remained unclear and furthermore it was uncertain whether Rb had any intrinsic role in erythropoiesis. The former question was explored using several different approaches. One group found that the presence of a wild-type placenta in Rb-null embryos allowed their survival through the time of birth, although these animals did not appear to have completely normal development [23, 24]. Using in vitro culture approaches conflicting results regarding the requirement for Rb became apparent. Rb was found to be an intrinsic role in limiting the proliferative capacity of erythroid progenitors during terminal erythroid differentiation [25], or, alternatively, to be necessary in macrophages to properly support erythroid cells that develop in a niche interaction involving these two cell types, termed the "erythroid island" [26, 27]. More recent chimeric analysis suggested that Rb was necessary for in vivo stress responses and found a variety of variable phenotypes over time in hematopoiesis of chimeric animals [28]. Unfortunately, this work was limited because of the inability to distinguish cell-autonomous defects from a role in other cell types (such as macrophages) [28]. Additionally, some of this phenotypic variation may represent the presence of concomitant deletion of Rb in myeloid and niche cells causing a myeloproliferative-type disorder, as has been described [29].

The recent generation of conditional alleles of Rb and the application of Cre-lox technology has allow the development of complex in vivo models and a reanalysis of the role of Rb in a variety of biological processes. Utilizing conditional strategies, we and others have investigated the role that Rb plays in the regulation of hematopoietic stem cell biology and more recently sought to clarify the requirement for Rb during erythropoiesis. These models have revealed new insight not only into the biology of Rb and its role in hematopoiesis but also more broadly into the fundamental underlying processes that regulate hematopoietic stem cells and into the coupling of cell cycle exit to terminal differentiation.

Cell cycle regulation and hematopoietic stem cells

The continual production of blood cells is maintained by a small number of stem cells (HSCs), which reside in a specialized microenvironment in the adult BM, termed the niche [30–32]. The niche is a complex three dimensional system comprising cellular, extracellular and mineral components [33]. It is within the niche that HSCs divide and undertake cell fate decisions to constantly replenish hematopoiesis through the processes of differentiation, ultimately producing mature blood cells, and self-renewal which results in the production of more HSCs to replenish and maintain the HSC pool throughout life. The fine regulation of HSCs involves cell division coupled with appropriate intrinsic and extrinsic cues, the latter principally derived from the niche environment.

In steady state conditions HSCs are in a slowly dividing state, termed relative quiescence, with a cell division cycle in the mouse in the range of 2–4 wks [34, 35]. This is in contrast to the rapidly cycling hematopoietic progenitor cells, which are more committed to differentiation than HSCs. Engraftment of transplanted HSCs has been shown to be dependent on cell cycle status, with only cells in the non-cycling G0/G1 phase efficiently engrafting following transplantation and contributing to stable long-term hematopoiesis [36–39]. The slow cycling of HSCs acts to spare them from acute toxicity (such as chemotherapy), but may also prevent eradication of neoplastic cells [40, 41]. In part, the dramatic contrast in cell cycle status between stem and progenitor cells has led to the hypothesis that cell cycle regulation plays a fundamentally important role in stem cell fate determination. This hypothesis is supported by recent data demonstrating a slower rate of division in Hoxb4hiPbx1lo cells, which extensively self-renewal in vitro, compared to control cells [42]. It is essential for an HSC to undergo cell division if it is to self-renew, but how the cell division cycle is integrated into the process of self-renewal is unclear. It is also unknown as to whether cell cycle regulation represents an intrinsic or extrinsic modifier of HSC fate.

Negative regulators of both Cdk2 and Cdk4/6 activity, and therefore Rb function, have been demonstrated to have roles in regulating HSCs [43–47]. For the most part however these phenotypes have been relatively subtle, particularly when compared to hematopoietic phenotypes apparent after disruption of transcription factors such as C/EBPα [10] and Tel [9] amongst others, and are often apparent only after serial transplantation. The "Rb pathway" has also been implicated in phenotypes observed in both the Bmi1-/- and ATM-/- HSCs [48–50]. Rb was also described as being an important regulator of stem cell maintenance in the plant species Arabidopsis [51]. The interaction of cell cycle regulators with other factors such as Hoxb4 or telomerase deficiency has produced much more striking phenotypes than that observed for the cell cycle mutants in isolation [52, 53]. While clearly demonstrating that cell cycle modifiers have roles in regulating stem cells, particularly HSCs, the aforementioned studies have not been able to clearly discriminate between intrinsic or extrinsic contributions to HSC fate as all studies to date had utilized non-hematopoietic restricted mutant alleles. A recent study demonstrating that the p27Kip1-/-microenvironment mediates the myelo-lymphoid expansion observed in the p27Kip1-/-animals raises the possibility that the HSC expansion observed in p27Kip1-/-BM is extrinsic in nature [47, 54]. This result suggested that cell cycle regulators may play a role in regulating the competence of the hematopoietic niche, in addition to having potential intrinsic roles in HSC fate determination.

The hematopoietic stem cell niche

Recent studies have begun to characterize the nature of the adult BM niche [30–32, 55–57]. Two major cell types have been identified as being important components of the HSC niche, the bone-forming osteoblast and the blood vessel lining endothelial cell, although there is still debate as to the extent of the contribution of each of these cell types to the HSC niche. Studies have shown that extrinsic regulation of hematopoiesis and HSCs can occur via modulation of osteoblast number and function [56–58]. Endothelial cells have also recently been suggested to play a critical role as part of the HSC niche [55, 59]. A recent study suggested a common anatomical location for both osteoblast and endothelial cell types with respect to the niche, raising the possibility that they may collectively contribute to the function of the HSC niche [60]. Irrespective of the exact cellular composition of the niche, products of each of these cell types have been shown to have the potential to modulate HSC function. Additionally studies have shown that extrinsic regulation of homeostatic HSC numbers can be dominant to even very profound intrinsic cues in vivo [61, 62]. It is therefore of major importance to further understand the roles of the different cell types comprising the HSC niche (osteoblast, endothelial cell) and delineate their effects on HSC fate decisions. This includes defining the molecular regulators of the niche cells and exploring regulatory interactions between the hematopoietic cells and the non-hematopoietic derived microenvironment. Despite the recent advances, little is known about the molecular regulators of hematopoietic niche competence or the involvement of the niche in the initiation and maintenance of hematopoietic diseases.

A role for Rb in hematopoietic stem cell fate regulation

As a result of the embryonic lethality of Rb-deficient animals, somatic conditional inactivation or lineage restricted deletion of Rb is necessary to define its role in HSC fate. To enable analysis of the role of Rb specifically in HSCs, Rbfl/fl animals [63, 64] were crossed to the interferon inducible Mx1-Cre transgene [65]. Inducible somatic deletion in the adult has many advantages over non-inducible systems in the context of the analysis of HSCs, in particular the ability to transplant HSCs prior to gene inactivation which will restrict deletion to only the hematopoietic system. Using this approach we observed that HSC contribution to hematopoiesis was largely normal in the absence of Rb when the HSCs were supported by a wild-type microenvironment [66]. This result is consistent with that observed in chimeric Rb animals where hematopoietic development is essentially normal and Rb-deficient cells are capable of widespread contribution [21].

Whilst hematopoiesis was largely normal when Rb deficient cells were supported by a wild-type microenvironment a distinctly different phenotype was observed when Mx1-Cre Rbfl/fl animals were induced to delete Rb [29]. This experimental design does not restrict gene deletion to the hematopoietic cells, but also results in gene deletion in the hematopoietic microenvironment [56] and other organs of the animal [65]. These animals rapidly developed myeloproliferation, with a dramatic expansion of the numbers of neutrophils in the bone marrow and extramedullary erythro- and myelopoiesis in the spleen. Stem and primitive progenitor cells were also mobilized to the periphery and were found in the spleen and peripheral blood. This myeloproliferation was stable and present for the lifespan of the animals, which in our care was approximately 8 months. At this time the animals present with a phenotype consistent with hematopoietic failure with a hypocellular bone marrow filled with mature neutrophils and a drastic reduction in splenic hematopoiesis, however the animals also developed pituitary tumors. This striking and profound phenotype was not present in a wild-type microenvironment.

When bone marrow HSC frequency was determined in these animals it was found to be reduced 5 fold with a concomitant increase at extramedullary sites, suggesting an overall redistribution of cells away from the bone marrow environment. The HSCs obtained from the bone marrow were able to reconstitute hematopoiesis in wild-type recipients and could be serially transplanted suggesting that when supported by a wild-type microenvironment the HSC itself was able to self-renew and differentiate relatively normally. Whilst the HSCs were functionally normal they were failing to be retained in the bone marrow microenvironment, placing Rb as an extrinsic regulator of HSC fate. These studies were supportive of the interpretation that stem cell self-renewal can occur independently of Rb, consistent with that reported from the analysis of embryonic stem cell self-renewal and differentiative cell cycles [67, 68]. This observation is intriguing in light of the reacquisition of self-renewal potential during tumorigenesis. Human tumors are thought to near universally inactivate the "Rb pathway", and whilst it can not be assumed that the consequences of the mutations of various components of this pathway are equivalent, it does raise the possibility that inactivation of this pathway may facilitate reacquisition of the self-renewal program [5].

The failure to recapitulate myeloproliferation from Rb-deficient hematopoietic cells in a wild-type microenvironment implied a role for the microenvironment in its development. An Rb-deficient microenvironment did not cause myeloproliferation of wild-type hematopoietic cells, contrasting with that observed in the case of an RARγ-deficient microenvironment which was the sole cause of myeloproliferation in this model [69]. In Rb-deficient myeloproliferative animals a significant increase in ostecoclast number could be observed which correlated with a rapid loss of bone architecture and trabecular volume, both factors that have been implicated in the regulation of HSCs [56, 57, 70]. Based on these findings, we sought to determine if myeloid cells were required for the development of the myeloproliferation. Deletion of Rb from myeloid cells using Lysozyme-M-Cre did not result in myeloproliferation, but when combined with an Rb-deficient microenvironment a fatal myeloproliferation rapidly ensued [29]. This result demonstrated that interactions between hematopoietic cells and non-hematopoietic stromal elements could result in the development of myeloproliferation and additionally modulate HSC fate within the bone marrow microenvironment. The nature of the non-hematopoietic cell or cells responsible for this interaction are currently under investigation. Interestingly, studies using Vav-Cre to delete Rb reported a similar phenotype, although not as severe, to that we had observed with the Mx1-Cre based deletion of Rb [71]. Vav-Cre is known excise in both hematopoietic and vascular lineages, which directs attention to the role of the vasculature in the myeloproliferative phenotypes that were observed [72–74].

Several differences are observed between the data derived from Vav-Cre and Mx1-Cre mediated deletion of Rb in hematopoietic cells and HSCs. Daria et al observed a requirement for Rb in the stress response of HSCs and this has also previously been suggested in the context of the role of Rb in erythropoiesis [28, 75]. The timing of gene deletion is also relevant for interpreting the phenotypes observed in these two models. Whilst with Mx1-Cre, gene deletion is largely temporally controlled and can be restricted to the adult HSC, Vav-Cre is active from the genesis of HSCs. Mx1-Cre could also be restricted to the HSC and subsequent hematopoiesis through transplantation prior to deletion of Rb where as deletion of Rb with Vav occurs in utero in both HSCs and vasculature potentially disturbing the microenvironment in which the HSCs reside and expand during development prior to the shift in hematopoiesis to the intramedullary sites of bone [76, 77]. Also of note is that the cell division dynamics of HSCs change during development, from rapidly cycling and dividing cells during the fetal liver and early stages of life to relatively quiescent and more slowly cycling in the adult context [34, 35, 38, 78, 79]. Thus the role for Rb may be context dependent, both in terms of stress response and developmentally in the regulation of HSC fate.

Red blood cells, anemia and Rb

Cell cycle regulation in erythropoiesis

Each day an average adult human produces nearly 200 billion RBCs. To maintain the effective production of RBCs, a rapid proliferative expansion of early progenitors needs to occur. This expansion is followed by termination of proliferation and commencement of the complex biogenic program allowing production of the hemoglobin necessary for the oxygen-carrying capability of the RBCs. If any step in this process is disrupted, as occurs in numerous human diseases, then anemia results [80]. Modulation of erythroid cell cycle regulation has been exploited therapeutically, particularly in patients with diseases due to defects in hemoglobin structure or production, but more efficacious therapies will depend on an increased understanding of this process [81–83].

The variation in cellular proliferation during erythropoiesis has been well-studied at a descriptive level over many decades and the stages at which alterations in this process occur have been characterized [80, 84–86]. However, an in-depth understanding of the molecular control of this process is largely lacking. It is known that early bipotential megakaryocyte-erythroid progenitors (MEPs) and early erythroid progenitors (BFU-Es) proliferate at a relatively slow rate, but that this level of cell cycle progression is necessary to maintain the pool of more differentiated precursors. This has been demonstrated through the study of mutations that disrupt the activity of the c-Myb gene, suggesting a critical role for cell cycle regulation at these early stages of differentiation [87–89].

Following these early stages, a rapid rise in proliferation is observed at the colony-forming unit erythroid (CFU-E) stage of differentiation. After the CFU-E stage, the erythroid progenitors undergo three to five additional cell divisions whilst maturing into erythroblasts that then need to undergo terminal cell cycle exit at the G1 phase to facilitate complete differentiation. Ultimately the post-mitotic cells undergo further maturation and eventually enucleate to give rise to the functional RBCs that can enter the circulation and play a critical role in oxygen transport. Our understanding of how cell cycle exit is carried out during erythroblast maturation and how this is coupled to differentiation is limited. Studies in erythroid cell lines have suggested some molecular players involved in this process [90], but our understanding of the control of this process in vivo is incomplete. Since it is known that G1 exit frequently requires the activity of Rb to occur in other cell types [63, 91], it is important to understand the role that this gene plays in the process of erythropoiesis.

The role of Rb in erythropoiesis

In order to directly examine whether Rb had any intrinsic or potentially extrinsic role in erythropoiesis, we utilized conditional deletion of the gene in a variety of hematopoietic lineages [92]. We were able to delete Rb specifically within the erythroid lineage using the erythropoietin receptor knock-in GFPcre mouse line (EpoR-GFPcre) [93, 94]. We could also delete Rb within the macrophage and granulocyte lineage using Lysozyme-M-Cre [95] and somatically within the entire adult hematopoietic system using Mx1-Cre [65]. The results of this analysis indicated that Rb was necessary within the erythroid cells for normal erythropoiesis, but was dispensible in macrophages. This finding suggests that the use of in vitro reconstituted erythroid islands may not faithfully mimic physiological situations with the culture conditions used in these experiments [27]. Alternatively, it is possible that there may be compensation for the loss of Rb in macrophages in vivo.

Mice harboring an Rb deletion within the erythroid compartment showed a moderate anemia that remained stable throughout the life of the animals. After examining the etiology of this anemia, we found that it was caused by an impaired maturation of precursors within the bone marrow and spleen. There was an expansion of early erythroblasts, but these cells failed to efficiently mature. It was apparent that a failure to differentiate, termed ineffective erythropoiesis, occurred at the stage where cell cycle exit normally occurs. Using phenotypically stage-matched erythroid precursors from mutant animals and controls, we examined pathways that were either globally up- or downregulated to gain a better understanding of how this block in differentiation may occur and what the contribution of Rb was to the coupling of cell cycle exit to differentiation during erythropoiesis [96, 97].

Consistent with the role of Rb in cell cycle exit, there was a failure to repress S-phase genes and particularly E2F transcription factor targets in the Rb-null erythrocytes. Surprisingly the most downregulated gene sets were all components of the mitochondrial electron transport chain and oxidative phosphorylation (OXPHOS) pathways. This result suggested a link between cell cycle regulation and mitochondrial biogenesis in erythroid cells. In agreement with the gene expression data there was both a reduced mitochondrial mass and mitochondrial DNA content in the Rb-null erythroid cells. There are numerous examples demonstrating how primary defects in mitochondrial function or biogenesis can lead to ineffective erythropoiesis in both humans [98–100] and experimental animals [101–103]. Our studies using lineage restricted gene deletion to allow in vivo studies allowed us to find a previously unappreciated link between cell cycle regulation and modulation of mitochondrial function during cellular differentiation, which appears critical at the mid-maturation erythroblast stage. Our initial work suggested that this phenomenon was likely to be mediated by modulation of the PPARγ-coactivator (PGC) transcriptional axis. In particular, we obtained some evidence to suggest that reduced expression of PGC-1β may play a role in this phenotype. Much work still remains to be done to further characterize the link that was observed between cell cycle regulation and mitochondrial biogenesis. Similar observations have been made in the context of proliferating fibroblasts, where it was suggested that the molecular control of this process occurs through modulation of transcription factors known to interact with the PGC family of coactivators [104].

The phenotype observed in the erythroid Rb-null mice closely resembled the ineffective erythropoiesis that is seen in human myelodysplastic syndrome (MDS). It is interesting to note that defects in mitochondrial structure and function have frequently been seen in the erythroid cells in MDS [98, 99]. Concomitantly, cell cycle deregulation is thought to underlie the pathophysiology of MDS and the most frequently identified molecular defect in human MDS involves epigenetic silencing of the cell cycle inhibitory protein CDKN2B/p15INK4B [105, 106]. Additionally, the anemia present in the Rb-null mice cannot be corrected even in the presence of high-level wild-type hematopoietic chimerism, suggesting that this may be the type of lesion that could allow a clonal disease like MDS to result in an anemia [105]. Our findings suggest a potential link between these observations that may lead to more effectively targeted therapies. If these features of MDS are linked together, then it may be useful to target both of these lesions simultaneously to strike at the "Achilles' heel" of MDS. The insights gained from this work may also suggest candidate genes that could be involved in the pathogenesis of MDS.

Outstanding questions regarding Rb and erythropoiesis

We have been able to uncover an interesting nexus between cell cycle exit during erythropoiesis and the regulation of mitochondrial biogenesis, which is mediated by Rb. This insight allows us to gain a greater understanding of normal erythropoiesis. Additionally, it is likely that this work will allow us to gain insight into the pathogenesis of MDS and possibly other disease states where ineffective erythropoiesis occurs. Importantly, while cell cycle exit is impaired in the Rb-null erythroid cells, cell cycle exit continues to occur to some extent in these cells and a significant proportion of the erythrocytes mature and are functional in the peripheral blood. It will be important to delineate the factors that are responsible for this continued ability to exit the cell cycle in the absence of Rb. Moreover, while the aforementioned studies have largely focused on homeostatic adult erythropoiesis, it will be important to delineate whether the "Rb pathway" functions differently during the extensive expansion in red cell mass that occurs in the course of ontogeny [107]. It will also be interesting to examine how the genes that play a role in promoting the increased proliferation during the early stages of erythropoiesis are coupled to differentiation. For example, it is likely that cyclins D2 and D3 are coupled to the differentiation of these progenitors to allow coordinated proliferation and maturation of these cells [15]. It is clear that alterations in the differentiation kinetics of RBC progenitors can impact how differentiation occurs, as exemplified by alterations in globin gene expression resulting from treatment with S-phase inhibitors ([81]. We only have a descriptive understanding of these phenomena currently, but it is likely that further molecular links similar to those we have described are playing a critical role here. Recent evidence from human genetic studies indicates that genes like c-Myb may have an important impact on the differentiation characteristics of these cells [108–110]. The further study of cell cycle regulation in the seemingly "well understood" differentiation model of erythropoiesis is likely to yield many new and fruitful insights into the general molecular networks that coordinate differentiation and proliferation.

Conclusion

The use of lineage and temporally controlled somatic deletion strategies has allowed the development of complex in vivo models with which to study the roles of genes in both development and organ homeostasis. Questions previously unable to be studied regarding the role of Rb in the context of the adult HSCs and hematopoiesis can now begin to be addressed. Whilst helping to clarify previous ambiguity regarding the phenotype of loss of Rb, these studies have also revealed previously unrecognized roles for Rb in the regulation of HSCs and their microenvironment and in the regulation of mitochondrial function during terminal erythropoiesis.

References

  1. Sherr CJ: G1 phase progression: cycling on cue. Cell 1994, 79: 551–555. 10.1016/0092-8674(94)90540-1

    Article  CAS  PubMed  Google Scholar 

  2. Weinberg RA: The retinoblastoma protein and cell cycle control. Cell 1995, 81: 323–330. 10.1016/0092-8674(95)90385-2

    Article  CAS  PubMed  Google Scholar 

  3. Malumbres M, Barbacid M: To cycle or not to cycle: a critical decision in cancer. Nature Rev Cancer 2001, 1: 222–231. 10.1038/35106065

    Article  CAS  Google Scholar 

  4. Sherr CJ, Roberts JM: Living with or without cyclins and cyclin-dependent kinases. Genes Dev 2004, 18: 2699–2711. 10.1101/gad.1256504

    Article  CAS  PubMed  Google Scholar 

  5. Hanahan D, Weinberg RA: The hallmarks of cancer. Cell 2000, 100: 57–70. 10.1016/S0092-8674(00)81683-9

    Article  CAS  PubMed  Google Scholar 

  6. Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, Levine JE, Wang J, Hahn WC, Gilliland DG, Golub TR, Armstrong SA: Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 2006, 442: 818–822. 10.1038/nature04980

    Article  CAS  PubMed  Google Scholar 

  7. Mikkola HK, Klintman J, Yang H, Hock H, Schlaeger TM, Fujiwara Y, Orkin SH: Haematopoietic stem cells retain long-term repopulating activity and multipotency in the absence of stem-cell leukaemia SCL/tal-1 gene. Nature 2003, 421: 547–551. 10.1038/nature01345

    Article  CAS  PubMed  Google Scholar 

  8. Hock H, Hamblen MJ, Rooke HM, Schindler JW, Saleque S, Fujiwara Y, Orkin SH: Gfi-1 restricts proliferation and preserves functional integrity of haematopoietic stem cells. Nature 2004, 431: 1002–1007. 10.1038/nature02994

    Article  CAS  PubMed  Google Scholar 

  9. Hock H, Meade E, Medeiros S, Schindler JW, Valk PJ, Fujiwara Y, Orkin SH: Tel/Etv6 is an essential and selective regulator of adult hematopoietic stem cell survival. Genes Dev 2004, 18: 2336–2341. 10.1101/gad.1239604

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  10. Zhang P, Iwasaki-Arai J, Iwasaki H, Fenyus ML, Dayaram T, Owens BM, Shigematsu H, Levantini E, Huettner CS, Lekstrom-Himes JA, Akashi K, Tenen DG: Enhancement of hematopoietic stem cell repopulating capacity and self-renewal in the absence of the transcription factor C/EBP alpha. Immunity 2004, 21: 853–863. 10.1016/j.immuni.2004.11.006

    Article  CAS  PubMed  Google Scholar 

  11. Scott EW, Simon MC, Anastasi J, Singh H: Requirement of transcription factor PU.1 in the development of multiple hematopoietic lineages. Science 1994, 265: 1573–1577. 10.1126/science.8079170

    Article  CAS  PubMed  Google Scholar 

  12. Xie H, Ye M, Feng R, Graf T: Stepwise reprogramming of B cells into macrophages. Cell 2004, 117: 663–676. 10.1016/S0092-8674(04)00419-2

    Article  CAS  PubMed  Google Scholar 

  13. Nutt SL, Heavey B, Rolink AG, Busslinger M: Commitment to the B-lymphoid lineage depends on the transcription factor Pax5. Nature 1999, 401: 556–562. 10.1038/44076

    Article  CAS  PubMed  Google Scholar 

  14. Orkin SH: Diversification of haematopoietic stem cells to specific lineages. Nat Rev Genet 2000, 1: 57–64. 10.1038/35049577

    Article  CAS  PubMed  Google Scholar 

  15. Kozar K, Ciemerych MA, Rebel VI, Shigematsu H, Zagozdzon A, Sicinska E, Geng Y, Yu Q, Bhattacharya S, Bronson RT, Akashi K, Sicinski P: Mouse development and cell proliferation in the absence of D-cyclins. Cell 2004, 118: 477–491. 10.1016/j.cell.2004.07.025

    Article  CAS  PubMed  Google Scholar 

  16. Ciemerych MA, Kenney AM, Sicinska E, Kalaszczynska I, Bronson RT, Rowitch DH, Gardner H, Sicinski P: Development of mice expressing a single D-type cyclin. Genes Dev 2002, 16: 3277–3289. 10.1101/gad.1023602

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. McArthur GA, Foley KP, Fero ML, Walkley CR, Deans AJ, Roberts JM, Eisenman RN: MAD1 and p27(KIP1) cooperate to promote terminal differentiation of granulocytes and to inhibit Myc expression and cyclin E-CDK2 activity. Mol Cell Biol 2002, 22: 3014–3023. 10.1128/MCB.22.9.3014-3023.2002

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Lee EY, Chang CY, Hu N, Wang YC, Lai CC, Herrup K, Lee WH, Bradley A: Mice deficient for Rb are nonviable and show defects in neurogenesis and haematopoiesis. Nature 1992, 359: 288–294. 10.1038/359288a0

    Article  CAS  PubMed  Google Scholar 

  19. Clarke AR, Maandag ER, van RM, Lugt Nm, Valk M, Hooper ML, Berns A, te Riele H: Requirement for a functional Rb-1 gene in murine development. Nature 1992, 359: 328–330. 10.1038/359328a0

    Article  CAS  PubMed  Google Scholar 

  20. Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA, Weinberg RA: Effects of an Rb mutation in the mouse. Nature 1992, 359: 295–300. 10.1038/359295a0

    Article  CAS  PubMed  Google Scholar 

  21. Williams BO, Schmitt EM, Remington L, Bronson RT, Albert DM, Weinberg RA, Jacks T: Extensive contribution of Rb-deficient cells to adult chimeric mice with limited histopathological consequences. Embo J 1994, 13: 4251–4259.

    CAS  PubMed Central  PubMed  Google Scholar 

  22. Maandag EC, Valk M, Vlaar M, Feltkamp C, O'Brien J, van Roon M, Lugt N, Berns A, te Riele H: Developmental rescue of an embryonic-lethal mutation in the retinoblastoma gene in chimeric mice. Embo J 1994, 13: 4260–4268.

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Wu L, de Bruin A, Saavedra HI, Starovic M, Trimboli A, Yang Y, Opavska J, Wilson P, Thompson JC, Ostrowski MC, Rosol TJ, Woollett LA, Weinstein M, Cross JC, Robinson ML, Leone G: Extra-embryonic function of Rb is essential for embryonic development and viability. Nature 2003, 421: 942–947. 10.1038/nature01417

    Article  CAS  PubMed  Google Scholar 

  24. Wenzel PL, Wu L, de Bruin A, Chong JL, Chen WY, Dureska G, Sites E, Pan T, Sharma A, Huang K, Ridgway R, Mosaliganti K, Sharp R, Machiraju R, Saltz J, Yamamoto H, Cross JC, Robinson ML, Leone G: Rb is critical in a mammalian tissue stem cell population. Genes Dev 2007, 21: 85–97. 10.1101/gad.1485307

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Clark AJ, Doyle KM, Humbert PO: Cell-intrinsic requirement for pRb in erythropoiesis. Blood 2004, 104: 1324–1326. 10.1182/blood-2004-02-0618

    Article  CAS  PubMed  Google Scholar 

  26. Chasis JA: Erythroblastic islands: specialized microenvironmental niches for erythropoiesis. Curr Opin Hematol 2006, 13: 137–141. 10.1097/01.moh.0000219657.57915.30

    Article  PubMed  Google Scholar 

  27. Iavarone A, King ER, Dai XM, Leone G, Stanley ER, Lasorella A: Retinoblastoma promotes definitive erythropoiesis by repressing Id2 in fetal liver macrophages. Nature 2004, 432: 1040–1045. 10.1038/nature03068

    Article  CAS  PubMed  Google Scholar 

  28. Spike BT, Dirlam A, Dibling BC, Marvin J, Williams BO, Jacks T, Macleod KF: The Rb tumor suppressor is required for stress erythropoiesis. Embo J 2004, 23: 4319–4329. 10.1038/sj.emboj.7600432

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Walkley CR, Shea JM, Sims NA, Purton LE, Orkin SH: Rb Regulates Interactions between Hematopoietic Stem Cells and Their Bone Marrow Microenvironment. Cell 2007, 129: 1081–1095. 10.1016/j.cell.2007.03.055

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Schofield R: The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 1978, 4: 7–25.

    CAS  PubMed  Google Scholar 

  31. Trentin JJ: Hemopoietic microenvironments. Transplant Proc 1978, 10: 77–82.

    CAS  PubMed  Google Scholar 

  32. Wolf NS, Trentin JJ: Hemopoietic colony studies. V. Effect of hemopoietic organ stroma on differentiation of pluripotent stem cells. J Exp Med 1968, 127: 205–214. 10.1084/jem.127.1.205

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Adams GB, Scadden DT: The hematopoietic stem cell in its place. Nat Immunol 2006, 7: 333–337. 10.1038/ni1331

    Article  CAS  PubMed  Google Scholar 

  34. Bradford GB, Williams B, Rossi R, Bertoncello I: Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp Hematol 1997, 25: 445–453.

    CAS  PubMed  Google Scholar 

  35. Kiel MJ, He S, Ashkenazi R, Gentry SN, Teta M, Kushner JA, Jackson TL, Morrison SJ: Haematopoietic stem cells do not asymmetrically segregate chromosomes or retain BrdU. Nature 2007, 449: 238–242. 10.1038/nature06115

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. 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 the cell cycle. Blood 1997, 90: 4384–4393.

    CAS  PubMed  Google Scholar 

  37. Gothot A, 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 1998, 92: 2641–2649.

    CAS  PubMed  Google Scholar 

  38. Bowie MB, McKnight KD, Kent DG, McCaffrey L, Hoodless PA, Eaves CJ: Hematopoietic stem cells proliferate until after birth and show a reversible phase-specific engraftment defect. J Clin Invest 2006, 116: 2808–2816. 10.1172/JCI28310

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Passegue E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL: Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J Exp Med 2005, 202: 1599–1611. 10.1084/jem.20050967

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Lerner C, Harrison DE: 5-Fluorouracil spares hemopoietic stem cells responsible for long-term repopulation. Exp Hematol 1990, 18: 114–118.

    CAS  PubMed  Google Scholar 

  41. Hodgson GS, Bradley TR: Properties of haematopoietic stem cells surviving 5-fluorouracil treatment: evidence for a pre-CFU-S cell? Nature 1979, 281: 381–382. 10.1038/281381a0

    Article  CAS  PubMed  Google Scholar 

  42. Cellot S, Krosl J, Chagraoui J, Meloche S, Humphries RK, Sauvageau G: Sustained in vitro trigger of self-renewal divisions in Hoxb4hiPbx1(10) hematopoietic stem cells. Exp Hematol 2007, 35: 802–816. 10.1016/j.exphem.2007.02.013

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  43. Cheng T, Rodrigues N, Shen H, Yang Y, Dombkowski D, Sykes M, Scadden DT: Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 2000, 287: 1804–1808. 10.1126/science.287.5459.1804

    Article  CAS  PubMed  Google Scholar 

  44. van Os R, Kamminga LM, Ausema A, Bystrykh LV, Draijer DP, van Pelt K, Dontje B, de Haan G: A limited role for p21cip1/waf1 in maintaining normal hematopoietic stem cell functioning. Stem Cells 2006.

    Google Scholar 

  45. Stepanova L, Sorrentino BP: A limited role for p16Ink4a and p19Arf in the loss of hematopoietic stem cells during proliferative stress. Blood 2005, 106: 827–832. 10.1182/blood-2004-06-2242

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Janzen V, Forkert R, Fleming HE, Saito Y, Waring MT, Dombkowski DM, Cheng T, DePinho RA, Sharpless NE, Scadden DT: Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 2006, 443: 421–426.

    CAS  PubMed  Google Scholar 

  47. Walkley CR, Fero ML, Chien WM, Purton LE, McArthur GA: Negative cell-cycle regulators cooperatively control self-renewal and differentiation of haematopoietic stem cells. Nat Cell Biol 2005, 7: 172–178. 10.1038/ncb1214

    Article  CAS  PubMed  Google Scholar 

  48. Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, Morrison SJ, Clarke MF: Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 2003, 423: 302–305. 10.1038/nature01587

    Article  CAS  PubMed  Google Scholar 

  49. Lessard J, Sauvageau G: Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature 2003, 423: 255–260. 10.1038/nature01572

    Article  CAS  PubMed  Google Scholar 

  50. Ito K, Hirao A, Arai F, Matsuoka S, Takubo K, Hamaguchi I, Nomiyama K, Hosokawa K, Sakurada K, Nakagata N, Ikeda Y, Mak TW, Suda T: Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 2004, 431: 997–1002. 10.1038/nature02989

    Article  CAS  PubMed  Google Scholar 

  51. Wildwater M, Campilho A, Perez-Perez JM, Heidstra R, Blilou I, Korthout H, Chatterjee J, Mariconti L, Gruissem W, Scheres B: The RETINOBLASTOMA-RELATES Gene Regulates Stem Cell Maintenance in Arabidopsis Roots. Cell 2005, 123: 1337–1349. 10.1016/j.cell.2005.09.042

    Article  CAS  PubMed  Google Scholar 

  52. Miyake N, Brun AC, Magnusson M, Miyake K, Scadden DT, Karlsson S: HOXB4-induced self-renewal of hematopoietic stem cells is significantly enhanced by p21 deficiency. Stem Cells 2006, 24: 653–661. 10.1634/stemcells.2005-0328

    Article  CAS  PubMed  Google Scholar 

  53. Choudhury AR, Ju Z, Djojosubroto MW, Schienke A, Lechel A, Schaetzlein S, Jiang H, Stepczynska A, Wang C, Buer J, Lee HW, von Zglinicki T, Ganser A, Schirmacher P, Nakauchi H, Rudolph KL: Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat Genet 2007, 39: 99–105. 10.1038/ng1937

    Article  CAS  PubMed  Google Scholar 

  54. Chien WM, Rabin S, Macias E, Miliani de Marval PL, Garrison K, Orthel J, Rodriguez-Puebla M, Fero ML: Genetic mosaics reveal both cell-autonomous and cell-nonautonomous function of murine p27Kip1. Proc Natl Acad Sci USA 2006, 103: 4122–4127. 10.1073/pnas.0509514103

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  55. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ: SLAM family receptors distinguish hematopoietic stem and progrnitor cells and reveal endothelial niches for stem cells. Cell 2005, 121: 1109–1121. 10.1016/j.cell.2005.05.026

    Article  CAS  PubMed  Google Scholar 

  56. Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, Ross J, Haug J, Johnson T, Feng JQ, Harris S, Wiedemann LM, Mishina Y, Li L: Identification of the haematopoietic stem cell niche and control of the niche size. Nature 2003, 425: 836–841. 10.1038/nature02041

    Article  CAS  PubMed  Google Scholar 

  57. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT: Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 2003, 425: 841–846. 10.1038/nature02040

    Article  CAS  PubMed  Google Scholar 

  58. Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL: Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood 2004, 103: 3258–3264. 10.1182/blood-2003-11-4011

    Article  CAS  PubMed  Google Scholar 

  59. Sugiyama T, Kohara H, Noda M, Nagasawa T: Maintenance of the Hematopoietic Stem Cell Pool by CXCL12-CXCR4 Chemokine Signalling in Bone Marrow Stromal Cell Niches. Immunity 2006, 25: 977–988. 10.1016/j.immuni.2006.10.016

    Article  CAS  PubMed  Google Scholar 

  60. Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, Tagliafico E, Ferrari S, Robey PG, Riminucci M, Bianco P: Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007, 131: 324–336. 10.1016/j.cell.2007.08.025

    Article  CAS  PubMed  Google Scholar 

  61. Krosl J, Beslu N, Mayotte N, Humphries RK, Sauvageau G: The competitive nature of HOXB4-transduced HSC is limited by PBX1: the generation of ultra-competitive stem cells retaining full differentiation potential. Immunity 2003, 18: 561–571. 10.1016/S1074-7613(03)00090-6

    Article  CAS  PubMed  Google Scholar 

  62. Antonchuk J, Sauvageau G, Humphries RK: HOXB4-induced expansion of adult hematopoietic stem cells ex vivo. Cell 2002, 109: 39–45. 10.1016/S0092-8674(02)00697-9

    Article  CAS  PubMed  Google Scholar 

  63. Sage J, Miller AL, Perez-Mancera PA, Wysocki JM, Jacks T: Acute mutation of retinoblastoma is sufficient for cell cycle re-entry. Nature 2003, 424: 223–228. 10.1038/nature01764

    Article  CAS  PubMed  Google Scholar 

  64. MacPherson D, Sage J, Crowley D, Trumpp A, Bronson RT, Jacks T: Conditional mutation of Rb causes cell cycle defects without apoptosis in the central nervous system. Mol Cell Biol 2003, 23: 1044–1053. 10.1128/MCB.23.3.1044-1053.2003

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  65. Kuhn R, Schwenk F, Aguet M, Rajewsky K: Inducible gene targeting in mice. Science 1995, 269: 1427–1429. 10.1126/science.7660125

    Article  CAS  PubMed  Google Scholar 

  66. Walkley CR, Orkin SH: Rb is dispensable for self-renewal and multilineage differentiation of adult hematopoietic stem cells. Proc Natl Acad Sci USA 2006, 103: 9057–9062. 10.1073/pnas.0603389103

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  67. Stead E, White J, Faast R, Conn S, Goldstone S, Rathjen J, Dhingra U, Rathjen P, Walker D, Dalton S: Pluripotent cell division cycles are driven by ectopic Cdk2, cyclin A/E and E2F activities. Oncogene 2002, 21: 8320–8333. 10.1038/sj.onc.1206015

    Article  CAS  PubMed  Google Scholar 

  68. White J, Stead E, Faast R, Conn S, Cartwright P, Dalton S: Developmental activation of the Rb-E2F pathway and establishment of cell cycle-regulated cyclin-dependent kinase activity during embryonic stem cell differentiation. Mol Biol Cell 2005, 16: 2018–2027. 10.1091/mbc.E04-12-1056

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  69. Walkley CR, Olsen GH, Dworkin S, Fabb SA, Swann J, McArthur GA, Westmoreland SV, Chambon P, Scadden DT, Purton LE: A Microenvironment-Induced Myeloproliferative Syndrome Caused by Retinoic Acid Receptor gamma Deficiency. Cell 2007, 129: 1097–1110. 10.1016/j.cell.2007.05.014

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  70. Kollet O, Dar A, Shivtiel S, Kalinkovich A, Lapid K, Sztainberg Y, Tesio M, Samstein RM, Goichberg P, Spiegel A, Elson A, Lapidot T: Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med 2006, 12: 657–664. 10.1038/nm1417

    Article  CAS  PubMed  Google Scholar 

  71. Daria D, Filippi MD, Knudsen ES, Faccio R, Li Z, Kalfa T, Geiger H: The retinoblastoma tumor suppressor is a critical intrinsic regulator for hematopoietic stem and progenitor cells under stress. Blood 2008, 111: 1894–1902. 10.1182/blood-2007-02-071746

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Ogilvy S, Elefanty AG, Visvader J, Bath ML, Harris AW, Adams JM: Transcriptional regulation of vav, a gene expressed throughout the hematopoietic compartment. Blood 1998, 91: 419–430.

    CAS  PubMed  Google Scholar 

  73. Georgiades P, Ogilvy S, Duval H, Licence DR, Charnock-Jones DS, Smith SK, Print CG: VavCre transgenic mice: a tool for mutagenesis in hematopoietic and endothelial lineages. Genesis 2002, 34: 251–256. 10.1002/gene.10161

    Article  CAS  PubMed  Google Scholar 

  74. de Boer J, Williams A, Skavdis G, Harker N, Coles M, Tolaini M, Norton T, Williams K, Roderick K, Potocnik AJ, Kioussis D: Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur J Immunol 2003, 33: 314–325. 10.1002/immu.200310005

    Article  CAS  PubMed  Google Scholar 

  75. Spike BT, Macleod KF: The Rb tumor suppressor in stress responses and hematopoietic homeostasis. Cell Cycle 2005, 4: 42–45.

    Article  CAS  PubMed  Google Scholar 

  76. Orkin SH, Zon LI: SnapShot: hematopoiesis. Cell 2008, 132: 712.

    Article  CAS  PubMed  Google Scholar 

  77. Morrison SJ, Spradling AC: Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell 2008, 132: 598–611. 10.1016/j.cell.2008.01.038

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  78. Sato T, Laver JH, Ogawa M: Reversible expression of CD34 by murine hematopoietic cells. Blood 1999, 94: 2548–2554.

    CAS  PubMed  Google Scholar 

  79. Ito T, Tajima F, Ogawa M: Developmental changes of CD34 expression by murine hematopoietic stem cells. Exp Hematol 2000, 28: 1269–1273. 10.1016/S0301-472X(00)00535-X

    Article  CAS  PubMed  Google Scholar 

  80. Nathan DG, Orkin SH, Look AT, Ginsburg D: Nathan and Oski's hematology of infancy and childhood. 6th edition. Philadelphia, Pa.: Saunders; 2003.

    Google Scholar 

  81. Papayannopoulou T, Torrealba de Ron A, Veith R, Knitter G, Stamatoyannopoulos G: Arabinosylcytosine induces fetal hemoglobin in baboons by perturbing erythroid cell differentiation kinetics. Science 1984, 224: 617–619. 10.1126/science.6200940

    Article  CAS  PubMed  Google Scholar 

  82. Letvin NL, Linch DC, Beardsley GP, McIntyre KW, Miller BA, Nathan DG: Influence of cell cycle phase-specific agents on simian fetal hemoglobin synthesis. J Clin Invest 1985, 75: 1999–2005. 10.1172/JCI111918

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  83. Stamatoyannopoulos G: Control of globin gene expression during development and erythroid differentiation. Exp Hematol 2005, 33: 259–271. 10.1016/j.exphem.2004.11.007

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  84. Zhang J, Socolovsky M, Gross AW, Lodish HF: Role of Ras signaling in erythroid differentiation of mouse fetal liver cells: functional analysis by a flow cytometry-based novel culture system. Blood 2003, 102: 3938–3946. 10.1182/blood-2003-05-1479

    Article  CAS  PubMed  Google Scholar 

  85. Gregory CJ, Eaves AC: Human marrow cells capable of erythropoietic differentiation in vitro: definition of three erythroid colony responses. Blood 1977, 49: 855–864.

    CAS  PubMed  Google Scholar 

  86. Gregory CJ, Eaves AC: Three stages of erythropoietic progenitor cell differentiation distinguished by a number of physical and biologic properties. Blood 1978, 51: 527–537.

    CAS  PubMed  Google Scholar 

  87. Mukai HY, Motohashi H, Ohneda O, Suzuki N, Nagano M, Yamamoto M: Transgene insertion in proximity to the c-myb gene disrupts erythroid-megakaryocytic lineage bifurcation. Mol Cell Biol 2006, 26: 7953–7965. 10.1128/MCB.00718-06

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  88. Emambokus N, Vegiopoulos A, Harman B, Jenkinson E, Anderson G, Frampton J: Progression through key stages of haemopoiesis is dependent on distinct threshold levels of c-Myb. Embo J 2003, 22: 4478–4488. 10.1093/emboj/cdg434

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  89. Mucenski ML, McLain K, Kier AB, Swerdlow SH, Schreiner CM, Miller TA, Pietryga DW, Scott WJ Jr, Potter SS: A functional c-myb gene is required for normal murine fetal hepatic hematopoiesis. Cell 1991, 65: 677–689. 10.1016/0092-8674(91)90099-K

    Article  CAS  PubMed  Google Scholar 

  90. Rylski M, Welch JJ, Chen YY, Letting DL, Diehl JA, Chodosh LA, Blobel GA, Weiss MJ: GATA-1-mediated proliferation arrest during erythroid maturation. Mol Cell Biol 2003, 23: 5031–5042. 10.1128/MCB.23.14.5031-5042.2003

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  91. Classon M, Harlow E: The retinoblastoma tumour suppressor in development and cancer. Nat Rev Cancer 2002, 2: 910–917. 10.1038/nrc950

    Article  CAS  PubMed  Google Scholar 

  92. Sankaran VG, Orkin SH, Walkley CR: Rb intrinsically promotes erythropoiesis by coupling cell cycle exit with mitochondrial biogenesis. Genes Dev 2008, 22: 463–475. 10.1101/gad.1627208

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  93. Heinrich AC, Pelanda R, Klingmuller U: A mouse model for visualization and conditional mutations in the erythroid lineage. Blood 2004, 104: 659–666. 10.1182/blood-2003-05-1442

    Article  CAS  PubMed  Google Scholar 

  94. Dumitriu B, Patrick MR, Petschek JP, Cherukuri S, Klingmuller U, Fox PL, Lefebvre V: Sox6 cell-autonomously stimulates erythroid cell survival, proliferation, and terminal maturation and is thereby an important enhancer of definitive erythropoiesis during mouse development. Blood 2006, 108: 1198–1207. 10.1182/blood-2006-02-004184

    Article  CAS  PubMed  Google Scholar 

  95. Clausen BE, Burkhardt C, Reith W, Renkawitz R, Forster I: Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res 1999, 8: 265–277. 10.1023/A:1008942828960

    Article  CAS  PubMed  Google Scholar 

  96. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP: Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 2005, 102: 15545–15550. 10.1073/pnas.0506580102

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  97. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstrale M, Laurila E, Houstis N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, Spiegelman B, Lander ES, Hirschhorn JN, Altshuler D, Groop LC: PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 2003, 34: 267–273. 10.1038/ng1180

    Article  CAS  PubMed  Google Scholar 

  98. Fontenay M, Cathelin S, Amiot M, Gyan E, Solary E: Mitochondria in hematopoiesis and hematological diseases. Oncogene 2006, 25: 4757–4767. 10.1038/sj.onc.1209606

    Article  CAS  PubMed  Google Scholar 

  99. Greenberg PL, Young NS, Gattermann N: Myelodysplastic syndromes. Hematology Am Soc Hematol Educ Program 2002, 136–161.

    Google Scholar 

  100. Rotig A, Cormier V, Blanche S, Bonnefont JP, Ledeist F, Romero N, Schmitz J, Rustin P, Fischer A, Saudubray JM, et al.: Pearson's marrow-pancreas syndrome. A multisystem mitochondrial disorder in infancy. J Clin Invest 1990, 86: 1601–1608. 10.1172/JCI114881

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  101. Inoue S, Yokota M, Nakada K, Miyoshi H, Hayashi J: Pathogenic mitochondrial DNA-induced respiration defects in hematopoietic cells result in anemia by suppressing erythroid differentiation. FEBS Lett 2007, 581: 1910–1916.

    Article  CAS  PubMed  Google Scholar 

  102. Craven SE, French D, Ye W, de Sauvage F, Rosenthal A: Loss of Hspa9b in zebrafish recapitulates the ineffective hematopoiesis of the myelodysplastic syndrome. Blood 2005, 105: 3528–3534. 10.1182/blood-2004-03-1089

    Article  CAS  PubMed  Google Scholar 

  103. Firkin FC: Mitochondrial lesions in reversible erythropoietic depression due to chloramphenicol. J Clin Invest 1972, 51: 2085–2092. 10.1172/JCI107015

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  104. Wang C, Li Z, Lu Y, Du R, Katiyar S, Yang J, Fu M, Leader JE, Quong A, Novikoff PM, Pestell RG: Cyclin D1 repression of nuclear respiratory factor 1 integrates nuclear DNA synthesis and mitochondrial function. Proc Natl Acad Sci USA 2006, 103: 11567–11572. 10.1073/pnas.0603363103

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  105. Corey SJ, Minden MD, Barber DL, Kantarjian H, Wang JC, Schimmer AD: Myelodysplastic syndromes: the complexity of stem-cell diseases. Nat Rev Cancer 2007, 7: 118–129. 10.1038/nrc2047

    Article  CAS  PubMed  Google Scholar 

  106. Boultwood J, Wainscoat JS: Gene silencing by DNA methylation in haematological malignancies. Br J Haematol 2007, 138: 3–11. 10.1111/j.1365-2141.2007.06604.x

    Article  CAS  PubMed  Google Scholar 

  107. McGrath K, Palis J: Ontogeny of erythropoiesis in the mammalian embryo. Curr Top Dev Biol 2008, 82: 1–22.

    Article  CAS  PubMed  Google Scholar 

  108. Uda M, Galanello R, Sanna S, Lettre G, Sankaran VG, Chen W, Usala G, Busonero F, Maschio A, Albai G, Piras MG, Sestu N, Lai S, Dei M, Mulas A, Crisponi L, Naitza S, Asunis I, Deiana M, Nagaraja R, Perseu L, Satta S, Cipollina MD, Sollaino C, Moi P, Hirschhorn JN, Orkin SH, Abecasis GR, Schlessinger D, Cao A: Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of beta-thalassemia. Proc Natl Acad Sci USA 2008, 105: 1620–1625. 10.1073/pnas.0711566105

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  109. Thein SL, Menzel S, Peng X, Best S, Jiang J, Close J, Silver N, Gerovasilli A, Ping C, Yamaguchi M, Wahlberg K, Ulug P, Spector TD, Garner C, Matsuda F, Farrall M, Lathrop M: Intergenic variants of HBS1L-MYB are responsible for a major quantitative trait locus on chromosome 6q23 influencing fetal hemoglobin levels in adults. Proc Natl Acad Sci USA 2007, 104: 11346–11351. 10.1073/pnas.0611393104

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  110. Lettre G, Sankaran VG, Bezerra MA, Araujo AS, Uda M, Sanna S, Cao A, Schlessinger D, Costa FF, Hirschhorn JN, Orkin SH: DNA polymorphisms at the BCL11A, HBS1L-MYB, and {beta}-globin loci associate with fetal hemoglobin levels and pain crises in sickle cell disease. Proc Natl Acad Sci USA 2008.

    Google Scholar 

Download references

Acknowledgements

The authors would like to thank Drs. L Purton, D. Nathan and M. Weiss for helpful discussion; Dr. T. Jacks for the generous provision of the conditional Rb animals and those who have generously shared Cre lines. V.G.S. was supported by MSTP and NRSA awards from the NIH. C.R.W. is a Special Fellow of the Leukemia & Lymphoma Society and S.H.O. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Stuart H Orkin.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CRW, VGS and SHO drafted the manuscript. All authors read and approved the final manuscript.

Carl R Walkley, Vijay G Sankaran contributed equally to this work.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Walkley, C.R., Sankaran, V.G. & Orkin, S.H. Rb and hematopoiesis: stem cells to anemia. Cell Div 3, 13 (2008). https://doi.org/10.1186/1747-1028-3-13

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1747-1028-3-13

Keywords