Skip to main content
Molecular Endocrinology logoLink to Molecular Endocrinology
. 2009 Nov 24;24(1):1–10. doi: 10.1210/me.2009-0332

Minireview: Nuclear Receptors, Hematopoiesis, and Stem Cells

John P Chute 1, Joel R Ross 1, Donald P McDonnell 1
PMCID: PMC2802897  PMID: 19934345

Abstract

Nuclear receptors (NRs) regulate a panoply of biological processes, including the function and development of cells within the hematopoietic and immune system, such as erythrocytes, monocytes, and lymphocytes. Significantly less is known regarding the function of NRs in regulating the fate of hematopoietic stem cells (HSCs), the self-renewing, pluripotent cells that give rise to the entirety of the blood and immune systems throughout the lifetime of an individual. Several recent studies suggest, either directly or indirectly, a role for members of the NR family in regulating the differentiation and self-renewal of HSCs, embryonic stem cells, and induced pluripotent stem cells. Herein, we review in detail the function of specific NRs in controlling HSC and other stem cell fate and propose a framework through which these observations can be translated into therapeutic amplification of HSCs for clinical purposes.


Nuclear receptors regulate hematopoietic and embryonic stem cell fate determinations and represent attractive targets for pharmacologic modulation to augment cellular therapy and tissue regeneration.

Introduction to Hematopoiesis and HSC Regulation

Hematopoietic stem cells (HSCs) possess the unique capacity to self-renew and give rise to the entirety of the blood and immune systems throughout the lifetime of an individual (Fig. 1) (1,2). Unlike other tissue-specific stem cells, hematopoietic stem and progenitor cells can be readily isolated to near purity using immunohistochemical staining and fluorescence activated cell sorting and characterized via well-established functional assays (Fig. 1). For example, murine bone marrow (BM) HSCs are lineage negative, c-kit+, sca-1+, and lack CD34 expression (34KLS cells), and BM HSC numbers can be estimated via competitive transplantation of limiting doses of donor BM 34KLS cells into lethally irradiated congenic recipient mice (competitive repopulating assay) (1,2,3,4). HSC self-renewal is regulated by both cell-intrinsic and -extrinsic pathways but the mechanisms controlling this process remain incompletely understood (1,2,3,4,5,6). Several transcription factors, including HoxB4, β-catenin, HES1, and GFI-1, are known to regulate the HSC self-renewal and differentiation program (Fig. 2) (3,4,5,6). Several of these targets are thought to be ligand inducible. For example, β-catenin signaling can be activated via treatment of murine HSCs with soluble Wnt3a, resulting in HSC expansion in culture (3). HES1 signaling can be also induced via binding of a Notch ligand (e.g. Jagged or Delta1) to the Notch receptor on HSCs, resulting in HSC self-renewal (5,7). Recently, treatment of murine HSCs with prostaglandin E2 (8) or angiopoietin-like protein-2 or -3 has been shown to induce significant expansions of repopulating HSCs in culture (9). Less is known regarding the contribution of nuclear receptors (NRs), ligand-inducible transcription factors, in regulating HSC fate determinations (10,11). However, given the contribution of NRs to a variety of biological processes and the availability of pharmacological ligands to modulate NR activities, further study of NRs in hematopoiesis is merited. Herein, we summarize the established functions of NRs in regulating hematopoiesis and HSC fate and highlight how manipulation of these receptors with small molecule ligands may have clinical utility in hematopoietic cell expansion and augmentation of hematopoiesis in vivo (Table 1).

Figure 1.

Figure 1

Overview of the hematopoietic hierarchy. The spectrum of hematopoietic stem and progenitor cells and their progeny are represented along with the phenotype and functional assays used to characterize the stem and progenitor cell pool. LTHSC, Long-term repopulating hematopoietic stem cell; STHSC, short-term repopulating hematopoietic stem cell; MPP, multipotent progenitor cell; CMP, common myeloid progenitor cell; MEP, megakaryocyte-erythroid progenitor cell; GMP, granulocyte monocyte progenitor cell; CLP, common lymphoid progenitor cell; RBC, red blood cell; 34-KLS, CD34- c-kit+lineage- sca-1+ cells; CRU, competitive repopulating unit assay; CFU-Spleen, colony forming unit-spleen assay; CFC, colony-forming cell assay, d 14.

Figure 2.

Figure 2

Schematic overview of major HSC self-renewal pathways. The most well-characterized signaling pathways and transcription factors involved in regulating HSC self-renewal are shown. BMP, Bone morphogenetic protein; NICD, notch intracellular domain; RBPJ, recombinant signal binding protein; LEF, lymphoid enhancer binding protein; GFI-1, growth factor independent 1; HOX, homeobox proteins.

Table 1.

NRs that regulate hematopoiesis and stem cell fate hematopoiesis

Receptor Function References
RARα Myeloid differentiation 20,21,27,28,29,30,33
HSC differentiation (human) 27,34
RARγ HSC self-renewal 10,11
RXR HSC differentiation 34
Inhibits erythroid differentiation 34,35
TRα Erythroid differentiation 38,40,41,42
GR Erythroid progenitor expansion 43,45,46,47,48,50
Stress erythropoiesis 43
ERβ Inhibition of B lymphopoiesis 51,52,53,54,55,56
Inhibition of early myelopoiesis 51,52
AR Erythroid progenitor expansion 60,63
Inhibition of B lymphopoiesis 65
VDR Monocyte differentiation 69
Th1 immune response 66,70
PPARγ Myeloid differentiation 34
Inhibition of erythoid differentiation 75
Nur77 + Nor1 HSC differentiation 79
ES cell differentiation and iPS
 Esrrb Represses ES cell differentiation 86,89
 SF-1 Maintains ES cell pluripotency 86,92,93
 DAX1 Repression of ES cell differentiation 86,95,96
 GCNF ES cell differentiation 86,97,98,99,100
 Esrrb Induced pluripotency (iPS) in adult cells 103

Retinoid Receptors

Retinoic acid (RA) is a vitamin A derivative with well-described effects on development and cellular differentiation (12,13,14). There are three isoforms of the retinoic acid receptor (RAR) (α, γ, β), which act as ligand-inducible transcription factors with distinct functions (15,16,17). The RARs are the most well-characterized NRs with regard to regulation of hematopoiesis and hematopoietic stem cell (HSC) function (10,18,19). This field of study was catalyzed by the observations of Breitman et al. (20,21) that incubation with all-trans retinoic acid (ATRA) caused the differentiation of both a human promyelocytic leukemia cell line (HL-60) and primary human promyelocytic cells in culture. Subsequently, it was shown that acute promyelocytic leukemia cells carry a t15;17(q21-q11-22) translocation, resulting in the expression of a promyelocytic leukemia (PML)-RARα fusion protein that functions as a dominant-negative inhibitor of both promyelocytic leukemia and RARα at their respective target genes (22,23,24). Treatment of patients with ATRA, combined with chemotherapy, induces complete remission of acute promyelocytic leukemia and is curative in many cases (25,26).

The role of retinoid signaling in regulating normal hematopoietic progenitor cell differentiation is also well established. ATRA stimulates the proliferation of myeloid progenitors (27) and induces the granulocytic differentiation of myeloid progenitors (28), and introduction of a dominant-negative RARα construct into a hematopoietic progenitor cell line suppresses neutrophil and monocyte development (29). Activation of RARα with ATRA has also been shown to inhibit the proliferation of human embryonic hematopoietic progenitor cells and adult cobblestone-area forming cells in culture and promote the apoptosis of human CD34+ cells (30,31,32). Although RARα is not required for granulocytic differentiation to occur (33), this receptor clearly plays an important role in inducing hematopoietic progenitor cell differentiation.

In addition to promoting the differentiation of myeloid progenitor cells, RARs regulate both murine and human HSC differentiation (10,11,27). Treatment of murine c-kit+sca-1+lin (KSL) stem/progenitor cells in culture with ATRA has been shown to enhance the maintenance of murine repopulating HSCs in vitro (10). Purton et al. (11) further demonstrated that ATRA-mediated maintenance of murine HSCs in culture was dependent upon RARγ and independent of RARα, suggesting that RARγ is an important positive regulator of murine HSC self-renewal (Fig. 2). Consistent with these findings, the authors also showed that RARγ−/− mice contain significantly less BM HSCs compared with wild-type mice (11). Interestingly, Chute et al. (27) reported that pharmacological inhibition of the enzyme, aldehyde dehydrogenase 1, which is required for the intracellular production of retinoids, caused a 3- to 4-fold expansion of human HSCs in culture. These results suggested perhaps opposing effects of retinoids in regulating HSC fate in mice and humans or that inhibition of aldehyde dehydrogenase 1 modified HSC fate via a mechanism independent of the retinoid receptors (27).

The role of retinoic X receptor (RXR) in regulating HSC fate has also recently been addressed (34). It has been previously shown that RXR functions as a negative regulator of erythropoiesis (35) but Safi et al. (34) demonstrated that human HSCs could be maintained in culture via the addition of a small molecule, LG1506, which antagonized RXR:RAR signaling. LG1506 was shown to bind RXRs, the obligate partners of RARs, facilitating an allosteric event that resulted in inhibition of RAR, within the RXR:RAR heterodimer. Although deletion of RXRα was not associated with any hematopoietic defects in mice (36), more global inhibition of all three RXRs, as was accomplished pharmacologically, inhibited the differentiation of human HSCs in culture (34). Taken together, these data suggest that pharmacological antagonism of retinoid signaling inhibited human HSC differentiation and enhanced the maintenance of human HSCs in culture. It is plausible that signaling via RAR and RXR mediate distinct differentiation events in mice and humans to explain the observations by Purton et al. (10,11) and Chute et al. (27) and Safi et al. (34). Nonetheless, it is clear that RAR and RXR have important and diverse functions in regulating murine and human HSC self-renewal and differentiation events. As such, these receptors represent excellent targets for therapeutic manipulation to expand HSCs for clinical purposes.

Thyroid Hormone Receptor (TR)

TRα is a member of the class II NRs (37). In the absence of ligand, TRs bind to specific thyroid response elements (TREs) within target genes and, as a consequence of their ability to interact with NR corepressors, they actively suppress gene transcription (38,39). Upon binding an agonist such as thyroid hormone (T3), the receptor undergoes a conformational change that facilitates the dissociation of corepressors and binding of coactivators; the binding of coactivators results in transcriptional activation (38). TRα is expressed by hematopoietic and erythroid progenitors (40), and hypothyroidism is clinically accompanied by a reduced red blood cell mass and normocytic anemia (41). The avian erythroblastosis virus (AEV) oncoprotein, v-ErbA, represents a mutated, oncogenic form of TRα (c-ErbA/TRα). The addition of T3 activates the cErbA/TRα receptor in erythroblasts, blocks self-renewal, and induces synchronous, terminal differentiation in culture (38). Conversely, vErbA cooperates with ligand-activated c-kit receptor to induce the self-renewal and arrest of differentiation of primary avian erythroblasts, resulting in a fatal erythroleukemia (38). Taken together, Bauer et al. (38) demonstrated that cErb/TRα represents a ligand-operated molecular switch, regulating the balance between erythroblast self-renewal and differentiation.

Recently, Kendrick et al. (42) verified the essential function of TRα in regulating erythropoiesis by characterizing the phenotype of TRα−/− mice. TRα mice had significantly reduced numbers of erythroid progenitor cells in the fetal liver, decreased hematocrits as adults, and a diminished erythroid stress response to hemolytic anemia (42). Although no defect was identified in HSC or progenitor cell numbers in the adult TRα−/− mice, these studies confirmed the essential function of TRα in regulating erythropoiesis both during development and in the adult.

Glucocorticoid Receptor (GR)

The importance of the GR in regulating erythropoiesis has long been inferred from the clinical observation that patients who lack glucocorticoid production (Addison’s disease) have a normocytic anemia (43,44). Similarly, patients with Cushing’s syndrome, in which there are excess circulating glucocorticoids, have elevated hemoglobin and hematocrit levels (43,44). Experimentally, Golde et al. (45) first demonstrated that dexamethasone promotes the generation of murine erythroid colony-forming cells (45), and glucocorticoids also enhance erythroid progenitor cell proliferation in response to limiting amounts of erythropoietin (46). The GR is expressed by erythroid progenitor cells, and activation of GR with dexamethasone induces the proliferation and expansion of erythroid progenitors while maintaining their colony-forming capacity and delaying the terminal differentiation of erythrocytes (47). Importantly, these effects of GR require cooperation with the erythropoietin receptor and c-kit activation in order for erythroid progenitor expansion to occur (47). It has also been shown that this expansion of erythroid progenitors requires the DNA-binding and transactivation function of the GR (48,49). Interestingly, Stellaci et al. (50) recently demonstrated GR-mediated inhibition of terminal erythroid differentiation is mediated via interference with erythropoietin-mediated signaling.

Importantly, GR−/− mice are not viable, but examination of erythropoiesis in GR−/− embryos demonstrates no obvious defect in erythropoiesis (43). However, studies of GR dim/dim mice vs. wild-type mice demonstrate an essential role for GR in the stress erythroid response to blood loss or hemolysis (43). GR dim/dim mice showed no increase in colony-forming unit-erythroid (CFU-E) in response to hemolysis and no increase in red blood cell count, hemoglobin, or hematocrit in response to hypoxia, whereas wild-type mice demonstrated strong erythroid responses to both stresses (43). Taken together, these data illustrate an important function of GR in regulating the erythroid progenitor response to stress. No obvious alterations in HSC content or function were observed in GR dim/dim mice compared with wild-type mice, suggesting that GR does not play an essential role in regulating HSC fate.

Estrogen Receptor (ER)

It has long been recognized that women have significantly increased risk of autoimmune diseases compared with men (51). However, a mechanistic link between estrogen receptor activation and immune function has only recently been elucidated. The loss of estrogen in ovariectomized mice is associated with splenomegaly, increased BM CFU-GEMM, BFU-E, and B lymphocytes (51,52). Conversely, administration of exogenous estrogen decreases B lymphopoiesis (51,53) and, surprisingly, was shown to inhibit B lymphoid and myeloid differentiation of BM HSCs (54,55). Estrogen exerts its effects through ERα and ERβ (51). ERα and ERβ are both expressed by microenvironment cells in the BM, but ERα−/− mice have a normal hematopoietic profile with the exception of a reduction in BM B lymphocyte subsets (51,56). Moreover, administration of exogenous estrogen to ERα−/− mice causes a decrease in B lymphopoiesis (51,56), suggesting that estrogen effects on hematopoietic progenitors may be mediated by ERβ.

ERβ−/− mice have also been examined for evidence of long-term hematopoietic defects (51). By age 1.5 yr, ERβ−/− mice have marked splenomegaly, BM myeloid hyperplasia, lymphadenopathy, and leukocyte infiltration in the lungs and liver, consistent with a lymphoid blast crisis of chronic myelogenous leukemia (51). Taken together, these data suggest that ERβ is an important negative regulator of hematopoietic progenitor cells (51). In light of these observations, it has been suggested that ERβ agonists may be useful in the treatment of patients with acute and chronic leukemia (51,57).

Androgen Receptor (AR)

The AR is expressed in all reproductive tissues in men, as well as muscle, skin, bone, and hematopoietic cells (58). Healthy men maintain higher steady-state levels of hemoglobin and red blood cell mass compared with healthy women. During andropause, aging males can develop a normocytic anemia that is associated with a decline in free testosterone levels and can be corrected via testosterone administration (59). However, the mechanisms through which androgens stimulate erythropoiesis are poorly understood (60). Early animal studies demonstrated that several testosterone derivatives could cause an increase in erythropoietin production in the kidneys (61) and the erythropoietic effects of testosterone administration in mice could be abolished by nephrectomy (62). However, more recent studies have not confirmed that the effects of androgens on erythropoiesis are erythropoietin dependent (60). Alternately, BM erythroid progenitor cells express AR, and the addition of androgens to culture of primary erythroid progenitor cells results in a significant expansion of erythroid progenitors without substantial terminal differentiation (63). In the same study, it was shown that the addition of T3 to erythroid progenitor cultures caused terminal differentiation of the erythroid progenitors. Taken together, these data demonstrate that the expansion of erythroid progenitors and their terminal differentiation are regulated in a coordinated manner by AR and TR activation (63).

Homozygous deletion of AR in male mice is grossly associated with a female phenotype and a significant reduction in trabecular bone but without formal description of effects on erythropoiesis in these animals (64). An unexpected finding in AR knockout mice was a significant expansion of the B cell pool and splenic enlargement caused by the expansion of B progenitor cells in the BM (65). These data indicate that AR functions in hematopoiesis beyond its role in promoting erythropoiesis, perhaps as a negative regulator of B lymphopoiesis. There is no information as to the function of AR in regulating HSC function or content in vitro or in vivo, but AR−/− mice have normocellular BM, suggesting that AR is not an essential regulator of hematopoiesis (64).

Vitamin D Receptor (VDR)

The VDR is a transcription factor that mediates the actions of its cognate ligand, 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3], in regulating calcium homeostasis (66). However, VDR is widely expressed in many tissues including hematopoietic progenitor cells (67,68) and culture of human CD34+ hematopoietic progenitor cells with 1,25-(OH)2D3 induces massive monocyte commitment in vitro via activation of HoxA10 and MafB, a master regulator of monocyte differentiation (69). Independently, it was shown that 1,25-(OH)2D3 inhibits the proliferation and cytokine production of T cells in culture (66,70). Interestingly, BM HSCs from VDR knockout mice can be induced to generate differentiated monocytes, but these mice have an impaired Th1 cell response (66). Taken together, these results demonstrate that VDR is not essential for monocytic differentiation of HSCs but may be required for the normal Th1 immune response (66).

Peroxisome Proliferator-Activated Receptor (PPAR)

PPARγ is an NR that has deterministic function in regulating adipogenesis and metabolism (71). Humans with diminished PPARγ activity have features of insulin resistance, dyslipidemia, and hypertension, and rosiglitazone, a PPARγ agonist, is used to augment insulin sensitivity and lower blood glucose levels in patients with type II diabetes mellitus (72,73,74). PPARγ ligands, troglitazone and pioglitazone, have been shown to suppress the proliferation and delay the maturation of primary erythroid progenitor cells (75). More recently, Prost et al. (76) reported the unexpected finding that human and simian immunodeficiency viruses inhibited hematopoietic progenitor cell growth via induction of an accessory viral protein, negative factor, and the actions of negative factor were dependent upon the presence of PPARγ in the hematopoietic progenitor cells. Moreover, the authors showed that treatment of mice or macaque monkeys with rosiglitazone × 15 d in vivo caused a significant decrease in the number of BM colony-forming cells (CFCs) recovered compared with untreated control animals (76). The inhibitory actions of PPARγ on hematopoietic progenitor cell growth were demonstrated to be mediated by inhibition of signal transducer and activator of transcription 5 signaling in the target cells (76).

More recently, Safi et al. (34) studied the activity of several PPAR agonists on the proliferation and expansion of human CD34+CD38lin HSCs in culture. The addition of clofibrate, a PPARα agonist, to human HSCs had no effect on total cell, CD34+, or CD34+CD38 progenitor cell expansion or CFC content in liquid suspension with thrombopoietin, stem cell factor, and Flt-3 ligand (34). Similarly, the addition of carbaprostacyclin, a PPARδ agonist, also had no effect on human stem/progenitor cell content in culture. Conversely, the addition of rosiglitazone, a PPARγ agonist, caused a decline in primitive CD34+CD38 cells and a significant increase in production of committed CFCs in culture compared with cytokines alone. Taken together, these results indicate that activation of PPARγ induces the myeloid differentiation of human HSCs rather than HSC self-renewal. It remains to be determined whether PPARγ is essential for myeloid maturation of HSCs because homozygous deletion of PPARγ is embryonic lethal in mice, and the hematopoietic phenotype of heterozygous PPARγ+/− mice has not been described (77,78). Because PPARγ is a heterodimeric partner of RXR and studies by Safi et al. demonstrated that antagonism of RXR blocked the differentiation of HSCs in response to cytokines, it will be important to verify whether RXR mediates actions on hematopoiesis via PPARγ signaling events. Preliminary studies by Safi et al. (34) suggested that PPARγ was not involved in mediating effects of RXR on HSC differentiation.

Orphan Receptors

The homologous orphan nuclear receptors, Nur77 and Nor-1, have been shown to promote the apoptosis of T and B cells (79,80,81), the apoptosis of nonhematopoietic epithelial cells in response to chemotherapy, and the proliferation of epithelial cells in response to growth factors (79,80,81). Lee et al. (82) showed that deletion of Nur77 alone caused minimal T cell functional defects, and Ponnio et al. (83) reported that deletion of Nor-1 caused defective development of the inner ear. However, mice that are deficient in both Nur77 and Nor-1 develop a marked expansion of phenotypic long-term HSCs with a block in differentiation, leading to acute myelogenous leukemia and early death (79). The authors also demonstrated that Nur77−/−;Nor1−/− mice displayed decreased expression of JunB and c-Jun, which are involved in the normal differentiation of myeloid progenitor cells (79,84,85). Taken together, these results suggest a role for Nur77 and Nor-1 as positive regulators of HSC differentiation that perhaps mediate effects via JunB and c-Jun signaling (79). It remains to be determined whether genetic enforcement or pharmacological activation of Nur77 or Nor-1 signaling can induce the differentiation of HSCs.

NRs and Embryonic Stem Cells

In addition to their role in regulating hematopoiesis, several NRs function in regulating the maintenance and differentiation of embryonic stem (ES) cells (86) The estrogen-related receptor β (Esrrβ) subfamily of NRs contains three isoforms (α, β, γ). Luo et al. (87) and Pettersson et al. (88) reported that mice lacking Esrrβ have impaired trophoblast stem cell differentiation and failure of placenta development. After short hairpin RNA-induced silencing of Esrrβ, ES cells differentiated in culture, suggesting that Esrrβ repressed ES cell differentiation (89). Transcriptional analysis demonstrated that knockdown of Esrrβ altered the expression of a distinct family of genes compared with those regulated by the pluripotency genes Oct4, Sox2, and Nanog (86,89). These results suggested that Esrrβ represses ES cell differentiation via a pathway that is independent of Oct4-, Sox2-, and Nanog-mediated control of ES cell pluripotency (86,89).

Steroidogenic factor (SF-1) is an orphan NR that is expressed in steroidogenic tissues including the adrenal cortex, Leydig cells, Sertoli cells, the pituitary gland, and in the inner cell mass of mouse blastocysts (86,90,91). The proximal promoter of Oct4 contains a consensus SF-1 response motif, and when SF-1 is overexpressed, a 3-fold increase in Oct4 promoter activity is observed (86,92,93). RA-induced differentiation of ES cells causes a loss of expression of both SF-1 and Oct4, suggesting a role for SF-1 in the maintenance of ES cell pluripotency (86,92,93). Similarly, the liver receptor homolog-1 (LRH-1), another orphan nuclear receptor, is expressed by ES cells and the inner cell mass of the blastocyst (86,94) and binds to response elements in the Oct4 promoter region (86,94). LRH-1 knockout mice die at d 6.5–d 9.5, and both LRH-1 and Oct4 are simultaneously down-regulated in response to the differentiative effects of RA (86,92).

The dosage-sensitive sex reversal, adrenal hypoplasia congenital, locus on the X-chromosome, gene 1 (DAX1) is an orphan nuclear receptor that is also critical in initiation of steroidogenesis in development (86,95). Disruption of DAX1 by RNA interference or conditional knockout in ES cells caused ES cell differentiation (96), and treatment of ES cells with RA caused a decrease in DAX1 expression in parallel with decline in Oct4 (96). Taken together, these data suggest a role for DAX1 in repressing the differentiation of ES cells and a functional relationship with RA signaling in regulating ES cell differentiation.

The orphan NR germ cell nuclear factor (GCNF) is essential for normal embryonic development and survival and proper formation of the anterior-posterior axis (86,97,98). RA induces GCNF expression in ES cells, and GCNF binds to a conserved region in the Oct4 promoter, thereby repressing Oct4 expression and promoting ES cell differentiation (99). Examination of GCNF−/− ES cells confirmed that GCNF is required for RA-mediated repression of Oct4, Nanog, and Sox2 during ES cell differentiation (99). Interestingly, GCNF appears to mediate repression of Oct4 expression via modulation of DNA methylation (100). Taken together, these data indicate that GCNF is essential for the repression of pluripotency genes and in the initiation of differentiation of ES cells (86,97,98,99,100). In addition to Esrrβ, SF-1, LRH-1, DAX-1, and GCNF, systematic gene expression profiling of NRs has demonstrated diametric expression of certain NRs between murine and human ES cells (101). This dataset provides an important resource for understanding and testing the precise roles of NRs in murine and human ES cells (101).

NRs and Induced Pluripotent Stem Cells (iPS)

A major advancement in cell biology was the demonstration that adult fibroblasts could be reprogrammed to pluripotency (iPS) via retroviral-mediated transduction of the transcription factors Oct4, Sox2, C-myc, and Klf4 (102,103). Recently, Feng et al. (104) reported that the orphan NR, Esrrβ, could function in replacement of Klf4 in this process such that retroviral infection of fibroblasts with Esrrβ, Oct4, and Sox2 became pluripotent with equal multilineage differentiation capacity to cells infected with Oct4, Sox2, C-myc, and Klf4. The authors also showed that Esrrβ induced the expression of several ES cell-specific genes (104). Taken together, these results demonstrate that an NR, Esrrβ, can be targeted to reprogram somatic cells (104).

Synopsis

Although NRs are not generally considered dominant regulators of hematopoiesis, this review illustrates that several NRs have precise and complementary roles in regulating virtually all cell types within the hematopoietic hierarchy (Fig. 3). For example, HSC self-renewal and differentiation (RARγ and RXR), myelopoiesis (RARα, PPARγ, ERβ, Nur-77/Nor-1), erythropoiesis (AR, GR, TRα, RXR), B lymphopoiesis (ERβ, AR), and T lymphopoiesis (VDR) are all regulated by the action of NRs (Fig. 3). Recent studies have also shown deterministic functions of NRs and orphan NRs in controlling ES cell pluripotency and differentiation and iPS. As our ability to measure HSC and ES function in vitro and in vivo continues to evolve, additional roles for NRs in regulating HSC regeneration and tissue stem cells in general will be identified. Because NRs are particularly amenable to manipulation with small molecule ligands, they represent attractive targets for the therapeutic expansion of HSCs, or iPS cells, and to augment hematopoiesis in vivo.

Figure 3.

Figure 3

Schematic overview of NR regulation in hematopoiesis. NRs that have activating function are shown in green and repressive actions are shown in red. LTHSC, long-term repopulating hematopoietic stem cell; STHSC, short-term repopulating hematopoietic stem cell; MPP, multipotent progenitor cell; CMP, common myeloid progenitor cell; MEP, megakaryocyte-erythroid progenitor cell; GMP, granulocyte-monocyte progenitor cell; CLP, common lymphoid progenitor cell; RBC, red blood cell.

Footnotes

This work was supported, in part, by funding from the National Institutes of Allergy and Infectious Diseases Grant AI067798-01 (to J.P.C.).

Disclosure Summary: The authors have nothing to disclose.

First Published Online November 24, 2009

Abbreviations: AR, Androgen receptor; ATRA, all-trans retinoic acid; BM, bone marrow; CFC, colony-forming cell; DAX1, adrenal hypoplasia congenital, locus on the X-chromosome, gene 1; ER, estrogen receptor; ES, embryonic stem; Essr, estrogen-related receptor; GCNF, germ cell nuclear factor; GR, glucocorticoid receptor; HSC, hematopoietic stem cell; iPS, induced pluripotent stem cells; LRH-1, liver receptor homolog-1; NR, nuclear receptor; PPAR, peroxisome proliferator-activated receptor; RA, retinoic acid; RAR, retinoic acid receptor; SF-1, steroidogenic factor 1; TR, thyroid hormone receptor; VDR, vitamin D receptor.

References

  1. Zon LI 2008 Intrinsic and extrinsic control of haematopoietic stem-cell self-renewal. Nature 453:306–313 [DOI] [PubMed] [Google Scholar]
  2. Orkin SH, Zon LI 2008 SnapShot: hematopoiesis. Cell 132:712 [DOI] [PubMed] [Google Scholar]
  3. Reya T, Duncan AW, Ailles L, Domen J, Scherer DC, Willert K, Hintz L, Nusse R, Weissman IL 2003 A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature 423:409–414 [DOI] [PubMed] [Google Scholar]
  4. Antonchuk J, Sauvageau G, Humphries RK 2002 HOXB4- induced expansion of adult hematopoietic stem cells ex vivo. Cell 109:39–45 [DOI] [PubMed] [Google Scholar]
  5. Kunisato A, Chiba S, Nakagami-Yamaguchi E, Kumano K, Saito T, Masuda S, Yamaguchi T, Osawa M, Kageyama R, Nakauchi H, Nishikawa M, Hirai H 2003 HES-1 preserves purified hematopoietic stem cells ex vivo and accumulates side population cells in vivo. Blood 101:1777–1783 [DOI] [PubMed] [Google Scholar]
  6. Hock H, Hamblen MJ, Rooke HM, Schindler JW, Saleque S, Fujiwara Y, Orkin SH 2004 Gfi-1 restricts proliferation and preserves functional integrity of haematopoietic stem cells. Nature 431:1002–1007 [DOI] [PubMed] [Google Scholar]
  7. Neves H, Weerkamp F, Gomes AC, Naber BA, Gameiro P, Becker JD, Lúcio P, Clode N, van Dongen JJ, Staal FJ, Parreira L 2006 Effects of Delta1 and Jagged1 on early human hematopoiesis: correlation with expression of Notch signaling related genes in CD34+ cells. Stem Cells 24:1328–1337 [DOI] [PubMed] [Google Scholar]
  8. North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, Lord AM, Weber GJ, Bowman TV, Jiang IH, Grosser T, Fitzgerald GA, Daley GQ, Orkin SH, Zon LI 2007 Prostaglandin E2 regulates vertebrate hematopoietic stem cell homeostasis. Nature 447:1007–1011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Zhang CC, Kaba M, Ge G, Xie K, Tong W, Hug C, Lodish HF 2006 Angiopoietin-like proteins stimulate ex vivo expansion of hematopoietic stem cells. Nat Med 12:240–245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Purton LE, Bernstein ID, Collins SJ 2000 All-trans retinoic acid enhances the long term repopulating activity of cultured hematopoietic stem cells. Blood 95:470–477 [PubMed] [Google Scholar]
  11. Purton LE, Dworkin S, Olsen GH, Walkley CR, Fabb SA, Collins SJ, Chambon P 2006 RARγ is critical for maintaining a balance between hematopoietic stem cell self-renewal and differentiation. J Exp Med 203:1283–1293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Campo-Paysaa F, Marlétaz F, Laudet V, Schubert M 2008 Retinoic acid signaling in development: tissue specific functions and evolutionary origins. Genesis 46:640–656 [DOI] [PubMed] [Google Scholar]
  13. Goldie LC, Lucitti JL, Dickinson ME, Hirschi KK 2008 Cell signaling directing the formation and function of hemogenic endothelium during murine embryogenesis. Blood 112:3194–3204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Motohashi T, Aoki H, Chiba K, Yoshimura N, Kunisada T 2007 Multipotent cell fate of neural crest like cells derived from embryonic stem cells. Stem Cells 25:402–410 [DOI] [PubMed] [Google Scholar]
  15. Lufkin T, Lohnes D, Mark M, Dierich A, Gorry P, Gaub MP, LeMeur M, Chambon P 1993 High postnatal lethality and testes degeneration in retinoic acid receptor α mutant mice. Proc Natl Acad Sci USA 90:7225–7229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Zelent A, Mendelsohn C, Kastner P, Krust A, Garnier JM, Ruffenach F, Leroy P, Chambon P 1991 Differentially expressed isoforms of the mouse retinoic acid receptor β generated by usage of two promoters and alternative splicing. EMBO J 10:71–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Lohnes D, Kastner P, Dierich A, Mark M, LeMeur M, Chambon P 1993 Function of retinoic acid receptor γ in the mouse. Cell 73:643–658 [DOI] [PubMed] [Google Scholar]
  18. Collins SJ 2008 Retinoic acid receptors, hematopoiesis and leukemogenesis. Curr Opin Hematol 15:346–351 [DOI] [PubMed] [Google Scholar]
  19. Tsai S, Collins SJ 1993 A dominant negative retinoic acid receptor blocks neutrophil differentiation at the promyelocyte stage. Proc Natl Acad Sci USA 90:7153–7157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Breitman TR, Selonick SE, Collins SJ 1980 Induction of differentiation of the human promyelocytic leukemia cell line (HL60) by retinoic acid. Proc Natl Acad Sci USA 77:2936–2940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Breitman TR, Collins SJ, Keene BR 1981 Terminal differentiation of human promyelocytic leukemia cells in primary culture in response to retinoic acid. Blood 57:1000–1004 [PubMed] [Google Scholar]
  22. Huang W, Sun GL, Li XS, Cao Q, Lu Y, Jang GS, Zhang FQ, Chai JR, Wang ZY, Waxman S 1993 Acute promyelocytic leukemia: clinical relevance of two major PML-RAR α isoforms and detection of minimal residual disease by retrotranscriptase/polymerase chain reaction to predict relapse. Blood 82:1264–1269 [PubMed] [Google Scholar]
  23. Larson RA, Kondo K, Vardiman JW, Butler AE, Golomb HM, Rowley JD 1984 Evidence for a 15;17 translocation in every patient with acute promyelocytic leukemia. Am J Med 76:827–841 [DOI] [PubMed] [Google Scholar]
  24. de Thé H, Chomienne C, Lanotte M, Degos L, Dejean A 1990 The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor α gene to a novel transcribed locus. Nature 347:558–561 [DOI] [PubMed] [Google Scholar]
  25. Huang ME, Ye YC, Chen SR, Chai JR, Lu JX, Zhoa L, Gu LJ, Wang ZY 1988 Use of all-trans retinoic acid in treatment of acute promyelocytic leukemia. Blood 72:567–572 [PubMed] [Google Scholar]
  26. Tallman M, Anderson J, Schiffer C, Appelbaum F, Feusner J, Ogden A, Shepherd L, Willman C, Bloomfield C, Rowe J, Wiernick P 1997 All-trans retinoic acid in acute promyelocytic leukemia. N Engl J Med 337:1639 [DOI] [PubMed] [Google Scholar]
  27. Chute JP, Muramoto GG, Whitesides J, Colvin M, Safi R, Chao NJ, McDonnell DP 2006 Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc Natl Acad Sci, USA 103:11707–11712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Kastner P, Chan S 2001 Function of RARα during the maturation of neutrophils. Oncogene 20:7178–7185 [DOI] [PubMed] [Google Scholar]
  29. Tsai S, Bartelmez S, Heyman R, Damm K, Evans R, Collins SJ 1992 A mutated retinoic acid receptor-α exhibiting dominant-negative activity alters the lineage development of a multipotent hematopoietic cell line. Genes Dev 6:2258–2269 [DOI] [PubMed] [Google Scholar]
  30. Tocci A, Parolini I, Gabbianelli M, Testa U, Luchetti L, Samoggia P, Masella B, Russo G, Valtieri M, Peschle C 1996 Dual action of retinoic acid on human embryonic/fetal hematopoiesis: blockade of primitive progenitor proliferation and shift from multipotent/erythroid/monocytic to granulocytic differentiation program. Blood 88:2878–2888 [PubMed] [Google Scholar]
  31. Sammons J, Ahmed N, Khokher MA, Hassan HT 2000 Mechanisms mediating the inhibitory effect of all-trans retinoic acid on primitive hematopoietic stem cells in long-term bone marrow culture. Stem Cells 18:214–219 [DOI] [PubMed] [Google Scholar]
  32. Josefsen D, Blomhoff HK, Lømo J, Blystad AK, Smeland EB 1999 Retinoic acid induces apoptosis of human CD34+ hematopoietic progenitor cells: involvement of retinoic acid receptors and retinoid X receptors depends on lineage commitment of the hematopoietic progenitor cells. Exp Hematol 27:642–653 [DOI] [PubMed] [Google Scholar]
  33. Zhang P, Nelson E, Radomska HS, Iwasaki-Arai J, Akashi K, Friedman AD, Tenen DG 2002 Induction of granulocytic differentiation by 2 pathways. Blood 99:4406–4412 [DOI] [PubMed] [Google Scholar]
  34. Safi R, Muramoto GG, Salter AB, Meadows S, Himburg H, Russell L, Daher P, Doan P, Leibowitz MD, Chao NJ, McDonnell DP, Chute JP 2009 Pharmacological manipulation of the RAR/RXR signaling pathway maintains the repopulating capacity of hematopoietic stem cells in culture. Mol Endocrinol 23:188–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Rusten LS, Dybedal I, Blomhoff HK, Blomhoff R, Smeland EB, Jacobsen SE 1996 The RAR-RXR as well as the RXR-RXR pathway is involved in signaling growth inhibition of human CD34+ erythroid progenitor cells. Blood 87:1728–1736 [PubMed] [Google Scholar]
  36. Ricote M, Snyder C, Leung H, Chen J, Chien K, Glass C 2006 Normal hematopoiesis after conditional targeting of RXRα in murine hematopoietic stem/progenitor cells. J Leuk Biol 80:850–861 [DOI] [PubMed] [Google Scholar]
  37. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM 1995 The nuclear receptor superfamily: the second decade. Cell 83:835–839 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Bauer A, Mikulits W, Lagger G, Stengl G, Brosch G, Beug H 1998 The thyroid hormone receptor functions as a ligand-operated developmental between proliferation and differentiation of erythroid progenitors. EMBO J 17:4291–4303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Chen JD, Evans RM 1995 A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 377:454–457 [DOI] [PubMed] [Google Scholar]
  40. Grymula K, Paczkowska E, Dziedziejko V, Báskiewicz-Masiuk M, Kawa M, Baumert B, Celewicz Z, Gawrych E, Machaliński B 2007 The influence of 3,3′,5-triiodo-L-thyronine on human hematopoiesis. Cell Prolif 40:302–315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Popovic WJ, Brown JE, Adamson JW 1977 The influence of thyroid hormones on in vitro erythropoiesis. Mediated by a receptor with β adrenergic properties. J Clin Invest 60:907–913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Kendrick TS, Payne CJ, Epis MR, Schneider JR, Leedman PJ, Klinken SP, Ingley E 2008 Erythroid defects in TRα−/− mice. Blood 111:3245–3248 [DOI] [PubMed] [Google Scholar]
  43. Bauer A, Tronche F, Wessely O, Kellendonk C, Reichardt HM, Steinlein P, Schütz G, Beug H 1999 The glucocorticoid receptor is required for stress erythropoiesis. Genes Dev 13:2996–3002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Miller W, Tyrell J 1995 The adrenal cortex. In Fehlig P, Baxter J, Frohman L, eds. Endocrinology and metabolism. New York: McGraw-Hill, Inc.; 555–711 [Google Scholar]
  45. Golde DW, Bersch N, Cline MJ 1976 Potentiation of erythropoiesis in vitro by dexamethasone. J Clin Invest 57:57–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Udupa KB, Crabtree HM, Lipschitz DA 1986 In vitro culture of proerythroblasts: characterization of proliferative response to erythropoietin and steroids. Br J Haematol 62:705–714 [DOI] [PubMed] [Google Scholar]
  47. von Lindern M, Zauner W, Mellitzer G, Steinlein P, Fritsch G, Huber K, Löwenberg B, Beug H 1999 The glucocorticoid receptor cooperates with the erythropoietin receptor and c-kit to enhance and sustain proliferation of erythroid progenitors in vitro. Blood 94:550–559 [PubMed] [Google Scholar]
  48. Wessely O, Deiner EM, Beug H, von Lindern M 1997 The glucocorticoid receptor is a key regulator of the decision between self renewal and differentiation in erythroid progenitors. EMBO J 16:267–280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O, Bock R, Gass P, Schmid W, Herrlich P, Angel P, Schütz G 1998 DNA binding of the glucocorticoid receptor is not essential for survival. Cell 93:531–541 [DOI] [PubMed] [Google Scholar]
  50. Stellacci E, Di Noia A, Di Baldassarre A, Migliaccio G, Battistini A, Migliaccio AR 2009 Interaction between the glucocorticoid and erythropoietin receptors in human erythroid cells. Exp Hematol 37:559–572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Shim GJ, Wang L, Andersson S, Nagy N, Kis LL, Zhang Q, Mäkelä S, Warner M, Gustafsson JA 2003 Disruption of the estrogen receptor β gene in mice causes myeloproliferative disease resembling chronic myelogenous leukemia with lymphoid blast crisis. Proc Natl Acad Sci USA 100:6694–6699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Erben RG, Raith S, Eberle J, Stangassinger M 1998 Ovariectomy augments B lymphopoiesis and generation of monocyte-macrophage precursors in rat bone marrow. Am J Physiol 274:E476–E483 [DOI] [PubMed] [Google Scholar]
  53. Medina KL, Smithson G, Kincade PW 1993 Suppression of B lymphopoiesis in normal pregnancy. J Exp Med 178:1507–1515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Medina KL, Garrett KP, Thompson LF, Rossi MI, Payne KJ, Kincade PW 2001 Identification of very early lymphoid precursors in bone marrow and their regulation by estrogen. Nat Immunol 2:718–724 [DOI] [PubMed] [Google Scholar]
  55. Kouro T, Medina KL, Ortani K, Kincade PW 2001 Characteristics of early murine B-lymphocyte precursors and their direct sensitivity to negative regulators. Blood 97:2708–2715 [DOI] [PubMed] [Google Scholar]
  56. Thurmond TS, Murante FG, Staples JE, Silverstone AE, Korach KS, Gasiewicz TA 2000 Role of estrogen receptor α in hematopoietic stem cell development and B lymphocyte maturation in the male mouse. Endocrinology 141:2309–2318 [DOI] [PubMed] [Google Scholar]
  57. Parker BW, Kaur G, Nieves-Neira W, Taimi M, Kohlhagen G, Shimizu T, Losiewicz MD, Pommier Y, Sausville EA, Senderowicz AM 1998 Early induction of apoptosis in hematopoietic cell lines after exposure to flavoperidol. Blood 91:458–465 [PubMed] [Google Scholar]
  58. Bagatell CJ, Bremner WJ 1996 Androgens in men—uses and abuses. N Engl J Med 334:707–714 [DOI] [PubMed] [Google Scholar]
  59. Bain J 2001 Andropause: testosterone replacement therapy for aging men. Can Fam Physician 47:91–97 [PMC free article] [PubMed] [Google Scholar]
  60. Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh AB, Bhasin S 2008 Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. J Clin Endocrinol Metab 93:914–919 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Paulo LG, Fink GD, Roh BL, Fisher JW 1974 Effects of several androgens and steroid metabolites on erythropoietin production in the isolated perfused dog kidney. Blood 43:39–47 [PubMed] [Google Scholar]
  62. Schooley J, Garcia J 1965 Some properties of serum obtained from rabbits immunized with human urinary erythropoietin. Blood 20:204 [PubMed] [Google Scholar]
  63. Leberbauer C, Boulmé F, Unfried G, Huber J, Beug H, Müllner EW 2005 Different steroids co-regulate long-term expansion versus terminal differentiation in primary human erythroid progenitors. Blood 105:85–94 [DOI] [PubMed] [Google Scholar]
  64. Shiina H, Matsumoto T, Sato T, Igarashi K, Miyamoto J, Takemasa S, Sakari M, Takada I, Nakamura T, Metzger D, Chambon P, Kanno J, Yoshikawa H, Kato S 2006 Premature ovarian failure in androgen receptor-deficient mice. Proc Natl Acad Sci USA 103:224–229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Ellis TM, Moser MT, Le PT, Flanigan RC, Kwon ED 2001 Alterations in peripheral B cells and B cell progenitors following androgen ablation in mice. Int Immunol 13:553–558 [DOI] [PubMed] [Google Scholar]
  66. O'Kelly J, Hisatake J, Hisatake Y, Bishop J, Norman A, Koeffler HP 2002 Normal myelopoiesis but abnormal T lymphocyte responses in Vitamin D receptor knockout mice. J Clin Invest 109:1091–1099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Reichel H, Koeffler HP, Norman AW 1989 The role of the Vitamin D endocrine system in health and disease. N Engl J Med 320:980–991 [DOI] [PubMed] [Google Scholar]
  68. Kizaki M, Norman AW, Bishop JE, Lin CW, Karmakar A, Koeffler HP 1991 1,25-Dihydroxyvitamin D3 receptor mRNA: expression in hematopoietic cells. Blood 77:1238–1247 [PubMed] [Google Scholar]
  69. Gemelli C, Orlandi C, Zanocco Marani T, Martello A, Vignudelli T, Ferrari F, Montanari M, Parenti S, Testa A, Grande A, Ferrari S 2008 The vitamin D3/HoxA10 pathway supports MafB function during the monocyte differentiation of human CD34+ hematopoietic progenitors. J Immunol 181:5660–5672 [DOI] [PubMed] [Google Scholar]
  70. Lemire JM, Adams JS, Kermani-Arab V, Bakke AC, Sakai R, Jordan SC 1985 1,25 Dihydroxyvitamin D3 suppresses human T helper/inducer lymphocyte activity in vitro. J Immunol 134:3032–3035 [PubMed] [Google Scholar]
  71. Freedman BD, Lee EJ, Park Y, Jameson JL 2005 A dominant negative peroxisome proliferator-activated receptor γ knock-in mouse exhibits features of the metabolic syndrome. J Biol Chem 280:17118–17125 [DOI] [PubMed] [Google Scholar]
  72. Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, Malsen GL, Williams TD, Lewis H, Shafer AJ, Chatterjee VK, O'Rahilly S 1999 Dominant negative mutations in human PPAR γ associated with severe insulin resistance, diabetes and hypertension. Nature 402:880–883 [DOI] [PubMed] [Google Scholar]
  73. Yki-Järvinen H 2004 Thiazolidinediones. N Engl J Med 351: 1106–1118 [DOI] [PubMed] [Google Scholar]
  74. Chang L, Villacorta L, Zhang J, Garcia-Barrio MT, Yang K, Hamblin M, Whitesall SE, D'Alecy LG, Chen YE 2009 Vascular smooth muscle cell-selective peroxisome proliferator-activated receptor γ deletion leads to hypotension. Circulation 119:2161–2169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Nagasawa E, Abe Y, Nishimura J, Yanase T, Nawata H, Muta K 2005 Pivotal role of peroxisome proliferator-activated receptor γ in regulation of erythroid progenitor cell proliferation and differentiation. Exp Hematol 33:857–864 [DOI] [PubMed] [Google Scholar]
  76. Prost S, Le Dantec M, Augé S, Le Grand R, Derdouch S, Auregan G, Déglon N, Relouzat F, Aubertin AM, Maillere B, Dusanter-Fourt I, Kirszenbaum M 2008 Human and simian immunodeficiency viruses deregulate hematopoiesis through a NEF/PPARγ/STAT5 signaling pathway in macaques. J Clin Invest 118:1765–1775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR, Koder A, Evans RM 1999 PPARγ is required for placental, cardiac and adipose tissue development. Mol Cell 4:585–595 [DOI] [PubMed] [Google Scholar]
  78. Rieusset J, Seydoux J, Anghel SI, Escher P, Michalik L, Soon Tan N, Metzger D, Chambon P, Wahli W, Desvergne B 2004 Altered growth in male peroxisome proliferator-activated receptor γ (PPAR γ) heterozygous mice: involvement of PPARγ in a negative feedback regulation of growth hormone action. Mol Endocrinol 18:2363–2377 [DOI] [PubMed] [Google Scholar]
  79. Mullican SE, Zhang S, Konopleva M, Ruvolo V, Andreeff M, Milbrandt J, Conneely OM 2007 Abrogation of nuclear receptors Nr4a3 and Nr4a1 leads to development of acute myeloid leukemia. Nat Med 13:730–735 [DOI] [PubMed] [Google Scholar]
  80. Rajpal A, Cho YA, Yelent B, Koza-Taylor PH, Li D, Chen E, Whang M, Kang C, Turi TG, Winoto A 2003 Transcriptional activation of known and novel apoptotic pathways by Nur77 orphan steroid receptor. EMBO J 22:6526–6536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Lee JM, Lee KH, Weidner M, Osborne BA, Hayward SD 2002 Epstein Barr virus EBNA2 blocks Nur77-mediated apoptosis. Proc Natl Acad Sci USA 99:11878–11883 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Lee SL, Wesselschmidt RL, Linette GP, Kanagawa O, Russell JH, Milbrandt J 1995 Unimpaired thymic and peripheral T cell death in mice lacking the nuclear receptor NGFI-B (Nur77). Science 269:532–535 [DOI] [PubMed] [Google Scholar]
  83. Ponnio T, Burton Q, Pereira FA, Wu DK, Conneely OM 2002 The nuclear receptor Nor-1 is essential for proliferation of the semicircular canals of the mouse inner ear. Mol Cell Biol 22:935–945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Steidl U, Rosenbauer F, Verhaak RG, Gu X, Ebralidze A, Otu HH, Klippel S, Steidl C, Bruns I, Costa DB, Wagner K, Aivado M, Kobbe G, Valk PJ, Passegué E, Leibermann TA, Delwel R, Tenen DG 2006 Essential role of Jun family transcription factors in PU.1 knockdown induced leukemic stem cells. Nat Genet 38:1269–1277 [DOI] [PubMed] [Google Scholar]
  85. Passegué E, Jochum W, Schorpp-Kistner M, Möhle-Steinlein U, Wagner EF 2001 Chronic myeloid leukemia with increased granulocyte progenitors in mice lacking JunB expression in the myeloid lineage. Cell 104:21–32 [DOI] [PubMed] [Google Scholar]
  86. Mullen EM, Gu P, Cooney AJ 2007 Nuclear receptors in regulation of mouse ES cell pluripotency and differentiation. PPAR Res 2007:61563–61572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Luo J, Sladek R, Bader JA, Matthyssen A, Rossant J, Giguère V 1997 Placental abnormalities in mice lacking the orphan nuclear receptor ERR-β. Nature 388:778–782 [DOI] [PubMed] [Google Scholar]
  88. Pettersson K, Svensson K, Mattsson R, Carlsson B, Ohlsson R, Berkenstam A 1996 Expression of a novel member of estrogen response element-binding nuclear receptors is restricted to the early stages of chorion formation during mouse embryogenesis. Mech Dev 54:211–223 [DOI] [PubMed] [Google Scholar]
  89. Ivanova N, Dobrin R, Lu R, Kotenko I, Levorse J, DeCoste C, Schafer X, Lun Y, Lemischka IR 2006 Dissecting self-renewal in stem cells with RNA interference. Nature 442:533–538 [DOI] [PubMed] [Google Scholar]
  90. Ikeda Y, Shen WH, Ingraham HA, Parker KL 1994 Developmental expression of mouse steroidogenic factor-1, an essential regulator of the steroid hydroxylases. Mol Endocrinol 8:654–662 [DOI] [PubMed] [Google Scholar]
  91. Sadovsky Y, Crawford PA, Woodson KG, Polish JA, Clements MA, Tourtellotte LM, Simburger K, Milbrandt J1995 Mice deficient in the orphan receptor steroidogenic factor-1 lack adrenal glands and gonads but express P450 side chain cleavage enzyme in the placenta and have normal embryonic serum levels of cortiocosteroids. Proc Natl Acad Sci USA 92:10939–10943 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Gu P, Goodwin B, Chung AC, Xu X, Wheeler DA, Price RR, Galardi C, Peng L, Latour AM, Koller BH, Gossen J, Kliewer SA, Cooney AJ 2005 Orphan nuclear receptor LRH-1 is required to maintain Oct4 expression at the epiblasts stage of embryonic development. Mol Cell Biol 25:3492–3505 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Yang HM, Do HJ, Kim DK, Park JK, Chang WK, Chung HM, Choi SY, Kim JH 2007 Transcriptional regulation of human Oct4 by steroidogenic factor-1. J Cell Biochem 101:1198–1209 [DOI] [PubMed] [Google Scholar]
  94. Paré JF, Malenfant D, Courtemanche C, Jacob-Wagner M, Roy S, Allard D, Bélanger L 2004 The fetoprotein transcription factor (FTF) is essential to embryogenesis and cholesterol homeostasis and is regulated by a DR4 response element. J Biol Chem 279:21206–21216 [DOI] [PubMed] [Google Scholar]
  95. Burris TP, Guo W, McCabe ER 1996 The gene responsible for adrenal congenital, DAX-1, encodes a nuclear hormone receptor that defines a new class within the superfamily. Recent Prog Horm Res 51:241–260 [PubMed] [Google Scholar]
  96. Niakan KK, Davis EC, Clipsham RC, Jiang M, DeHart DB, Sulik KK, McCabe ER 2006 Novel role for the orphan nuclear receptor DAX-1 in embryogenesis, different from steroidogenesis. Mol Genet Metab 88:261–271 [DOI] [PubMed] [Google Scholar]
  97. Chung AC, Katz D, Pereira FA, Jackson KJ, DeMayo FJ, Cooney AJ, O'Malley BW 2001 Loss of orphan receptor germ cell nuclear factor function results in ectopic development of the tail bud and a novel posterior truncation. Mol Cell Biol 21:663–677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. David R, Joos TO, Dreyer C 1998 Anteroposterior patterning and organogenesis of Xenopus laevis require a correct dose of germ cell nuclear factor (xGCNF). Mech Dev 79:137–152 [DOI] [PubMed] [Google Scholar]
  99. Gu P, LeMenuet D, Chung AC, Mancini M, Wheeler DA, Cooney AJ 2005 Orphan nuclear receptor GCNF is required for the repression of pluripotency genes during retinoic acid induced embryonic stem cell differentiation. Mol Cell Biol 25:8507–8519 [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
  100. Gu P, Le Menuet D, Chung AC, Cooney AJ 2006 Differential recruitment of methylated CpG binding domains by the orphan receptor GCNF initiates the repression and silencing of Oct4 expression. Mol Cell Biol 26:9471–9483 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  101. Mangelsdorf D 2009 Expression profiling of nuclear receptors in human and mouse embryonic stem cells. NURSA Datasets March (www.nursa.org/10.1621/datasets.05005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Takahashi K, Yamanaka S 2006 Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663–676 [DOI] [PubMed] [Google Scholar]
  103. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, Hochedlinger K, Bernstein B, Jaenisch R 2007 In vitro reprogramming of fibroblasts into a pluripotent ES cell-like state. Nature 448:318–324 [DOI] [PubMed] [Google Scholar]
  104. Feng B, Jiang J, Kraus P, Ng JH, Heng JC, Chan YS, Yaw LP, Zhang W, Loh YH, Han J, Vega VB, Cacheux-Rataboul V, Lim B, Lufkin T, Ng HH 2009 Reprogramming of fibroblasts into induced pluripotent stem cells with orphan nuclear receptor Esrrb. Nat Cell Biol 11:197–203 [DOI] [PubMed] [Google Scholar]

Articles from Molecular Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES