In the human genome, 43 different genes are found that encode proteins belonging to the family of the POK (poxvirus and zinc finger and Krüppel)/ZBTB (zinc finger and broad complex, tramtrack, and bric à brac) factors. Generally considered transcriptional repressors, several of these genes play fundamental roles in cell lineage fate decision in various tissues, programming specific tasks throughout the life of the organism. Here, we focus on functions of leukemia/lymphoma-related factor/POK erythroid myeloid ontogenic factor, which is probably one of the most exciting and yet enigmatic members of the POK/ZBTB family.

The Drosophila broad complex, tramtrack, and bric à brac factors (BTB) are all zinc finger (ZF) transcriptional repressors characterized by BTB, a unique N-terminal domain. These genes play fundamental roles during Drosophila development, including metamorphosis, central nervous system organization,1,2  ommatidial cell development,3  ovary morphogenesis,4  homeotic transformation of the bristle pattern of tarsal segments,5  and wing and limb formation.4,6 

This family of genes has expanded over time to comprise, in mammals, a group of 43 different BTB/poxvirus and zinc finger (POZ)-ZF (BTB alias POZ-ZF) transcription factors playing key functions in a spectrum of diverse biological processes such as cell cycle progression, DNA damage responses, apoptosis, cell fate determination, and a multitude of developmental processes.7  Accordingly, dysfunction of vertebrate POZ-ZF proteins such as promyelocytic leukemia ZF (PLZF), B-cell lymphoma 6 (BCL6), hypermethylated in cancer 1, ZBTB7, and Fanconi anemia ZF (FAZF/PLZP) has been linked to developmental disorders and tumorigenesis.7 

Here we focus our attention on leukemia/lymphoma-related factor (LRF)/Pokemon (POK [POZ and Krüppel] erythroid myeloid ontogenic factor), one of the most intriguing members of the BTB/POZ-ZF family, which has reached prominence in view of its pleiotropic role in the control of critical processes within the hemopoietic compartment and beyond.

The transcription factor LRF (also known as Pokemon, osteoclast-derived ZF, FBI-1, and ZBTB7A) is characterized by a peculiar protein structure shared by 43 POK proteins in humans.8,9  This family of proteins contains a POZ/BTB domain at the N terminus and multiple Krüppel-type ZFs at the C terminus (Figure 1A). Although the POZ/BTB domain is involved in protein–protein interactions such as homo- and possibly heterodimerization and multimerization of POK family members (Figure 1B), the ZF region mediates sequence-specific binding to DNA elements (Figure 1B). Finally, the poorly conserved hinge region between the POZ and ZF domains, as well as the C terminus at the end of the ZFs domains, are often the targets of posttranslational modifications responsible for the regulation of protein function (Figure 1A).10-18 

Figure 1

LRF protein structure, modifications, and interactions. (A) LRF protein domains and posttranslational modifications. (B) LRF protein interactions.

Figure 1

LRF protein structure, modifications, and interactions. (A) LRF protein domains and posttranslational modifications. (B) LRF protein interactions.

Close modal

Multiple POK proteins, LRF included, have been shown to act as transcriptional repressors by directly binding specific consensus sequences on DNA and interacting with corepressors such as NCoR, SMRT, and Sin3a via the POZ domain at the N terminus.19,20  For its part, LRF preferentially binds to the GC-rich sequence [(G/A)(C/A)GACCCC], as has been revealed by cyclic amplification and selection of target analysis,21  gel-shift assay,22  and chromatin immunoprecipitation sequencing.23  Ultimately, this binding leads to the recruitment of histone deacetylases to gene promoters and results in a closed chromatin conformation that is refractory to transcription. Recently, however, the inventory of LRF/Pokemon interactors has been expanded to include other transcription factors such as tumor protein p53 (TP53), androgen receptor, specificity protein 1, BCL6, sex determining region Y-box 9 (SOX9), and growth factor independent 1, thereby suggesting an indirect transcriptional repressive activity of LRF on specific subclasses of genes (Figure 1B).10-18  These findings in turn highlight LRF as a key node for the transcriptional regulation of fundamental pathways involved in cell cycle control, apoptosis, and cell fate decision.

Erythroid

The role for POK family proteins such as LRF, BCL6, and PLZF in tumorigenesis was initially inferred from genetic studies in human hemopoietic malignancies and was finally confirmed through the study of genetically engineered knockout and transgenic mouse models.21,24-29 

An additional and striking observation derived from these mouse models, however, was the realization of the central role played by POK family proteins in a variety of developmental processes. For instance, Bcl6-deficient mice lack formation of germinal centers (GCs) and display a profound Th2-type inflammatory response, demonstrating the key role played by BCL6 in the regulation of lymphocyte differentiation.24  In addition, mice lacking Plzf expression display defects in limb morphogenesis and germline stem cell maintenance resulting from alterations in cell proliferation, apoptosis, and self-renewal.9,30 

Studies regarding the developmental functions of LRF have likewise brought unique insight into the cellular pathways regulated by this protein, especially in the hematopoietic cell lineages (Figure 2).31,32 Lrf−/− embryos exhibit an embryonic lethality around 16.5 days postcoitum as a consequence of extensive anemia.31  Pandolfi and colleagues defined a strong apoptotic induction of late-stage erythroblasts as the primary cause of lethality in Lrf-null embryos.31  This cell death response occurs despite an intact erythropoietin signaling pathway and in an Arf/p53-independent manner and is associated with strong upregulation of the proapoptotic factor BCL2 interacting mediator of cell death (Bim).32  Importantly, these studies identified LRF as a new direct transcriptional target of GATA1 that is essential to the transcriptional repression of the proapoptotic factor Bim, thus defining a novel transcriptional cascade for the suppression of apoptosis during erythroid cell fate decision (Figure 3).32 

Figure 2

LRF in the hematopoietic cell lineages. LRF regulates hematopoiesis by playing specific roles in different cell lineages.

Figure 2

LRF in the hematopoietic cell lineages. LRF regulates hematopoiesis by playing specific roles in different cell lineages.

Close modal
Figure 3

LRF in the erythrocytes lineage. (A) LRF promotes erythrocyte differentiation. (B) GATA1-dependent LRF upregulation drives a potent antiapoptotic activity during the late stage of erythroblast differentiation through (C) BIM transcriptional repression.

Figure 3

LRF in the erythrocytes lineage. (A) LRF promotes erythrocyte differentiation. (B) GATA1-dependent LRF upregulation drives a potent antiapoptotic activity during the late stage of erythroblast differentiation through (C) BIM transcriptional repression.

Close modal

It is worth noting that erythroid Krüppel-like factor (Eklf, also known as Klf1) upregulates Lrf expression in mouse fetal liver, where erythropoiesis takes place at the late embryonic stage.33  Furthermore, Cantor and colleagues recently showed that Gata1 and Lrf cooccupy regulatory elements of key erythroid genes,34  suggesting that Gata1 not only trans-activates the Lrf gene32  but also functions with LRF on erythroid genes regulation.34 

Myeloid

Although LRF is abundantly expressed in normal and malignant myeloid cells, hematopoietic-specific Lrf conditional knockout mice (LrfFlox/Flox;Mx1-Cre) did not exhibit a gross defect in the numbers of mature myeloid lineage cells in peripheral blood or bone marrow; however, mature myeloid cells (Gr-1+CD11b+c-Kit) were barely detectable in Lrf/ fetal livers.35  Furthermore, a slight but significant reduction in the numbers of granulocyte-macrophage progenitors was observed in Lrf/ fetal liver and in LrfFlox/Flox;Mx1-Cre bone marrow.31  It remains to be clarified whether Lrf intrinsically functions during myeloid development in a developmental stage–specific manner or does so in a non-cell-autonomous fashion (eg, niche effects).

Lymphoid

Inactivation of LRF dictates dramatic consequences in the lymphoid compartment. Cre-lox mediated Lrf inactivation at hematopoietic stem cell (HSC)/progenitor stages in adult mice (LrfFlox/Flox;Mx1-Cre) leads to development and accumulation of CD4/8 double-positive T-cells in the bone marrow at the expense of B lymphopoiesis.31  The number of pro-B, pre-B, and immature B-cells is drastically reduced, but prepro-B-cells, which resemble normal thymic CD4/8 double-negative T-cells, accumulate.31  Interestingly, LrfFlox/Flox;Mx1-Cre mice phenocopied mice overexpressing the intracellular domain of Notch1, in that ectopic CD4/CD8 double-positive cell development was induced at the expense of B-cell development in the bone marrow.36  Furthermore, treatment of LrfFlox/Flox;Mx1-Cre mice with γ secretase inhibitors, which block Notch signaling, did rescue aberrant lymphoid development, suggesting that LRF antagonizes the Notch pathway at the HSC/progenitor level.31 

Our understanding of the role of LRF in peripheral T-cell differentiation continues to evolve. Observations of the thymus have shown LRF to be suppressed in CD4/8 double-positive T cells there and re-expressed in CD4 or CD8 single-positive T cells via as-yet-unknown mechanisms, although LRF deficiency was not observed to grossly affect CD4/CD8 T-cell differentiation.31  Further light was shed on these findings when a new and unexpected role for LRF in peripheral T-cell function was recently revealed.37 

Previous reports had suggested that Thpok (T-helper-inducing POZ/Kruppel like factor; also known as ZBTB7B) functions as a master regulator of CD4+ T-cells, as in its absence, CD4+CD8+ double-positive T-cells preferentially differentiate into CD8+ T− cells in the thymus.38,39  It was therefore presumed that Thpok was essential not only for CD4+ T-cell differentiation but also for T helper (Th) cell functions. Carpenter et al37  demonstrated, however, that Thpok is dispensable for many features of Th cell differentiation and that Lrf promotes Th cell gene expression in Thpok-deficient cells. In this study, analysis of Lrf/Thpok double-knockout T cells revealed that mutant cells fail to express Th cell genes or undergo Th cell differentiation in vivo, suggesting that these 2 transcription factors critically regulate Th cell gene expression in a cooperative fashion (Figure 4). However, the precise mechanisms by which LRF and THPOK maintain Th cell genes transcription remain elusive.

Figure 4

LRF roles in lymphoid differentiation. LRF promotes B-cell lineage by repressing Notch activity in early lymphoid precursors, whereas LRF regulates mature B-cell lineage fate and GC formation through distinct mechanisms.

Figure 4

LRF roles in lymphoid differentiation. LRF promotes B-cell lineage by repressing Notch activity in early lymphoid precursors, whereas LRF regulates mature B-cell lineage fate and GC formation through distinct mechanisms.

Close modal

Finally, LRF is also highly expressed in GC B-cells and non-Hodgkin lymphoma tissues,21,40  implying that LRF may also function in GC formation and lymphomagenesis, as does BCL6. Indeed, GC-B cells are dramatically reduced in B-cell-specific Lrf conditional knockout mice (LrfFlox/Flox;Mb1-Cre) after immunization with T-cell-dependent antigens.40  Although Bcl6 knockout mice exhibit complete loss of GC formation,24  a few GC B-cells were observed, and overall GC structures remain intact in LrfFlox/Flox;Mb1-Cre mice. The tumor suppressor p19Arf, a transcriptional target of LRF,21  was significantly upregulated in LRF-deficient GC B-cells, and de-repression of the p19Arf gene accounts for, at least in part, the impaired proliferation and increased apoptosis seen in LRF-deficient GC B-cells.40  Notably, despite its critical role in lymphoid lineage fate determination at the HSC/progenitor stages, LRF was found to be dispensable to the maintenance of immature B cells in the bone marrow of LrfFlox/Flox;Mb1-Cre mice. These findings were also consistent with the fact that treatment with γ secretase inhibitor restored normal B-cell development in LrfFlox/Flox;Mx1-Cre mice.31 LrfFlox/Flox;Mb1-Cre mice also exhibit an increase in marginal zone B (MZB) cell numbers and a concomitant decrease in follicular B (FOB) cell numbers, suggesting that LRF-mediated signals favor FOB fate at the branching point for the FOB vs MZB fate decision and regulate the balance between FOB and MZB development (Figure 4).40 

HSCs: LRF’s role in cell fate decision goes beyond cell autonomy

Cell fate determination by positional cues is a fundamental mechanism in animal and plant development. Physiologically, a non-cell-autonomous trait occurs whenever a cell population instructs other cells, via direct contact or secreted molecules, how to develop or behave.

The stem cell niche represents a classic example of cell fate determination dictated by a non-cell-autonomous mechanism. A stem cell niche is defined as an environment in which stem cells are not only hosted but also supported in their self-renewal41  and are instructed to differentiate into specific cell lineages. In mammals, stem cell niches have been described in many different organs: HSC niches in the bone marrow,42  the epithelial stem cell niche in the skin,43  the intestinal stem cell niche,44  neural stem cell niches in the brain,45  and the germ-line stem cell niche, which was identified in mouse testis.46 

HSCs and their bone marrow niche together make up one of the most studied microenvironment systems. It is widely believed that the regulation of the balance between HSC self-renewal and differentiation is mainly dependent on the cross talk between cell-autonomous pathways intrinsically wired into HSCs and cell nonautonomous signaling through the interaction of HSCs, with the different types of cells forming the bone marrow niche (Figure 5).

Figure 5

Cell nonautonomous function of LRF in LT-HSCs differentiation. (A) Cell nonautonomous inhibition of the Notch pathway and block of LT-HSCs differentiation through (B) LRF-dependent downregulation of Dll4 in the erythroblastic islands.

Figure 5

Cell nonautonomous function of LRF in LT-HSCs differentiation. (A) Cell nonautonomous inhibition of the Notch pathway and block of LT-HSCs differentiation through (B) LRF-dependent downregulation of Dll4 in the erythroblastic islands.

Close modal

Regardless of their physical localization in the bone marrow, bone marrow niche components such as endothelial cells, mesenchymal cells, mesenchymal stem cells, osteoblasts, and osteoblast progenitors provide membrane-bound and secreted factors that promote the quiescence, self-renewal, migration, and differentiation of HSCs (Figure 5). Accordingly, the cellular composition of the bone marrow niche and its biological function is under intense scrutiny, as the specific contributions of different cellular residents to these processes are not yet completely delineated.

In a recent manuscript published in Blood,35  Maeda’s group provided the first evidence that differentiated hematopoietic cells, the progeny of HSCs, have the capacity to in turn regulate the fate and function of their parents (ie, undifferentiated HSCs) in a hematopoietic cell-autonomous but not HSC-autonomous circuit (Figure 5). In this study, LrfFlox/Flox;Mx1-Cre conditional mice exhibited a substantial decline in the numbers of long-term hematopoietic stem cells (LT-HSCs) and Flt3+ lymphoid biased multipotential progenitors, with a concomitant expansion of T-cell precursors. Importantly, this defect was observed to be primarily Notch1-dependent, as Notch1 loss rescued HSC defects seen in LrfFlox/Flox;Mx1-Cre mice, suggesting that Notchhigh+ CD34 LT-HSCs are hypersensitive to Notch signaling caused by LRF deficiency and that the signal is transmitted mainly through Notch1 in the most primitive LT-HSCs.35 

Δ-like 4 (Dll4) is a nonredundant Notch1 ligand in the thymus47  that is expressed primarily in endothelial cells and thymic epithelial cells, but not (or at very low levels, if any) in hematopoietic cells.35,48-50  Strikingly, clusters of Dll4-positive erythroblasts were evident in the bone marrow of LrfFlox/Flox;Mx1-Cre mice35  (Figure 5). Furthermore, in vivo anti-Dll4 treatment restored HSC numbers in LrfFlox/Flox;Mx1-Cre mice.35  These results prompted the authors to investigate the erythroblasts in the bone marrow as a potential source of the non-cell-autonomous T-cell instructive signal responsible for the LT-HSCs phenotype.

To prove this in vivo, they generated LrfFlox/Flox;ErGFP-Cre mice in which GFP/Cre fusion gene expression and its consequent Lrf inactivation were limited to the erythroblasts. Accordingly, they observed that LrfFlox/Flox;ErGFP-Cre mice strikingly phenocopied the LrfFlox/Flox;Mx1-Cre mice, showing a significant reduction in the bone marrow of the total number of LT-HSCs concomitant with aberrant T-cell differentiation and the block of B-cell development (Figure 5).35 

The role of Notch in adult HSC homeostasis remains highly controversial, as detailed in recent reviews.51-54  The study by Lee et al35  demonstrates that Notch1/Dll4-mediated signals are normally suppressed by LRF in the bone marrow, preventing HSCs from premature T-cell differentiation.

Osteoclasts

Osteoclast precursors derive from hematopoietic cells (monocytes/macrophages) that are prompted to differentiate into osteoclasts upon contact with osteoblasts or stromal cells.55  During a screening focused on cloning new factors involved in the osteoclast cell lineage specification, Kukita and colleagues identified a new transcription factor that they called osteoclast-derived ZFs, the ortholog of human LRF in the rat.56  Osteoclast differentiation is a multistep process starting with the differentiation of a monocytic cell to an osteoclast precursor, proceeding toward a committed osteoclast, and finishing with the fusion of mononuclear-committed osteoclasts into a polarized and resorbing multinucleated osteoclast (Figure 6). Spleen focus forming virus proviral integration oncogene and microphtalmia-associated transcription factor family members, as well as macrophage colony-stimulating factor, are key factors responsible for the initiation of the osteoclastogenesis; however, it is only after the induction of RANK (receptor activator of NF-κB) signals that the monocytes could transform into early osteoclast precursors.57-62  Once activated by the RANK signal, the osteoclast precursors become committed and engage the differentiation/polarization step, which requires NFATc1 (nuclear factor of activated T cells cytoplasmic 1), Rous sarcoma tyrosine kinase pp660 activation and αvβ3-integrin expression. Polarization permits the firm attachment of the mature osteoclasts to the bone cells, a fundamental prerequisite necessary to initiate their function (Figure 6).55 

Figure 6

LRF role in osteoclastogenesis. (A-B) LRF transcriptional inhibition of Nfatc1 during the early stage of osteoclast differentiation. (A-C) LRF is an essential cofactor for NFATc1 transcriptional activity during osteoclast terminal differentiation.

Figure 6

LRF role in osteoclastogenesis. (A-B) LRF transcriptional inhibition of Nfatc1 during the early stage of osteoclast differentiation. (A-C) LRF is an essential cofactor for NFATc1 transcriptional activity during osteoclast terminal differentiation.

Close modal

In a follow-up publication, Kukita and colleagues finely described the function of LRF in osteoclastogenesis.63  Taking advantage of a previous analysis of a transgenic mouse model overexpressing LRF in osteoclast progenitors under the Ctsk gene promoter, these authors linked the high levels of LRF to fragile bones and significantly lower bone mineral content and bone mineral density, as well as to a marked increase in the number of osteoclasts, but not osteoblasts.63  Mechanistically, Kukita and colleagues defined LRF as a positively regulated downstream target gene of RANKL signaling that is essential to the elevation of levels of NFATc1. Interestingly, they also showed that LRF decreases p21 expression in precursor cells but increases expression of p21 and p27 in osteoclasts, suggesting different roles for LRF in proliferating osteoclast precursors and mature osteoclasts.63 

In a recent publication, however, Takayanagi’s group has unveiled new and important insights into the function of LRF in osteoclastogenesis.64  Conditional Lrf gene deletion during the early or late stage of osteoclast development in 2 different engineered mouse models (LrfFlox/Flox;Mx1-Cre and LrfFlox/Flox;Ctsk-Cre) has revealed a biphasic role for LRF during osteoclastogenesis. In particular, LRF is found to repress osteoclast differentiation by acting as a transcriptional repressor of the Nfatc1 gene in the early phase of osteoclasts development while also acting as a coactivator of NFATc1 in controlling the genes required for the resorbing activity in terminally differentiated osteoclasts (Figure 6).64 

Although further studies will be necessary to reconcile the apparent contradiction between the results described by Takayanagi’s group and the working model proposed by Kukita and colleagues, both works contribute valuable detail to the picture of LRF as a stage-specific gene playing distinct roles in osteoclast lineage commitment.

The famous dictum known as Occam’s razor warns that “Pluralitas non est ponenda sine necessitate” (ie, “Plurality should not be posited without necessity”). What we know of LRF, however, would seem to surprisingly contradict this generally sound advice. Although it would have been tempting to hypothesize that LRF could regulate cellular differentiation in such pleiotropic manner through a unifying mechanism, this turned out not to be the case.

Research has instead revealed an unexpected plurality of roles, pathways, and functions for this transcription factor, both within and beyond the hemopoietic compartment, so that now we must contend with a complex model in which the protein controls differentiation by partnering with different key players in an utterly tissue- and context-dependent manner.

In the hemopoietic compartment it is obvious that LRF exerts its activity through very different mechanisms: BIM transcriptional repression in erythrocyte terminal differentiation, Notch repression in the early lymphoid cell fate decision, ARF transcriptional repression for the GC B-cell differentiation, NFATc1 transcriptional repression during the early steps of osteoclast cell lineage, and an increase of NFATc1 transcriptional activity for osteoclast terminal differentiation.

Furthermore, recent publications have also described this gene as profoundly involved in chondrocyte, adipocyte, and oligodendrocyte cell lineages through the regulation of completely independent pathways.

Chondrogenesis also can be considered the earliest phase of skeletal development. In skeletal cells, SOX9, together with 2 other SOX family members, L-SOX5 and SOX6, favor chondrogenesis over osteogenesis through the negative regulation of RUNX2,65-67  a potent osteogenic inducer, and by driving cartilage formation through the transcription of collagen types 2, 9, and 11 (Col2a1, Col9a1, Col11a1); Aggrecan; and the cartilage oligomeric matrix protein (COMP) genes.68-71  The COMP promoter is composed of a 30-bp negative regulator element and a 51-bp positive regulatory element. Although the 51-bp positive regulatory element contains 3 putative binding sites for SOX9, the 30-pb negative regulator element contains a binding site for LRF.68,72  To correctly establish the chondrocyte differentiation program, the LRF-SIN3a-histone deacetylase 1 repressor complex must dissociate from the COMP promoter to permit the binding of the SOX9/p300/CBP activator complex to the positive regulatory element in the COMP promoter, which induces the transcription of the COMP gene.68,72  Accordingly, micromasses of C3H10T1/2 (mesenchymal-like stem cells) overexpressing LRF are unable to differentiate toward chondrocytes when treated with bone morphogenetic protein 2,68,72  thereby defining LRF as a potent negative regulator of chondrocyte lineage fate decision, more than a simple transcriptional regulator of specific chondrocyte genes.

Although clearly distinct from chondrocytes, adipocytes are nevertheless another cell lineage derived from multipotent mesenchymal stem cells and mesenchymal precursors that reside both in the bone marrow as well as in the adipose tissues of the body. Recent observations of in vitro adipocyte differentiation from primary mesenchyme cells as well as from the 3T3L1 cell line have revealed much of the detail of this stepwise sequential process. On reaching confluence, cells arrest their cell cycle at the G1/S phase boundary. Under treatment with dexamethasone, indomethacin, and insulin, the cells then complete 2 cycles of cell division known as mitotic clonal expansion, and finally, after a second growth arrest, preadipocytes undergo terminal differentiation and acquire all the characteristics of mature adipocytes.73 

The genetic determinants of this process are likewise being unraveled. In a screening focused on identifying new genes critically involved in human and mouse adipogenesis, Laudes and colleagues74  identified LRF as the second most upregulated gene during mitotic clonal expansion, being second only to fatty acid binding protein 4 (FABP-4). Accordingly, 3T3L1 cells that overexpress LRF also exhibit accelerated lipid accumulation and early terminal differentiation capacity.74,75  Although the molecular mechanism through which LRF promotes adipocyte differentiation remains a matter of debate, it has been proposed that LRF would exert its central role through the concomitant transcriptional repression of cyclin A and E2F-4.75 

Finally, the Allen Brain Atlas describes a widespread distribution of LRF mRNA in embryonic mouse central nervous system, suggesting a potential role for LRF in neural and/or glial development. Accordingly, Armstrong’s group76  has recently demonstrated that knockout mouse models characterized by specific Lrf gene loss conditionally in the oligodendrocyte progenitors (OPs) at P7 are characterized by a strong increase in the number of NG2+ OPs and decreases in CC1+ mature oligodendrocytes at P28.76  This imbalance is mainly a result of the inhibition of OP differentiation into oligodendrocytes without any evident effect on OP proliferation or oligodendrocyte cell death. These in vitro results are again intriguing and are in line with the pleiotropic role of LRF in the control of cell fate determination, whereas the molecular mechanisms underlying this process have yet to be defined.

A flurry of recent studies have conclusively proved the thesis that tumor suppressor genes and proto-oncogenes act as key factors regulating cell fate decision and tissue differentiation not only during embryonic development but also in adult tissues. Accordingly, the reactivation of such pathways as HEDGEHOG, WNT, NOTCH, SOX, and TGF-β is now seen as a turning point in tumor development, progression, and drug resistance in a range of malignancies.

Multiple members of the POK protein family align perfectly with this scenario, as they have been described as key regulators of important developmental processes and have shown striking positive or negative roles in neoplastic transformation.

Plzf-null mice, for instance, display severe defects in invariant natural killer T-cell differentiation, as well as in limb development and germ stem cell maintenance.77-81  Taking advantage of Plzf knockout mouse models, it has been also demonstrated, however, that PLZF acts as a tumor suppressor gene in the context of acute promyelocytic leukemia,28  whereas transgenic models have defined the causality of PLZF gene chromosomal translocation t(11;17) in human acute promyelocytic leukemia.29  Accordingly, transduction of cell lines with a PLZF construct results in consistent G1-phase cell cycle arrest resulting from PLZF-mediated transcriptional repression of the proto-oncogene c-myc.27 

A second compelling example of POK family involvement in cancer pathogenesis is represented by BCL6. Both in vitro and in vivo mouse model studies have demonstrated the essential role of BCL6 in B-cell GC establishment and/or maintenance,24,82  as well as the ability of BCL6 to act as a potent oncogene in non-Hodgkin’s lymphoma through its ability to repress the expression of tumor suppressors and DNA-damage-sensing genes such as TP53 and ATR.83,84 

POK proteins are therefore key developmental regulators and important players in the pathogenesis of human cancer that are found to act either as oncogenes or oncosuppressors, and LRF is not an exception.

LRF may play an even more complex and multifaceted role in tumorigenesis than has been described for other POK family members because of its critical role in a plethora of different lineage fate decisions and terminal cell differentiation. Transgenic mice overexpressing LRF in the immature B- and T-cell compartments (lckEμ-Lrf) offered the first in vivo demonstration of a proto-oncogenic role for this gene.21 lckEμ-Lrf transgenic mice develop an aggressive and fatal T-cell lymphoblastic lymphoma/leukemia, strongly suggesting that LRF, similar to BCL6, may drive human lymphoma when ectopically overexpressed, as in non-Hodgkin lymphoma and nodular lymphocyte-predominant Hodgkin lymphoma21,85  (Julie Teruya-Feldstein, T.M., and P.P.P., manuscript in preparation). Furthermore, primary mouse embryonic fibroblasts generated from Lrf-null mouse embryos were found to be refractory to transformation by multiple classical oncogene combinations such as E1A+Ras or Myc+Ras.21  A remarkable feature of cultured Lrf-null mouse embryonic fibroblasts is their premature growth arrest and senescence on passaging when compared with wild-type cells. This premature senescence response is associated with a marked overexpression of the p19Arf tumor suppressor and subsequent Trp53 activation.

Accordingly, the promoters of both human and mouse Arf genes contain functional binding sites for, and are strictly regulated by, LRF. Thus, LRF is able to regulate expression of a key gene involved in p53 pathway activity, suggesting a molecular basis for the oncogenic role of the protein in lymphoma development.

Finally, shRNA-mediated LRF knockdown results in toxicity to a subset of B-cell lymphoma cell lines.37  Given that most lymphoma cell lines harbor the mutation in the ARF-p53 pathway, it is likely that LRF could also exert its oncogenic function through p53-independent mechanisms. Notably, it has recently been reported that LRF is overexpressed in non–small cell lung carcinomas in breast and ovarian cancer,86-90  reinforcing the idea of LRF as a proto-oncogene with multiple oncogenic activities.

Intriguingly, however, the ability of LRF to promote terminal differentiation in so many different cell lineages by antagonizing a number of oncogenic pathways such as SOX9, NOTCH, E2F4, or Cyclin A, together with a novel and key function of LRF in the control of genomic stability (Liu Xue Song et al, manuscript in preparation), suggest a range of potential oncosuppressive functions for this protein in specific cell systems and tumor types.91-96  Accordingly, for instance, downregulation of LRF in primary advanced prostate cancer accompanied by heterozygous genetic loss of the LRF gene in castration-resistant metastatic advanced prostate cancer have been recently described in subgroups of patients and directly linked to tumor progression and androgen deprivation resistance (G.W. et al, manuscript submitted December 2012; A.L. et al, manuscript submitted December 2012). In line with a possible context-dependent oncosuppressive function of LRF, although LRF is ectopically overexpressed in human non-Hodgkin lymphoma and nodular lymphocyte-predominant Hodgkin lymphoma,21  Reed-Sternberg cells in classical Hodgkin lymphoma show low to absent LRF expression (Julie Teruya-Feldstein, T.M., and P.P.P., manuscript in preparation).

In such a tumor-suppressive role, loss of LRF could therefore bestow to the cancer cell a progenitor/stem cell–like state by causing a block in terminal cellular differentiation (Figure 7).

Figure 7

LRF in tumorigenesis. Examples of context-dependent and dose-dependent oncogenic or oncosuppressive functions of LRF.

Figure 7

LRF in tumorigenesis. Examples of context-dependent and dose-dependent oncogenic or oncosuppressive functions of LRF.

Close modal

What is truly remarkable and rather unique regarding LRF is the realization that this transcription factor is essential for the proper cell fate decision of virtually every tissue in which it is expressed, both positively and negatively, through its ability to modulate a number of different pathways.

It is a matter of fact, as supported by numerous in vitro and in vivo studies, that LRF controls fundamental pathways in erythroid, lymphoid, myeloid, osteoclast, adipocytic, chondrocytic, and glial cell lineage fate decisions by partnering with or opposing distinct and context-dependent transcriptional players. These findings in turn suggest unexpected implications regarding the tissue- and context-dependent oncogenic or oncosuppressive roles of this enigmatic and critical transcription factor.

The authors thank Thomas Garvey for insightful editing.

A.L. was supported in part by a fellowship from the Istituto Toscano Tumori. This work was supported by grants from the National Institutes of Health, National Institute of Allergy and Infectious Diseases (R01 AI084905 04 to T.M.) and National Cancer Institute (R01 CA102142-7 to P.P.P.).

Contribution: A.L., J.G., G.W., T.M., and P.P.P. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Takahiro Maeda, Brigham and Women's Hospital, 1 Blackfan Circle, 02215 Boston, MA; e-mail: tmaeda@partners.org; and Pier Paolo Pandolfi, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, 02215 Boston, MA; e-mail: ppandolf@bidmc.harvard.edu.

1
Restifo
 
LL
White
 
K
Mutations in a steroid hormone-regulated gene disrupt the metamorphosis of the central nervous system in Drosophila.
Dev Biol
1991
, vol. 
148
 
1
(pg. 
174
-
194
)
2
Karim
 
FD
Guild
 
GM
Thummel
 
CS
The Drosophila Broad-Complex plays a key role in controlling ecdysone-regulated gene expression at the onset of metamorphosis.
Development
1993
, vol. 
118
 
3
(pg. 
977
-
988
)
3
Xiong
 
WC
Montell
 
C
tramtrack is a transcriptional repressor required for cell fate determination in the Drosophila eye.
Genes Dev
1993
, vol. 
7
 
6
(pg. 
1085
-
1096
)
4
Sahut-Barnola
 
I
Godt
 
D
Laski
 
FA
, et al. 
Drosophila ovary morphogenesis: analysis of terminal filament formation and identification of a gene required for this process.
Dev Biol
1995
, vol. 
170
 
1
(pg. 
127
-
135
)
5
Godt
 
D
Couderc
 
JL
Cramton
 
SE
, et al. 
Pattern formation in the limbs of Drosophila: bric à brac is expressed in both a gradient and a wave-like pattern and is required for specification and proper segmentation of the tarsus.
Development
1993
, vol. 
119
 
3
(pg. 
799
-
812
)
6
Maeng
 
O
Son
 
W
Chung
 
J
, et al. 
The BTB/POZ-ZF transcription factor dPLZF is involved in Ras/ERK signaling during Drosophila wing development.
Mol Cells
2012
, vol. 
33
 
5
(pg. 
457
-
463
)
7
Lee
 
SU
Maeda
 
T
POK/ZBTB proteins: an emerging family of proteins that regulate lymphoid development and function.
Immunol Rev
2012
, vol. 
247
 
1
(pg. 
107
-
119
)
8
Kelly
 
KF
Daniel
 
JM
POZ for effect—POZ-ZF transcription factors in cancer and development.
Trends Cell Biol
2006
, vol. 
16
 
11
(pg. 
578
-
587
)
9
Costoya
 
JA
Functional analysis of the role of POK transcriptional repressors.
Brief Funct Genomics Proteomics
2007
, vol. 
6
 
1
(pg. 
8
-
18
)
10
Cui
 
J
Yang
 
Y
Zhang
 
C
, et al. 
FBI-1 functions as a novel AR co-repressor in prostate cancer cells.
Cell Mol Life Sci
2011
, vol. 
68
 
6
(pg. 
1091
-
1103
)
11
Davies
 
JM
Hawe
 
N
Kabarowski
 
J
, et al. 
Novel BTB/POZ domain zinc-finger protein, LRF, is a potential target of the LAZ-3/BCL-6 oncogene.
Oncogene
1999
, vol. 
18
 
2
(pg. 
365
-
375
)
12
Choi
 
WI
Kim
 
Y
Kim
 
Y
, et al. 
Eukaryotic translation initiator protein 1A isoform, CCS-3, enhances the transcriptional repression of p21CIP1 by proto-oncogene FBI-1 (Pokemon/ZBTB7A).
Cell Physiol Biochem
2009
, vol. 
23
 
4-6
(pg. 
359
-
370
)
13
Lee
 
DK
Suh
 
D
Edenberg
 
HJ
, et al. 
POZ domain transcription factor, FBI-1, represses transcription of ADH5/FDH by interacting with the zinc finger and interfering with DNA binding activity of Sp1.
J Biol Chem
2002
, vol. 
277
 
30
(pg. 
26761
-
26768
)
14
Roh
 
HE
Lee
 
MN
Jeon
 
BN
, et al. 
Regulation of pokemon 1 activity by sumoylation.
Cell Physiol Biochem
2007
, vol. 
20
 
1-4
(pg. 
167
-
180
)
15
Beausoleil
 
SA
Jedrychowski
 
M
Schwartz
 
D
, et al. 
Large-scale characterization of HeLa cell nuclear phosphoproteins.
Proc Natl Acad Sci USA
2004
, vol. 
101
 
33
(pg. 
12130
-
12135
)
16
Dephoure
 
N
Zhou
 
C
Villén
 
J
, et al. 
A quantitative atlas of mitotic phosphorylation.
Proc Natl Acad Sci USA
2008
, vol. 
105
 
31
(pg. 
10762
-
10767
)
17
Mayya
 
V
Han
 
DK
Phosphoproteomics by mass spectrometry: insights, implications, applications and limitations.
Expert Rev Proteomics
2009
, vol. 
6
 
6
(pg. 
605
-
618
)
18
Cantin
 
GT
Yi
 
W
Lu
 
B
, et al. 
Combining protein-based IMAC, peptide-based IMAC, and MudPIT for efficient phosphoproteomic analysis.
J Proteome Res
2008
, vol. 
7
 
3
(pg. 
1346
-
1351
)
19
David
 
G
Alland
 
L
Hong
 
SH
, et al. 
Histone deacetylase associated with mSin3A mediates repression by the acute promyelocytic leukemia-associated PLZF protein.
Oncogene
1998
, vol. 
16
 
19
(pg. 
2549
-
2556
)
20
Dhordain
 
P
Lin
 
RJ
Quief
 
S
, et al. 
The LAZ3(BCL-6) oncoprotein recruits a SMRT/mSIN3A/histone deacetylase containing complex to mediate transcriptional repression.
Nucleic Acids Res
1998
, vol. 
26
 
20
(pg. 
4645
-
4651
)
21
Maeda
 
T
Hobbs
 
RM
Merghoub
 
T
, et al. 
Role of the proto-oncogene Pokemon in cellular transformation and ARF repression.
Nature
2005
, vol. 
433
 
7023
(pg. 
278
-
285
)
22
Pessler
 
F
Hernandez
 
N
Flexible DNA binding of the BTB/POZ-domain protein FBI-1.
J Biol Chem
2003
, vol. 
278
 
31
(pg. 
29327
-
29335
)
23
Wang
 
J
Zhuang
 
J
Iyer
 
S
, et al. 
Sequence features and chromatin structure around the genomic regions bound by 119 human transcription factors.
Genome Res
2012
, vol. 
22
 
9
(pg. 
1798
-
1812
)
24
Ye
 
BH
Cattoretti
 
G
Shen
 
Q
, et al. 
The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation.
Nat Genet
1997
, vol. 
16
 
2
(pg. 
161
-
170
)
25
Koken
 
MH
Reid
 
A
Quignon
 
F
, et al. 
Leukemia-associated retinoic acid receptor alpha fusion partners, PML and PLZF, heterodimerize and colocalize to nuclear bodies.
Proc Natl Acad Sci USA
1997
, vol. 
94
 
19
(pg. 
10255
-
10260
)
26
Lo Coco
 
F
Ye
 
BH
Lista
 
F
, et al. 
Rearrangements of the BCL6 gene in diffuse large cell non-Hodgkin’s lymphoma.
Blood
1994
, vol. 
83
 
7
(pg. 
1757
-
1759
)
27
McConnell
 
MJ
Chevallier
 
N
Berkofsky-Fessler
 
W
, et al. 
Growth suppression by acute promyelocytic leukemia-associated protein PLZF is mediated by repression of c-myc expression.
Mol Cell Biol
2003
, vol. 
23
 
24
(pg. 
9375
-
9388
)
28
He
 
LZ
Bhaumik
 
M
Tribioli
 
C
, et al. 
Two critical hits for promyelocytic leukemia.
Mol Cell
2000
, vol. 
6
 
5
(pg. 
1131
-
1141
)
29
Rego
 
EM
Ruggero
 
D
Tribioli
 
C
, et al. 
Leukemia with distinct phenotypes in transgenic mice expressing PML/RAR alpha, PLZF/RAR alpha or NPM/RAR alpha.
Oncogene
2006
, vol. 
25
 
13
(pg. 
1974
-
1979
)
30
Barna
 
M
Pandolfi
 
PP
Niswander
 
L
Gli3 and Plzf cooperate in proximal limb patterning at early stages of limb development.
Nature
2005
, vol. 
436
 
7048
(pg. 
277
-
281
)
31
Maeda
 
T
Merghoub
 
T
Hobbs
 
RM
, et al. 
Regulation of B versus T lymphoid lineage fate decision by the proto-oncogene LRF.
Science
2007
, vol. 
316
 
5826
(pg. 
860
-
866
)
32
Maeda
 
T
Ito
 
K
Merghoub
 
T
, et al. 
LRF is an essential downstream target of GATA1 in erythroid development and regulates BIM-dependent apoptosis.
Dev Cell
2009
, vol. 
17
 
4
(pg. 
527
-
540
)
33
Hodge
 
D
Coghill
 
E
Keys
 
J
, et al. 
A global role for EKLF in definitive and primitive erythropoiesis.
Blood
2006
, vol. 
107
 
8
(pg. 
3359
-
3370
)
34
Yu
 
M
Riva
 
L
Xie
 
H
, et al. 
Insights into GATA-1-mediated gene activation versus repression via genome-wide chromatin occupancy analysis.
Mol Cell
2009
, vol. 
36
 
4
(pg. 
682
-
695
)
35
Lee
 
SU
Maeda
 
M
Ishikawa
 
Y
, et al. 
LRF-mediated Dll4 repression in erythroblasts is necessary for hematopoietic stem cell maintenance.
Blood
2013
, vol. 
121
 
6
(pg. 
918
-
929
)
36
Pui
 
JC
Allman
 
D
Xu
 
L
, et al. 
Notch1 expression in early lymphopoiesis influences B versus T lineage determination.
Immunity
1999
, vol. 
11
 
3
(pg. 
299
-
308
)
37
Carpenter
 
AC
Grainger
 
JR
Xiong
 
Y
, et al. 
The transcription factors Thpok and LRF are necessary and partly redundant for T helper cell differentiation.
Immunity
2012
, vol. 
37
 
4
(pg. 
622
-
633
)
38
Sun
 
G
Liu
 
X
Mercado
 
P
, et al. 
The zinc finger protein cKrox directs CD4 lineage differentiation during intrathymic T cell positive selection.
Nat Immunol
2005
, vol. 
6
 
4
(pg. 
373
-
381
)
39
He
 
X
He
 
X
Dave
 
VP
, et al. 
The zinc finger transcription factor Th-POK regulates CD4 versus CD8 T-cell lineage commitment.
Nature
2005
, vol. 
433
 
7028
(pg. 
826
-
833
)
40
Sakurai
 
N
Maeda
 
M
Lee
 
SU
, et al. 
The LRF transcription factor regulates mature B cell development and the germinal center response in mice.
J Clin Invest
2011
, vol. 
121
 
7
(pg. 
2583
-
2598
)
41
Schofield
 
R
The relationship between the spleen colony-forming cell and the haemopoietic stem cell.
Blood Cells
1978
, vol. 
4
 
1-2
(pg. 
7
-
25
)
42
Wilson
 
A
Oser
 
GM
Jaworski
 
M
, et al. 
Dormant and self-renewing hematopoietic stem cells and their niches.
Ann N Y Acad Sci
2007
, vol. 
1106
 (pg. 
64
-
75
)
43
Fuchs
 
E
Finding one’s niche in the skin.
Cell Stem Cell
2009
, vol. 
4
 
6
(pg. 
499
-
502
)
44
Yen
 
TH
Wright
 
NA
The gastrointestinal tract stem cell niche.
Stem Cell Rev
2006
, vol. 
2
 
3
(pg. 
203
-
212
)
45
Doetsch
 
F
A niche for adult neural stem cells.
Curr Opin Genet Dev
2003
, vol. 
13
 
5
(pg. 
543
-
550
)
46
Spradling
 
A
Drummond-Barbosa
 
D
Kai
 
T
Stem cells find their niche.
Nature
2001
, vol. 
414
 
6859
(pg. 
98
-
104
)
47
Koch
 
U
Fiorini
 
E
Benedito
 
R
, et al. 
Delta-like 4 is the essential, nonredundant ligand for Notch1 during thymic T cell lineage commitment.
J Exp Med
2008
, vol. 
205
 
11
(pg. 
2515
-
2523
)
48
Shutter
 
JR
Scully
 
S
Fan
 
W
, et al. 
Dll4, a novel Notch ligand expressed in arterial endothelium.
Genes Dev
2000
, vol. 
14
 
11
(pg. 
1313
-
1318
)
49
Krebs
 
LT
Xue
 
Y
Norton
 
CR
, et al. 
Notch signaling is essential for vascular morphogenesis in mice.
Genes Dev
2000
, vol. 
14
 
11
(pg. 
1343
-
1352
)
50
Duarte
 
A
Hirashima
 
M
Benedito
 
R
, et al. 
Dosage-sensitive requirement for mouse Dll4 in artery development.
Genes Dev
2004
, vol. 
18
 
20
(pg. 
2474
-
2478
)
51
Liu
 
J
Sato
 
C
Cerletti
 
M
, et al. 
Notch signaling in the regulation of stem cell self-renewal and differentiation.
Curr Top Dev Biol
2010
, vol. 
92
 (pg. 
367
-
409
)
52
Pajcini
 
KV
Speck
 
NA
Pear
 
WS
Notch signaling in mammalian hematopoietic stem cells.
Leukemia
2011
, vol. 
25
 
10
(pg. 
1525
-
1532
)
53
Bigas
 
A
D’Altri
 
T
Espinosa
 
L
The Notch pathway in hematopoietic stem cells.
Curr Top Microbiol Immunol
2012
, vol. 
360
 (pg. 
1
-
18
)
54
Sandy
 
AR
Jones
 
M
Maillard
 
I
Notch signaling and development of the hematopoietic system.
Adv Exp Med Biol
2012
, vol. 
727
 (pg. 
71
-
88
)
55
Udagawa
 
N
Takahashi
 
N
Akatsu
 
T
, et al. 
Origin of osteoclasts: mature monocytes and macrophages are capable of differentiating into osteoclasts under a suitable microenvironment prepared by bone marrow-derived stromal cells.
Proc Natl Acad Sci USA
1990
, vol. 
87
 
18
(pg. 
7260
-
7264
)
56
Kukita
 
A
Kukita
 
T
Ouchida
 
M
, et al. 
Osteoclast-derived zinc finger (OCZF) protein with POZ domain, a possible transcriptional repressor, is involved in osteoclastogenesis.
Blood
1999
, vol. 
94
 
6
(pg. 
1987
-
1997
)
57
Yasuda
 
H
Shima
 
N
Nakagawa
 
N
, et al. 
Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL.
Proc Natl Acad Sci USA
1998
, vol. 
95
 
7
(pg. 
3597
-
3602
)
58
Kong
 
YY
Yoshida
 
H
Sarosi
 
I
, et al. 
OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis.
Nature
1999
, vol. 
397
 
6717
(pg. 
315
-
323
)
59
Luchin
 
A
Suchting
 
S
Merson
 
T
, et al. 
Genetic and physical interactions between Microphthalmia transcription factor and PU.1 are necessary for osteoclast gene expression and differentiation.
J Biol Chem
2001
, vol. 
276
 
39
(pg. 
36703
-
36710
)
60
Tondravi
 
MM
McKercher
 
SR
Anderson
 
K
, et al. 
Osteopetrosis in mice lacking haematopoietic transcription factor PU.1.
Nature
1997
, vol. 
386
 
6620
(pg. 
81
-
84
)
61
Teitelbaum
 
SL
Ross
 
FP
Genetic regulation of osteoclast development and function.
Nat Rev Genet
2003
, vol. 
4
 
8
(pg. 
638
-
649
)
62
Insogna
 
KL
Sahni
 
M
Grey
 
AB
, et al. 
Colony-stimulating factor-1 induces cytoskeletal reorganization and c-src-dependent tyrosine phosphorylation of selected cellular proteins in rodent osteoclasts.
J Clin Invest
1997
, vol. 
100
 
10
(pg. 
2476
-
2485
)
63
Kukita
 
A
Kukita
 
T
Nagata
 
K
, et al. 
The transcription factor FBI-1/OCZF/LRF is expressed in osteoclasts and regulates RANKL-induced osteoclast formation in vitro and in vivo.
Arthritis Rheum
2011
, vol. 
63
 
9
(pg. 
2744
-
2754
)
64
Tsuji-Takechi
 
K
Negishi-Koga
 
T
Sumiya
 
E
, et al. 
Stage-specific functions of leukemia/lymphoma-related factor (LRF) in the transcriptional control of osteoclast development.
Proc Natl Acad Sci USA
2012
, vol. 
109
 
7
(pg. 
2561
-
2566
)
65
Lengner
 
CJ
Hassan
 
MQ
Serra
 
RW
, et al. 
Nkx3.2-mediated repression of Runx2 promotes chondrogenic differentiation.
J Biol Chem
2005
, vol. 
280
 
16
(pg. 
15872
-
15879
)
66
Zhou
 
G
Zheng
 
Q
Engin
 
F
, et al. 
Dominance of SOX9 function over RUNX2 during skeletogenesis.
Proc Natl Acad Sci USA
2006
, vol. 
103
 
50
(pg. 
19004
-
19009
)
67
Dy
 
P
Wang
 
W
Bhattaram
 
P
, et al. 
Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth plate chondrocytes.
Dev Cell
2012
, vol. 
22
 
3
(pg. 
597
-
609
)
68
Liu
 
CJ
Zhang
 
Y
Xu
 
K
, et al. 
Transcriptional activation of cartilage oligomeric matrix protein by Sox9, Sox5, and Sox6 transcription factors and CBP/p300 coactivators.
Front Biosci
2007
, vol. 
12
 (pg. 
3899
-
3910
)
69
Nagy
 
A
Kénesi
 
E
Rentsendorj
 
O
, et al. 
Evolutionarily conserved, growth plate zone-specific regulation of the matrilin-1 promoter: L-Sox5/Sox6 and Nfi factors bound near TATA finely tune activation by Sox9.
Mol Cell Biol
2011
, vol. 
31
 
4
(pg. 
686
-
699
)
70
Aza-Carmona
 
M
Shears
 
DJ
Yuste-Checa
 
P
, et al. 
SHOX interacts with the chondrogenic transcription factors SOX5 and SOX6 to activate the aggrecan enhancer.
Hum Mol Genet
2011
, vol. 
20
 
8
(pg. 
1547
-
1559
)
71
Dvir-Ginzberg
 
M
Gagarina
 
V
Lee
 
EJ
, et al. 
Regulation of cartilage-specific gene expression in human chondrocytes by SirT1 and nicotinamide phosphoribosyltransferase.
J Biol Chem
2008
, vol. 
283
 
52
(pg. 
36300
-
36310
)
72
Liu
 
CJ
Prazak
 
L
Fajardo
 
M
, et al. 
Leukemia/lymphoma-related factor, a POZ domain-containing transcriptional repressor, interacts with histone deacetylase-1 and inhibits cartilage oligomeric matrix protein gene expression and chondrogenesis.
J Biol Chem
2004
, vol. 
279
 
45
(pg. 
47081
-
47091
)
73
Zhang
 
Y
Khan
 
D
Delling
 
J
Tobiasch
 
E
Mechanisms underlying the osteo- and adipo-differentiation of human mesenchymal stem cells.
ScientificWorldJournal
2012
, vol. 
2012
 pg. 
793823
 
74
Laudes
 
M
Christodoulides
 
C
Sewter
 
C
, et al. 
Role of the POZ zinc finger transcription factor FBI-1 in human and murine adipogenesis.
J Biol Chem
2004
, vol. 
279
 
12
(pg. 
11711
-
11718
)
75
Laudes
 
M
Bilkovski
 
R
Oberhauser
 
F
, et al. 
Transcription factor FBI-1 acts as a dual regulator in adipogenesis by coordinated regulation of cyclin-A and E2F-4.
J Mol Med (Berl)
2008
, vol. 
86
 
5
(pg. 
597
-
608
)
76
Dobson
 
NR
Moore
 
RT
Tobin
 
JE
, et al. 
Leukemia/lymphoma-related factor regulates oligodendrocyte lineage cell differentiation in developing white matter.
Glia
2012
, vol. 
60
 
9
(pg. 
1378
-
1390
)
77
Kovalovsky
 
D
Uche
 
OU
Eladad
 
S
, et al. 
The BTB-zinc finger transcriptional regulator PLZF controls the development of invariant natural killer T cell effector functions.
Nat Immunol
2008
, vol. 
9
 
9
(pg. 
1055
-
1064
)
78
Costoya
 
JA
Hobbs
 
RM
Barna
 
M
, et al. 
Essential role of Plzf in maintenance of spermatogonial stem cells.
Nat Genet
2004
, vol. 
36
 
6
(pg. 
653
-
659
)
79
Barna
 
M
Hawe
 
N
Niswander
 
L
, et al. 
Plzf regulates limb and axial skeletal patterning.
Nat Genet
2000
, vol. 
25
 
2
(pg. 
166
-
172
)
80
Hobbs
 
RM
Seandel
 
M
Falciatori
 
I
, et al. 
Plzf regulates germline progenitor self-renewal by opposing mTORC1.
Cell
2010
, vol. 
142
 
3
(pg. 
468
-
479
)
81
Hobbs
 
RM
Fagoonee
 
S
Papa
 
A
, et al. 
Functional antagonism between Sall4 and Plzf defines germline progenitors.
Cell Stem Cell
2012
, vol. 
10
 
3
(pg. 
284
-
298
)
82
Dent
 
AL
Shaffer
 
AL
Yu
 
X
, et al. 
Control of inflammation, cytokine expression, and germinal center formation by BCL-6.
Science
1997
, vol. 
276
 
5312
(pg. 
589
-
592
)
83
Phan
 
RT
Dalla-Favera
 
R
The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells.
Nature
2004
, vol. 
432
 
7017
(pg. 
635
-
639
)
84
Ranuncolo
 
SM
Polo
 
JM
Dierov
 
J
, et al. 
Bcl-6 mediates the germinal center B cell phenotype and lymphomagenesis through transcriptional repression of the DNA-damage sensor ATR.
Nat Immunol
2007
, vol. 
8
 
7
(pg. 
705
-
714
)
85
Wlodarska
 
I
Nooyen
 
P
Maes
 
B
, et al. 
Frequent occurrence of BCL6 rearrangements in nodular lymphocyte predominance Hodgkin lymphoma but not in classical Hodgkin lymphoma.
Blood
2003
, vol. 
101
 
2
(pg. 
706
-
710
)
86
Vredeveld
 
LC
Rowland
 
BD
Douma
 
S
, et al. 
Functional identification of LRF as an oncogene that bypasses RASV12-induced senescence via upregulation of CYCLIN E.
Carcinogenesis
2010
, vol. 
31
 
2
(pg. 
201
-
207
)
87
Qu
 
H
Qu
 
D
Chen
 
F
, et al. 
ZBTB7 overexpression contributes to malignancy in breast cancer.
Cancer Invest
2010
, vol. 
28
 
6
(pg. 
672
-
678
)
88
Apostolopoulou
 
K
Pateras
 
IS
Evangelou
 
K
, et al. 
Gene amplification is a relatively frequent event leading to ZBTB7A (Pokemon) overexpression in non-small cell lung cancer.
J Pathol
2007
, vol. 
213
 
3
(pg. 
294
-
302
)
89
Zhao
 
ZH
Wang
 
SF
Yu
 
L
, et al. 
Overexpression of Pokemon in non-small cell lung cancer and foreshowing tumor biological behavior as well as clinical results.
Lung Cancer
2008
, vol. 
62
 
1
(pg. 
113
-
119
)
90
Jiang
 
L
Siu
 
MK
Wong
 
OG
, et al. 
Overexpression of proto-oncogene FBI-1 activates membrane type 1-matrix metalloproteinase in association with adverse outcome in ovarian cancers.
Mol Cancer
2010
, vol. 
9
 pg. 
318
 
91
Miller
 
SJ
Jessen
 
WJ
Mehta
 
T
, et al. 
Integrative genomic analyses of neurofibromatosis tumours identify SOX9 as a biomarker and survival gene.
EMBO Mol Med
2009
, vol. 
1
 
4
(pg. 
236
-
248
)
92
Grabher
 
C
von Boehmer
 
H
Look
 
AT
Notch 1 activation in the molecular pathogenesis of T-cell acute lymphoblastic leukaemia.
Nat Rev Cancer
2006
, vol. 
6
 
5
(pg. 
347
-
359
)
93
Sjölund
 
J
Manetopoulos
 
C
Stockhausen
 
MT
, et al. 
The Notch pathway in cancer: differentiation gone awry.
Eur J Cancer
2005
, vol. 
41
 
17
(pg. 
2620
-
2629
)
94
Zhou
 
CH
Ye
 
LP
Ye
 
SX
, et al. 
Clinical significance of SOX9 in human non-small cell lung cancer progression and overall patient survival.
J Exp Clin Cancer Res
2012
, vol. 
31
 pg. 
18
 
95
Clemons
 
NJ
Wang
 
DH
Croagh
 
D
, et al. 
Sox9 drives columnar differentiation of esophageal squamous epithelium: a possible role in the pathogenesis of Barrett’s esophagus.
Am J Physiol Gastrointest Liver Physiol
2012
, vol. 
303
 
12
(pg. 
G1335
-
G1346
)
96
Wang
 
L
He
 
S
Yuan
 
J
, et al. 
Oncogenic role of SOX9 expression in human malignant glioma.
Med Oncol
2012
, vol. 
29
 
5
(pg. 
3484
-
3490
)
Sign in via your Institution