Skip to main content
Cell Cycle logoLink to Cell Cycle
. 2012 Sep 15;11(18):3358–3369. doi: 10.4161/cc.21277

The BTB-ZF transcription factors

Owen M Siggs 1,2,*, Bruce Beutler 3
PMCID: PMC3466544  PMID: 22894929

Abstract

The BTB-ZF (broad-complex, tramtrack and bric-à-brac - zinc finger) proteins are encoded by at least 49 genes in mouse and man and commonly serve as sequence-specific silencers of gene expression. This review will focus on the known physiological functions of mammalian BTB-ZF proteins, which include essential roles in the development of the immune system. We discuss their function in terminally differentiated lymphocytes and the progenitors that give rise to them, their action in hematopoietic malignancy and roles beyond the immune system.

Keywords: BTB, hematopoiesis, lymphoma, lymphopoiesis, zinc finger

Introduction

The zinc finger (ZF) motif was first identified in Xenopus laevis transcription factor IIIA protein1 and was so named because it contained zinc and grasped DNA. The consensus sequence of this particular motif [two cysteines in close proximity to two histidines (C2H2)], has come to be recognized as one of the most common DNA-binding motifs, encoded by 3% of all human genes.2

Some of the earliest C2H2 ZF genes were found in Drosophila melanogaster, cloned from the developmental mutants tramtrack,3 broad-complex4 and bric-à-brac.5 The Broad-complex and Tramtrack proteins were noted to share N-terminal homology,4 a finding that was later extended to mammalian ZF proteins ZBTB14 and ZBTB25 (respectively known as ZF5 and KUP at the time).6 Originally known as the ZiN (Zinc finger N-terminal) motif, it was also recognized to exist in the Vaccinia virus protein KBTB1 (then known as A55).6 A longer region of homology was recognized among other Drosophila proteins5,7 and in a number of poxvirus proteins,8 leading to the use of the abbreviations BTB (for Broad-complex, Tramtrack and Bric-à-brac) or POZ (for Poxvirus and Zinc finger) for this newly recognized domain.

The BTB domain is approximately 120 amino acids in length,8 typically present in a single copy, and associated with a variety of other domains, including C2H2 zinc finger and Kelch domains.9 BTB domain-encoding genes are found among eukaryotic and viral genomes yet are absent from prokaryotic genomes (with the exception of an endosymbiotic bacterium10). The core molecular function of the BTB domain is to promote protein-protein interactions, although the consequences of these interactions will depend upon the broader context of the protein.9 The BTB domain of SKP1, for example, recruits target recognition modules to the Cullin E3 ubiquitin ligase complex.11,12

Of a predicted 156 human genes that encode BTB domain-containing proteins, 49 also possess a series of C2H2 zinc fingers (Fig. 1). These proteins are commonly known as BTB-ZF or POK proteins (for POZ/Krüppel-like, after the defining N-terminal POZ domain and the C2H2 zinc fingers characteristic of the Drosophila segmentation protein Krüppel).13 Apart from appearances in viral genomes, BTB-ZF proteins appear to be restricted to the genomes of higher eukaryotes and are most numerous in vertebrates. One colorful example in Drosophila is the fruitless gene which, when mutated, causes male flies to indiscriminately court both females and males.14,15

graphic file with name cc-11-3358-g1.jpg

Figure 1. Domain structure and phylogeny of 49 human BTB-ZF proteins. All contain a single BTB domain and between 2 and 14 C2H2 ZF domains, with PATZ1 and ZBTB24 also containing an AT-hook domain. Note that ZBTB12B is a pseudogene, ZBTB8OS does not encode a BTB-ZF protein, and that ANKFY1 has a FYVE-type rather than a C2H2 ZF domain. Amino acid sequences of each of the BTB-ZF proteins were obtained from Uniprot, and phylogenetic analysis was performed using Phylogeny.fr134: (i) Amino acid sequences were aligned with Multiple Sequence Comparison by Log-Expectation (MUSCLE, v. 3.7; default settings). (ii) The maximum likelihood method in PhyML (v. 3.0, aLRT) was used to generate the phylogenetic tree from the multiple sequence alignment. The WAG substitution model was selected assuming an estimated proportion of invariant sites (of 0.000) and 4 gamma-distributed rate categories to account for rate heterogeneity across sites. The gamma shape parameter was estimated directly from the data (gamma = 1.158). Reliability for internal branch was assessed using the Approximate Likelihood Ratio Test (aLRT; SH-Like). (iii) Graphical representation of the phylogenetic tree was performed with TreeDyn (v. 198.3) in the radial style of DrawTree from the PHYLIP package (v. 3.66).

Since the discovery of mammalian BTB-ZF proteins in leukemic translocations, much has been learned about the function of these proteins. For example, the DNA-binding ZF motifs determine sequence specificity, while the BTB domain promotes oligomerization and the recruitment of transcriptional regulators.8 These are typically repressors of transcription such as N-CoR (nuclear receptor corepressor) and SMRT (silencing mediator for retinoid and thyroid hormone receptor),16,17 but also include activators such as p300.18 Transcriptional regulation by BTB-ZF proteins is often required for normal development of lymphocytes, while other members regulate fertility, skeletal morphogenesis, and/or neurological development (Table 1).

Table 1. BTB-ZF-encoding genes and the phenotypes associated with their mutation.

# human symbol mouse symbol synonyms ZFs phenotypes
1
ZBTB1
Zbtb1
ZNF909
8
immune/hematopoietic99
2
ZBTB2
Zbtb2
ZNF437
4
 
3
ZBTB3
Zbtb3
 
2
 
4
ZBTB4
Zbtb4
KAISO-L1, ZNF903
6
 
5
ZBTB5
Zbtb5
 
2
 
6
ZBTB6
Zbtb6
ZID, ZNF482
4
 
7
ZBTB7A
Zbtb7a
FBI-1, LRF, pokemon, ZBTB7, ZNF857A
4
immune/hematopoietic,92-94 embryogenesis,32 tumorigenesis32
8
ZBTB7B
Zbtb7b
c-Krox, Th-POK, ZBTB15, ZFP67, ZNF857B
4
immune/hematopoietic17,72,73,77,78
9
ZBTB7C
Zbtb7c
ZBTB36, ZNF857C
4
tumorigenesis34
10
ZBTB8A
Zbtb8a
BOZF1, ZBTB8, ZNF916A
2
 
11
ZBTB8B
Zbtb8b
ZNF916B
2
 
12
ZBTB9
Zbtb9
ZNF919
2
 
13
ZBTB10
Zbtb10
RINZF
2
 
14
ZBTB11
Zbtb11
ZNF913
12
 
15
ZBTB12
Zbtb12
G10, NG35, Bat9
4
 
16
ZFP161
Zfp161
ZBTB14, ZNF478, ZF5
5
 
17
ZBTB16
Zbtb16
PLZF, ZNF145
9
immune/hematopoietic,79-84,91 tumorigenesis,21,23 skeleton,103,104,106 reproductive103,107,108
18
ZBTB17
Zbtb17
MIZ1, pHZ-67, ZNF151, ZNF60
13
immune/hematopoietic,96,97 embryogenesis,95 integument136
19
ZNF238
Zfp238
C2H2–171, RP58, TAZ-1, ZBTB18
4
nervous120
20
PATZ1
Patz1
MAZR, PATZ, RIAZ, ZBTB19, ZNF278, ZSG
7
immune/hematopoietic,76 reproductive112
21
ZBTB20
Zbtb20
DPZF, ODA-8S, ZNF288
5
nervous,121 liver/biliary122,123
22
ZNF295
Zfp295
ZBTB21
8
 
23
ZBTB22
Zbtb22
BING1, fruitless, ZBTB22A, ZNF297, ZNF297A
3
 
24
GZF1
Gzf1
ZBTB23, ZNF336
10
 
25
ZBTB24
Zbtb24
BIF1, PATZ2, ZNF450
8
immune/hematopoietic127,128
26
ZBTB25
Zbtb25
KUP, ZNF46
2
 
27
ZBTB26
Zbtb26
ZNF481
4
 
28
BCL6
Bcl6
BCL5, BCL6A, LAZ3, ZBTB27, ZNF51
6
immune/hematopoietic,43-45,49-58,137 tumorigenesis,24-26,28 reproductive,113 skeleton64
29
BCL6B
Bcl6b
BAZF, ZBTB28, ZNF62
5
immune/hematopoietic89,90,137
30
HIC1
Hic1
ZBTB29, ZNF901
5
embryogenesis,124 tumorigenesis37
31
HIC2
Hic2
HRG22, ZBTB30, ZNF907
5
 
32
MYNN
Mynn
SBBIZ1, ZBTB31, ZNF902
8
 
33
ZBTB32
Zbtb32
PLZP, FAXF, FAZF, Rog, TZFP, ZNF538
3
immune/hematopoietic87,88
34
ZBTB33
Zbtb33
KAISO, ZNF-kaiso, ZNF348
3
tumorigenesis33
35
ZBTB34
Zbtb34
ZNF918
3
 
36
ZBTB37
Zbtb37
ZNF908
3
 
37
ZBTB38
Zbtb38
CIBZ, ZNF921
10
 
38
ZBTB39
Zbtb39
ZNF922
8
 
39
ZBTB40
Zbtb40
ZNF923
12
 
40
ZBTB41
Zbtb41
FRBZ1, ZNF924
14
 
41
ZBTB42
Zbtb42
ZNF925
4
 
42
ZBTB43
Zbtb43
ZBTB22B, ZNF-X, ZNF297B
3
 
43
ZBTB44
Zbtb44
BTBD15, ZNF851
4
 
44
ZBTB45
Zbtb45
ZNF499
4
 
45
ZBTB46
Zbtb46
BTBD4, RINZF, ZNF340, zDC
2
immune/hematopoietic100
46
ZBTB47
Zfp651
ZNF651
9
 
47
ZBTB48
Zbtb48
HKR3, ZNF855
11
 
48
ZBTB49
Zbtb49
ZNF509
7
 
49 ZNF131 Zfp131   6  

Hematopoietic Malignancy

The first mammalian BTB-ZF proteins to be characterized in detail emerged from the study of chromosomal translocations in human leukemias. Of the translocations associated with acute promyelocytic leukemia, all of them involve the retinoic acid receptor α gene (RARA) on chromosome 17. In 95% of cases, RARA is fused to the promyelocytic leukemia gene on chromosome 15; in a subset of the remaining cases, RARA forms a fusion with the promyelocytic leukemia zinc finger gene (PLZF, now known as ZBTB16) on chromosome 11.19 The t(11;17) translocation has the worst prognosis of all, since it is the only one resistant to the standard treatment for acute promyelocytic leukemia: all-trans retinoic acid therapy.20

ZBTB16 encodes a number of ZF motifs,21 and, unlike many other translocations, both ZBTB16 reciprocal translocation products are required for oncogenesis. RARα-PLZF by itself is not oncogenic in mice, and although PLZF-RARα causes a chronic myelogenous leukemia-like disease in isolation,22 RARα-PLZF is required for the development of a disease more similar to acute promyelocytic leukemia.23 The action of RARα-PLZF is likely to be a dominantly interfering one, since deletion of ZBTB16 has the same effect in the context of PLZF-RARα.23

B cell lymphoma is a second example of a BTB-ZF gene rearrangement associated with hematopoietic malignancy. ~40% of diffuse large B cell lymphomas, as well as 5–10% of follicular lymphomas, are associated with translocations of chromosome 3q27 within the region of an actively transcribed gene. This gene, named B cell lymphoma 6 (BCL6) after the disease associated with it, encodes a number of ZF domains;24-26 only later was it realized that both BCL6 and PLZF also shared a BTB domain.7,8 The majority of BCL6 breakpoints occur around its noncoding first exon, leading to promoter substitution and deregulated expression of an otherwise normal BCL6 protein,27 which is sufficient to cause B cell lymphoma when expressed in mice.28 Following tyrosine kinase inhibitor treatment, BCL6 is also overexpressed in BCR-ABL1 acute lymphoblastic leukemia cells at levels similar to those seen in B cell lymphomas with BCL6 translocations.29 BCL6 is therefore an attractive target for the treatment of both B cell lymphoma and BCR-ABL1 leukemia. Targeting the BTB domain of BCL6 by peptide inhibition may be one approach, and has been shown to induce cell cycle arrest and apoptosis by disrupting corepressor recruitment.30 BTB-specific interference might be equally useful for the treatment of other BTB-ZF-associated malignancies.

The BTB-ZF leukemia-related factor (LRF) protein, encoded by ZBTB7A (a paralog of ZBTB7B and ZBTB7C), is known to interact with BCL631 and can induce T cell leukemia in mice when overexpressed by an lck/Eμ enhancer/promoter.32 LRF-deficient mouse fibroblasts are also resistant to oncogene-mediated transformation.32 Similarly, deletion of Zbtb33 can delay tumorigenesis,33 while the ZBTB7C protein is required for fibroblast proliferation and promotes tumorigenesis when overexpressed.34

HIC1 (hypermethylated in cancer 1) is a candidate tumor suppressor gene underexpressed in many cancers,35 including hematological ones.36 Heterozygous Hic1-knockout mice develop a variety of neoplasms with age (lymphomas in ~25% of cases)37 and exhibit accelerated tumorigenesis on a variety of sensitized backgrounds,38-40 presumably through SIRT1-mediated inactivation of p53.41 Finally, while not a tumor suppressor itself, FAZF is an interacting partner of both Fanconi anemia group C protein (FANCC) and PLZF42 and may, therefore, play a role in tumorigenesis of Fanconi anemia and/or acute promyelocytic leukemia.

Lymphopoiesis and Immunity

BCL6: A master regulator of the germinal center response.

As one of the first mammalian BTB-ZF genes to be discovered, Bcl6 was also one of the first to be targeted in the mouse genome. Germinal centers were conspicuously absent from Bcl6 mutant mice due in part to a cell-intrinsic failure of germinal center B cell differentiation.43-45 Immunoglobulin class switching and somatic hypermutation, which are frequent events in germinal center B cells, would otherwise induce a DNA damage-associated apoptotic response. BCL6 allows germinal center B cells to tolerate these events by actively repressing transcription of TP53, the DNA damage sensor ATR and the cell cycle arrest gene CDKN1A.46-48 Given that p53 is so frequently inactivated in cancer, these findings also offered an explanation for how BCL6 activation could lead to B cell lymphoma.

More recently it has become clear that BCL6 is also required for the differentiation of follicular helper T cells, which support germinal center formation,49-51 and for follicular regulatory T cells and follicular helper NKT cells.52-54 Several other hematopoietic lineages require BCL6 for their development, including memory T cells (CD4+ and CD8+)55-57 and conventional dendritic cells (CD4+ and CD8α+).58

BCL6 also actively counteracts the differentiation of other cell lineages. While BCL6 is necessary for the formation of germinal center B cells, their progression to immunoglobulin-secreting plasma cells requires the transcriptional regulator BLIMP-1 (encoded by Prdm1).59,60 Prdm1 itself is directly repressed by BCL6;61-63 only after Bcl6 expression is extinguished can plasma cell differentiation occur. Osteoclasts, which are derived from hematopoietic precursors in the bone marrow, also depend upon the Bcl6/Prdm1 axis. BCL6 is a suppressor of osteoclastogenesis by default, while BLIMP-1 promotes it, likely via direct repression of the Bcl6 promoter by BLIMP-1.64

Another defining phenotype of the original Bcl6-knockout mice was their development of a lethal Th2-type inflammatory disease.43,44 Neither IL-4 nor STAT6 was required for disease development,65 which was driven by non-lymphoid cells,66 although BCL6 was required for STAT6-dependent IgE class switching.67 The critical inflammatory cell type appears to be macrophages, in which BCL6-mediated repression of proinflammatory chemokines and cytokines is lost,66,68 driving macrophage proliferation68 and promoting Th17 differentiation.69

Th-POK: Thymic lineage commitment and effector T cell function.

One of the key stages of T cell lineage commitment occurs in the thymus, where CD4+CD8+ thymocytes diverge into either the CD4+ helper or CD8+ cytotoxic lineage. The molecular basis of this choice had been a mystery for many years, until the serendipitous discovery of a spontaneous mouse mutant, hd (helper T cell deficient). hd mice entirely lack CD4 single positive cells in the thymus70 and T cells which would otherwise be destined for the CD4 lineage (e.g., those with an MHC class II-restricted TCR transgene or those developing in MHC class I-deficient hosts) are diverted to the CD8 lineage in hd mice, implying that CD8 was the default lineage in the absence of the hd gene product.71 The hd mutation was eventually revealed to be a missense allele of Zbtb7b, encoding the BTB-ZF protein Th-POK72 (also known as cKrox).73 Zbtb7b, which is normally silenced by Runx transcription factor complexes in CD8 cells,74 does not appear to be essential for CD4 specification but is required to prevent Runx-dependent commitment to the CD8 lineage.75 PATZ1 (POZ-, AT hook- and zinc finger-containing protein 1), another BTB-ZF protein, also contributes to silencing of the Zbtb7b locus in CD8+ cells.76

Th-POK is required for the normal maturation of invariant NKT cells17,77 as well as for the optimal development of γδ T cells.78 PLZF is also essential for the development of a variety of innate T cells, including effector NKT cells79-81 and innate γδ T cells.82-84 The effect of PLZF on innate γδ T cell development is not an intrinsic one, however, and instead requires the secretion of IL-4 by PLZF+ NKT cells.85,86

At least two BTB-ZF proteins have been shown to be involved in TCR-mediated proliferation, with varying roles. PLZP-deficient T cells proliferate more in response to TCR and cytokine receptor stimulation,87,88 while anti-CD3-induced proliferation of naïve, but not memory CD4+ T cells, is partially reduced in BCL6B-deficient mice.89 Conversely, the recall response of virus-specific memory CD8+ T cells is BCL6B-dependent, while their primary response is not.90 In the broader context of viral infection, PLZF plays an important role in the induction of type I interferon-inducible gene expression. Through an interaction with the histone deacetylase HDAC1 and the promyelocytic leukemia protein PML, PLZF binds to the promoter regions of interferon-inducible genes and promotes their expression.91 Accordingly, PLZF-deficient NK cells have impaired function, and PLZF-deficient mice show a heightened sensitivity to viral infection.91

LRF and MIZ1: T vs B lineage commitment and beyond.

T and B lymphocytes arise from common lymphoid precursors in the bone marrow. Within these cells, the action of at least two BTB-ZF proteins influences the choice of lineage: LRF, encoded by Zbtb7a, and MIZ1, encoded by Zbtb17. While germline deletion of Zbtb7a is lethal to the developing embryo,32,92 fetal liver chimera and conditional deletion experiments established that LRF was essential for fetal and adult B cell development.92 In the conditional LRF mutants, failure of B cell development was associated with ectopic development of CD4+CD8+ cells in the bone marrow. Treating Zbtb7a mutant progenitors with a Notch1 inhibitor could partially correct this developmental imbalance, implying that LRF promotes B cell lineage commitment by counteracting Notch1.92

LRF is important beyond the T/B lineage split, with B cell-specific deletion of Zbtb7a resulting in the generation of more marginal zone B cells but fewer follicular and germinal center B cells.93 The embryonic lethality of LRF-deficient mice is also associated with profound anemia.32,92 GATA1, a key transcriptional regulator of erythropoiesis, was found to activate the expression of Zbtb7a, which, in turn, suppressed expression of the gene encoding the proapoptotic protein Bim.94 Accordingly, deficiency of Bim could correct the anemia in Zbtb7a mutant embryos and prolong their survival in utero.94

Germline deletion of Zbtb17 (encoding the MIZ1 protein) is also lethal in utero, but at a much earlier stage than Zbtb7a (E7.5) due to a failure of gastrulation.95 Zbtb17 mutant cells can nevertheless contribute to myeloid and erythroid lineages in ES cell chimeras yet were not represented in T or B lymphocytes.96 Hematopoietic conditional deletion of Zbtb17 using a Vav-cre transgene also led to deficiencies of T and B cells, with respective blocks at the early T cell progenitor and pre-pro-B stages of development due to insufficient IL-7R signaling.96,97 The block in IL-7R signaling could be explained in part by the overexpression of Socs1, which MIZ1 would otherwise repress. Inhibition of Socs1 could rescue lymphoid development to a small extent in vitro, with more substantial improvements seen after Bcl2 transgene expression (in combination with Ebf1 overexpression for B cells).96,97

ZBTB1: A determinant of lymphoid development.

One of the more recently characterized BTB-ZF members is ZBTB1. Like many other BTB-ZF proteins, ZBTB1 is a potent transcriptional repressor,98 whose localization and repressive activity is regulated by SUMOylation.98 Forward genetic screening in mice revealed that a missense mutation in the BTB domain of ZBTB1 (C74R) leads to a profound T cell deficiency, with milder deficiencies of B and NK cells.99 When placed in competition with wild-type cells, however, Zbtb1 mutant cells fail to generate any lymphocytes at all, while myelopoiesis is unperturbed.99

ZBTB46: A marker of conventional DCs.

One of the most highly expressed genes in mouse conventional dendritic cells (cDCs) is Zbtb46, a BTB-ZF gene also expressed in endothelial cells and erythroid precursors.100 Knock-in alleles at the Zbtb46 locus have allowed the creation of cDC-specific reporters, distinguishing them from other CD11c+ phagocytes and allowing lineage-specific ablation.100,101 Zbtb46 is not essential for their differentiation, but may be important for their full functional maturation, as expression of the receptors for G-CSF and leukemia inhibitory factor persist in its absence.100 Retroviral overexpression of Zbtb46 promotes cDC development at the expense of granulocytes and can even overcome the cDC developmental block in Irf8 mutant progenitors.100 This effect was dependent upon the tandem ZF motifs, as a ZF deletion mutant could not enhance cDC development.

Functions of BTB-ZF Proteins Beyond Hematopoiesis

Limb patterning.

Long before BTB-ZF proteins were known, the effects of a particular BTB-ZF mutation had been closely studied. Green’s luxoid is a spontaneous mouse mutant characterized by skeletal abnormalities similar to the luxate mutant.102,103 The observed limb patterning defects in this model are a consequence of a nonsense mutation in Zbtb16 (encoding PLZF), are recapitulated in a Zbtb16-knockout strain104 and are influenced by the transcriptional regulator Gli3.105 Similar skeletal abnormalities, including an absence of thumbs, were observed in a PLZF-deficient patient, who inherited an 11q23 deletion in trans with a recessive missense mutation of ZBTB16.106

Fertility.

A second defining feature of luxoid mutants was their infertility, which has since been explained by a failure of spermatogonial stem cell self-renewal,107,108 potentially due to the inhibition of mTORC1109 or the stem cell antigen Kit.110 These pleiotropic effects of PLZF are likely to be due to BTB-mediated interactions with distinct proteins, as they can be separated by a point mutation in the BTB domain.111 Genital hypoplasia is also a feature of human PLZF deficiency,106 suggesting the conservation of both functions in man. Similar to PLZF, loss of PATZ1 causes male infertility due to a loss of spermatogonial stem cells;112 germ cell apoptosis is also a feature of BCL6 mutant males.113 Sterility is also a feature of at least two Drosophila BTB-ZF mutants, ken and barbie, characterized by loss of terminalia114,115 and mutations in the fly ortholog of ZBTB22, fruitless, which causes male sterility due to behavioral defects, rather than a failure to develop germ cells or genitalia.14,15

Neurological and other developmental roles.

A more common function for Drosophila BTB-ZF proteins is the regulation of neuronal development. Tramtrack itself is required to prevent transformation of neuronal support cells into neurons,116 while fruitless, abrupt, lola and chinmo are all examples of Drosophila BTB-ZF mutants that affect neurological development when mutated.14,117-119 Similarly, mutant alleles of mouse Zfp238 exhibit cortical and hippocampal dysplasia.120 Zbtb20 is required for the development of clusters of hippocampal neurons121 as well as for glucose metabolism and development of the liver,122 where it suppresses transcription of the hepatocyte proliferation gene AFP.123 Miller-Dieker syndrome, caused by a multigene chromosomal deletion spanning HIC1, is associated with mental retardation and a variety of developmental abnormalities, some of which are recapitulated in homozygous Hic1 mutant embryos.124 KAISO, encoded by Zbtb33, is redundant during embryonic development in mice,33 yet it is essential in Xenopus.125 This may be explained in part by the compensatory activity of ZBTB4 and ZBTB38, which, like KAISO, bind to methylated DNA and repress transcription.126

BTB-ZF proteins of obscure physiological function.

The vast majority of BTB-ZF genes have not been studied in germline mutant mice, yet their expression patterns hint that many others may be important for hematopoiesis and other developmental pathways (Fig. 4). One alluring candidate is ZBTB24, which is mutated in immunodeficiency, centromeric instability and facial anomalies syndrome type 2 (ICF2).127,128 ZBTB24 mutant patients often succumb to recurrent respiratory or gastrointestinal infections, although the nature of their immunodeficiency has only been linked to a deficiency of immunoglobulins, usually in the presence of normal T and B lymphocyte frequencies.129 ICF2 patients share features of their disorder with ICF1 patients, who carry mutations in the DNA methyltransferase 3B gene (DNMT3B).130 One characteristic feature is centromeric instability, commonly associated with hypomethylated juxtacentromeric regions of chromosomes 1, 9 and 16. How this leads to the clinical features of ICF is unknown, as is the means by which ZBTB24 prevents chromosomal instability. Along with PATZ1, ZBTB24 is also one of only two mammalian BTB-ZF proteins recognized to contain an AT-hook domain, a DNA-binding motif capable of interacting with the minor groove of AT-rich sequences.131

graphic file with name cc-11-3358-g4.jpg

Figure 4. Collective display of mouse BTB-ZF gene expression data. Relative tissue-specific transcript expression data were obtained from BioGPS135 and are representative of multiple transcript probes where appropriate. Gray columns indicate hematopoietic tissues, while black columns are non-hematopoietic. pDC, plasmacytoid dendritic cell; BMDM, bone marrow-derived macrophage; LPS, lipopolysaccharide; GMP, granulocyte-monocyte progenitor; MEP, megakaryocyte-erythrocyte progenitor; HSC, hematopoietic stem cell.

Given that mutant alleles of almost every BTB-ZF gene already exist in mouse ES cells132 and in chemically-induced mutant archives,133 it is only a matter of time before we learn the non-redundant functions of every BTB-ZF protein. Rare loss-of-function alleles of the same genes will also exist in the human population and may emerge in homozygous or compound heterozygous form as genomes of human patients are sequenced at a rapidly increasing pace.

Summary

A host of physiological functions have emerged for the mammalian BTB-ZF proteins, from embryogenesis to the development of specialized lymphocyte effector cells. (Fig. 2) As new terminal branches of lymphocyte differentiation are revealed, so too are new functions for BTB-ZF proteins. Transcriptional suppression appears to be a unifying functional trait, (Fig. 3) orchestrated by BTB-mediated recruitment of chromatin remodeling factors to ZF-defined recognition sites, and there is increasing evidence that individual BTB-ZF proteins can influence multiple developmental pathways. The functions of most remain unknown, and for the ones that are known, the proteins they interact with and the genes they regulate are mostly a mystery.

graphic file with name cc-11-3358-g2.jpg

Figure 2. Schematic of BTB-ZF protein functions in hematopoiesis and immunity. Labels outside cells represent a requirement for the BTB-ZF protein during development. Labels inside cells represent a requirement in effector function. See text for additional details.

graphic file with name cc-11-3358-g3.jpg

Figure 3. Transcriptional circuits involving mammalian BTB-ZF proteins. Examples shown are from pathways established in mammalian primary cells, question marks indicate that a direct interaction is yet to be demonstrated. See text for additional details.

Acknowledgments

We thank Vickie Webster for her expert assistance with figures and Anne Murray for proofreading and preparation of the phylogenetic tree. We also apologize to those authors whose work could not be cited within our limitations. This work was supported by BAA-NIH-NIAID-DAIT and The Bill and Melinda Gates Foundation.

Footnotes

References

  • 1.Miller J, McLachlan AD, Klug A. Repetitive zinc-binding domains in the protein transcription factor IIIA from Xenopus oocytes. EMBO J. 1985;4:1609–14. doi: 10.1002/j.1460-2075.1985.tb03825.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Klug A. The discovery of zinc fingers and their development for practical applications in gene regulation and genome manipulation. Annu Rev Biochem. 2010;79:213–31. doi: 10.1017/S0033583510000089. [DOI] [PubMed] [Google Scholar]
  • 3.Harrison SD, Travers AA. The tramtrack gene encodes a Drosophila finger protein that interacts with the ftz transcriptional regulatory region and shows a novel embryonic expression pattern. EMBO J. 1990;9:207–16. doi: 10.1002/j.1460-2075.1990.tb08097.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.DiBello PR, Withers DA, Bayer CA, Fristrom JW, Guild GM. The Drosophila Broad-Complex encodes a family of related proteins containing zinc fingers. Genetics. 1991;129:385–97. doi: 10.1093/genetics/129.2.385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Godt D, Couderc JL, Cramton SE, Laski FA. 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;119:799–812. doi: 10.1242/dev.119.3.799. [DOI] [PubMed] [Google Scholar]
  • 6.Numoto M, Niwa O, Kaplan J, Wong KK, Merrell K, Kamiya K, et al. Transcriptional repressor ZF5 identifies a new conserved domain in zinc finger proteins. Nucleic Acids Res. 1993;21:3767–75. doi: 10.1093/nar/21.16.3767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zollman S, Godt D, Privé GG, Couderc JL, Laski FA. The BTB domain, found primarily in zinc finger proteins, defines an evolutionarily conserved family that includes several developmentally regulated genes in Drosophila. Proc Natl Acad Sci USA. 1994;91:10717–21. doi: 10.1073/pnas.91.22.10717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bardwell VJ, Treisman R. The POZ domain: a conserved protein-protein interaction motif. Genes Dev. 1994;8:1664–77. doi: 10.1101/gad.8.14.1664. [DOI] [PubMed] [Google Scholar]
  • 9.Stogios PJ, Downs GS, Jauhal JJ, Nandra SK, Privé GG. Sequence and structural analysis of BTB domain proteins. Genome Biol. 2005;6:R82. doi: 10.1186/gb-2005-6-10-r82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Perez-Torrado R, Yamada D, Defossez PA. Born to bind: the BTB protein-protein interaction domain. Bioessays. 2006;28:1194–202. doi: 10.1002/bies.20500. [DOI] [PubMed] [Google Scholar]
  • 11.Pintard L, Willis JH, Willems A, Johnson JL, Srayko M, Kurz T, et al. The BTB protein MEL-26 is a substrate-specific adaptor of the CUL-3 ubiquitin-ligase. Nature. 2003;425:311–6. doi: 10.1038/nature01959. [DOI] [PubMed] [Google Scholar]
  • 12.Xu L, Wei Y, Reboul J, Vaglio P, Shin TH, Vidal M, et al. BTB proteins are substrate-specific adaptors in an SCF-like modular ubiquitin ligase containing CUL-3. Nature. 2003;425:316–21. doi: 10.1038/nature01985. [DOI] [PubMed] [Google Scholar]
  • 13.Wieschaus E, Nusslein-Volhard C, Kluding H. Krüppel, a gene whose activity is required early in the zygotic genome for normal embryonic segmentation. Dev Biol. 1984;104:172–86. doi: 10.1016/0012-1606(84)90046-0. [DOI] [PubMed] [Google Scholar]
  • 14.Ryner LC, Goodwin SF, Castrillon DH, Anand A, Villella A, Baker BS, et al. Control of male sexual behavior and sexual orientation in Drosophila by the fruitless gene. Cell. 1996;87:1079–89. doi: 10.1016/S0092-8674(00)81802-4. [DOI] [PubMed] [Google Scholar]
  • 15.Ito H, Fujitani K, Usui K, Shimizu-Nishikawa K, Tanaka S, Yamamoto D. Sexual orientation in Drosophila is altered by the satori mutation in the sex-determination gene fruitless that encodes a zinc finger protein with a BTB domain. Proc Natl Acad Sci USA. 1996;93:9687–92. doi: 10.1073/pnas.93.18.9687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Huynh KD, Bardwell VJ. The BCL-6 POZ domain and other POZ domains interact with the co-repressors N-CoR and SMRT. Oncogene. 1998;17:2473–84. doi: 10.1038/sj.onc.1202197. [DOI] [PubMed] [Google Scholar]
  • 17.Wang L, Carr T, Xiong Y, Wildt KF, Zhu J, Feigenbaum L, et al. The sequential activity of Gata3 and Thpok is required for the differentiation of CD1d-restricted CD4+ NKT cells. Eur J Immunol. 2010;40:2385–90. doi: 10.1002/eji.201040534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Staller P, Peukert K, Kiermaier A, Seoane J, Lukas J, Karsunky H, et al. Repression of p15INK4b expression by Myc through association with Miz-1. Nat Cell Biol. 2001;3:392–9. doi: 10.1038/35070076. [DOI] [PubMed] [Google Scholar]
  • 19.Chen SJ, Chen Z, Chen A, Tong JH, Dong S, Wang ZY, et al. Occurrence of distinct PML-RAR-alpha fusion gene isoforms in patients with acute promyelocytic leukemia detected by reverse transcriptase/polymerase chain reaction. Oncogene. 1992;7:1223–32. [PubMed] [Google Scholar]
  • 20.Licht JD, Chomienne C, Goy A, Chen A, Scott AA, Head DR, et al. Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11;17) Blood. 1995;85:1083–94. [PubMed] [Google Scholar]
  • 21.Chen Z, Brand NJ, Chen A, Chen SJ, Tong JH, Wang ZY, et al. Fusion between a novel Krüppel-like zinc finger gene and the retinoic acid receptor-alpha locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia. EMBO J. 1993;12:1161–7. doi: 10.1002/j.1460-2075.1993.tb05757.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.He LZ, Guidez F, Tribioli C, Peruzzi D, Ruthardt M, Zelent A, et al. Distinct interactions of PML-RARalpha and PLZF-RARalpha with co-repressors determine differential responses to RA in APL. Nat Genet. 1998;18:126–35. doi: 10.1038/ng0298-126. [DOI] [PubMed] [Google Scholar]
  • 23.He LZ, Bhaumik M, Tribioli C, Rego EM, Ivins S, Zelent A, et al. Two critical hits for promyelocytic leukemia. Mol Cell. 2000;6:1131–41. doi: 10.1016/S1097-2765(00)00111-8. [DOI] [PubMed] [Google Scholar]
  • 24.Baron BW, Nucifora G, McCabe N, Espinosa R, 3rd, Le Beau MM, McKeithan TW. Identification of the gene associated with the recurring chromosomal translocations t(3;14)(q27;q32) and t(3;22)(q27;q11) in B-cell lymphomas. Proc Natl Acad Sci USA. 1993;90:5262–6. doi: 10.1073/pnas.90.11.5262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kerckaert JP, Deweindt C, Tilly H, Quief S, Lecocq G, Bastard C. LAZ3, a novel zinc-finger encoding gene, is disrupted by recurring chromosome 3q27 translocations in human lymphomas. Nat Genet. 1993;5:66–70. doi: 10.1038/ng0993-66. [DOI] [PubMed] [Google Scholar]
  • 26.Ye BH, Lista F, Lo Coco F, Knowles DM, Offit K, Chaganti RS, et al. Alterations of a zinc finger-encoding gene, BCL-6, in diffuse large-cell lymphoma. Science. 1993;262:747–50. doi: 10.1126/science.8235596. [DOI] [PubMed] [Google Scholar]
  • 27.Ye BH, Chaganti S, Chang CC, Niu H, Corradini P, Chaganti RS, et al. Chromosomal translocations cause deregulated BCL6 expression by promoter substitution in B cell lymphoma. EMBO J. 1995;14:6209–17. doi: 10.1002/j.1460-2075.1995.tb00311.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cattoretti G, Pasqualucci L, Ballon G, Tam W, Nandula SV, Shen Q, et al. Deregulated BCL6 expression recapitulates the pathogenesis of human diffuse large B cell lymphomas in mice. Cancer Cell. 2005;7:445–55. doi: 10.1016/j.ccr.2005.03.037. [DOI] [PubMed] [Google Scholar]
  • 29.Duy C, Hurtz C, Shojaee S, Cerchietti L, Geng H, Swaminathan S, et al. BCL6 enables Ph+ acute lymphoblastic leukaemia cells to survive BCR-ABL1 kinase inhibition. Nature. 2011;473:384–8. doi: 10.1038/nature09883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Polo JM, Dell’Oso T, Ranuncolo SM, Cerchietti L, Beck D, Da Silva GF, et al. Specific peptide interference reveals BCL6 transcriptional and oncogenic mechanisms in B-cell lymphoma cells. Nat Med. 2004;10:1329–35. doi: 10.1038/nm1134. [DOI] [PubMed] [Google Scholar]
  • 31.Davies JM, Hawe N, Kabarowski J, Huang QH, Zhu J, Brand NJ, et al. Novel BTB/POZ domain zinc-finger protein, LRF, is a potential target of the LAZ-3/BCL-6 oncogene. Oncogene. 1999;18:365–75. doi: 10.1038/sj.onc.1202332. [DOI] [PubMed] [Google Scholar]
  • 32.Maeda T, Hobbs RM, Merghoub T, Guernah I, Zelent A, Cordon-Cardo C, et al. Role of the proto-oncogene Pokemon in cellular transformation and ARF repression. Nature. 2005;433:278–85. doi: 10.1038/nature03203. [DOI] [PubMed] [Google Scholar]
  • 33.Prokhortchouk A, Sansom O, Selfridge J, Caballero IM, Salozhin S, Aithozhina D, et al. Kaiso-deficient mice show resistance to intestinal cancer. Mol Cell Biol. 2006;26:199–208. doi: 10.1128/MCB.26.1.199-208.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Jeon BN, Kim MK, Choi WI, Koh DI, Hong SY, Kim KS, et al. KR-POK interacts with p53 and represses its ability to activate transcription of p21WAF1/CDKN1A. Cancer Res. 2012;72:1137–48. doi: 10.1158/0008-5472.CAN-11-2433. [DOI] [PubMed] [Google Scholar]
  • 35.Wales MM, Biel MA, el Deiry W, Nelkin BD, Issa JP, Cavenee WK, et al. p53 activates expression of HIC-1, a new candidate tumour suppressor gene on 17p13.3. Nat Med. 1995;1:570–7. doi: 10.1038/nm0695-570. [DOI] [PubMed] [Google Scholar]
  • 36.Issa JP, Zehnbauer BA, Kaufmann SH, Biel MA, Baylin SB. HIC1 hypermethylation is a late event in hematopoietic neoplasms. Cancer Res. 1997;57:1678–81. [PubMed] [Google Scholar]
  • 37.Chen WY, Zeng X, Carter MG, Morrell CN, Chiu Yen RW, Esteller M, et al. Heterozygous disruption of Hic1 predisposes mice to a gender-dependent spectrum of malignant tumors. Nat Genet. 2003;33:197–202. doi: 10.1038/ng1077. [DOI] [PubMed] [Google Scholar]
  • 38.Chen W, Cooper TK, Zahnow CA, Overholtzer M, Zhao Z, Ladanyi M, et al. Epigenetic and genetic loss of Hic1 function accentuates the role of p53 in tumorigenesis. Cancer Cell. 2004;6:387–98. doi: 10.1016/j.ccr.2004.08.030. [DOI] [PubMed] [Google Scholar]
  • 39.Briggs KJ, Corcoran-Schwartz IM, Zhang W, Harcke T, Devereux WL, Baylin SB, et al. Cooperation between the Hic1 and Ptch1 tumor suppressors in medulloblastoma. Genes Dev. 2008;22:770–85. doi: 10.1101/gad.1640908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mohammad HP, Zhang W, Prevas HS, Leadem BR, Zhang M, Herman JG, et al. Loss of a single Hic1 allele accelerates polyp formation in Apc(Δ716) mice. Oncogene. 2011;30:2659–69. doi: 10.1038/onc.2010.633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chen WY, Wang DH, Yen RC, Luo J, Gu W, Baylin SB. Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses. Cell. 2005;123:437–48. doi: 10.1016/j.cell.2005.08.011. [DOI] [PubMed] [Google Scholar]
  • 42.Hoatlin ME, Zhi Y, Ball H, Silvey K, Melnick A, Stone S, et al. A novel BTB/POZ transcriptional repressor protein interacts with the Fanconi anemia group C protein and PLZF. Blood. 1999;94:3737–47. [PubMed] [Google Scholar]
  • 43.Ye BH, Cattoretti G, Shen Q, Zhang J, Hawe N, de Waard R, et al. The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation. Nat Genet. 1997;16:161–70. doi: 10.1038/ng0697-161. [DOI] [PubMed] [Google Scholar]
  • 44.Dent AL, Shaffer AL, Yu X, Allman D, Staudt LM. Control of inflammation, cytokine expression, and germinal center formation by BCL-6. Science. 1997;276:589–92. doi: 10.1126/science.276.5312.589. [DOI] [PubMed] [Google Scholar]
  • 45.Fukuda T, Yoshida T, Okada S, Hatano M, Miki T, Ishibashi K, et al. Disruption of the Bcl6 gene results in an impaired germinal center formation. J Exp Med. 1997;186:439–48. doi: 10.1084/jem.186.3.439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Phan RT, Dalla-Favera R. The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature. 2004;432:635–9. doi: 10.1038/nature03147. [DOI] [PubMed] [Google Scholar]
  • 47.Ranuncolo SM, Polo JM, Dierov J, Singer M, Kuo T, Greally 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;8:705–14. doi: 10.1038/ni1478. [DOI] [PubMed] [Google Scholar]
  • 48.Phan RT, Saito M, Basso K, Niu H, Dalla-Favera R. BCL6 interacts with the transcription factor Miz-1 to suppress the cyclin-dependent kinase inhibitor p21 and cell cycle arrest in germinal center B cells. Nat Immunol. 2005;6:1054–60. doi: 10.1038/ni1245. [DOI] [PubMed] [Google Scholar]
  • 49.Johnston RJ, Poholek AC, DiToro D, Yusuf I, Eto D, Barnett B, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science. 2009;325:1006–10. doi: 10.1126/science.1175870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Nurieva RI, Chung Y, Martinez GJ, Yang XO, Tanaka S, Matskevitch TD, et al. Bcl6 mediates the development of T follicular helper cells. Science. 2009;325:1001–5. doi: 10.1126/science.1176676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yu D, Rao S, Tsai LM, Lee SK, He Y, Sutcliffe EL, et al. The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity. 2009;31:457–68. doi: 10.1016/j.immuni.2009.07.002. [DOI] [PubMed] [Google Scholar]
  • 52.Chang PP, Barral P, Fitch J, Pratama A, Ma CS, Kallies A, et al. Identification of Bcl-6-dependent follicular helper NKT cells that provide cognate help for B cell responses. Nat Immunol. 2012;13:35–43. doi: 10.1038/ni.2166. [DOI] [PubMed] [Google Scholar]
  • 53.Linterman MA, Pierson W, Lee SK, Kallies A, Kawamoto S, Rayner TF, et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 2011;17:975–82. doi: 10.1038/nm.2425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med. 2011;17:983–8. doi: 10.1038/nm.2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ichii H, Sakamoto A, Hatano M, Okada S, Toyama H, Taki S, et al. Role for Bcl-6 in the generation and maintenance of memory CD8+ T cells. Nat Immunol. 2002;3:558–63. doi: 10.1038/ni802. [DOI] [PubMed] [Google Scholar]
  • 56.Ichii H, Sakamoto A, Kuroda Y, Tokuhisa T. Bcl6 acts as an amplifier for the generation and proliferative capacity of central memory CD8+ T cells. J Immunol. 2004;173:883–91. doi: 10.4049/jimmunol.173.2.883. [DOI] [PubMed] [Google Scholar]
  • 57.Ichii H, Sakamoto A, Arima M, Hatano M, Kuroda Y, Tokuhisa T. Bcl6 is essential for the generation of long-term memory CD4+ T cells. Int Immunol. 2007;19:427–33. doi: 10.1093/intimm/dxm007. [DOI] [PubMed] [Google Scholar]
  • 58.Ohtsuka H, Sakamoto A, Pan J, Inage S, Horigome S, Ichii H, et al. Bcl6 is required for the development of mouse CD4+ and CD8α+ dendritic cells. J Immunol. 2011;186:255–63. doi: 10.4049/jimmunol.0903714. [DOI] [PubMed] [Google Scholar]
  • 59.Turner CA, Jr., Mack DH, Davis MM. Blimp-1, a novel zinc finger-containing protein that can drive the maturation of B lymphocytes into immunoglobulin-secreting cells. Cell. 1994;77:297–306. doi: 10.1016/0092-8674(94)90321-2. [DOI] [PubMed] [Google Scholar]
  • 60.Shapiro-Shelef M, Lin KI, McHeyzer-Williams LJ, Liao J, McHeyzer-Williams MG, Calame K. Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity. 2003;19:607–20. doi: 10.1016/S1074-7613(03)00267-X. [DOI] [PubMed] [Google Scholar]
  • 61.Shaffer AL, Yu X, He Y, Boldrick J, Chan EP, Staudt LM. BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control. Immunity. 2000;13:199–212. doi: 10.1016/S1074-7613(00)00020-0. [DOI] [PubMed] [Google Scholar]
  • 62.Tunyaplin C, Shaffer AL, Angelin-Duclos CD, Yu X, Staudt LM, Calame KL. Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation. J Immunol. 2004;173:1158–65. doi: 10.4049/jimmunol.173.2.1158. [DOI] [PubMed] [Google Scholar]
  • 63.Reljic R, Wagner SD, Peakman LJ, Fearon DT. Suppression of signal transducer and activator of transcription 3-dependent B lymphocyte terminal differentiation by BCL-6. J Exp Med. 2000;192:1841–8. doi: 10.1084/jem.192.12.1841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Miyauchi Y, Ninomiya K, Miyamoto H, Sakamoto A, Iwasaki R, Hoshi H, et al. The Blimp1-Bcl6 axis is critical to regulate osteoclast differentiation and bone homeostasis. J Exp Med. 2010;207:751–62. doi: 10.1084/jem.20091957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Dent AL, Hu-Li J, Paul WE, Staudt LM. T helper type 2 inflammatory disease in the absence of interleukin 4 and transcription factor STAT6. Proc Natl Acad Sci USA. 1998;95:13823–8. doi: 10.1073/pnas.95.23.13823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Toney LM, Cattoretti G, Graf JA, Merghoub T, Pandolfi PP, Dalla-Favera R, et al. BCL-6 regulates chemokine gene transcription in macrophages. Nat Immunol. 2000;1:214–20. doi: 10.1038/79749. [DOI] [PubMed] [Google Scholar]
  • 67.Harris MB, Chang CC, Berton MT, Danial NN, Zhang J, Kuehner D, et al. Transcriptional repression of Stat6-dependent interleukin-4-induced genes by BCL-6: specific regulation of iepsilon transcription and immunoglobulin E switching. Mol Cell Biol. 1999;19:7264–75. doi: 10.1128/mcb.19.10.7264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yu RY, Wang X, Pixley FJ, Yu JJ, Dent AL, Broxmeyer HE, et al. BCL-6 negatively regulates macrophage proliferation by suppressing autocrine IL-6 production. Blood. 2005;105:1777–84. doi: 10.1182/blood-2004-08-3171. [DOI] [PubMed] [Google Scholar]
  • 69.Mondal A, Sawant D, Dent AL. Transcriptional repressor BCL6 controls Th17 responses by controlling gene expression in both T cells and macrophages. J Immunol. 2010;184:4123–32. doi: 10.4049/jimmunol.0901242. [DOI] [PubMed] [Google Scholar]
  • 70.Dave VP, Allman D, Keefe R, Hardy RR, Kappes DJ. HD mice: a novel mouse mutant with a specific defect in the generation of CD4(+) T cells. Proc Natl Acad Sci USA. 1998;95:8187–92. doi: 10.1073/pnas.95.14.8187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Keefe R, Dave V, Allman D, Wiest D, Kappes DJ. Regulation of lineage commitment distinct from positive selection. Science. 1999;286:1149–53. doi: 10.1126/science.286.5442.1149. [DOI] [PubMed] [Google Scholar]
  • 72.He X, He X, Dave VP, Zhang Y, Hua X, Nicolas E, et al. The zinc finger transcription factor Th-POK regulates CD4 versus CD8 T-cell lineage commitment. Nature. 2005;433:826–33. doi: 10.1038/nature03338. [DOI] [PubMed] [Google Scholar]
  • 73.Sun G, Liu X, Mercado P, Jenkinson SR, Kypriotou M, Feigenbaum L, et al. The zinc finger protein cKrox directs CD4 lineage differentiation during intrathymic T cell positive selection. Nat Immunol. 2005;6:373–81. doi: 10.1038/ni1183. [DOI] [PubMed] [Google Scholar]
  • 74.Setoguchi R, Tachibana M, Naoe Y, Muroi S, Akiyama K, Tezuka C, et al. Repression of the transcription factor Th-POK by Runx complexes in cytotoxic T cell development. Science. 2008;319:822–5. doi: 10.1126/science.1151844. [DOI] [PubMed] [Google Scholar]
  • 75.Egawa T, Littman DR. ThPOK acts late in specification of the helper T cell lineage and suppresses Runx-mediated commitment to the cytotoxic T cell lineage. Nat Immunol. 2008;9:1131–9. doi: 10.1038/ni.1652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sakaguchi S, Hombauer M, Bilic I, Naoe Y, Schebesta A, Taniuchi I, et al. The zinc-finger protein MAZR is part of the transcription factor network that controls the CD4 versus CD8 lineage fate of double-positive thymocytes. Nat Immunol. 2010;11:442–8. doi: 10.1038/ni.1860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Engel I, Hammond K, Sullivan BA, He X, Taniuchi I, Kappes D, et al. Co-receptor choice by V alpha14i NKT cells is driven by Th-POK expression rather than avoidance of CD8-mediated negative selection. J Exp Med. 2010;207:1015–29. doi: 10.1084/jem.20090557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Park K, He X, Lee HO, Hua X, Li Y, Wiest D, et al. TCR-mediated ThPOK induction promotes development of mature (CD24-) gammadelta thymocytes. EMBO J. 2010;29:2329–41. doi: 10.1038/emboj.2010.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Savage AK, Constantinides MG, Han J, Picard D, Martin E, Li B, et al. The transcription factor PLZF directs the effector program of the NKT cell lineage. Immunity. 2008;29:391–403. doi: 10.1016/j.immuni.2008.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kovalovsky D, Uche OU, Eladad S, Hobbs RM, Yi W, Alonzo E, et al. The BTB-zinc finger transcriptional regulator PLZF controls the development of invariant natural killer T cell effector functions. Nat Immunol. 2008;9:1055–64. doi: 10.1038/ni.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Constantinides MG, Picard D, Savage AK, Bendelac A. A naive-like population of human CD1d-restricted T cells expressing intermediate levels of promyelocytic leukemia zinc finger. J Immunol. 2011;187:309–15. doi: 10.4049/jimmunol.1100761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kreslavsky T, Savage AK, Hobbs R, Gounari F, Bronson R, Pereira P, et al. TCR-inducible PLZF transcription factor required for innate phenotype of a subset of gammadelta T cells with restricted TCR diversity. Proc Natl Acad Sci USA. 2009;106:12453–8. doi: 10.1073/pnas.0903895106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Alonzo ES, Gottschalk RA, Das J, Egawa T, Hobbs RM, Pandolfi PP, et al. Development of promyelocytic zinc finger and ThPOK-expressing innate gamma delta T cells is controlled by strength of TCR signaling and Id3. J Immunol. 2010;184:1268–79. doi: 10.4049/jimmunol.0903218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gordon SM, Carty SA, Kim JS, Zou T, Smith-Garvin J, Alonzo ES, et al. Requirements for eomesodermin and promyelocytic leukemia zinc finger in the development of innate-like CD8+ T cells. J Immunol. 2011;186:4573–8. doi: 10.4049/jimmunol.1100037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Weinreich MA, Odumade OA, Jameson SC, Hogquist KA. T cells expressing the transcription factor PLZF regulate the development of memory-like CD8+ T cells. Nat Immunol. 2010;11:709–16. doi: 10.1038/ni.1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Verykokakis M, Boos MD, Bendelac A, Kee BL. SAP protein-dependent natural killer T-like cells regulate the development of CD8(+) T cells with innate lymphocyte characteristics. Immunity. 2010;33:203–15. doi: 10.1016/j.immuni.2010.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Piazza F, Costoya JA, Merghoub T, Hobbs RM, Pandolfi PP. Disruption of PLZP in mice leads to increased T-lymphocyte proliferation, cytokine production, and altered hematopoietic stem cell homeostasis. Mol Cell Biol. 2004;24:10456–69. doi: 10.1128/MCB.24.23.10456-10469.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kang BY, Miaw SC, Ho IC. ROG negatively regulates T-cell activation but is dispensable for Th-cell differentiation. Mol Cell Biol. 2005;25:554–62. doi: 10.1128/MCB.25.2.554-562.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Takamori M, Hatano M, Arima M, Sakamoto A, Fujimura L, Hartatik T, et al. BAZF is required for activation of naive CD4 T cells by TCR triggering. Int Immunol. 2004;16:1439–49. doi: 10.1093/intimm/dxh144. [DOI] [PubMed] [Google Scholar]
  • 90.Manders PM, Hunter PJ, Telaranta AI, Carr JM, Marshall JL, Carrasco M, et al. BCL6b mediates the enhanced magnitude of the secondary response of memory CD8+ T lymphocytes. Proc Natl Acad Sci USA. 2005;102:7418–25. doi: 10.1073/pnas.0501585102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Xu D, Holko M, Sadler AJ, Scott B, Higashiyama S, Berkofsky-Fessler W, et al. Promyelocytic leukemia zinc finger protein regulates interferon-mediated innate immunity. Immunity. 2009;30:802–16. doi: 10.1016/j.immuni.2009.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Maeda T, Merghoub T, Hobbs RM, Dong L, Maeda M, Zakrzewski J, et al. Regulation of B versus T lymphoid lineage fate decision by the proto-oncogene LRF. Science. 2007;316:860–6. doi: 10.1126/science.1140881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sakurai N, Maeda M, Lee SU, Ishikawa Y, Li M, Williams JC, et al. The LRF transcription factor regulates mature B cell development and the germinal center response in mice. J Clin Invest. 2011;121:2583–98. doi: 10.1172/JCI45682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Maeda T, Ito K, Merghoub T, Poliseno L, Hobbs RM, Wang G, et al. LRF is an essential downstream target of GATA1 in erythroid development and regulates BIM-dependent apoptosis. Dev Cell. 2009;17:527–40. doi: 10.1016/j.devcel.2009.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Adhikary S, Peukert K, Karsunky H, Beuger V, Lutz W, Elsässer HP, et al. Miz1 is required for early embryonic development during gastrulation. Mol Cell Biol. 2003;23:7648–57. doi: 10.1128/MCB.23.21.7648-7657.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kosan C, Saba I, Godmann M, Herold S, Herkert B, Eilers M, et al. Transcription factor miz-1 is required to regulate interleukin-7 receptor signaling at early commitment stages of B cell differentiation. Immunity. 2010;33:917–28. doi: 10.1016/j.immuni.2010.11.028. [DOI] [PubMed] [Google Scholar]
  • 97.Saba I, Kosan C, Vassen L, Möröy T. IL-7R-dependent survival and differentiation of early T-lineage progenitors is regulated by the BTB/POZ domain transcription factor Miz-1. Blood. 2011;117:3370–81. doi: 10.1182/blood-2010-09-310680. [DOI] [PubMed] [Google Scholar]
  • 98.Matic I, Schimmel J, Hendriks IA, van Santen MA, van de Rijke F, van Dam H, et al. Site-specific identification of SUMO-2 targets in cells reveals an inverted SUMOylation motif and a hydrophobic cluster SUMOylation motif. Mol Cell. 2010;39:641–52. doi: 10.1016/j.molcel.2010.07.026. [DOI] [PubMed] [Google Scholar]
  • 99.Siggs OM, Li X, Xia Y, Beutler B. ZBTB1 is a determinant of lymphoid development. J Exp Med. 2012;209:19–27. doi: 10.1084/jem.20112084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Satpathy AT, Kc W, Albring JC, Edelson BT, Kretzer NM, Bhattacharya D, et al. Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages. J Exp Med. 2012;209:1135–52. doi: 10.1084/jem.20120030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Meredith MM, Liu K, Darrasse-Jeze G, Kamphorst AO, Schreiber HA, Guermonprez P, et al. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J Exp Med. 2012;209:1153–65. doi: 10.1084/jem.20112675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Carter T. The genetics of luxate mice. I. morphological abnormalities of heterozygotes and homozygotes. J Genet. 1951;50:277–99. doi: 10.1007/BF02996224. [DOI] [PubMed] [Google Scholar]
  • 103.Green MC. Luxoid - a new hereditary leg and foot abnormality in the house mouse. J Hered. 1955;46:91–9. [Google Scholar]
  • 104.Barna M, Hawe N, Niswander L, Pandolfi PP. Plzf regulates limb and axial skeletal patterning. Nat Genet. 2000;25:166–72. doi: 10.1038/76014. [DOI] [PubMed] [Google Scholar]
  • 105.Barna M, Pandolfi PP, Niswander L. Gli3 and Plzf cooperate in proximal limb patterning at early stages of limb development. Nature. 2005;436:277–81. doi: 10.1038/nature03801. [DOI] [PubMed] [Google Scholar]
  • 106.Fischer S, Kohlhase J, Böhm D, Schweiger B, Hoffmann D, Heitmann M, et al. Biallelic loss of function of the promyelocytic leukaemia zinc finger (PLZF) gene causes severe skeletal defects and genital hypoplasia. J Med Genet. 2008;45:731–7. doi: 10.1136/jmg.2008.059451. [DOI] [PubMed] [Google Scholar]
  • 107.Buaas FW, Kirsh AL, Sharma M, McLean DJ, Morris JL, Griswold MD, et al. Plzf is required in adult male germ cells for stem cell self-renewal. Nat Genet. 2004;36:647–52. doi: 10.1038/ng1366. [DOI] [PubMed] [Google Scholar]
  • 108.Costoya JA, Hobbs RM, Barna M, Cattoretti G, Manova K, Sukhwani M, et al. Essential role of Plzf in maintenance of spermatogonial stem cells. Nat Genet. 2004;36:653–9. doi: 10.1038/ng1367. [DOI] [PubMed] [Google Scholar]
  • 109.Hobbs RM, Seandel M, Falciatori I, Rafii S, Pandolfi PP. Plzf regulates germline progenitor self-renewal by opposing mTORC1. Cell. 2010;142:468–79. doi: 10.1016/j.cell.2010.06.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Filipponi D, Hobbs RM, Ottolenghi S, Rossi P, Jannini EA, Pandolfi PP, et al. Repression of kit expression by Plzf in germ cells. Mol Cell Biol. 2007;27:6770–81. doi: 10.1128/MCB.00479-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ching YH, Wilson LA, Schimenti JC. An allele separating skeletal patterning and spermatogonial renewal functions of PLZF. BMC Dev Biol. 2010;10:33. doi: 10.1186/1471-213X-10-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Fedele M, Franco R, Salvatore G, Paronetto MP, Barbagallo F, Pero R, et al. PATZ1 gene has a critical role in the spermatogenesis and testicular tumours. J Pathol. 2008;215:39–47. doi: 10.1002/path.2323. [DOI] [PubMed] [Google Scholar]
  • 113.Kojima S, Hatano M, Okada S, Fukuda T, Toyama Y, Yuasa S, et al. Testicular germ cell apoptosis in Bcl6-deficient mice. Development. 2001;128:57–65. doi: 10.1242/dev.128.1.57. [DOI] [PubMed] [Google Scholar]
  • 114.Castrillon DH, Gönczy P, Alexander S, Rawson R, Eberhart CG, Viswanathan S, et al. Toward a molecular genetic analysis of spermatogenesis in Drosophila melanogaster: characterization of male-sterile mutants generated by single P element mutagenesis. Genetics. 1993;135:489–505. doi: 10.1093/genetics/135.2.489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Lukacsovich T, Yuge K, Awano W, Asztalos Z, Kondo S, Juni N, et al. The ken and barbie gene encoding a putative transcription factor with a BTB domain and three zinc finger motifs functions in terminalia development of Drosophila. Arch Insect Biochem Physiol. 2003;54:77–94. doi: 10.1002/arch.10105. [DOI] [PubMed] [Google Scholar]
  • 116.Salzberg A, D’Evelyn D, Schulze KL, Lee JK, Strumpf D, Tsai L, et al. Mutations affecting the pattern of the PNS in Drosophila reveal novel aspects of neuronal development. Neuron. 1994;13:269–87. doi: 10.1016/0896-6273(94)90346-8. [DOI] [PubMed] [Google Scholar]
  • 117.Hu S, Fambrough D, Atashi JR, Goodman CS, Crews ST. The Drosophila abrupt gene encodes a BTB-zinc finger regulatory protein that controls the specificity of neuromuscular connections. Genes Dev. 1995;9:2936–48. doi: 10.1101/gad.9.23.2936. [DOI] [PubMed] [Google Scholar]
  • 118.Giniger E, Tietje K, Jan LY, Jan YN. lola encodes a putative transcription factor required for axon growth and guidance in Drosophila. Development. 1994;120:1385–98. doi: 10.1242/dev.120.6.1385. [DOI] [PubMed] [Google Scholar]
  • 119.Zhu S, Lin S, Kao CF, Awasaki T, Chiang AS, Lee T. Gradients of the Drosophila Chinmo BTB-zinc finger protein govern neuronal temporal identity. Cell. 2006;127:409–22. doi: 10.1016/j.cell.2006.08.045. [DOI] [PubMed] [Google Scholar]
  • 120.Okado H, Ohtaka-Maruyama C, Sugitani Y, Fukuda Y, Ishida R, Hirai S, et al. The transcriptional repressor RP58 is crucial for cell-division patterning and neuronal survival in the developing cortex. Dev Biol. 2009;331:140–51. doi: 10.1016/j.ydbio.2009.04.030. [DOI] [PubMed] [Google Scholar]
  • 121.Xie Z, Ma X, Ji W, Zhou G, Lu Y, Xiang Z, et al. Zbtb20 is essential for the specification of CA1 field identity in the developing hippocampus. Proc Natl Acad Sci USA. 2010;107:6510–5. doi: 10.1073/pnas.0912315107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Sutherland AP, Zhang H, Zhang Y, Michaud M, Xie Z, Patti ME, et al. Zinc finger protein Zbtb20 is essential for postnatal survival and glucose homeostasis. Mol Cell Biol. 2009;29:2804–15. doi: 10.1128/MCB.01667-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Xie Z, Zhang H, Tsai W, Zhang Y, Du Y, Zhong J, et al. Zinc finger protein ZBTB20 is a key repressor of alpha-fetoprotein gene transcription in liver. Proc Natl Acad Sci USA. 2008;105:10859–64. doi: 10.1073/pnas.0800647105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Carter MG, Johns MA, Zeng X, Zhou L, Zink MC, Mankowski JL, et al. Mice deficient in the candidate tumor suppressor gene Hic1 exhibit developmental defects of structures affected in the Miller-Dieker syndrome. Hum Mol Genet. 2000;9:413–9. doi: 10.1093/hmg/9.3.413. [DOI] [PubMed] [Google Scholar]
  • 125.Kim SW, Park JI, Spring CM, Sater AK, Ji H, Otchere AA, et al. Non-canonical Wnt signals are modulated by the Kaiso transcriptional repressor and p120-catenin. Nat Cell Biol. 2004;6:1212–20. doi: 10.1038/ncb1191. [DOI] [PubMed] [Google Scholar]
  • 126.Filion GJ, Zhenilo S, Salozhin S, Yamada D, Prokhortchouk E, Defossez PA. A family of human zinc finger proteins that bind methylated DNA and repress transcription. Mol Cell Biol. 2006;26:169–81. doi: 10.1128/MCB.26.1.169-181.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.de Greef JC, Wang J, Balog J, den Dunnen JT, Frants RR, Straasheijm KR, et al. Mutations in ZBTB24 are associated with immunodeficiency, centromeric instability, and facial anomalies syndrome type 2. Am J Hum Genet. 2011;88:796–804. doi: 10.1016/j.ajhg.2011.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Chouery E, Abou-Ghoch J, Corbani S, El Ali N, Korban R, Salem N, et al. A novel deletion in ZBTB24 in a Lebanese family with immunodeficiency, centromeric instability, and facial anomalies syndrome type 2. Clin Genet. 2011 doi: 10.1111/j.1399-0004.2011.01783.x. [DOI] [PubMed] [Google Scholar]
  • 129.Hagleitner MM, Lankester A, Maraschio P, Hultén M, Fryns JP, Schuetz C, et al. Clinical spectrum of immunodeficiency, centromeric instability and facial dysmorphism (ICF syndrome) J Med Genet. 2008;45:93–9. doi: 10.1136/jmg.2007.053397. [DOI] [PubMed] [Google Scholar]
  • 130.Xu GL, Bestor TH, Bourc’his D, Hsieh CL, Tommerup N, Bugge M, et al. Chromosome instability and immunodeficiency syndrome caused by mutations in a DNA methyltransferase gene. Nature. 1999;402:187–91. doi: 10.1038/46214. [DOI] [PubMed] [Google Scholar]
  • 131.Reeves R, Nissen MS. The A.T-DNA-binding domain of mammalian high mobility group I chromosomal proteins. A novel peptide motif for recognizing DNA structure. J Biol Chem. 1990;265:8573–82. [PubMed] [Google Scholar]
  • 132.Skarnes WC, Rosen B, West AP, Koutsourakis M, Bushell W, Iyer V, et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature. 2011;474:337–42. doi: 10.1038/nature10163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Gondo Y. Trends in large-scale mouse mutagenesis: from genetics to functional genomics. Nat Rev Genet. 2008;9:803–10. doi: 10.1038/nrg2431. [DOI] [PubMed] [Google Scholar]
  • 134.Dereeper A, Guignon V, Blanc G, Audic S, Buffet S, Chevenet F, et al. Phylogeny.fr: robust phylogenetic analysis for the non-specialist. Nucleic Acids Res 2008; 36(Web Server issue):W465-9; 10.1093/nar/gkn180;. [DOI] [PMC free article] [PubMed]
  • 135.Lattin JE, Schroder K, Su AI, Walker JR, Zhang J, Wiltshire T, et al. Expression analysis of G Protein-Coupled Receptors in mouse macrophages. Immunome Res. 2008;4:5. doi: 10.1186/1745-7580-4-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Gebhardt A, Kosan C, Herkert B, Möröy T, Lutz W, Eilers M, et al. Miz1 is required for hair follicle structure and hair morphogenesis. J Cell Sci. 2007;120:2586–93. doi: 10.1242/jcs.007104. [DOI] [PubMed] [Google Scholar]
  • 137.Broxmeyer HE, Sehra S, Cooper S, Toney LM, Kusam S, Aloor JJ, et al. Aberrant regulation of hematopoiesis by T cells in BAZF-deficient mice. Mol Cell Biol. 2007;27:5275–85. doi: 10.1128/MCB.01967-05. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cell Cycle are provided here courtesy of Taylor & Francis

RESOURCES