Phenotypic Mutation 'scant' (pdf version)
Allelescant
Mutation Type missense
Chromosome12
Coordinate76,432,235 bp (GRCm39)
Base Change T ⇒ C (forward strand)
Gene Zbtb1
Gene Name zinc finger and BTB domain containing 1
Synonym(s) C430003J21Rik
Chromosomal Location 76,417,040-76,443,724 bp (+) (GRCm39)
MGI Phenotype PHENOTYPE: Mice homozygous for an ENU induced mutation exhibit abnormal thymus, B cell, and T cell differentiation, and reduced numbers of T, B, and NK cells in the spleen. [provided by MGI curators]
Accession Number

NCBI RefSeq: NM_178744; MGI: 2442326

MappedYes 
Limits of the Critical Region 75096672 - 80094577 bp
Amino Acid Change Cysteine changed to Arginine
Institutional SourceBeutler Lab
Gene Model not available
AlphaFold Q91VL9
SMART Domains Protein: ENSMUSP00000041955
Gene: ENSMUSG00000033454
AA Change: C74R

DomainStartEndE-ValueType
BTB 24 121 1.01e-16 SMART
ZnF_C2H2 216 242 2.17e1 SMART
low complexity region 359 368 N/A INTRINSIC
ZnF_C2H2 421 443 3.38e1 SMART
ZnF_C2H2 534 554 1.4e1 SMART
ZnF_C2H2 578 600 2.02e-1 SMART
ZnF_C2H2 606 628 6.23e-2 SMART
ZnF_C2H2 634 656 1.62e0 SMART
ZnF_C2H2 662 684 1.08e-1 SMART
ZnF_C2H2 686 709 1.36e-2 SMART
Predicted Effect probably damaging

PolyPhen 2 Score 0.998 (Sensitivity: 0.27; Specificity: 0.99)
(Using ENSMUST00000042779)
Meta Mutation Damage Score Not available question?
Is this an essential gene? Possibly essential (E-score: 0.698) question?
Phenotypic Category Autosomal Recessive
Candidate Explorer Status loading ...
Single pedigree
Linkage Analysis Data
Penetrance 100% 
Alleles Listed at MGI

All alleles(141) : Targeted(5) Gene trapped(136)

Lab Alleles
AlleleSourceChrCoordTypePredicted EffectPPH Score
IGL01891:Zbtb1 APN 12 76432435 missense probably damaging 1.00
IGL02097:Zbtb1 APN 12 76433371 missense probably damaging 1.00
IGL02328:Zbtb1 APN 12 76433450 missense possibly damaging 0.75
IGL02496:Zbtb1 APN 12 76432169 missense possibly damaging 0.76
IGL03270:Zbtb1 APN 12 76432289 missense possibly damaging 0.59
Limited UTSW 12 76432601 missense probably damaging 0.99
Occasional UTSW 12 76433784 missense probably damaging 1.00
Old_friend UTSW 12 76432665 missense probably damaging 0.96
R0893:Zbtb1 UTSW 12 76432113 missense probably damaging 1.00
R1317:Zbtb1 UTSW 12 76433573 missense probably benign 0.00
R1525:Zbtb1 UTSW 12 76433206 missense probably benign
R1761:Zbtb1 UTSW 12 76432595 nonsense probably null
R2920:Zbtb1 UTSW 12 76432619 missense possibly damaging 0.83
R5307:Zbtb1 UTSW 12 76433014 missense probably damaging 1.00
R5718:Zbtb1 UTSW 12 76433698 missense probably benign
R5975:Zbtb1 UTSW 12 76433049 missense possibly damaging 0.88
R6484:Zbtb1 UTSW 12 76432665 missense probably damaging 0.96
R6493:Zbtb1 UTSW 12 76433247 missense probably benign
R6513:Zbtb1 UTSW 12 76432604 missense possibly damaging 0.55
R6904:Zbtb1 UTSW 12 76432985 nonsense probably null
R6948:Zbtb1 UTSW 12 76432601 missense probably damaging 0.99
R8725:Zbtb1 UTSW 12 76432646 missense probably damaging 1.00
R9202:Zbtb1 UTSW 12 76433784 missense probably damaging 1.00
R9303:Zbtb1 UTSW 12 76432773 missense probably damaging 0.98
R9305:Zbtb1 UTSW 12 76432773 missense probably damaging 0.98
X0028:Zbtb1 UTSW 12 76432073 missense probably damaging 1.00
Z1191:Zbtb1 UTSW 12 76432023 missense probably benign
Mode of Inheritance Autosomal Recessive
Local Stock Live Mice, Sperm, gDNA
MMRRC Submission 036487-MU
Last Updated 2019-02-11 5:32 PM by Diantha La Vine
Record Created unknown
Record Posted 2011-12-28
Phenotypic Description

The scanT phenotype was initially identified in a flow cytometry screen looking for aberrant percentages of immune cell types in blood from G3 mice homozygous for mutations induced by N-ethyl-N-nitrosourea (ENU) (Figure 1A) (1). Homozygous scanT animals displayed a severe reduction in CD4+ and CD8+ T cells, which express T cell receptor αβ subunits. This observation was later extended to T cells including γδ T and natural killer T (NKT) cells (Figure 1B) (1). The thymi of these animals are greatly reduced, and an examination of early T cell development revealed a lack of early thymic precursors (Figure 2A,B) (1).  In the spleens of the mutant mice, the numbers of all lymphocyte lineages (T, B and NK) were reduced while the number of myeloid cells was not (1).

 

Further examination of B cell development in the scanT bone marrow found that there was an accumulation of B cell progenitors at the Hardy Fraction C stage, with a subsequent reduction of progenitors at the later stages (1).  Furthermore, transitional and follicular B cells were reduced in the mutant spleen.  The mutant B cells were able to respond to a T independent immunization, indicating that the B cell defect was developmental rather than functional (1).

To confirm that the mutation in Zbtb1 caused the scanT phenotype, wild-type Zbtb1 was retrovirally transduced into scanT hematopoietic progenitors.  Following transduction, GFP+ T cells were recovered from these progenitors indicating that thymic development was restored (1).

The generation of radiation chimeras was used to determine whether the mutant phenotype was hematopoietic or non-hematopoietic in origin.  All blood lymphocytes (with the exception of CD11b+ myeloid cells) were wild-type donor-derived, indicating that the Zbtb1 mutant lymphoid progenitors were unable to compete (1).  The numbers (and percentages) of early hematopoietic progenitors, including common lymphoid progenitors (CLPs), were comparable between heterozygous and homozygous scanT mice (1).  The competitive disadvantage in the B and NK cell lineages as well as the degree of embryonic development observed in the scanT mice indicate that Zbtb1 has a specialized role in determining lymphoid fate.

Nature of Mutation
The scanT phenotype was mapped to Chromosome 12 at D12Mit157 with a peak LOD score of 4.13 by crossing the index mouse to a C3H female and intercrossing the F1 offspring (Figure 3A). Sequencing of the coding regions present in the critical region identified a T to C transition at position 15196 in the Genbank genomic region NC_000078 for the Zbtb1 gene on Chromosome 12 (Figure 3B). The mutation corresponds to nucleotide 701 of the Zbtb1 transcript in exon 2 of 2 total exons.
 
15181 AAGATAAGTGCTGAGTGCTTTGATCTCATTTTG
69    -K--I--S--A--E--C--F--D--L--I--L-
 
The mutated nucleotide is indicated in red lettering, and results in a cysteine to arginine substitution at amino acid 74 of the ZBTB1 protein.
Illustration of Mutations in
Gene & Protein
Protein Prediction
Figure 4. Domain structures of selected BTB-ZF proteins displaying N-terminal BTB domains and a variable number of C2H2 zinc fingers. The shaded proteins have roles in T cell development and function. The structure of LRF is very similar to that of Th-POK, while the BTB domain of ZBTB1 most closely resembles that of MIZ-1. MAZR also contains two AT hooks (not shown). Sequence numbering is for murine proteins. The amino acid altered by the scanT mutation results in a cysteine to arginine substitution at amino acid 74. This image is interactive; click on an allele to view more information about other Zbtb1 mutations.
 

The ZBTB1 protein contains 713 amino acids and is a member of the POK (POZ and Krüppel) or BTB-ZF family of transcription factors (2). Between 40-60 genes encoding POK proteins exist in mammalian genomes (2-4). BTB-ZF family members contain a combination of two domains: an N-terminal BTB (Broad complex, Tramtrack, and Bric à brac) or POZ (Poxviruses and Zinc-finger) domain, and the C2H2 Krüppel -type zinc finger motif named for its resemblance to the Drosophila melanogaster segmentation protein Krüppel (Figure 4). Most of these factors play roles in cell proliferation and development, and many have also been implicated as oncogenes or tumor suppressors in cancer (4). Mouse ZBTB1 is 94% identical to its human homologue.  

 
 

The BTB/POZ domain is a highly conserved domain of approximately 100 amino acids that contains a dimerization interface, a possible oligomerization surface, and surfaces for interactions with other factors, such as nuclear corepressors and histone deacetylases (HDACs) (3;5). Based on its protein-protein interaction abilities, a variety of functional roles have been identified for the domain, including transcriptional repression, cytoskeleton regulation, tetramerization and gating of ion channels, and protein ubiquitination/degradation (3). The most conserved region of the BTB domain consists of a core of 5 α-helices flanked by 3 short β-sheets, but various BTB-containing protein families display N- or C-terminal extensions to this core fold. Most members of the BTB-ZF family, including BCL6 (B-cell lymphoma 6), PZLF (promyelocytic leukemia zinc finger) and LRF (leukemia/lymphoa-related factor), have an N-terminal extension containing an additional β-sheet and α-helix that have been shown to form part of a homodimer interface (β1 and α1) (5-8). However, ZBTB1 is missing the N-terminal β -sheet, and has a BTB domain sequence that is most similar to that of MIZ-1 (Myc-interacting zinc-finger protein) with 43.6% identity (Figure 5). The structure of the MIZ-1 BTB domain has been solved (9), and can be described as an association of two dimers to form a tetramer. The two dimeric interfaces of the four MIZ-1 BTB subunits (A:B and C:D) resemble the structures of BCL6, PZLF and LRF homodimers including a hydrophobic interface of α-helices at the center of the structure formed by close packing of α1, α2 and α3 from each subunit (Figure 6A). For BCL6, PZLF and LRF, two β-sheet interfaces at the bottom of this core are formed by the interaction of β1 of one chain with β5 of its partner (Figure 6B) (5-8). This interaction is absent from the MIZ-1 structure due to the absence of β1, but otherwise the dimeric structure is retained. Tetramerization of the MIZ-1 BTB domain results from the association of the dimers at the solvent-exposed β3-β2-β4 fold located at the top of each subunit (A:D and B:C). Association of the dimers results in the displacement of an outer β4 strand from one of the subunits of each dimer, generating a tetramer with two five-stranded β-sheet interfaces. The two displaced β4 strands adopt an α-helical conformation and are then referred to as αx (Figure 6A). The tetramer is stabilized by a salt bridge between aspartic acids located at the N-termini of β4B/D and lysines at the N-termini of α2A/C (9). The BTB domain can also promote heterodimerization, and several BTB-ZF proteins are able to physically interact with each other or with other BTB-containing proteins (3;10). Such interactions include BCL6 with BAZF (BCL6 associated zinc finger) and MIZ-1 (11;12), PZLF with FAZF (Fanconi anemia zinc finger) (13), and MAZR (Myc-associated zinc finger protein-related factor) with the transcription factor, BACH2 (BTB and CNC homolog 2) (14).

 

Surface features of BTB-ZF BTB domains determine interactions with specific non-BTB partners. For instance, BCL6 recruits the SMRT (silencing mediator for retinoid and thyroid hormone receptor) corepressor to lateral grooves that extend from the dimer interface across the bottom of the molecule and involves the β1-sheet (7;15) (Figure 6B). A charged pocket that lies at the top between the BCL6, PZLF and LRF dimer subunits has also been implicated in corepressor recruitment. The depth and charge of this pocket, as well as differences in the lateral grooves, determines the specificity of binding partners for various BTB-ZF transcription factors (5-7). In MIZ-1, tetramerization blocks access to the charged pocket. This, along with the lack of the β1-sheet, suggests that MIZ-1 may not be able to bind to corepressors (9). Many of the amino acids that have been determined to be necessary for dimerization and corepressor binding in BTB-ZF factors are mostly conserved in ZBTB1. These include Leu 11, Arg 18, Asp 25 and Lys 39 (15;16). The latter two residues form the charged pocket in BCL6, PZLF and LRF (5-8)

BTB-ZF proteins generally interact with DNA via C2H2-type zinc finger motifs, the most common DNA-binding motifs found in eukaryotes (17). The number of zinc fingers varies widely protein to protein. ZBTB1 contains 8-9 predicted C2H2-type zinc finger motifs according to Uniprot (Q91VL9), and analysis using SMART (Simple Modular Architecture Research Tool). These occur at amino acids 216-242, 421-443, 448-470, 534-556, 578-600, 606-628, 634-656, 662-684, 686-709. The C2H2 or classical zinc finger contains the consensus sequence (F/Y)-X-C-X2-5-C-X3-(F/Y)-X5-Ψ-X2-H-X3-4-H, where X is any amino acid and Ψ is any hydrophobic residue (18). C2H2-type zinc fingers fold into a ββα structure and coordinate a zinc ion using the two conserved cysteine and histidine residues. Natural variants of this motif containing a cysteine as the final zinc-chelating residue (C2HC) also fold into the same structure. C2H2 zinc fingers can be divided into three groups based on the number and pattern of fingers: triple- C2H2 (binds a single ligand), multiple-adjacent- C2H2 (binds multiple ligands), and separated paired- C2H2 (19). C2H2 zinc fingers typically recognize DNA sequences by binding to the major groove of DNA via residues -1,2,3, and 6 of the α-helix, spanning 3-4 bases of DNA (18;19). An important determinant of specific DNA binding are the amino acid side chains present at these positions (20;21), but conserved linkers between adjacent fingers also play roles by interacting with the phosphate backbone of the DNA and by contacting the C-terminus of the adjacent α-helix through a hydrogen bond (21;22). These motifs have also been shown to bind to both RNA and protein substrates in many proteins (17;19)

Several BTB-ZF factors, including PLZF, BCL6, FAZF (Fanconi anemia zinc finger), MAZR and LRF, display the ability to interact with multiple protein partners through their zinc fingers (17). These protein-protein interactions typically involve binding to factors present in large transcriptional complexes such as transcription factors, nuclear receptors and corepressors. BTB-ZF proteins can also bind to each other through their zinc fingers, in addition to BTB-domain heterodimerization. BCL6 is able to bind to both PZLF and LRF through its zinc fingers (23;24). Different combinations of zinc fingers in these proteins have been shown to be involved in binding to different partners (17).         

In most BTB-ZF proteins, the zinc fingers are located near the C-terminus and are connected to the BTB domain by an unstructured, flexible linker domain. Thus, dimerized BTB proteins are likely to bind two promoter sites via their zinc fingers, but the exact spacing and orientation of these sites may not be critical depending on the length of the linker region (3). Despite not being well conserved, these domains may still have important functions by interacting with proteins that are part of a transcriptional repression or activation complex. Such interactions include the binding of BCL6 to the corepressor SIN3A (SIN3 homolog A, transcription regulator-yeast) and PZLF binding to the corepressor ETO (eight twenty-one) (16;25)

ZBTB1 can be phosphorylated on Ser 355 and Thr 356 (26). A putative phosphorylation site exists at Thr 169.

The scanT mutation results in a cysteine to arginine change in the predicted α3 helix of the BTB domain. It is unknown if the affected protein is expressed and localized normally.

Expression/Localization

Zbtb1 gene expression is upregulated during early mouse embryogenesis at roughly embryonic day (E) 9.5 in the mid-foregut (27)In situ hybridization in adult mouse brain shows low levels of expression with highest levels present in the medulla and pons (Allen Brain Atlas). According to SymAtlas, Zbtb1 mRNA is highly expressed in hematopoietic stem cells (HSCs), T cells and mast cells in the mouse, with moderate expression in myeloid cells, osteoblasts and B cells. In humans, ZBTB1 mRNA is expressed in many tissues especially in the heart, liver, skeletal muscle, adrenal cortex, and ganglions. Expression is seen in some lymphoma cell lines.

Background

The founding invertebrate BTB-ZF members are all transcriptional repressors, each regulating different Drosophila developmental processes. The broad-complex (BR-C) factors are essential for Drosophila metamorphosis (28;29). Tramtrack (Ttk) regulates compound eye development (30), and Bric à brac (Bab) is necessary for ovary morphogenesis and limb formation (31;32). In vertebrates, many of these transcription factors appear to mediate transcriptional repression as well and are linked to developmental and tumorigenic processes (4). The developmental roles of BTB-ZF proteins are supported primarily through knockout approaches in mice and other model systems, while roles in tumorigenesis were identified by specific chromosomal aberrations and/or epigenetic modifications involving human genes encoding these factors. For example, PZLF was initially identified due to a chromosomal translocation event that results in a direct fusion of PZLF to the retinoic acid receptor α (RARα) and causes acute promyelocytic leukemia (APL) (33). BCL6 was also identified by being involved in chromosomal translocations that cause certain types of B cell lymphomas (34). The major functions of many of the well-studied vertebrate BTB-ZF transcription factors are listed in Table 1 [please see (2;4;10;35-38) and references therein]. Most BTB-ZF proteins appear to modulate transcription by recruiting transcriptional corepressors such as SIN3A, SMRT and NCoR1 (nuclear receptor corepressor 1), which in turn recruit HDACs resulting in histone deacetylation and transcriptional repression.

 
Table 1. Vertebrate BTB-ZF transcription factors*
 
Protein
Gene
Repressor/Activator
Functions
MIZ-1
Zbtb17
Both
Gastrulation; cell cycle progression; regulator of Myc-mediated gene regulation; interacts with BCL6 in germinal center B cells
HIC-1
Hic1
Repressor
Tumor suppressor; craniofacial development; DNA damage response; inhibits Wnt signaling
Kaiso
Zbtb33
Both
Gastrulation; Wnt signaling; synapse formation
APM-1
Zbtb7c
Repressor
Tumor suppressor; cell growth inhibition
LRF, Pokemon
Zbtb7a
Repressor
Proto-oncogene; stimulates HIV1 Tat activity; erythroid differentiation; represses T cell fate and promotes B cell development
PLZF
Zbtb16
Repressor
Limb development; gene regulation in APL; germ cell maintenance; appropriate differentiation of NKT and γδ T cell subsets
 
Th-POK, cKrox, Zfp67
Zbtb7B, hcKrox
Repressor
CD4+ T cell fate
MAZR
Zn278, Patz
Repressor
Represses CD8 expression during T cell development
BCL6
Bcl6, Znf51
Repressor
GC formation; germ cell maintenance; regulation of B cell differentiation and B cell lymphomas; regulation of T cell subsets
BCL6b, BAZF
Znf62, Bcl6b
Repressor
Activation of naïve T cells?
Memory T cell responses
FAZF, PLZP, ROG, TZFP
Zbtb32, Fazf
Repressor
T cell proliferation; cytokine production; HSC proliferation
* Shaded cells indicates proteins involved in T cell development
 

The majority of T cell development occurs in the thymus, but as thymic progenitor T cells lack self-renewing capacity, T lymphopoiesis requires periodic import of hemataopoietic progenitors from the blood. All blood cell types originate from multipotent, self-renewing hematopoietic stem cells (HSCs), which first differentiate into non-renewing multipotent progenitors (MPPs) through two intermediate steps known as long-term HSCs (LT-HSCs) and short-term HSCs (ST-HSCs) [reviewed by (39;40)]. Factors implicated in stem cell renewal and differentiation include the receptor tyrosine kinases Flt3 (FMS-like tyrosine kinase 3) and c-Kit (see the records for warmflash and Pretty2). Historically, MPPs have been divided into common myeloid progenitors (CMPs) and common lymphoid progenitors (CLPs) in the bone marrow (BM). CMPs give rise to cells of the myeloid lineage, while CLPs give rise to B, T and NK (natural killer) cell lymphocytes. Interleukin (IL)-7 /IL-7R signaling is critical for both B and T cell development, and CLPs were initially identified based on their expression of IL-7Rα (41-43). The current model of HSC differentiation suggests that erythroid potential is lost from MPPs, but myeloid potential is retained in progenitors of lymphocytes after the CLP stage and is only gradually lost through successive stages of differentiation (Figure 7). The stages of lymphoid development in the BM are identified by the expression of specific cell surface markers and the developmental potential of these cells in vitro and in vivo [reviewed by (39;40)], and include early lymphoid progenitors (ELPs) that express high levels of Flt3 and no VCAM-1 (vascular cell adhesion molecule-1;typically expressed on more primitive cell types), CLPs, CLP-2s and a rare BM population known as CTPs (committed T cell progenitors) that are able to differentiate into T cells outside of the thymus (44)

Although CLPs retain the potential to differentiate into T cells (39), recent evidence suggests that the hematopoietic cells that settle the thymus come from the earlier ELP stage as the earliest T lineage progenitors (ETPs) in the thymus retain myeloid potential and are phenotypically similar to ELPs in the BM (45-47). A subset of ELPs was shown to express CCR9, the receptor for the thymus-expressed chemokine (TECK) or CCL25, which is specifically expressed in the thymus (48;49). The expression of CCR9 on ELPs in the BM may enable these cells to populate the thymus, and recent studies suggest that both CCR9 and CCR7, which recognizes CCL19 and CCL21, together recruit hematopoietic progenitors to the adult thymus (50). Additionally, recruitment of adult thymic progenitors to the thymus is also regulated by the adhesion molecule P-selectin and its ligand PSGL1 (51). Once relocated to the thymus, T cell development goes through several stages leading to the generation of mature T cells including the double negative (DN) stages DN1-4, which lack expression of the T cell coreceptors CD4 and CD8. The most immature DN1 cells are the ETPs, which expess high levels of Kit, but are heterogeneous for other markers including CCR9 and Flt3. The DN2 stage is defined by the expression of both CD44 and the cell surface marker CD25 (the α-chain of the IL-2 receptor), while the DN3 stage is defined by downregulation of CD44 (39;40). Commitment to the T cell lineage is only complete at the DN3 stage where the cells undergo extensive DNA rearrangements at the β, γ, and δ loci in order to express functional TCR chains (52;53). After progressing through the DN4 stage (CD44-CD25-), αβ T cells then express both CD4 and CD8 and are known as double positive (DP) cells. They then go through positive selection to become CD4+ or CD8+ T cells. By contrast, most γδ T cells remain DN as they mature (54). CD4+ T cells or T helper cells go on to become several distinct subsets of T cells including T helper cell subsets Th1, Th2, Th3, Th17 and follicular helper (TFH) cells (please see the record for sanroque), as well as regulatory T cells (Tregs). Other T cell types include natural killer T (NKT) cells, which express receptors normally associated with NK cells and can rapidly produce large amounts of cytokines upon stimulation (55).

One of the factors essential for T cell specification and throughout T cell development is the Notch signaling pathway [reviewed by (56)]. The Notch system consists of interactions between type I membrane receptors (Notch receptors) and the Delta-like (Dll) and Jagged membrane-bound ligands. Notch proteins are synthesized as single precursor proteins that are cleaved in the Golgi, generating an extracellular subunit (NEC) that is noncovalently linked to the transmembrane/cytoplasmic subunit (NTM). Notch signaling is initiated by ligand receptor interaction, which induces a second cleavage of Notch near the TM domain mediated by ADAM-type metalloproteases (see the record for wavedX) followed by a third cleavage within the TM. The last cleavage allows the Notch intracellular domain (NICD) to localize to the nucleus and heterodimerize with the transcription factor RBP-J (recombining binding protein suppressor of hairless). Once bound to RBP-J, NICD recruits coactivators such as Mastermind-like 1 (MAML-1), which in turn recruit MED8-mediator transcription activation complexes (see the record for zeitgeist) to the promoters of Notch target genes. In most cells, these target genes include the transcriptional repressor Hes1 (hairy enhancer of split 1), Nrarp (Notch regulated ankyrin repeat protein), Deltex1, and Notch1 itself (57). T cell lineage genes are also directly activated by Notch including Ptcra (encoding the pre-TCRα chain) and Cd25. During T cell development, the strength of Notch signaling determines whether early progenitor populations in the BM adopt a B versus a T cell fate with high levels of Notch signaling favoring T cell differentiation (58). Inducible inactivation of Notch1 or RBP-J results in a block in T cell development accompanied by the accumulation of ectopic B cells in the thymus (59;60). Interference with Notch signaling by transgenic expression of Notch inhibitors including Fringe, Deltex1, or Nrarp also blocks T cell development in the thymus (56)

In addition to its role in T cell development, a connection between the activation of Notch and leukemia has been recognized.  It is speculated that Notch signaling in B-cell malignancies could be the result of expression of ligands on neighboring cells, of interactions between tumor cells as well as an interaction of the tumor cell with the microenvironment (61).  Evidence suggests that Notch signaling is essential for cross-talk between multiple myeloma cells and their environment (61).  In patients with T-cell acute lymphoblastic leukemia (T-ALL), a chromosomal translocation of Notch1 has been reported (62;63).  There is also a high rate of Notch1 activating mutations in T-ALL, indicating that Notch1 expression is linked to the development of T-ALL (64).  In addition to the role of Notch signaling in T-ALL, approximate 30% of patients with T-lymphotropic virus type I (HTLF-I)-associated adult T-cell leukemia (ATL) have activating Notch1 mutations (65).  The activating mutations are linked to single-substitution mutations that lead to reduced degradation and stabilization of activated Notch (65).  Furthermore, mutations and insertions in activated Notch lead to increased expression of Notch in cancer cells (66).  Furthermore, cancer-causing mutations in proteins that regulate the expression of Notch1 (e.g. CDC4/Fbw7) have been reported (67;68)

In addition to Notch, a core group of transcription factors are important for T cell determination [reviewed by (69)]. These include factors that are recurrently used during T cell differentiation like GATA-binding protein 3 (GATA3), MYB, basic helix-loop-helix E proteins such as E2A and HEB (HeLa E-box binding factor), high mobility group factor T cell factor 1 (TCF1), the zinc finger growth factor independent 1 transcription repressor (GFI1), the zinc finger Ikaros proteins, and runt-related transcription factor (RUNX) along with its binding partner core binding factor β (CBFβ). Other transcription factors such as the ETS (E26-transformation specific) family factor PU.1 are stage-specific. PU.1 switches from being essential for pre-thymic precursor development to an antagonist of T cell differentiation during the DN2 stages (70-73).  Along with PU.1, Ikaros, RUNX-CBFβ and E2A (and/or HEB) are needed to initiate early T cell differentiation in progenitor cells and are all necessary for the earliest stages of T cell development (74-76). Following this stage, GATA3 is required for differentiation into ETPs while conditional deletions of Gata3 at later stages during T cell development blocks β-selection and the generation of CD4+ T cells (77-79). TCF1 and GFI1 are specifically needed to generate DN2 cells (80;81). The zinc-finger transcription factor BCL-11b, HEBalt (HEB, alternative form) and GLI2 (glioma-associated oncogene 2), a transcription factor involved in the hedgehog (Hh) signaling pathway are all turned on at this stage. HEBalt and Hh signaling appear to be necessary for DN2 cells, but BCL-11b is required specifically for αβ T cells (82-85). MYB has critical functions at multiple stages of T cell development, although loss of MYB has little effect on γδ T cells (86-88). In addition to being necessary for the earliest stages of T cell development, RUNX-CBFβ is required to generate DN3 and CD8+ T cells (75;82).

Six different BTB-ZF factors are now known to play essential roles during the development and function of T cells (10;36;38), while a seventh, LRF, promotes B cell differentiation in early lymphoid progenitors of the BM by inhibiting Notch signaling and repressing T cell fate (35) (Table 1). All of these proteins are known transcriptional repressors and have been shown to bind DNA. Despite conservation of key residues in their BTB domains, these factors display diverse DNA target sequences and have divergent roles in T cell development and function (10;35;36;38;89;90)Bcl6-/- mice have increased Th2-specific cytokine responses and inflammation (91), reduced numbers of certain memory T cell subsets (92), and defective differentiation of T cells into TFH cells (89;90). BAZF, which is highly homologous to BCL6, may act together with BCL6 to repress certain target genes including some normally activated by signal transducer and activator of transcription 6 (STAT6), which promotes Th2 cell differentiation (91;93). BAZF also appears to control a subset of CD8+ memory T cell responses (94), and may be important for the proliferative responses of naïve T cells upon stimulation (95). MAZR regulates early T cell development by directly repressing Cd8 transcription in DN thymocytes (96), while PZLF is necessary for the proper differentiation of innate-like T cells including invariant NKT cells and specific subsets of γδ T cells (36;38). Mice with mutations in Zbtb7b, encoding Th-POK (T-helper-inducing POZ/Krüppel-like factor),display reduced numbers of CD4+ cells, while transgenic overexpression of Th-POK in the thymus redirects all positively selected T cells into the CD4+ lineage (79;97-100). Th-POK is involved in a negative feed back loop with RUNX transcription factors, which repress CD4 expression and allow CD8+ T cell development (82;98;99), and is downstream of GATA3 (79).  Th-POK also plays overlapping roles with PZLF in the differentiation of subsets of CD4-expressing NKT and γδT cells (36-38).  Finally, FAZF interacts with GATA3 and represses GATA3-mediated transactivation in CD4+ T cells (101;102). FAZF-deficient mice display increased T cell proliferation, cytokine production, and altered HSC homeostasis (102;103).

Putative Mechanism

As described above, several BTB-ZF transcription factors have been shown to play important roles at different stages of T cell development from very early stages with LRF to late stage specification of different T cell subsets. The phenotype of scanT homozygous animals shows that ZBTB1 is essential during lymphoid development, possibly because the mutation causes ZBTB1 to fail to proliferate, to fail to migrate to the thymus, or both (1).  There are several steps where ZBTB1 function may be critical. ZBTB1 may be necessary to promote Notch signaling at the ELP/CLP stage that is necessary for the commitment of these cells to the T cell lineage, and thus may play an opposing role to the one displayed by LRF. In the BM, LRF is able to repress the activation of Notch target genes in CLPs by the moderate levels of Notch ligands expressed in BM stromal cells, while the high expression of Notch ligands in the thymus overcomes LRF repression and allows the specification of T cell fate (35). Since BTB-ZF proteins are typically involved in transcriptional repression, the mechanism by which ZBTB1 may promote Notch signaling could involve repression of Notch inhibitors such as Fringe, Deltex1 or Nrarp. However, many of these proteins are themselves induced by Notch signaling (56). As Notch receptors are typically rapidly degraded by E3-ubiquitin ligases (including Fbw7 in T cells) (104), it is also possible that ZBTB1 may somehow repress this activity to extend Notch signaling in early T cells.

If ZBTB1 is involved in mediating Notch signals in T cell progenitors, it may also act at subsequent stages of T cell development as Notch signaling continues to be critical for committment to the T cell lineage in the thymus. Notch signaling is less critical for the development of γδ T cells as opposed to αβ T cells (54), making it likely that ZBTB1 functions prior to lineage commitment to this cell fate (during DN2 to DN3). Alternatively, ZBTB1 may be necessary to upregulate the expression of homing signals on T cell progenitor cells such as CCR7, CCR9 or PSGL1, thus allowing their migration to the thymus. In addition, ZBTB1 may be necessary to promote the migration of early T cell progenitors in the thymus as multiple chemokines, including CXCL12 (also known as stromal cell-derived factor 1, SDF-1), CCL19, CCL21, and CCL25, have been shown to be important in the homing of developing thymocytes to their specific microenvironment (105). DN thymocytes deficient for the chemokine receptor CXCR4, which recognizes CXCL12, fail to migrate to the cortex and do not differentiate beyond the DN stages (106). However, animals that lack both CCR7 and CCR9, displayed normal thymic cellularity despite severe reductions in the numbers of early thymic progenitors (50), unlike the phenotype observed in scanT animals. 

It is also possible that ZBTB1 plays a T cell-extrinsic role necessary for T cell development. The thymi of scanT animals are very small and suggests the possibility of aberrant thymus development, which would critically affect the maturation of most T cells [reviewed by (105)]. The thymus is divided into the cortex and the medulla with the majority of early T cell development, including positive selection, occurring in the cortex (and the subcapsular zone), while negative selection and the induction of self-tolerance occur in the medulla. Initial T cell differentiation until the DN3 stage is promoted by Notch-mediated signals delivered by binding of the Notch receptor by Delta ligands and is supported by IL-7, both signals derived from cortical thymic epithelial cells (cTECs) (41;43;59). Conversely, immature DN thymocytes also promote the differentiation of thymic stromal cells and trigger the formation of the cortical-epithelial environment in the thymus. Mice homozygous for the human CD3εtransgene overexpress CD3ε during T cell development (see the record for tumormouse), and display a complete block at the DN1 stage in the thymus with differentiation affected even earlier prior to the development of NK cells (107). The thymi of these animals are characterized by very low cellularity and the presence of cysts (108;109). In addition, animals with blocked T cell development due to defects in Notch signaling display a severe reduction of thymus size (59;60).

Despite the conservation of key residues in the BTB domain of ZBTB1, the amino acid altered by the scanT mutation is not conserved with other BTB-containing proteins (Figure 5). Nevertheless, the severe T cell phenotype observed in scanT animals suggests that this residue is important for protein activity, and it is likely that the substitution of an arginine at this position will affect the structure and folding of the ZBTB1 BTB domain.  Other than the severe T cell phenotype observed in scanT mice these animals do not exhibit any other abnormalities. As no other mutations in Zbtb1 have been found in any vertebrate species, it is not clear if the scanT allele is null, hypomorphic or is even a gain-of-function allele.

Primers Primers cannot be located by automatic search.
Genotyping
ScanT genotyping is performed by amplifying the region containing the mutation using PCR, followed by sequencing of the amplified region to detect the single nucleotide transition. 
 
Primers
ScanT(F): 5’- TACAGAGGCTCAGGCATGTGACTG -3’
ScanT (R): 5’- AGGTAAAGCTCCGTCCAAAGCG  -3’
 
PCR program
1) 95°C             2:00
2) 95°C             0:30
3) 56°C             0:30
4) 72°C             1:00
5) repeat steps (2-4) 29X
6) 72°C             7:00
7) 4°C               ∞
 
Primers for sequencing
ScanT _seq(F): 5'- GCTGTATCGCTATCGACGAC -3'
ScanT _seq(R): 5’- CGAGACACACGTTCTTTTGG -3’
 
The following sequence of 1397 nucleotides (NCBI Mouse Genome Build 37.1, Chromosome 12, bases 77,485,484 to 77,486,880) is amplified:
 
                        tacagaggc tcaggcatgt gactgggttg ggtgtgtggt gtgtgtatgc
agtggaggcc ggcggtcagc ttgggtgttg ggttgggtgt gtggtgtgtg tgtgcagtgg
aggccagcgg tcagcctcag gtgttgttct tgggtgctgt ctaccttgtt ttttgagaca
gggtttcttg ttggcttaga actttccacg taggctagcc ggttgcagca ggtctcacgg
gtctgtccca tgcgcagctt tgtaaaatgt gtgtttctgg tggtcctcat gcttcctcat
tcgcttatgt ggatacaggg tctcactgag ttgccttggc tagcctggaa cttactctgt
agaccaggct ggtctggaac tcatagagac ctgcctgcct ctgcctcctg agtgtggcta
taagggacta aagactgtcc cagcacccca gacacgtcag ggcctggccc ttatgcttct
cagataagca ctccaccaac tgagaggtct tgggtttaga tttgttttta attttaaaaa
aaaattacgg attttgcttc tgattccttt tttgttcagt atgagaaaat tgaataaatg
actaggtatt ttcatcttca gacttagtag aaaaccatgc tttgattgtc atagcaaagg
tacacaagtg aaagtcatgc tcaagggtcc cttttgaagt gccttcctgc cgcctgaccg
ttacgttttg ttttgcatag gtcgagtcag cagaagatgg caaagcccag ccacagcagc
tatgtcctgc agcagctcaa caaccaaaga gagtggggct ttctctgtga ctgctgtatc
gctatcgacg acatctactt ccaagcacac aaagccgtcc tggctgcctg tagctcctac
tttaggatgt ttttcatgaa ccatcagcac agtactgcac aactgaacct cagcaacatg
aagataagtg ctgagtgctt tgatctcatt ttgcagttca tgtatttagg aaagatcatg
accgctcctt ccagtttcga gcagttcaaa gtggcaatga actacctcca gctgtacaat
gttcctgact gcttggaaga catacaggac gcagactgtt ctagttcaaa gtgttcgtct
tctgcctcca gcaggcagag cagcaagatg atttttgggg tgagaatgta tgaagacacg
gtggctagaa atggcaatga agccaacagg tggtgtgcgg agcccagttc aacggtgaat
acgccgcatc acagagaacc tgaggaagag tctttgcagt tggccaactt ccccgagccg
ctgtttgatg tgtgtaaaaa aagttctgtg tccaaattat ctactccaaa agaacgtgtg
tctcgacgct ttggacggag ctttacct
 
Primer binding sites are underlined; sequencing primer binding sites are highlighted in gray; the mutated T is indicated in red.
References
Science Writers Nora G. Smart, Anne Murray
Illustrators Nora G. Smart, Diantha La Vine
AuthorsOwen M. Siggs, Bruce Beutler