Fig 1 - uploaded by Hiromitsu Nakauchi
Content may be subject to copyright.
Structure of mouse IgH locus. ( A ) Schematic representation of mouse IgH gene after recombination of the variable region. Boxes and circles indicate exons and enhancers, respectively. C α 3 Ј E and HS3 contain identical putative MAREs. ( B ) Comparison of the putative MAREs in the IgH C α 3 Ј E and HS3 with NF-E2-type MARE and TRE. 

Structure of mouse IgH locus. ( A ) Schematic representation of mouse IgH gene after recombination of the variable region. Boxes and circles indicate exons and enhancers, respectively. C α 3 Ј E and HS3 contain identical putative MAREs. ( B ) Comparison of the putative MAREs in the IgH C α 3 Ј E and HS3 with NF-E2-type MARE and TRE. 

Source publication
Article
Full-text available
Maf family transcription factors are important regulators in various differentiation systems. Putative Maf recognition elements (MAREs) are found in the 3' enhancer region of the immunoglobulin heavy chain (IgH) gene. These elements are bound in B-cell extracts by a heterodimeric protein complex containing both Bach2 and a small Maf protein. Analys...

Contexts in source publication

Context 1
... Furthermore, the two MAREs within the 3 Ј enhancer of the mouse IgH gene are bound by Bach2– small-Maf heterodimers in B-cell extracts (Figure 2). Finally, we show here that Bach2 is expressed at high levels in immature B cells but not in terminally differentiated B cells that express high levels of the IgH gene (plasmacytes; Figure 4). A similar expression profile of Bach2 was also evident in human B-lineage cells (E.Ito, T.Toki and K.Igarashi, unpublished observation). Since Bach2 does not act as a transcriptional activator and rather represses activity of the IgH 3 Ј enhancer regions (Figure 6), we interpret our findings to show that Bach2 functions by inhibiting premature initiation of robust transcription of the IgH locus in undifferentiated B cells. Consistent with this model, the IgH 3 Ј enhancer is inactive before terminal differentiation where Bach2 is expressed abundantly, but is very active in plasmacytoma or myeloma cells, in which Bach2 is not expressed (Madisen and Groudine, 1994; this study). The present results established that Bach2 represses the IgH 3 Ј enhancer by forming a heterodimer with a small Maf protein, because synergistic repression with MafK of the enhancer activity was lost upon deletion of the leucine zipper from Bach2 (Figure 6D). However, the MARE within the HS3 is not the only target cis -element. This was indicated by the results that deletion of the HS3 MARE did not completely abolish cooperative transcription repression by Bach2 and MafK (Figure 6E). In this regard, it should be noted there are several other TREs within HS1, HS2, HS3 and HS4 (Pettersson et al. , 1990; Lieberson et al. , 1991; Matthias and Baltimore, 1993; Madisen and Groudine, 1994; Grant et al. , 1995; Chauveau and Cogne, 1996). Because MARE is closely related to TRE, some of these TREs may actually be a MARE and bind Bach2–small-Maf heterodimer within cells. Indeed, Maf family members can bind to sequences that diverge considerably from the consensus MARE (Ho et al. , 1996). We are currently trying to identify other Bach2 target sites within the 3 Ј enhancer regions. We examined the effect of the HS3 MARE deletion in pro-B and pre-B cells because there was a possibility that the deletion would relieve repression and activate HS1234 enhancer activity in these early-stage cells. However, the HS3 MARE deletion did not cause activation of the reporter gene expression in these cells (data not shown). As the HS1234 is more than 6 kb in length and contains many cis -elements, its enhacer activity may not reflect function of a singular cis -element. Rather, output may be determined as a combination of activities of positive and negative cis - and trans -elements. Regarding repression, another candidate effector is BASP/Pax5 which is known to repress IgH 3 Ј enhancer activity in early B cells (Singh and Birshtein, 1993). An interesting possibility is that Bach2 could modulate the activity of the IgH locus through its BTB domain, which is one of the hallmarks of Bach proteins and distinguishes them from other p45-related factors (Oyake et al. , 1996). The BTB domain has been implicated in transcription repression by the transcription factors BCL6 and PLZF (Chen et al. , 1993; Ye et al. , 1993). These proteins interact with co-repressor molecules like SMRT through the BTB domain (Dhordain et al. , 1997; Grignani et al. , 1998; Lin et al. , 1998). Bach2 indeed binds to SMRT in a yeast two hybrid assay (Figure 7), raising the possibility that it recruits the co-repressor to the IgH locus. In the transfection assay, Bach2 repressed the IgH 3 Ј HS1234 mini-enhancer reporter gene that contains multiple binding sites for various B cell transcription factors. Such a dominant effect of Bach2 might be explained by its interaction with co-repressors like SMRT. We examined effect of histone deacetylase inhibitor trichostatin A, and found that it does not abrogate transcription repression by Bach2 (A.Muto and K.Igarashi, unpublished observation). Because trichostatin A inhibits some, but not all, of histone deacetylase (Carmen et al ., 1996), further studies are necessary to reveal the mechanism by which Bach2 represses transcription. It will also be interesting to analyze whether Bach2 participates in the function of the IgH 3 Ј enhancer in terms of altering the chromatin structure of the locus, thereby contributing to the LCR activity of this element (Madisen and Groudine, 1994). BTB-domain-containing proteins have previously been shown to be involved in regulating chromatin structure (Albagli et al. , 1995). We showed recently that Bach1 is a candidate molecule that binds to the β -globin LCR. Bach1 functions as a novel type of architectural transcription factor that mediates interactions among multiple MAREs within the β -globin LCR depending on the protein interaction through the BTB domain (Igarashi et al ., 1998). In this sense, it should be noted that IgH 3 Ј LCR also contains at least two separated MAREs (Figure 1) and other putative target sites. Hence Bach2 may orchestrate assembly of a regulatory complex on this region by mediating interactions among the cis -elements. Previous results suggest that stem cells express various genes at low levels that later become up-regulated when the stem cells differentiate into a specific hematopoietic cell lineage (Hu et al. , 1997). The commitment and differentiation of stem cells, which express these ‘differen- tiation markers’ at low levels, toward a particular cell lineage occurs by the induction and repression of specific sets of genes (Hu et al. , 1997). From this point of view, the up-regulation of Bach2 during differentiation of stem cells into B220 ϩ /IgM – bone marrow cells, as well as its high expression in B220 B31-1 cells, indicates a potential role of Bach2 in lineage commitment of B cells. Several transcription factor genes have been shown to be essential for proper development of B cells (Fitzsimmons and Hagman, 1996). Among these factors, EBF and BSAP/ Pax5 show a very similar expression pattern to that of Bach2 (Barberis et al. , 1990; Hagman et al. , 1993). Expression of these proteins is restricted to the B cell lineage; they are turned on at an early stage of B cell development and down-regulated upon maturation to plasma cells. Ikaros, another hematopoietic-specific transcription factor, is required for development of both B and T cells, and is expressed in the stem-cell fraction as well (Georgopoulos, 1997). Bach2 may act together with these B- and lymphoid-specific transcription factors, leading to the differentiation of B cells from uncommitted stem cells. In conclusion, the present study has identified Bach2 as a B cell-specific component of a small Maf heterodimer and implicated Bach2 in the regulation of B cell-specific gene expression. The results thus provide crucial inform- ation and together with further studies will enhance our ability to understand how the development of B cells is regulated by members of the bZip family of transcription ...
Context 2
... family transcription factors possess a conserved basic- region leucine zipper (bZip) domain which mediates protein–protein interactions and DNA binding (Nishizawa et al ., 1989; Kataoka et al ., 1993). While c-Maf, MafB and NRL contain putative transcription activation domains (Nishizawa et al. , 1989; Swaroop et al. , 1992; Kataoka et al. , 1994a), MafF, MafK and MafG lack canonical trans -activation domains (Andrews et al. , 1993b; Fujiwara et al. , 1993; Igarashi et al. , 1994, 1995b; Kataoka et al. , 1995; Blank et al. , 1997). MafF, MafG and MafK are essentially composed of bZip domains and are collectively referred to as the small Maf family proteins. Various dimeric combinations of Maf family proteins bind in vitro to a DNA sequence motif called T-MARE (TGCTGA G / C TCAGCA) containing a 12- O -tetradecanoyl- phorbol-13-acetate (TPA)-responsive element (TRE), TGA G / C TCA (Kataoka et al. , 1994a,b, 1995). Maf family proteins are emerging as important regulators of cell differentiation in various systems (Blank and Andrews, 1997; Motohashi et al. , 1997). The hematopoietic cell-specific transcription factor NF-E2 is a heterodimer formed between the erythroid- and mega- karyocyte-specific bZip protein, p45 NF-E2 and one of the small Maf family proteins (Andrews et al. , 1993a,b; Ney et al. , 1993; Igarashi et al. , 1994). Resulting heterodimers bind to a NF-E2 consensus site (TGCTGA G / C TCA T / C ), which is related to T-MARE (Andrews et al. , 1993a,b; Igarashi et al. , 1994). A simple nomenclature of NF-E2 binding sites as Maf recognition elements (MAREs, including T-MARE and its derivatives) was recently sug- gested (Motohashi et al. , 1997). Besides generating NF- E2, the expression patterns of the small Maf proteins suggest that they also function in other cell lineages and tissues (Fujiwara et al. , 1993; Igarashi et al. , 1995b; Motohashi et al. , 1996). Fos and several p45-related bZip factors such as Nrf1 (LCR-F1/TCF-11), Nrf2 (ECH), Bach1 and Bach2 have been shown to form heterodimers with the small Maf proteins and bind to the MARE in vitro (Chan et al. , 1993; Caterina et al. , 1994; Luna et al. , 1994; Moi et al. , 1994; Itoh et al. , 1995; Oyake et al. , 1996; Toki et al. , 1997; Johnsen et al. , 1998). However, the precise role of these potential heterodimeric combinations during development and cell differentiation has remained elusive. Among these bZip proteins, Bach proteins may play mechanistically distinct roles as MARE-binding proteins, since they bear a BTB/POZ domain (Oyake et al ., 1996). Several lines of evidence suggest that the BTB domain may be involved in the regulation of chromatin structure (Albagli et al ., 1995). MARE-like elements are found in the regulatory regions of an increasing number of genes (Kataoka et al. , 1994b). Of particular interest, we noticed that TREs within the 3 Ј enhancer region of the immunoglobulin heavy chain (IgH) gene (Pettersson et al. , 1990; Matthias and Baltimore, 1993; Madisen and Groudine, 1994) resemble MAREs. Since immunoglobulin genes are regulated by B-cell- specific transcription factors (Fitzsimmons and Hagman, 1996; Arulampalam et al. , 1997), the presence of MARE- like elements in the IgH enhancer region suggests the presence of B-cell-specific MARE effector proteins. In this study, we examined MARE-binding activity in B cells. The results presented here indicate that Bach2 functions as a B-cell- and developmental-stage-specific partner for a small Maf protein and that the heterodimer interacts with the MAREs. Interestingly, co-expression of Bach2 and MafK repressed activity of the IgH 3 Ј enhancer. Hence, the Bach2–small-Maf heterodimer may represent the first example of a cell-lineage-restricted negative effector of MAREs, and might be involved in the repression of immunoglobulin genes at earlier stages of B-cell differentiation. Several TREs have been identified in the 3 enhancer/ locus control region (LCR) of the immunoglobulin heavy chain (IgH) gene (Pettersson et al. , 1990; Lieberson et al. , 1991; Matthias and Baltimore, 1993; Madisen and Groudine, 1994; Grant et al. , 1995; Chauveau and Cogne, 1996). At least two of them, located in the C α 3 Ј E and HS3, are identical to the MARE consensus sequence (Figure 1). It should be noted that C α 3 Ј E is composed of an inverted repeat of HS3 (Chauveau and Cogne, 1996), and hence it contains an identical MARE. To examine whether Maf dimers bind to these elements in B cells, we carried out electrophoretic mobility shift assays (EMSAs) using nuclear extracts from various B cell lines (Figure 2). The DNA probe which we used is a 29 bp fragment that was derived from IgH HS3 and contains one putative MARE. As shown in Figure 2A, EMSA and competition assays revealed the presence of only one specific DNA-binding protein complex in the nuclear extracts prepared from the pro-B cell line 63-12 and from the mature B cell line BAL17 (Figure 2A, lanes 2–5). An oligonucleotide containing mutations in the MARE failed to compete with the bound complex (Figure 2A, lanes 6 and 7). Furthermore, the mutated DNA did not generate a corresponding protein–DNA complex when it was radiolabeled and incubated with the nuclear extracts (Figure 2A, lanes 8 and 9). These results established specific binding of the complex to the MARE. This MARE-binding complex was not detected using nuclear extracts from the plasmacytoma cell line J558L (Figure 2B, lane 4), indicating its stage- specific activity. To reveal constituents of the complex, we utilized antibodies that recognize various factors that could bind to a MARE. Formation of the specific protein– DNA complex was inhibited by anti-Bach2 as well as by anti-small-Maf antisera (Figure 2B, lanes 5, 6, 12 and 13). On the other hand, preimmune sera did not show any effect on complex formation (Figure 2C, lanes 6–7). Furthermore, anti-Fos antibodies, which react with c-Fos, FosB, Fra-1 and Fra-2, or anti-Jun antibodies, which recognize c-Jun, JunB and JunD, did not have any effects (Figure 2B, lanes 15–16; data not shown). Finally, involve- ment of Bach2 in the MARE-binding complex was confirmed using an anti-Bach2 monoclonal antibody (Figure 2C): addition of the Bach2 monoclonal antibody super- shifted the MARE-binding complex. These results estab- lished that the TRE within the 3 enhancers C 3 E and HS3 of the IgH gene is actually a MARE and bound by a heterodimer of Bach2 and one or another of the small Maf proteins in B-cell extracts. We have reported previously that expression of bach2 mRNA in mice is restricted to the brain and spleen. To determine the possible role played by Bach2 during hematopoiesis, we isolated total RNA samples from bone marrow, thymus and spleen of adult mice as well as from fetal livers (13.5 days post-coitus embryos), and performed RNA blotting analysis (Figure 3A). Hematopoietic cells in the adult bone marrow revealed a 2.5-fold higher level of bach2 mRNA expression compared with that of other hematopoietic tissues or brain. To determine the cell-lineage specificity of Bach2 expression in vivo , we fractionated hematopoietic cells from bone marrow, spleen and thymus into various lineages using lineage-specific monoclonal antibodies (mAbs) and made comparisons by RT–PCR (Figure 3B). Only the B220-positive (B220 ϩ ) B cells isolated from bone marrow showed a high level of Bach2 expression. Mac-1 ϩ cells (mono-macrophage lineage) did not express Bach2. These results clearly established that Bach2 is a B-cell-restricted transcription factor. The relative expression levels in bone marrow and spleen suggests that Bach2 is expressed during the earlier stages of B-cell differentiation. This possibility has been addressed by fractionating the B220 ϩ cells into two populations, depending on the cell surface expression of IgM. The B220 ϩ /IgM – fraction contains pro- and pre-B cells, whereas the B220 ϩ /IgM ϩ fraction contains immature- and mature-B cells (Hardy et al ., 1991; Rolink and Melchers, 1991). As a source of mature B-cell population we also assessed B220 ϩ cells from spleen . As shown in Figure 3C, the B220 ϩ /IgM – fraction, which represents an early stage of B-cell lineage development, showed the highest level of expression of bach2 mRNA, whereas the B220 ϩ /IgM ϩ bone marrow cells and B220 ϩ spleen cells expressed less bach2 mRNA. The mitogen lipopolysaccha- ride (LPS) is known to induce proliferation and differentiation of resting B cells. Upon LPS treatment of spleen B cells, Bach2 expression was further down-regulated (to 50%, Figure 3D). Taken together, these results indicated that Bach2 expression decreases during the maturation of B cells. The relationship between Bach2 expression and B-cell development was further examined using various B cell lines at different stages of B-cell differentiation (Figure 4A). Among the cell lines examined, B31-1 is a stroma- dependent B-cell line and is at the earliest stage (N.Yanai and M.Obinata, unpublished observations). When cultured with the stromal cell line TBR31-1, B31-1 cells are a mixture of B220 – and B220 ϩ fractions, both of which express the pre-pro-B-cell-surface-marker-pattern like c-Kit ϩ , CD43-, HSA (CD24) – and IgM – (N.Yanai and M.Obinata, ...
Context 3
... the B220 ϩ /IgM ϩ bone marrow cells and B220 ϩ spleen cells expressed less bach2 mRNA. The mitogen lipopolysaccha- ride (LPS) is known to induce proliferation and differentiation of resting B cells. Upon LPS treatment of spleen B cells, Bach2 expression was further down-regulated (to 50%, Figure 3D). Taken together, these results indicated that Bach2 expression decreases during the maturation of B cells. The relationship between Bach2 expression and B-cell development was further examined using various B cell lines at different stages of B-cell differentiation (Figure 4A). Among the cell lines examined, B31-1 is a stroma- dependent B-cell line and is at the earliest stage (N.Yanai and M.Obinata, unpublished observations). When cultured with the stromal cell line TBR31-1, B31-1 cells are a mixture of B220 – and B220 ϩ fractions, both of which express the pre-pro-B-cell-surface-marker-pattern like c-Kit ϩ , CD43-, HSA (CD24) – and IgM – (N.Yanai and M.Obinata, unpublished observations). RT–PCR analysis revealed that bach2 was expressed abundantly at the earliest stage of B-cell development (i.e. B31-1 on TBR31-1; Figure 4A) and, in addition, in the stroma- independent pre-B, pro-B, immature-B and mature-B cell lines. In contrast, it was not detected in two different plasmacytoma cell lines, X63/0 (X63-Ag8.653) and J558L. Two stromal cell lines (TBR31-1 and ST-2) expressed the bach2 gene at a low level, raising the possibility that Bach2 may play some roles in stromal cells as well. Immunoblotting analysis with an anti-Bach2 antiserum confirmed that Bach2 is expressed in the pro-B, pre-B, immature-B and mature-B cell lines, and that it is absent in the plasmacytoma cell lines (Figure 4B). Furthermore, the expression profile of Bach2 in the different B-cell lines correlates well with the presence or absence of the MARE-binding complex, as shown in the EMSA studies (Figure 2). On the other hand, the anti-small-Maf antibody reacted with an ~20 kDa antigen which was present in all of the B-cell lines including plasmacytoma, as well as in the murine erythroleukemic (MEL) cells, in which mafK is known to be expressed (Andrews et al ., 1993b; Igarashi et al ., 1995a,b; Figure 4C; data not shown). Hence, Bach2 determines the B-cell lineage and stage-specificity of MARE effectors by interacting with more broadly expressed small Maf proteins. Such a B-cell- and stage- specific Bach2 heterodimer will most probably play an important role during B-cell development and differentiation. To determine at which point Bach2 expression commences during the differentiation of B cells, we examined hematopoietic stem cells (c-Kit ϩ /Sca-1 ϩ /CD34 low or negative/ lineage markers negative). A single cell of the stem cell fraction was shown previously to be able to reconstitute bone marrow cells in lethally irradiated mice (Osawi et al ., 1996). Expression of Bach2 was compared with those in more differentiated c-Kit ϩ /Lin – cells, which include progenitor cells of various lineages as well as in lineage- markers-positive (Lin ϩ ) differentiated cells. As shown in Figure 5A, a significant level of bach2 mRNA could be detected in the stem cell fraction, whereas the c-Kit ϩ /Lin – cells expressed Bach2 at low levels. Considering the fact that the Lin ϩ fraction contained differentiated cells of various lineages and that Bach2 expression in this fraction is restricted to the B220 ϩ cells, the expression level in the stem-cell fraction was estimated to be relatively low compared with expression in B220 cells in the Lin ϩ fraction. B220 is known as one of the earliest markers expressed in the B-cell lineage (Li et al. , 1996). To determine the timing of Bach2 induction during B-cell development, we took advantage of the fact that the B31-1 cells, grown on the TBR31-1 stromal cells, are a mixture of B220 – and B220 ϩ cells, and that B220 – cells give rise to B220 ϩ cells in vitro (N.Yanai and M.Obinata, unpublished observations). Each population was purified and examined for the presence of Bach2 protein by immunoblot analysis. As shown in Figure 5B, expression of Bach2 in the less- differentiated B220 – fraction is more abundant than it is in the B220 ϩ fraction, suggesting that B220 – B31-1 cells, which are already committed to the B-cell lineage, express Bach2 at a high level. Taken together, these results indicate that Bach2 is expressed at low levels in uncommitted hematopoietic stem cells, and that its expression is up- regulated during, or soon after, commitment of stem cells to the B-cell lineage. The commitment to other hematopoietic cell lineages might then lead to the down- regulation of bach2 . To examine an effect of Bach2 on the IgH 3 enhancer activity, the HS1, –2, –3 and –4 were cloned in combination as described previously, downstream of the luciferase gene under the control of IgH promoter (Figure 6A). HS3 contains an inverted repeat of the C α 3 Ј E and carries an identical MARE that binds Bach2–small-Maf heterodimer (Figure 1). Hence, the HS1234 reporter plasmid contains at least one functional MARE. The HS1234 mini-enhancer was shown previously to be active in plasmacytoma cells but relatively inactive in pre-B cells (Madisen and Groudine, 1994). Accordingly, the HS1234 mini-enhancer failed to activate IgH promoter-driven reporter gene expression in pro-B- and pre-B-cell lines (Figure 6B). In contrast, the ability of the HS1234 to enhance transcription from the IgH promoter in plasmacytoma cells was evident, as shown in Figure 6B (compare lanes 9 and 10). Its stimulating activity was lower in mature B cells than in plasmacytoma cells (compare lanes 8 and 10). To examine the regulatory role of Bach2 in B-cell differentiation, we carried out co-transfection experiments (Figure 6C). In this experiment, we used 0.1 μ g of reporter plasmid (1 μ g was used in Figure 6B). Co-transfection of Bach2-expression plasmid into the plasmacytoma cells repressed reporter gene activity driven by the HS1234 (Figure 6C, lane 6). Expression of MafK alone resulted in only weak inhibition of the reporter gene activity (Figure 6C, lane 9). Interestingly, co-transfection of both Bach2- and MafK-expression plasmids resulted in more efficient repression that virtually abolished the effect of the HS1234 enhancer (Figure 6C, lanes 7 and 8), indicating that Bach2 and MafK cooperatively repressed gene expression. Neither of them showed significant repression of reporter gene in the absence of the HS1234, indicating that effects of Bach2 and MafK were mediated by HS1234. To verify the interaction of Bach2 and MafK in the observed cooperative transcription repression, we examined a Bach2 derivative that lacked the leucine zipper (Bach2 ∆ zip), and hence could not form a heterodimer with a small Maf protein (Figure 6D). In this experiment, we used 0.1 μ g of Bach2-expression plasmid per transfection (0.9 μ g was used in Figure 6C). Both wild-type Bach2 and Bach2 ∆ zip repressed the reporter gene activity to 60% in the absence of MafK. However, Bach2 ∆ zip did not exert synergistic transcription repression with MafK, whereas Bach2 did exert such an effect (compare lanes 4 and 6). The results indicated that a heterodimer of Bach2 and MafK was responsible for the observed cooperative repression of transcription. The residual repression activity of Bach2 ∆ zip that was independent of dimer formation may be due to its interaction with other proteins through remaining regions such as the BTB domain. Such an interaction could inhibit the function of other transcription factors that are involved in enhancer activity of the HS1234. In order to examine roles of the MARE within HS3 in the context of the large cis -regulatory region, we deleted the MARE from the 6.6 kb HS1234 mini-enhancer ( ∆ MARE). As shown in Figure 6E, the deletion caused a modest decrease in the reporter gene expression (compare lanes 1 and 5). Co-expression of Bach2 and MafK still showed some synergistic repression of such a reporter gene activity, even though its extent was reduced. Based on these observations, we could draw two conclusions. First, in plasmacytoma cells, the enhancer function of the HS1234 is partly dependent on the MARE within the HS3. Secondly, the HS3 MARE is not the sole target of the Bach2–small-Maf heterodimer. Other functional MAREs appear to be present within the IgH 3 Ј enhancer regions (see Discussion). The results described in Figure 6D and E, taken together, strongly implicated Bach2 as a negative regulator of IgH 3 Ј enhancer that functions by forming a heterodimer with small Maf proteins. Since several BTB-domain-containing proteins have been shown to bind to co-repressors like SMRT, and because we had shown that Bach2 functions as a transcription repressor, we examined whether Bach2 interacts with SMRT in a yeast two-hybrid system. Bach2 was fused to the GAL4 DNA binding domain, and the fusion protein was then expressed in yeast cells. The Bach2 fusion protein did not activate the GAL4-dependent HIS3 reporter gene (Figure 7). However, when it was co-expressed together with SMRT–GAL4 activation domain fusion protein, the Bach2–GAL4 fusion activated the reporter gene, resulting in histidine autotrophy. In contrast to the Bach2 fusion, the Bach1 fusion did not show any interaction with SMRT, as evidenced by its failure to support yeast growth in the presence of the SMRT fusion. These results thus indicate that Bach2 but not Bach1 binds specifically to SMRT. The results of the RT–PCR assay and EMSA, shown in this study, have established that Bach2 functions in B cells as a major MARE-binding factor. In terms of cell differentiation, B cell is the second hematopoietic cell lineage in which partners of MARE-effectors have been identified, with a precedent being NF-E2 p45 in erythroid cells (Andrews et al. , 1993a,b; Igarashi et al. , 1994). This is somewhat surprising in view of our previous observation that, among various hematopoietic cell lines tested, Bach2 ...

Similar publications

Article
Full-text available
We have reported previously that antisense c-jun overcomes a block of Friend erythroleukemia cells to differentiation suggesting that the factor c-Jun may be an important negative regulator of erythroid differentiation. The recently described erythroid transcription factor NF-E2 plays an important role in the regulation of the transcription of glob...

Citations

... A single SCF FBXO22 E3 complex, therefore, appears responsible for ubiquitinating both protomers of a BACH1 dimer ( Figure S3B). Mammalian BACH1 has a close paralog, BACH2, which is primarily expressed in the brain and spleen with well-documented roles in regulating immune cell differentiation [35][36][37] . ...
Preprint
Full-text available
The transcription factor BACH1 regulates heme homeostasis and oxidative stress responses and promotes cancer metastasis upon aberrant accumulation. Its stability is controlled by two F-box protein ubiquitin ligases, FBXO22 and FBXL17. Here we show that the homodimeric BTB domain of BACH1 functions as a previously undescribed quaternary structure degron, which is deciphered by the two F-box proteins via distinct mechanisms. After BACH1 is released from chromatin by heme, FBXO22 asymmetrically recognizes a cross-protomer interface of the intact BACH1 BTB dimer, which is otherwise masked by the co-repressor NCOR1. If the BACH1 BTB dimer escapes the surveillance by FBXO22 due to oxidative modifications, its quaternary structure integrity is probed by a pair of FBXL17, which simultaneously engage and remodel the two BTB protomers into E3-bound monomers for ubiquitination. By unveiling the multifaceted regulatory mechanisms of BACH1 stability, our studies highlight the abilities of ubiquitin ligases to decode high-order protein assemblies and reveal therapeutic opportunities to block cancer invasion via compound-induced BACH1 destabilization.
... As a transcriptional repressor, Bach2 was initially reported to play a key role in the B-cell lineage, and subsequent studies identified its function in the fate of the T-cell lineage as well [8]. Bach2 is abundantly expressed in the early stages of T-cell differentiation and expressed only sparsely in the terminal differentiation of T cells [7,10]. Compared with their naïve counterparts, CD8 + T, antigen-experienced, effector, and memory CD4 + T cells express lower levels of Bach2 [8]. ...
Article
Full-text available
T and B cells participate in the development of systemic lupus erythematosus (SLE). BTB and CNC homology 2 (Bach2) is an irreplaceable regulator in the T and B lineages that helps to maintain immune homeostasis. However, the function of Bach2 in the pathogenesis of SLE has not been studied in depth. Flow cytometry and qRT‒PCR were used to assess Bach2 levels, bisulfite sequencing PCR (BSP) was used to measure the methylation level, and silencing by electroporation and stimulation with a cytokine concentration gradient were used to investigate the effect of Bach2 on T cells. Bach2 expression was elevated in the helper T‐cell subsets (Tfh, Th1, Th2, Th17 and Treg cells) of SLE patients and negatively correlated with disease severity and autoantibody levels. CD4+ T cells from SLE patients had decreased methylation levels in the Bach2 promoter region. Silencing Bach2 in CD4+ T cells induced increases in the CD19+ B‐cell count, plasmablasts and secretion of IgG by prompting the secretion of cytokines. The activation signals CD3/CD28, IL‐6 and IL‐21 upregulated Bach2 expression in CD4+ T cells. The regulation of Bach2 by cytokines and T‐cell activation signals in CD4+ T cells was shown to act on B cells and play a protective role against SLE. This article is protected by copyright. All rights reserved
... Furthermore, BACH2 is a transcriptional factor connected to the maturation of B-cell specificity and the establishment of germinal centers [125][126][127], and its transcription is activated by IKAROS. In pre-B-ALL, chronic myeloid leukemia, and Ph-positive ALL cells, it performs as a tumor suppressor, controls the pre-BCR checkpoint and promotes apoptosis in response to oxidative stress [128][129][130]. In the case of IKZF1 deletion, BACH2 expression levels drop, resulting in a lower disease-free survival in pediatric ALL patients [108]. ...
Article
Full-text available
Citation: Conserva, M.R.; Redavid, I.; Anelli, L.; Zagaria, A.; Tarantini, F.; Cumbo, C.; Tota, G.; Parciante, E.; Coccaro, N.; Minervini, C.F.; et al. IKAROS in Acute Leukemia: A Positive Influencer or a Mean Hater? Int. J. Mol. Sci. 2023, 24, 3282. Abstract: One key process that controls leukemogenesis is the regulation of oncogenic gene expression by transcription factors acting as tumor suppressors. Understanding this intricate mechanism is crucial to elucidating leukemia pathophysiology and discovering new targeted treatments. In this review, we make a brief overview of the physiological role of IKAROS and the molecular pathway that contributes to acute leukemia pathogenesis through IKZF1 gene lesions. IKAROS is a zinc finger transcription factor of the Krüppel family that acts as the main character during hematopoiesis and leukemogenesis. It can activate or repress tumor suppressors or oncogenes, regulating the survival and proliferation of leukemic cells. More than 70% of Ph+ and Ph-like cases of acute lymphoblastic leukemia exhibit IKZF1 gene variants, which are linked to worse treatment outcomes in both childhood and adult B-cell precursor acute lymphoblastic leukemia. In the last few years, much evidence supporting IKAROS involvement in myeloid differentiation has been reported, suggesting that loss of IKZF1 might also be a determinant of oncogenesis in acute myeloid leukemia. Considering the complicated "social" network that IKAROS manages in hematopoietic cells, we aim to focus on its involvement and the numerous alterations of molecular pathways it can support in acute leukemias.
... These results imply that NRF2 has a critical role in maintaining HSPC function through quiescence and self-renewal and by extension, differentiation. It was also recognized from Nrf2 loss-of-function mice that NRF2 does not control lineage specification in hematopoiesis unlike other CNC family genes such as Nfe2p45 (Kuroha et al, 1998;Motohashi et al, 2010) or Bach2 (Muto et al, 1998). ...
Article
Full-text available
Significance: The transcription factor Nrf2 (NF-E2-related factor 2) plays an important role as a master regulator of the cellular defense system by activating transcriptional programs of Nrf2 target genes encoding multiple enzymes related to cellular redox balance and xenobiotic detoxication. Comprehensive transcriptional analyses continue to reveal an ever-broadening range of Nrf2 target genes, demonstrating the sophistication and diversification of Nrf2 biological signatures beyond its canonical cytoprotective roles. Recent advances: Accumulating evidence indicates that Nrf2 has a strong association with the regulation of cell fates by influencing key processes of cellular transitions in the three major phases of the life cycle of the cell (i.e., cell birth, cell differentiation, and cell death). The molecular integration of Nrf2 signaling into this regulatory program occurs through a wide range of Nrf2 target genes encompassing canonical functions and those manipulating cell fate pathways. Critical issues: A singular focus on Nrf2 signaling for dissecting its' actions limits in-depth understanding of its intersection with the molecular machinery of cell fate determinations. Compensatory responses of downstream pathways governed by Nrf2 executed by a variety of transcription factors and multifactorial signaling crosstalk require further exploration. Future directions: Further investigations using optimized in vivo models and active engagement of overarching approaches to probe the interplay of widespread pathways are needed to study the properties and capabilities of Nrf2 signaling as a part of a large network within the cell fate regulatory domain.
... Nucleuslocated BACH2 is mostly bound to genes together with its heterodimer partner MafK. 27 Using the TFSearch and PROMO 3.0 online tools, we found 2 independent binding sties on the Rip1 promoter (−766/−762, Rip1-1; −76/−72, Rip1-2) and Rip3 promoter (−1112/−1106, Rip3-1; −137/−132, Rip3-2) for BACH2 ( Figure S5E); however, this method failed to predict binding sites of BACH2 on the Mlkl promoter. Chromatin immunoprecipitation assays showed that BACH2 and its partner MafK indeed bound to the specific promoter sites of Rip1 and Rip3 and that this binding activity was further enhanced in CB2R-overexpressed cells ( Figure 3F). ...
Article
Background: Diabetic heart dysfunction is a common complication of diabetes. Cell death is a core event that leads to diabetic heart dysfunction. However, the time sequence of cell death pathways and the precise time to intervene of particular cell death type remain largely unknown in the diabetic heart. This study aims to identify the particular cell death type that is responsible for diabetic heart dysfunction and to propose a promising therapeutic strategy by intervening in the cell death pathway. Methods: Type 2 diabetes models were established using db/db leptin receptor-deficient mice and high-fat diet/streptozotocin-induced mice. The type 1 diabetes model was established in streptozotocin-induced mice. Apoptosis and programmed cell necrosis (necroptosis) were detected in diabetic mouse hearts at different ages. G protein-coupled receptor-targeted drug library was searched to identify potential receptors regulating the key cell death pathway. Pharmacological and genetic approaches that modulate the expression of targets were used. Stable cell lines and a homemade phosphorylation antibody were prepared to conduct mechanistic studies. Results: Necroptosis was activated after apoptosis at later stages of diabetes and was functionally responsible for cardiac dysfunction. Cannabinoid receptor 2 (CB2R) was a key regulator of necroptosis. Mechanically, during normal glucose levels, CB2R inhibited S6 kinase-mediated phosphorylation of BACH2 at serine 520, thereby leading to BACH2 translocation to the nucleus, where BACH2 transcriptionally repressed the necroptosis genes Rip1, Rip3, and Mlkl. Under hyperglycemic conditions, high glucose induced CB2R internalization in a β-arrestin 2-dependent manner; thereafter, MLKL (mixed lineage kinase domain-like), but not receptor-interacting protein kinase 1 or 3, phosphorylated CB2R at serine 352 and promoted CB2R degradation by ubiquitin modification. Cardiac re-expression of CB2R rescued diabetes-induced cardiomyocyte necroptosis and heart dysfunction, whereas cardiac knockout of Bach2 diminished CB2R-mediated beneficial effects. In human diabetic hearts, both CB2R and BACH2 were negatively associated with diabetes-induced myocardial injuries. Conclusions: CB2R transcriptionally repressed necroptosis through interaction with BACH2; in turn, MLKL formed a negative feedback to phosphorylate CB2R. Our study provides the integrative view of a novel molecular mechanism loop for regulation of necroptosis centered by CB2R, which represents a promising alternative strategy for controlling diabetic heart dysfunction.
... Our main guess is that the IgH 3′RR is providing a higher specificity of expression for those stages corresponding to activated B-cells and GC B-cells and could thus provide optimal pre-malignant settings for lymphomas of the GC or post-GC type. It is indeed noticeable that the 3′RR not only includes transcriptional enhancers but also sites for transcriptional repression after binding factors such as Bach2, Mafk, and Pax5 [64][65][66][67]. In contrast, expression of a knock-in oncogene within the Igk locus yields expression at all B-cell stages under the influence of both the intronic Ek and the downstream kE3′ enhancer. ...
Article
Full-text available
Upregulated expression of the anti-apoptotic BCL2 oncogene is a common feature of various types of B-cell malignancies, from lymphoma to leukemia or myeloma. It is currently unclear how the various patterns of deregulation observed in pathology eventually impact the phenotype of malignant B cells and their microenvironment. Follicular lymphoma (FL) is the most common non-Hodgkin lymphoma arising from malignant germinal center (GC) B-cells, and its major hallmark is the t(14:18) translocation occurring in B cell progenitors and placing the BCL2 gene under the control of the immunoglobulin heavy chain locus regulatory region (IgH 3′RR), thus exposing it to constitutive expression and hypermutation. Translocation of BCL2 onto Ig light chain genes, BCL2 gene amplification, and other mechanisms yielding BCL2 over-expression are, in contrast, rare in FL and rather promote other types of B-cell lymphoma, leukemia, or multiple myeloma. In order to assess the impact of distinct BCL2 deregulation patterns on B-cell fate, two mouse models were designed that associated BCL2 and its full P1-P2 promoter region to either the IgH 3′RR, within a “3′RR-BCL2” transgene mimicking the situation seen in FL, or an Ig light chain locus context, through knock-in insertion at the Igκ locus (“Igκ-BCL2” model). While linkage to the IgH 3′ RR mostly yielded expression in GC B-cells, the Igκ-driven up-regulation culminated in plasmablasts and plasma cells, boosting the plasma cell in-flow and the accumulation of long-lived plasma cells. These data demonstrate that the timing and level of BCL2 deregulation are crucial for the behavior of B cells inside GC, an observation that could strongly impact the lymphomagenesis process triggered by secondary genetic hits.
... The transcription factor, BTB domain And CNC Homolog 2 (Bach2), was identified as a transcriptional repressor. It forms a heterodimer with Maf family proteins and binds to a DNA motif called T-MARE (TGCTGA G/C TCAGCA), a Maf recognition element (MARE), to regulate gene expression (Muto et al., 1998). The regulatory function of Bach2 is mediated through its interaction with the super-enhancers (SEs), and its aberrant expression is associated with a variety of autoimmune diseases as well as cancers (Afzali et al., 2017;Marroquí et al., 2014;Roychoudhuri et al., 2016b). ...
... Bach2 is expressed in CLP and represses genes of myeloid lineage to promote the development of cells in the lymphoid lineage (Itoh-Nakadai et al., 2014). Bach2 was first shown to be a B cell-intrinsic transcription factor that regulates B cell development through inhibiting the expression of Blimp-1 (encoded by Prdm1) (Muto et al., 1998;Ochiai et al., 2006). The rapid upregulation of Blimp-1 mediated by Bach2 deficiency promotes the terminal differentiation of B cells toward plasma cells even prior to class-switch recombination (CSR) (Muto et al., 2004). ...
... The online version of this article includes the following figure supplement(s) for figure 5: of the immunoglobulin heavy chain of activated p53 by competing with BCL6 for functional VDJ rearrangements (Muto et al., 1998;Swaminathan et al., 2013). Bach2 also suppressed the differentiation of activated B cells to plasma cells by inhibiting the expression of Blimp-1 (encoded by Prdm1 gene), which allowed CSR and somatic hypermutation to take place before becoming plasma cells Muto et al., 2004;Ochiai et al., 2006). ...
Article
Full-text available
BTB domain And CNC Homolog 2 (Bach2) is a transcription repressor that actively participates in T and B lymphocyte development, but it is unknown if Bach2 is also involved in the development of innate immune cells, such as natural killer (NK) cells. Here, we followed the expression of Bach2 during murine NK cell development, finding that it peaked in immature CD27 ⁺ CD11b ⁺ cells and decreased upon further maturation. Bach2 showed an organ and tissue-specific expression pattern in NK cells. Bach2 expression positively correlated with the expression of transcription factor TCF1 and negatively correlated with genes encoding NK effector molecules and those involved in the cell cycle. Lack of Bach2 expression caused changes in chromatin accessibility of corresponding genes. In the end, Bach2-deficiency resulted in increased proportions of terminally differentiated NK cells with increased production of granzymes and cytokines. NK cell-mediated control of tumor metastasis was also augmented in the absence of Bach2. Therefore, Bach2 is a key checkpoint protein regulating NK terminal maturation.
... Consistent with the function of E proteins as transcription activators, Peng et al. found more genes activated than repressed by E47-ER at both time points (168). Among them are three genes encoding transcriptional repressors, Cbfa2t3, Jdp2 and Bach2 (169)(170)(171). ...
Article
Full-text available
T cells develop in the thymus from lymphoid primed multipotent progenitors or common lymphoid progenitors into αβ and γδ subsets. The basic helix-loop-helix transcription factors, E proteins, play pivotal roles at multiple stages from T cell commitment to maturation. Inhibitors of E proteins, Id2 and Id3, also regulate T cell development while promoting ILC differentiation. Recent findings suggest that the thymus can also produce innate lymphoid cells (ILCs). In this review, we present current findings that suggest the balance between E and Id proteins is likely to be critical for controlling the bifurcation of T cell and ILC fates at early stages of T cell development.
... where BACH2 expression was found to be high (Muto et al., 1998). Further, the expression of Bach2 mRNA was observed to be highest in the early stage of B cell lineage development before decreasing during B cell maturation, with the lowest level of expression seen in terminally differentiated B cells. ...
... PCR genotyping of cells belonging to different lymphoid lineages from animals of different genotypes confirms the specificity of Cremediated excision of Bach2 in NK cells of Bach2 flox Ncr1 iCre+ mice only ( Figure 5.2). This was important to test because of the widely reported roles for BACH2 in multiple lymphoid lineages (Muto et al., 1998;Roychoudhuri et al., 2016;Roychoudhuri et al., 2013). Additionally, promiscuous Cre-deletion of Bach2 has previously been reported in other models (Grant et al., 2020). ...
Thesis
Natural killer (NK) cells are critical to immune surveillance against infections and cancer. Their role in immune surveillance requires that NK cells are present within tissues in a quiescent state. The functional maturation of NK cells is a tightly regulated process which is controlled by transcription factors (TFs), and multiple positive regulators of this process have been defined. However, mechanisms by which NK cells remain quiescent in tissues are incompletely elucidated. The transcriptional repressor BACH2 plays a critical role within the adaptive immune system but its function within innate lymphocytes has been unclear. The studies presented here show that BACH2 acts as an intrinsic negative regulator of NK cell maturation and function. BACH2 is expressed within developing and mature NK cells and promotes the maintenance of immature NK cells by restricting their maturation in the presence of tonic IL-15 signalling. Loss of BACH2 within NK cells results in accumulation of activated NK cells with unrestrained cytotoxic function and increased immune surveillance to pulmonary cancer metastasis. These findings establish a critical function of BACH2 as a negative regulator of innate cytotoxic function and tumour immune surveillance by NK cells.
... L'action de la 3'RR est régulée, entre autres, par des facteurs trans qui peuvent être recrutés sur les nombreux sites de fixations répartis sur les quatre enhancers (Pinaud et al. 2011a). En effet, en plus des sites de fixation pour les facteurs oct1 et oct2 présents sur les élements hs3a et hs3b, (Matthias & Baltimore 1993) on retrouve également des sites de liaisons pour les protéines µE2 et µE5 de la famille E2A ou encore des sites de fixation de protéines inhibitrices bloquant la transcription aux stades précoces avant les réarrangements permettant l'expression d'une Ig fonctionnelle (Muto et al. 1998). ...
Thesis
Full-text available
De nombreux lymphomes B matures sont caractérisés par une translocation oncogénique au locus des chaines lourdes des immunoglobulines (IgH). Elles surviennent, le plus souvent, lors des différents remaniements géniques du locus IgH qui ponctuent la vie du lymphocyte B. L'oncogène transloqué est alors dérégulé sous l'action des deux principaux activateurs transcriptionnels qui y résident: Eµ et 3'RR. Il a précédemment été montré que la 3'RR est l'acteur majeur régulant les commutations de classes (CSR) notamment en participant au recrutement de l'enzyme AID et donc des cassures doubles brins (DSBs).Au cours de ma thèse, l'analyse de lymphomes B développés par trois modèles murins portant des KI de c-myc en différentes positions du locus IgH a permis de montrer une coopération des deux éléments Eµ et 3'RR afin de promouvoir la lymphomagenèse B bien que la 3'RR suffit, à elle seule, à induire le développement de lymphomes B matures. Nous montrons que son absence perturbe le bon recrutement des facteurs de réparation lors de la résolution des DSBs suite à la CSR, suggérant que sa défaillance pourrait représenter un risque de translocation oncogènique au locus IgH.L'étude du mécanisme d'action de la 3'RR a montré une différence entre les cellules B normales dans lesquelles HDAC1 est recrutée par son élément central hs1.2 et les cellules B lymphomateuses issues de nos lignées murines où c'est l'HAT CBP qui est recrutée sur les éléments hs3a et hs3b de la 3'RR. L'utilisation de l'HDACi SAHA impacte significativement la prolifération B, ainsi que les processus de CSR et de synthèse d'Ig dans les cellules B normales mais a un effet inconsistant sur la prolifération in-vitro de nos lymphomes B murins.Enfin, le développement de souris KI CmycC homozygotes a permis la mise en évidence d’un nouveau modèle murin développant des lymphomes B présentant un profil cellulaire et moléculaire myélome-like, faisant de ces souris un modèle intéressant pour la mise au point de nouvelles stratégies thérapeutiques dans le traitement de myélomes humains.