scholarly journals Global transcriptional coactivators CREB-binding protein and p300 are highly essential collectively but not individually in peripheral B cells

Blood ◽  
2006 ◽  
Vol 107 (11) ◽  
pp. 4407-4416 ◽  
Author(s):  
Wu Xu ◽  
Tomofusa Fukuyama ◽  
Paul A. Ney ◽  
Demin Wang ◽  
Jerold Rehg ◽  
...  

Abstract CREB-binding protein (CBP) and its para-log p300 are transcriptional coactivators that physically or functionally interact with over 320 mammalian and viral proteins, including 36 that are essential for B cells in mice. CBP and p300 are generally considered limiting for transcription, yet their roles in adult cell lineages are largely unknown since homozygous null mutations in either gene or compound heterozygosity cause early embryonic lethality in mice. We tested the hypotheses that CBP and p300 are limiting and that each has unique properties in B cells, by using mice with Cre/LoxP conditional knockout alleles for CBP (CBPflox) and p300 (p300flox), which carry CD19Cre that initiates floxed gene recombination at the pro–B-cell stage. CD19Cre-mediated loss of CBP or p300 led to surprisingly modest deficits in B-cell numbers, whereas inactivation of both genes was not tolerated by peripheral B cells. There was a moderate decrease in B-cell receptor (BCR)–responsive gene expression in CBP or p300 homozygous null B cells, suggesting that CBP and p300 are essential for this signaling pathway that is crucial for B-cell homeostasis. These results indicate that individually CBP and p300 are partially limiting beyond the pro-B-cell stage and that other coactivators in B cells cannot replace their combined loss.

2002 ◽  
Vol 9 (1) ◽  
pp. 35-45 ◽  
Author(s):  
Zhe-Xiong Lian ◽  
Hiroto Kita ◽  
Tomoyuki Okada ◽  
Tom Hsu ◽  
Leonard D. Shultz ◽  
...  

Reductions in populations of both Pre-B cell (Hardy fractions D) and Pro-B cells (Hardy fractions B–C) have been described in association with murine lupus. Recent studies of B cell populations, based on evaluation of B cell differentiation markers, now allow the enumeration and enrichment of other stage specific precursor cells. In this study we report detailed analysis of the ontogeny of B cell lineage subsets in New Zealand black (NZB) and control strains of mice. Our data suggest that B cell development in NZB mice is partially arrested at the fraction A Pre–Pro B cell stage. This arrest at the Pre-Pro B cell stage is secondary to prolonged lifespan and greater resistance to spontaneous apoptosis. In addition, expression of the gene encoding the critical B cell development transcription factor BSAP is reduced in the Pre–Pro B cell stage in NZB mice. This impairment may influence subsequent B cell development to later stages, and thereby accounts for the down-regulation of the B cell receptor componentIgα(mb-1). Furthermore, levels of expression of theRug2, λ5andIgβ(B29) genes are also reduced in Pre–Pro B cells of NZB mice. The decreased frequency of precursor B cells in the Pre–Pro B cell population occurs at the most primitive stage of B cell differentiation.


2020 ◽  
Vol 11 (9) ◽  
Author(s):  
Ken-Hong Lim ◽  
Lih-Chyang Chen ◽  
Kate Hsu ◽  
Chia-Ching Chang ◽  
Chia-Yu Chang ◽  
...  

Abstract BAFF supports B-cell survival and homeostasis by activating the NF-κB pathway. While NF-κB is also involved in the priming signal of NLRP3 inflammasome, the role of BAFF in NLRP3 inflammasome regulation is unknown. Here we report BAFF engagement to BAFF receptor elicited both priming and activating signals for NLRP3 inflammasomes in primary B cells and B lymphoma cell lines. This induction of NLRP3 inflammasomes by BAFF led to increased NLRP3 and IL-1β expression, caspase-1 activation, IL-1β secretion, and pyroptosis. Mechanistically, BAFF activated NLRP3 inflammasomes by promoting the association of cIAP-TRAF2 with components of NLRP3 inflammasomes, and by inducing Src activity-dependent ROS production and potassium ion efflux. B-cell receptor (BCR) stimulation on the Lyn signaling pathway inhibited BAFF-induced Src activities and attenuated BAFF-induced NLRP3 inflammasome activation. These findings reveal an additional function of BAFF in B-cell homeostasis that is associated with BCR activities.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2524-2524
Author(s):  
Baohua Sun ◽  
Saradhi Mallampati ◽  
Yun Gong ◽  
Donghai Wang ◽  
M. James You ◽  
...  

Abstract Abstract 2524 Poster Board II-501 B cells constitute an integral part of the immune system. The development of mature B cells from hematopoietic stem cells is a complex process that is regulated in a hierarchical order by various proteins, particularly transcription factors. Sox4 is a SRY-related HMG box containing transcription factor and is known to be involved in B cell development. However, the role of Sox4 in various stages of B cell development has not been systematically investigated. In this study we used a conditional knockout mouse strain and studied the effect of Sox4 deletion in B lymphopoiesis in adult mice. We crossed the Sox4-floxed mice with different Cre mouse strains that were expected to delete the floxed Sox4 gene at different B cell developmental stages. These Cre strains included Vav-iCre (expressed in hematopoietic stem cell stage, starting from early embryos), MX1-Cre (expression in hematopoietic stem cells, induced by pIpC injection in adults), MB1-Cre (expressed in B cells, starting from early progenitor cells in embryos), and CD21-Cre (expressed in mature B cells). We demonstrated that deletion of Sox4 caused an arrest of B lymphopoiesis at the transition from pre-pro-B cell (fraction A) stage to pro-B cell stage (fraction B): fraction A cells are slightly reduced in number whereas fraction B and later stage cells are nearly absent. The pre-pro-B cells from the Sox4 knockout mice retain a population of AA4.1+ cells, which are considered to be developed into B cells. Deletion of Sox4 in early embryonic stage (Vav-iCre) or in adults (Mx1-Cre) results in a similar phenotype on B lymphopoiesis, except that peritoneal B1 cells appear to be affected with Vav-iCre, but not with Mx1-Cre. MB1-Cre gave rise to similar results as did Vav-iCre, but the arrest was not as dramatic as with Vav-iCre. CD21-Cre produced no significant difference in B cell phenotype. These data suggested that Sox4 is required for early B cell development at the transition from pre-pro-B cells to pro-B cells and is not required for mature B cells. We are currently investigating the transcription program of this transcription factor in B cell development. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 114 (40) ◽  
pp. E8411-E8420 ◽  
Author(s):  
Pei Tong ◽  
Alessandra Granato ◽  
Teng Zuo ◽  
Neha Chaudhary ◽  
Adam Zuiani ◽  
...  

Ig heavy chain (IgH) isotypes (e.g., IgM, IgG, and IgE) are generated as secreted/soluble antibodies (sIg) or as membrane-bound (mIg) B cell receptors (BCRs) through alternative RNA splicing. IgH isotype dictates soluble antibody function, but how mIg isotype influences B cell behavior is not well defined. We examined IgH isotype-specific BCR function by analyzing naturally switched B cells from wild-type mice, as well as by engineering polyclonal Ighγ1/γ1 and Ighε/ε mice, which initially produce IgG1 or IgE from their respective native genomic configurations. We found that B cells from wild-type mice, as well as Ighγ1/γ1 and Ighε/ε mice, produce transcripts that generate IgM, IgG1, and IgE in an alternative splice form bias hierarchy, regardless of cell stage. In this regard, we found that mIgμ > mIgγ1 > mIgε, and that these BCR expression differences influence respective developmental fitness. Restrained B cell development from Ighγ1/γ1 and Ighε/ε mice was proportional to sIg/mIg ratios and was rescued by enforced expression of the respective mIgs. In addition, artificially enhancing BCR signal strength permitted IgE+ memory B cells—which essentially do not exist under normal conditions—to provide long-lived memory function, suggesting that quantitative BCR signal weakness contributes to restraint of IgE B cell responses. Our results indicate that IgH isotype-specific mIg/BCR dosage may play a larger role in B cell fate than previously anticipated.


2008 ◽  
Vol 28 (8) ◽  
pp. 2815-2824 ◽  
Author(s):  
Simanta Pathak ◽  
Shibin Ma ◽  
Long Trinh ◽  
Runqing Lu

ABSTRACT Receptor editing is the primary means through which B cells revise antigen receptors and maintain central tolerance. Previous studies have demonstrated that interferon regulatory factor 4 (IRF-4) and IRF-8 promote immunoglobulin light-chain rearrangement and transcription at the pre-B stage. Here, the roles of IRF-4 and -8 in receptor editing were analyzed. Our results show that secondary rearrangement was impaired in IRF-4 but not IRF-8 mutant mice, suggesting that receptor editing is defective in the absence of IRF-4. The role of IRF-4 in receptor editing was further examined in B-cell-receptor (BCR) transgenic mice. Our results show that secondary rearrangement triggered by membrane-bound antigen was defective in the IRF-4-deficient mice. Our results further reveal that the defect in secondary rearrangement is more severe at the immunoglobulin λ locus than at the κ locus, indicating that IRF-4 is more critical for the λ rearrangement. We provide evidence demonstrating that the expression of IRF-4 in immature B cells is rapidly induced by self-antigen and that the reconstitution of IRF-4 expression in the IRF-4 mutant immature B cells promotes secondary rearrangement. Thus, our studies identify IRF-4 as a nuclear effector of a BCR signaling pathway that promotes secondary rearrangement at the immature B-cell stage.


2017 ◽  
Author(s):  
Jason B Wong ◽  
Susannah L Hewitt ◽  
Lynn M Heltemes-Harris ◽  
Malay Mandal ◽  
Kristen Johnson ◽  
...  

SUMMARYB-1a cells are long-lived, self-renewing innate like B cells that predominantly inhabit the peritoneal and pleural cavities. In contrast to conventional B-2 cells they have a receptor repertoire that is biased towards bacterial and self-antigens, promoting a rapid response to infection and clearing of apoptotic cells. Although B-1a cells are known to primarily originate from fetal tissues the mechanisms by which they arise has been a topic of debate for many years. Here we show that in the fetal liver (FL) versus bone marrow (BM) environment, reduced IL-7R/STAT5 levels promote immunoglobulin kappa (Igk) recombination at the early pro-B cell stage. As a result, B cells can directly generate a mature B cell receptor (BCR) and bypass the requirement for a pre-BCR and pairing with surrogate light chain (SLC). This ‘alternate pathway’ of development enables the production of B cells with self reactive, skewed specificity receptors that are peculiar to the B-1a compartment. Together our findings connect seemingly opposing models of B-1a cell development and explain how these cells acquire their unique properties.


2020 ◽  
Author(s):  
Inesa Thomsen ◽  
Natalia Kunowska ◽  
Roshni de Souza ◽  
Anne-Marie Moody ◽  
Greg Crawford ◽  
...  

ABSTRACTRUNX1 is a transcription factor that plays key roles in haematopoietic development and in adult haematopoiesis and lymphopoiesis. Here we report that RUNX1 is also involved in controlling the dynamics of cell cycle entry of naïve resting B cells in response to stimulation of the B cell receptor (BCR). Conditional knockout of Runx1 in mouse resting B cells resulted in accelerated entry of the cells into S-phase following BCR engagement. Our results indicate that Runx1 regulates the cyclin D2 (Ccnd2) gene, the immediate early genes, Fosl2, Atf3 and Egr2, and the Notch effector Rbpj, in B cells, reducing the rate at which transcription of these genes increases following BCR stimulation. RUNX1 interacts with the chromatin remodeller SRCAP, recruiting it to promoter and enhancer regions of the Ccnd2 gene. BCR-mediated activation triggers switching between binding of RUNX1 and its paralog RUNX3 and between SRCAP and the SWI/SNF remodelling complex member BRG1. We also find that RUNX1 regulates expression of a number of immunomodulatory genes in resting B cells. These include the interferon receptor subunit gene Ifnar1, which is upregulated in B cells from lupus patients, the Ptpn22 gene, which has been identified as a major lupus risk allele, and the Lrrk2 gene, which is mutated in familial Parkinson’s disease. The hyperresponsiveness of the Runx1 knockout B cells to antigen stimulation and its role in regulating a suite of genes that are known to be associated with autoimmune disease suggest that RUNX1 is a major regulator of B cell tolerance and autoimmunity.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4702-4702
Author(s):  
Lina A. Gugliotti ◽  
Kiran B. Sakhuja ◽  
Hongsheng Wang ◽  
Julia Pinkhasov ◽  
Paul E. Love ◽  
...  

Abstract Abstract 4702 The development of B lymphocytes and the process of lineage determination are initiated by expression of a set of transcriptional regulators leading to V(D)J recombination events initiated by double-strand DNA breaks. Subsequently, these recombinations form DNAs that permit transcription of immunoglobulin genes and translation of the corresponding mRNAs, first by joining the V(D)J DNA sequences, then by recombination, that generates various isotypes of immunoglobulins by class-switch recombination (CSR). Formation of R-loops, regions containing RNA/DNA hybrid and a displaced single-stranded DNA, have been shown to lead to recombination in bacteria, yeast, HeLa and chick cells. Expression in each of these cases of excess ribonuclease H1 (RNase H1), a class of enzymes that degrade RNA in RNA/DNA hybrids, has ameliorated the deleterious effects and decreased recombinational events associated with R-loop formation. R-loops have been observed following transcription of the switch regions that occurs during CSR. The possibility that R-loops are important in V(D)J recombination has not been addressed, and whether ribonucleases H (RNases H) play a role in this process is still uncertain. Transgenic (TG) mice that overexpress RNase H1 in B and T cells (M27F7) were employed in this study. FACS analysis of hematopoietic cells from TG mice revealed a decrease in pre-B cells in the bone marrow. The data indicate a block at the pro-B to pre-B stage of B cell development, which may be the result of apoptosis due to the failure to generate a productive VH-D-JH rearrangement and expression of the pre-B cell receptor. A few B cells that successfully passed these checkpoints predominately differentiated into marginal zone and B1a cells in the peripheral lymphoid organs of the TG mice. These data suggest that R-loops are important in H chain gene rearrangement. This research is supported by the Intramural Research Program of the National Institutes of Health, the Eunice Kennedy Shriver National Institute of Child Health and Human Development and the National Institute of Allergy and Infectious Diseases. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 913-913
Author(s):  
Baohua Sun ◽  
Saradhi Mallampati ◽  
Yun Gong ◽  
Donghai Wang ◽  
Véronique Lefebvre ◽  
...  

Abstract Abstract 913 The development of mature B cells from self-renewing pluripotent hematopoietic stem cells (HSCs) involves a complex process that is regulated in a hierarchical order by various proteins, particularly transcription factors. Sox4 is an SRY-related HMG-box-containing transcription factor and is known to be implicated in B cell development. However, Sox4's role in B cell development has not been systematically investigated. In this project, we used conditional knockout mouse strains to study the effect of Sox4 deletion on B lymphopoiesis in adult mice. To determine when in the B cell developmental process Sox4 is required, we crossed Sox4-floxed mice with 5 Cre-recombinase-expressing mouse strains that were expected to result in deletion of the floxed Sox4 gene at different developmental stages, as follows: Vav-Cre (HSC stage in early embryos), Mx1-Cre (HSC stage in adults, induced by pIpC injection), MB1-Cre (early progenitor B cell stage), CD19-Cre (pro-B cell stage), and CD21-Cre (mature B cell stage). We demonstrated that deletion of Sox4 caused arrest of B lymphopoiesis at the transition from pre-pro-B cell stage (fraction A) to pro-B cell stage (fraction B); fraction B and later-stage B cells were nearly absent. There was a significant reduction in the number of hematopoietic stem (LSK) cells upon Sox4 deletion, but the number of common lymphocyte progenitors (CLPs) was not significantly changed and that of fraction A cells was only slightly reduced, suggesting that reduction in HSCs was not directly responsible for the B cell deficiency. Deletion of Sox4 in the early embryonic stage (Vav-Cre) and in adults (Mx1-Cre) resulted in similar phenotypes with regard to B lymphopoiesis, except that the number of peritoneal B1 cells was reduced more significantly in Vav-Cre than in Mx1-Cre mice. MB1-Cre yielded results similar to those of Vav-Cre, but B lymphopoiesis arrest was not as dramatic. Sox4f/f CD19-Cre and Sox4f/f CD21-Cre mice produced no significant difference in B cell phenotype compared with Sox4f/+ CD19-Cre and Sox4f/+ CD21-Cre mice, respectively. These data suggest that Sox4 is required for early B cell development at the transition from pre-pro-B cells to pro-B cells and is not required for mature B cell development. To further understand the mechanism by which Sox4 deletion induced pro-B cell deficiency, we performed semi-quantitative RT-PCR on residual pro-B cells and showed that Sox4 deletion did not change the mRNA expression of the well-known genes involved in early B cell development (EBF1, E2A, Rag1, CXCR4, IL7R, EZH2, PAX5, FoxP1, STAT5), indicating that Sox4 might be functioning in early B lymphopoiesis through a novel mechanism. We next studied pro-B cell viability in Sox4f/+ and Sox4f/f Vav-Cre mice. We found that, when Sox4 was depleted, bone marrow pro-B cells, but not pre-pro-B cells, underwent remarkable apoptosis, as shown by increased annexin V staining. In accordance, the level of the antiapoptotic protein Bcl2 in Sox4-deleted pro-B cells was much lower than that in control pro-B cells. Introduction of Bcl2 transgenic gene into the Sox4 conditional knockout mice decreased pro-B cell apoptosis and partially rescued the Sox4-deficient B cell development. We also observed striking molecular communication between Sox4 and the c-Kit-mediated cell survival signaling pathway; after intraperitoneal injection of the c-Kit inhibitor imatinib, the apoptotic rate was much higher in Sox4f/+Vav-Cre mice than in Sox4+/+ mice, suggesting that reduction of Sox4 level sensitized the cells to the inhibitor. Our data suggest that Sox4 deletion markedly decreases pro-B cell viability by modulating Bcl2 and that the c-Kit signaling pathway and Sox4 function is interlinked. We are currently investigating Sox4 transcription program and the mechanism of Sox4-induced Bcl2 protein reduction in progenitor B cells. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 1 (S1) ◽  
pp. 10-10
Author(s):  
Nina Ran ◽  
Christoph Ellebrecht ◽  
Eun Jung Choi ◽  
Aimee Payne

OBJECTIVES/SPECIFIC AIMS: Pemphigus vulgaris (PV) is a potentially fatal blistering disease caused by autoantibodies to the keratinocyte adhesion protein desmoglein 3. Several other autoimmune diseases have defective B cell tolerance checkpoints, resulting in the accumulation of self-reactive and polyreactive B cells. METHODS/STUDY POPULATION: The present work aims to determine whether PV patients develop normal tolerance to self-antigens other than desmoglein 3, as a potential “first hit” in the development of autoimmunity. We use FACS to isolate single B cells at 4 developmental stages from 8 PV patients. We perform single-cell RT-PCR to amplify each B cell receptor, produce monoclonal antibodies, and screen these for autoreactivity using ELISA/IF to several self-antigens. At each B cell stage, we compare the frequencies of self-reactive and polyreactive B cells to those found in healthy controls. RESULTS/ANTICIPATED RESULTS: We anticipate similar frequencies between PV patients and controls, suggesting that the B cell repertoire in PV patients develops normally at early checkpoints. DISCUSSION/SIGNIFICANCE OF IMPACT: The absence of generalized reactivity would distinguish PV from other autoimmune diseases and would show that PV arises from a specific break in tolerance to a single self-antigen (desmoglein 3) during late B cell maturation. Such a result would further support PV as an ideal candidate for targeted immunotherapy.


Sign in / Sign up

Export Citation Format

Share Document