Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Adv Sci (Weinh) ; 10(28): e2206692, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37587835

ABSTRACT

Regulatory T (Treg) cells are inevitable to prevent deleterious immune responses to self and commensal microorganisms. Treg function requires continuous expression of the transcription factor (TF) FOXP3 and is divided into two major subsets: resting (rTregs) and activated (aTregs). Continuous T cell receptor (TCR) signaling plays a vital role in the differentiation of aTregs from their resting state, and in their immune homeostasis. The process by which Tregs differentiate, adapt tissue specificity, and maintain stable phenotypic expression at the transcriptional level is still inconclusivei. In this work, the role of BATF is investigated, which is induced in response to TCR stimulation in naïve T cells and during aTreg differentiation. Mice lacking BATF in Tregs developed multiorgan autoimmune pathology. As a transcriptional regulator, BATF is required for Treg differentiation, homeostasis, and stabilization of FOXP3 expression in different lymphoid and non-lymphoid tissues. Epigenetically, BATF showed direct regulation of Treg-specific genes involved in differentiation, maturation, and tissue accumulation. Most importantly, FOXP3 expression and Treg stability require continuous BATF expression in Tregs, as it regulates demethylation and accessibility of the CNS2 region of the Foxp3 locus. Considering its role in Treg stability, BATF should be considered an important therapeutic target in autoimmune disease.


Subject(s)
Autoimmune Diseases , T-Lymphocytes, Regulatory , Mice , Animals , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Receptors, Antigen, T-Cell/metabolism
2.
J Exp Med ; 219(11)2022 11 07.
Article in English | MEDLINE | ID: mdl-36048018

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are crucial for the maintenance of host-microbiota homeostasis in gastrointestinal mucosal tissues. The mechanisms that maintain lineage identity of intestinal ILC3s and ILC3-mediated orchestration of microbiota and mucosal T cell immunity are elusive. Here, we identified BATF as a gatekeeper of ILC3 homeostasis in the gut. Depletion of BATF in ILC3s resulted in excessive interferon-γ production, dysbiosis, aberrant T cell immune responses, and spontaneous inflammatory bowel disease (IBD), which was considerably ameliorated by the removal of adaptive immunity, interferon-γ blockade, or antibiotic treatment. Mechanistically, BATF directly binds to the cis-regulatory elements of type 1 effector genes, restrains their chromatin accessibility, and inhibits their expression. Conversely, BATF promotes chromatin accessibility of genes involved in MHCII antigen processing and presentation pathways, which in turn directly promotes the transition of precursor ILC3s to MHCII+ ILC3s. Collectively, our findings reveal that BATF is a key transcription factor for maintaining ILC3 stability and coordinating ILC3-mediated control of intestinal homeostasis.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Immunity, Innate , Lymphocytes , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Chromatin/metabolism , Homeostasis , Interferon-gamma/metabolism , Intestinal Mucosa , Mice
3.
Sci Immunol ; 7(67): eabc9934, 2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35030033

ABSTRACT

Activated group 2 innate lymphoid cells (ILC2s) accumulate and promote inflammatory resolution and tissue repair in host defense against acute respiratory viral infections. However, the heterogeneity of ILC2s in the lung and the mechanisms by which ILC2 cells contribute to tissue repair remain elusive. Using single-cell RNA sequencing, we identify a transcriptionally distinct ILC2 subset that showed enrichment for wound healing signature genes and the transcription factor BATF. BATF promotes the proliferation and function of ILC2s and restricts their plasticity during infection with influenza virus. In the absence of BATF, ILC2s lose their immune protective properties and acquire pathogenic ILC3-like functions, leading to persistent neutrophil infiltration, tissue damage, and respiratory failure. Mechanistically, BATF directly binds to the cis-regulatory elements of wound healing genes, maintains their chromatin accessibility, and promotes their expression. Last, BATF plays an important role in an IL-33­ST2 feed-forward loop that supports ILC2 cell identity and function. Collectively, our findings shed light on a BATF-dependent ILC2 program, thereby providing a potential therapeutic target for terminating detrimental inflammation during acute viral infection.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Immunity, Innate/immunology , Influenza, Human/immunology , Lung/immunology , Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
4.
Nat Immunol ; 22(8): 996-1007, 2021 08.
Article in English | MEDLINE | ID: mdl-34282329

ABSTRACT

During chronic viral infection, CD8+ T cells develop into three major phenotypically and functionally distinct subsets: Ly108+TCF-1+ progenitors, Ly108-CX3CR1- terminally exhausted cells and the recently identified CX3CR1+ cytotoxic effector cells. Nevertheless, how CX3CR1+ effector cell differentiation is transcriptionally and epigenetically regulated remains elusive. Here, we identify distinct gene regulatory networks and epigenetic landscapes underpinning the formation of these subsets. Notably, our data demonstrate that CX3CR1+ effector cells bear a striking similarity to short-lived effector cells during acute infection. Genetic deletion of Tbx21 significantly diminished formation of the CX3CR1+ subset. Importantly, we further identify a previously unappreciated role for the transcription factor BATF in maintaining a permissive chromatin structure that allows the transition from TCF-1+ progenitors to CX3CR1+ effector cells. BATF directly bound to regulatory regions near Tbx21 and Klf2, modulating their enhancer accessibility to facilitate the transition. These mechanistic insights can potentially be harnessed to overcome T cell exhaustion during chronic infection and cancer.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , T-Box Domain Proteins/genetics , T-Lymphocyte Subsets/cytology , Animals , Antigens, Ly/metabolism , CX3C Chemokine Receptor 1/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Kruppel-Like Transcription Factors/genetics , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/immunology
5.
Eur J Immunol ; 48(9): 1492-1505, 2018 09.
Article in English | MEDLINE | ID: mdl-29898247

ABSTRACT

BATF functions in T cells and B cells to control the host response to antigen and promote the production of class switched immunoglobulins. In this study, we demonstrate that BATF expression increases rapidly, and transiently, following B cell stimulation and use an inducible murine model of BATF deletion to show that this induction is necessary, and sufficient, for immunoglobulin (Ig) class switch recombination (CSR). We examine two genes (Nfil3 and miR155gh) that are positively regulated, and one gene (Wnt10a) that is negatively regulated by BATF during CSR. These genes play essential roles in CSR and each impacts the expression and/or function of the others. Our observations allow these targets of BATF regulation to be positioned in a network upstream of the activation of germline transcripts (GLT) from the IgH locus and of transcriptional activation of Aicda - the gene encoding the enzyme directing Ig gene rearrangements. This work extends the knowledge of the molecular control of CSR and, importantly, positions the induction and function of BATF as an early event in this process.


Subject(s)
B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/biosynthesis , Basic-Leucine Zipper Transcription Factors/metabolism , Immunoglobulin Class Switching/genetics , Immunoglobulin Isotypes/genetics , MicroRNAs/biosynthesis , Nerve Tissue Proteins/biosynthesis , Wnt Proteins/biosynthesis , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Cytidine Deaminase/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Transcriptional Activation/genetics
6.
PLoS One ; 10(12): e0145724, 2015.
Article in English | MEDLINE | ID: mdl-26717480

ABSTRACT

Acinar cells of the exocrine pancreas are tasked with synthesizing, packaging and secreting vast quantities of pro-digestive enzymes to maintain proper metabolic homeostasis for the organism. Because the synthesis of high levels of hydrolases is potentially dangerous, the pancreas is prone to acute pancreatitis (AP), a disease that targets acinar cells, leading to acinar-ductal metaplasia (ADM), inflammation and fibrosis-events that can transition into the earliest stages of pancreatic ductal adenocarcinoma. Despite a wealth of information concerning the broad phenotype associated with pancreatitis, little is understood regarding specific transcriptional regulatory networks that are susceptible to AP and the role these networks play in acinar cell and exocrine pancreas responses. In this study, we examined the importance of the acinar-specific maturation transcription factor MIST1 to AP damage and organ recovery. Analysis of wild-type and Mist1 conditional null mice revealed that Mist1 gene transcription and protein accumulation were dramatically reduced as acinar cells underwent ADM alterations during AP episodes. To test if loss of MIST1 function was primarily responsible for the damaged status of the organ, mice harboring a Cre-inducible Mist1 transgene (iMist1) were utilized to determine if sustained MIST1 activity could alleviate AP damage responses. Unexpectedly, constitutive iMist1 expression during AP led to a dramatic increase in organ damage followed by acinar cell death. We conclude that the transient silencing of Mist1 expression is critical for acinar cells to survive an AP episode, providing cells an opportunity to suppress their secretory function and regenerate damaged cells. The importance of MIST1 to these events suggests that modulating key pancreas transcription networks could ease clinical symptoms in patients diagnosed with pancreatitis and pancreatic cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation , Gene Silencing , Pancreatitis/genetics , Acinar Cells/metabolism , Acute Disease , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Mice , Models, Biological , Pancreatitis/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Transcription, Genetic
7.
Immunity ; 43(1): 107-19, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26141583

ABSTRACT

Distinct groups of innate lymphoid cells (ILCs) such as ILC1, ILC2, and ILC3 populate the intestine, but how these ILCs develop tissue tropism for this organ is unclear. We report that prior to migration to the intestine ILCs first undergo a "switch" in their expression of homing receptors from lymphoid to gut homing receptors. This process is regulated by mucosal dendritic cells and the gut-specific tissue factor retinoic acid (RA). This change in homing receptors is required for long-term population and effector function of ILCs in the intestine. Only ILC1 and ILC3, but not ILC2, undergo the RA-dependent homing receptor switch in gut-associated lymphoid tissues. In contrast, ILC2 acquire gut homing receptors in a largely RA-independent manner during their development in the bone marrow and can migrate directly to the intestine. Thus, distinct programs regulate the migration of ILC subsets to the intestine for regulation of innate immunity.


Subject(s)
Cell Movement/physiology , Intestinal Mucosa/immunology , Intestines/immunology , Lymphocyte Subsets/immunology , Tretinoin/metabolism , Animals , Cells, Cultured , Citrobacter rodentium/immunology , Dendritic Cells/immunology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Immunity, Innate , Intestines/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR/genetics , Receptors, CCR7/genetics
8.
J Clin Invest ; 123(11): 4641-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24216482

ABSTRACT

T helper 9 (Th9) cells are specialized for the production of IL-9, promote allergic inflammation in mice, and are associated with allergic disease in humans. It has not been determined whether Th9 cells express a characteristic transcriptional signature. In this study, we performed microarray analysis to identify genes enriched in Th9 cells compared with other Th subsets. This analysis defined a transcriptional regulatory network required for the expression of a subset of Th9-enriched genes. The activator protein 1 (AP1) family transcription factor BATF (B cell, activating transcription factor­like) was among the genes enriched in Th9 cells and was required for the expression of IL-9 and other Th9-associated genes in both human and mouse T cells. The expression of BATF was increased in Th9 cultures derived from atopic infants compared with Th9 cultures from control infants. T cells deficient in BATF expression had a diminished capacity to promote allergic inflammation compared with wild-type controls. Moreover, mouse Th9 cells ectopically expressing BATF were more efficient at promoting allergic inflammation than control transduced cells. These data indicate that BATF is a central regulator of the Th9 phenotype and contributes to the development of allergic inflammation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Interleukin-9/biosynthesis , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/classification , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Humans , Hypersensitivity/etiology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity, Immediate/genetics , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/metabolism , Infant , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interleukin-9/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Transcription, Genetic
9.
Immunity ; 39(5): 833-45, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24211184

ABSTRACT

Upon infection, CD8(+) T cells undergo a stepwise process of early activation, expansion, and differentiation into effector cells. How these phases are transcriptionally regulated is incompletely defined. Here, we report that interferon regulatory factor 4 (IRF4), dispensable for early CD8(+) T cell activation, was vital for sustaining the expansion and effector differentiation of CD8(+) T cells. Mechanistically, IRF4 promoted the expression and function of Blimp1 and T-bet, two transcription factors required for CD8(+) T cell effector differentiation, and simultaneously repressed genes that mediate cell cycle arrest and apoptosis. Selective ablation of Irf4 in peripheral CD8(+) T cells impaired antiviral CD8(+) T cell responses, viral clearance, and CD8(+) T cell-mediated host recovery from influenza infection. IRF4 expression was regulated by T cell receptor (TCR) signaling strength via mammalian target of rapamycin (mTOR). Our data reveal that IRF4 translates differential strength of TCR signaling into different quantitative and qualitative CD8(+) T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Interferon Regulatory Factors/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins/antagonists & inhibitors , Bcl-2-Like Protein 11 , Cell Differentiation , Cells, Cultured/cytology , Coculture Techniques , Cyclin-Dependent Kinase Inhibitor Proteins/antagonists & inhibitors , Dendritic Cells/immunology , Gene Expression Regulation , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Lymphocyte Activation , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Specific Pathogen-Free Organisms , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , TOR Serine-Threonine Kinases/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Viral Plaque Assay
10.
J Exp Med ; 210(3): 475-89, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23460729

ABSTRACT

CCR9 and α4ß7 are the major trafficking receptors for lymphocyte migration to the gut, and their expression is induced during lymphocyte activation under the influence of retinoic acid (RA). We report here that BATF (basic leucine zipper transcription factor, ATF-like), an AP-1 protein family factor, is required for optimal expression of CCR9 and α4ß7 by T helper cells. BATF-deficient (knockout [KO]) mice had reduced numbers of effector T and regulatory T cells in the intestine. The intestinal T cells in BATF KO mice expressed CCR9 and α4ß7 at abnormally low levels compared with their wild-type (WT) counterparts, and BATF KO CD4(+) T cells failed to up-regulate the expression of CCR9 and α4ß7 to WT levels in response to RA. Defective binding of RARα and histone acetylation at the regulatory regions of the CCR9 and Itg-α4 genes were observed in BATF KO T cells. As a result, BATF KO effector and FoxP3(+) T cells failed to populate the intestine, and neither population functioned normally in the induction and regulation of colitis. Our results establish BATF as a cellular factor required for normal expression of CCR9 and α4ß7 and for the homeostasis and effector functions of T cell populations in the intestine.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Intestines/immunology , Receptors, Lymphocyte Homing/analysis , T-Lymphocytes, Helper-Inducer/immunology , Tretinoin/pharmacology , Animals , Cell Movement , Cells, Cultured , Forkhead Transcription Factors/analysis , Immune Tolerance , Integrins/analysis , Integrins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR/analysis , Receptors, CCR/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , T-Lymphocytes, Helper-Inducer/drug effects
11.
BMC Immunol ; 14: 16, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23537103

ABSTRACT

BACKGROUND: BATF plays important roles in the function of the immune system. Batf null mice are deficient in both CD4+ Th17 cells and T follicular helper cells and possess an intrinsic B cell defect that leads to the complete absence of class switched Ig. In this study, Tg mice overexpressing BATF in T cells were used together with Batf null mice to investigate how altering levels of BATF expression in T cells impacts the development and function of a recently characterized population of iNKT cells expressing IL-17 (iNKT-17). RESULTS: BATF has a direct impact on IL-17 expression by iNKT cells. However, in contrast to the Th17 lineage where BATF activates IL-17 expression and leads to the expansion of the lineage, BATF overexpression restricts overall iNKT cell numbers while skewing the compartment in vivo and in vitro toward an iNKT-17 phenotype. CONCLUSIONS: This work is the first to demonstrate that BATF joins RORγt as the molecular signature for all IL-17 producing cells in vivo and identifies BATF as a component of the nuclear protein network that could be targeted to regulate IL-17-mediated disease. Interestingly, these studies also reveal that while the Il17a gene is a common target for BATF regulation in Th17 and iNKT-17 cells, this regulation is accompanied by opposite effects on the growth and expansion of these two cell lineages.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Interleukin-17/biosynthesis , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Animals , Cell Differentiation/immunology , Hybridomas/immunology , Lymphocyte Count , Mice
12.
Nat Immunol ; 13(10): 981-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22842344

ABSTRACT

The mechanisms that regulate the T(H)9 subset of helper T cells and diseases mediated by T(H)9 cells remain poorly defined. Here we found that the costimulatory receptor OX40 was a powerful inducer of T(H)9 cells in vitro and T(H)9 cell-dependent airway inflammation in vivo. In polarizing conditions based on transforming growth factor-ß (TGF-ß), ligation of OX40 inhibited the production of induced regulatory T cells and the T(H)17 subset of helper T cells and diverted CD4(+)Foxp3(-) T cells to a T(H)9 phenotype. Mechanistically, OX40 activated the ubiquitin ligase TRAF6, which triggered induction of the kinase NIK in CD4(+) T cells and the noncanonical transcription factor NF-κB pathway; this subsequently led to the generation of T(H)9 cells. Thus, our study identifies a previously unknown mechanism for the induction of T(H)9 cells and may have important clinical implications in allergic inflammation.


Subject(s)
OX40 Ligand/metabolism , Receptors, OX40/metabolism , Respiratory System/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4 Antigens/biosynthesis , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-9/biosynthesis , Interleukin-9/metabolism , Mice , NF-kappa B/metabolism , OX40 Ligand/immunology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Receptors, OX40/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/metabolism , TNF Receptor-Associated Factor 6/biosynthesis , TNF Receptor-Associated Factor 6/metabolism , Trans-Activators/immunology , Trans-Activators/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , NF-kappaB-Inducing Kinase
13.
Mol Cancer Res ; 9(3): 350-63, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21296860

ABSTRACT

Batf is a basic leucine zipper transcription factor belonging to the activator protein-1 superfamily. Batf expression is regulated following stimulation of both lymphoid and myeloid cells. When treated with leukemia inhibitory factor, mouse M1 myeloid leukemia cells commit to a macrophage differentiation program that is dependent on Stat3 and involves the induction of Batf gene transcription via the binding of Stat3 to the Batf promoter. RNA interference was employed to block Batf induction in this system and the cells failed to growth arrest or to terminally differentiate. Restoring Batf expression not only reversed the differentiation-defective phenotype but also caused the cells to display signs of spontaneous differentiation in the absence of stimulation. Efforts to define genetic targets of the Batf transcription factor in M1 cells led to the identification of c-myb, a proto-oncogene known to promote blood cell proliferation and to inhibit the differentiation of M1 cells. These results provide strong evidence that Batf mediates the differentiation-inducing effects of Stat3 signaling in M1 cells and suggest that Batf may play a similar role in other blood cell lineages where alterations to the Jak-Stat pathway are hallmarks of disrupted development and disease.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Leukemia, Myeloid/genetics , Myeloid Cells/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Gene Knockdown Techniques , Genes, myb/genetics , Growth Inhibitors/genetics , Interleukin-6/metabolism , Leukemia Inhibitory Factor/metabolism , Leukemia, Myeloid/metabolism , Mice , Proto-Oncogene Mas , RNA Interference , Signal Transduction , Tumor Cells, Cultured
14.
J Exp Med ; 207(5): 933-42, 2010 May 10.
Article in English | MEDLINE | ID: mdl-20421391

ABSTRACT

Batf belongs to the activator protein 1 superfamily of basic leucine zipper transcription factors that includes Fos, Jun, and Atf proteins. Batf is expressed in mouse T and B lymphocytes, although the importance of Batf to the function of these lineages has not been fully investigated. We generated mice (Batf(DeltaZ/DeltaZ)) in which Batf protein is not produced. Batf(DeltaZ/DeltaZ) mice contain normal numbers of B cells but show reduced numbers of peripheral CD4(+) T cells. Analysis of CD4(+) T helper (Th) cell subsets in Batf(DeltaZ/DeltaZ) mice demonstrated that Batf is required for the development of functional Th type 17 (Th17), Th2, and follicular Th (Tfh) cells. In response to antigen immunization, germinal centers were absent in Batf(DeltaZ/DeltaZ) mice and the maturation of Ig-secreting B cells was impaired. Although adoptive transfer experiments confirmed that this B cell phenotype can be driven by defects in the Batf(DeltaZ/DeltaZ) CD4(+) T cell compartment, stimulation of Batf(DeltaZ/DeltaZ) B cells in vitro, or by a T cell-independent antigen in vivo, resulted in proliferation but not class-switch recombination. We conclude that loss of Batf disrupts multiple components of the lymphocyte communication network that are required for a robust immune response.


Subject(s)
B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation , Basic-Leucine Zipper Transcription Factors/genetics , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , DNA Primers , Interleukins/genetics , Lymphocyte Count , Mice , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Sequence Deletion , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th2 Cells/immunology
15.
J Immunol ; 178(1): 58-66, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17182540

ABSTRACT

NKT cells are glycolipid-reactive lymphocytes that express markers and perform functions common to both T lymphocytes and NK cells. Although the genetic events controlling conventional T cell development are well defined, the transcription factors and genetic programs regulating NKT cell development are only beginning to be elucidated. Previously, we described the NKT cell-deficient phenotype of transgenic (Tg) mice constitutively expressing B cell-activating transcription factor (BATF), a basic leucine zipper protein and inhibitor of AP-1. In this study, we show that Tg BATF targets the majority of Valpha14Jalpha281 (Valpha14i(7)) NKT cells, regardless of CD4 expression and Vbeta gene usage. The residual NKT cells in the thymus of BATF-Tg mice are CD44(+), yet are slow to display the NK1.1 marker characteristic of mature cells. As a population, BATF-expressing NKT cells are TCRbeta/CD3epsilon(low), but express normal levels of CD69, suggesting a failure to expand appropriately following selection. Consistent with the sensitivity of NKT cells to BATF-induced changes in AP-1 activity, we detect a full complement of AP-1 basic leucine zipper proteins in wild-type NKT cells isolated from the thymus, spleen, and liver, and show that AP-1 DNA-binding activity and cytokine gene transcription are induced in NKT cells within a few hours of glycolipid Ag exposure. This study is the first to characterize AP-1 activity in NKT cells and implicates the integrity of this transcription factor complex in developmental events essential to the establishment of this unique T cell subset in the thymus.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/growth & development , Transcription Factor AP-1/physiology , Animals , Antigens, CD/analysis , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Differentiation, T-Lymphocyte/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Cytokines/genetics , Gene Expression Regulation, Developmental , Humans , Hyaluronan Receptors/analysis , Hyaluronan Receptors/metabolism , Killer Cells, Natural/chemistry , Lectins, C-Type , Liver/immunology , Mice , Mice, Transgenic , Receptor-CD3 Complex, Antigen, T-Cell/analysis , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Spleen/immunology , T-Lymphocyte Subsets/chemistry , Thymus Gland/immunology
16.
Biochemistry ; 45(32): 9857-65, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16893186

ABSTRACT

B-Myc is an endogenous, N-terminal homologue of transcription factor c-Myc that lacks the C-terminal DNA binding and protein dimerization domain of c-Myc. Clinical mutations in the c-Myc N-terminal region, and the subsequent misregulation of Myc, are implicated in the development of numerous human cancers. Myc functions to both activate and repress transcription by associating with multiple binding partners. We investigated the structural and dynamical properties of B-Myc, free or associated with the transactivation inhibitor, MM-1, and the activator, TBP, using NMR spectroscopy. B-Myc has no persistent tertiary structure, yet regions corresponding to Myc homology boxes 1 and 2 (MBI and MBII, respectively) have molten globule-like characteristics. B-Myc binds to MM-1 in a specific manner without becoming highly structured. The local regions of B-Myc involved in binding differ for MM-1 and TBP, and regions not identified by mutagenesis are found to be involved in MM-1 binding. The results provide new insights into Myc N-terminal protein-protein interactions. We propose a model for Myc regulation through differential involvement of MBI and MBII in the binding of Myc interacting proteins.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Amino Acid Sequence , Animals , Circular Dichroism , Humans , Mice , Models, Biological , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-myc/chemistry , Sequence Alignment
17.
J Immunoassay Immunochem ; 27(3): 207-12, 2006.
Article in English | MEDLINE | ID: mdl-16827223

ABSTRACT

The ability to correlate changes in antigen-reactive lymphocytes with disease will provide information needed to develop strategies for combating illness. One critical group of lymphocytes are the CD1-restricted T cells. It is desirable to use CD1 molecules in an array format to query CD1-restricted lymphocytes in humans. To investigate the feasibility of this technique, we employed mCD1d and alpha-galactosylceramide to demonstrate that-slide immobilized, CD1d-alpha-GalCer complexes capture an NKT cell hybridoma in the presence of a competitor. The success of this scheme represents the first step toward the development of CD1-antigen arrays that could be used to profile biological samples.


Subject(s)
Antigens, CD1/immunology , Immunoassay/methods , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD1d , Humans , Hybridomas/immunology , Mice
18.
Methods Enzymol ; 407: 322-35, 2006.
Article in English | MEDLINE | ID: mdl-16757335

ABSTRACT

Ras proteins function as signaling nodes that are activated by extracellular stimuli. On activation, Ras interacts with a spectrum of functionally diverse downstream effectors and stimulates a variety of downstream cytoplasmic signaling cascades that regulate cellular proliferation, differentiation, and apoptosis. In addition to the association of Ras with the plasma membrane, recent studies have established an association of Ras with Golgi membranes and showed that H-Ras and N-Ras are activated on endomembranes and signal to regulate downstream pathways. Whereas the effectors of signal transduction by activated, plasma membrane-localized Ras are well characterized, very little is known about the effectors used by Golgi-associated Ras. Recently, we have reported the identification of the first endomembrane Ras effector molecule, RAIN. This chapter details the methods used to study RAIN-Ras interaction and localization in vivo. In addition, we describe the tools and methods we have used to explore role of endogenous RAIN in endothelial cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , ras Proteins/physiology , Animals , COS Cells , Cattle , Cell Line , Chlorocebus aethiops , Endothelium, Vascular/cytology , Humans , Mice , NIH 3T3 Cells , Transfection
19.
Mol Biol Evol ; 23(8): 1480-92, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16731568

ABSTRACT

Dimeric basic leucine zipper (bZIP) factors constitute one of the most important classes of enhancer-type transcription factors. In vertebrates, bZIP factors are involved in many cellular processes, including cell survival, learning and memory, cancer progression, lipid metabolism, and a variety of developmental processes. These factors have the ability to homodimerize and heterodimerize in a specific and predictable manner, resulting in hundreds of dimers with unique effects on transcription. In recent years, several studies have described dimerization preferences for bZIP factors from different species, including Homo sapiens, Drosophila melanogaster, Arabidopsis thaliana, and Saccharomyces cerevisiae. Here, these findings are summarized as novel, graphical representations of closed, interacting protein networks. These representations combine phylogenetic information, DNA-binding properties, and dimerization preference. Beyond summarizing bZIP dimerization preferences within selected species, we have included annotation for a solitary bZIP factor found in the primitive eukaryote, Giardia lamblia, a possible evolutionary precursor to the complex networks of bZIP factors encoded by other genomes. Finally, we discuss the fundamental similarities and differences between dimerization networks within the context of bZIP factor evolution.


Subject(s)
Arabidopsis/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Drosophila melanogaster/genetics , Evolution, Molecular , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/chemistry , Dimerization , Giardia lamblia/genetics , Humans , Molecular Sequence Data , Phylogeny , Plant Proteins/chemistry , Plant Proteins/genetics
20.
Biochim Biophys Acta ; 1759(1-2): 4-12, 2006.
Article in English | MEDLINE | ID: mdl-16580748

ABSTRACT

Over the last 15 years, numerous studies have addressed the structural rules that regulate dimerization stability and dimerization specificity of the leucine zipper, a dimeric parallel coiled-coil domain that can either homodimerize or heterodimerize. Initially, these studies were performed with a limited set of B-ZIP proteins, sequence-specific DNA binding proteins that dimerize using the leucine zipper domain to bind DNA. A global analysis of B-ZIP leucine zipper dimerization properties can be rationalized using a limited number of structural rules [J.R. Newman, A.E. Keating, Comprehensive identification of human bZIP interactions with coiled-coil arrays, Science 300 (2003) 2097-2101]. Today, however, access to the genomic sequences of many different organisms has made possible the annotation of all B-ZIP proteins from several species and has generated a bank of data that can be used to refine, and potentially expand, these rules. Already, a comparative analysis of the B-ZIP proteins from Arabidopsis thaliana and Homo sapiens has revealed that the same amino acids are used in different patterns to generate diverse B-ZIP dimerization patterns [C.D. Deppmann, A. Acharya, V. Rishi, B. Wobbes, S. Smeekens, E.J. Taparowsky, C. Vinson, Dimerization specificity of all 67 B-ZIP motifs in Arabidopsis thaliana: a comparison to Homo sapiens B-ZIP motifs, Nucleic Acids Res. 32 (2004) 3435-3445]. The challenge ahead is to investigate the biological significance of different B-ZIP protein-protein interactions. Gaining insight at this level will rely on ongoing investigations to (a) define the role of target DNA on modulating B-ZIP dimerization partners, (b) characterize the B-ZIP transcriptome in various cells and tissues through mRNA microarray analysis, (c) identify the genomic localization of B-ZIP binding at a genomic level using the chromatin immunoprecipitation assay, and (d) develop more sophisticated imaging technologies to visualize dimer dynamics in single cells and whole organisms. Studies of B-ZIP family leucine zipper dimerization and the regulatory mechanisms that control their biological activities could serve as a paradigm for deciphering the biophysical and biological parameters governing other well-characterized protein-protein interaction motifs. This review will focus on the dimerization specificity of coiled-coil proteins, particularly the human B-ZIP transcription family that consists of 53 proteins that use the leucine zipper coiled-coil as a dimerization motif.


Subject(s)
Nuclear Proteins/chemistry , RNA-Binding Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Motifs , Amino Acid Sequence , DNA/metabolism , Dimerization , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Thermodynamics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...