Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 581
Filter
1.
Nat Immunol ; 18(2): 173-183, 2017 02.
Article in English | MEDLINE | ID: mdl-27992401

ABSTRACT

Most Foxp3+ regulatory T (Treg) cells develop in the thymus as a functionally mature T cell subpopulation specialized for immune suppression. Their cell fate appears to be determined before Foxp3 expression; yet molecular events that prime Foxp3- Treg precursor cells are largely obscure. We found that Treg cell-specific super-enhancers (Treg-SEs), which were associated with Foxp3 and other Treg cell signature genes, began to be activated in Treg precursor cells. T cell-specific deficiency of the genome organizer Satb1 impaired Treg-SE activation and the subsequent expression of Treg signature genes, causing severe autoimmunity due to Treg cell deficiency. These results suggest that Satb1-dependent Treg-SE activation is crucial for Treg cell lineage specification in the thymus and that its perturbation is causative of autoimmune and other immunological diseases.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes, Regulatory/physiology , Transcriptional Activation/immunology , Animals , Autoimmunity , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Immune Tolerance , Male , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Precursor Cells, T-Lymphoid/physiology
2.
Genes Dev ; 34(3-4): 149-165, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31919189

ABSTRACT

Differentiating neutrophils undergo large-scale changes in nuclear morphology. How such alterations in structure are established and modulated upon exposure to microbial agents is largely unknown. Here, we found that prior to encounter with bacteria, an armamentarium of inflammatory genes was positioned in a transcriptionally passive environment suppressing premature transcriptional activation. Upon microbial exposure, however, human neutrophils rapidly (<3 h) repositioned the ensemble of proinflammatory genes toward the transcriptionally permissive compartment. We show that the repositioning of genes was closely associated with the swift recruitment of cohesin across the inflammatory enhancer landscape, permitting an immediate transcriptional response upon bacterial exposure. We found that activated enhancers, marked by increased deposition of H3K27Ac, were highly enriched for cistromic elements associated with PU.1, CEBPB, TFE3, JUN, and FOSL2 occupancy. These data reveal how upon microbial challenge the cohesin machinery is recruited to an activated enhancer repertoire to instruct changes in chromatin folding, nuclear architecture, and to activate an inflammatory gene program.


Subject(s)
Cell Nucleus/immunology , Chromatin/immunology , Escherichia coli Infections/immunology , Neutrophils/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Cells, Cultured , Escherichia coli , Histones/metabolism , Humans
3.
Nat Immunol ; 16(7): 737-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26006015

ABSTRACT

Aire is a transcriptional regulator that induces the promiscuous expression of thousands of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs), a step critical for the induction of immunological self-tolerance. Studies have offered molecular insights into how Aire operates, but more comprehensive understanding of this process still remains elusive. Here we found abundant expression of the protein deacetylase Sirtuin-1 (Sirt1) in mature Aire(+) mTECs, wherein it was required for the expression of Aire-dependent TRA-encoding genes and the subsequent induction of immunological self-tolerance. Our study elucidates a previously unknown molecular mechanism for Aire-mediated transcriptional regulation and identifies a unique function for Sirt1 in preventing organ-specific autoimmunity.


Subject(s)
Central Tolerance/immunology , Sirtuin 1/immunology , Transcription Factors/immunology , Transcriptional Activation/immunology , Acetylation , Animals , Antigens/immunology , Central Tolerance/genetics , Epithelial Cells/immunology , Epithelial Cells/metabolism , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Organ Specificity/immunology , Protein Binding/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/immunology , AIRE Protein
4.
Nat Immunol ; 15(4): 365-372, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24584088

ABSTRACT

T lymphocytes responding to microbial infection give rise to effector cells that mediate acute host defense and memory cells that provide long-lived immunity, but the fundamental question of when and how these cells arise remains unresolved. Here we combined single-cell gene-expression analyses with 'machine-learning' approaches to trace the transcriptional 'roadmap' of individual CD8(+) T lymphocytes throughout the course of an immune response in vivo. Gene-expression signatures predictive of eventual fates could be discerned as early as the first T lymphocyte division and may have been influenced by asymmetric partitioning of the receptor for interleukin 2 (IL-2Rα) during mitosis. Our findings emphasize the importance of single-cell analyses in understanding fate determination and provide new insights into the specification of divergent lymphocyte fates early during an immune response to microbial infection.


Subject(s)
Adaptive Immunity , CD8-Positive T-Lymphocytes/immunology , Gene Expression Profiling/methods , Infections/immunology , Infections/microbiology , Receptors, Interleukin-2/metabolism , Single-Cell Analysis/methods , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cell Lineage/genetics , Computer Simulation , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitosis/genetics , Mitosis/immunology , Ovalbumin/genetics , Ovalbumin/immunology , Receptors, Interleukin-2/genetics , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/virology , Transcriptional Activation/immunology
5.
Nat Immunol ; 14(1): 27-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23179078

ABSTRACT

The E3 ligase ARIH2 has an unusual structure and mechanism of elongating ubiquitin chains. To understand its physiological role, we generated gene-targeted mice deficient in ARIH2. ARIH2 deficiency resulted in the embryonic death of C57BL/6 mice. On a mixed genetic background, the lethality was attenuated, with some mice surviving beyond weaning and then succumbing to an aggressive multiorgan inflammatory response. We found that in dendritic cells (DCs), ARIH2 caused degradation of the inhibitor IκBß in the nucleus, which abrogated its ability to sequester, protect and transcriptionally coactivate the transcription factor subunit p65 in the nucleus. Loss of ARIH2 caused dysregulated activation of the transcription factor NF-κB in DCs, which led to lethal activation of the immune system in ARIH2-sufficent mice reconstituted with ARIH2-deficient hematopoietic stem cells. Our data have therapeutic implications for targeting ARIH2 function.


Subject(s)
Dendritic Cells/immunology , Embryonic Development/immunology , Multiple Organ Failure/immunology , Ubiquitin-Protein Ligases/physiology , Animals , Cells, Cultured , Disease Models, Animal , Embryonic Development/genetics , Hematopoiesis/genetics , Humans , Immune System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Multiple Organ Failure/genetics , NF-kappa B/metabolism , Transcriptional Activation/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/immunology
6.
Nat Immunol ; 14(5): 514-22, 2013 May.
Article in English | MEDLINE | ID: mdl-23563688

ABSTRACT

Here we identified B cells as a major source of rapid, innate-like production of interleukin 17 (IL-17) in vivo in response to infection with Trypanosoma cruzi. IL-17(+) B cells had a plasmablast phenotype, outnumbered cells of the TH17 subset of helper T cells and were required for an optimal response to this pathogen. With both mouse and human primary B cells, we found that exposure to parasite-derived trans-sialidase in vitro was sufficient to trigger modification of the cell-surface mucin CD45, which led to signaling dependent on the kinase Btk and production of IL-17A or IL-17F via a transcriptional program independent of the transcription factors RORγt and Ahr. Our combined data suggest that the generation of IL-17(+) B cells may be a previously unappreciated feature of innate immune responses required for pathogen control or IL-17-mediated autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Chagas Disease/immunology , Glycoproteins/metabolism , Interleukin-17/immunology , Neuraminidase/metabolism , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/immunology , Animals , B-Lymphocytes/parasitology , Cell Proliferation , Cells, Cultured , Chagas Disease/genetics , Glycoproteins/genetics , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuraminidase/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/parasitology , Th17 Cells/immunology , Th17 Cells/parasitology , Transcriptional Activation/immunology
7.
Nat Immunol ; 14(12): 1237-46, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24185615

ABSTRACT

Induction of type I interferon is a central event of innate immunity, essential for host defense. Here we report that the transcription factor ELF4 is induced by type I interferon and upregulates interferon expression in a feed-forward loop. ELF4 deficiency leads to reduced interferon production, resulting in enhanced susceptibility to West Nile virus encephalitis in mice. After viral infection, ELF4 is recruited by STING, interacts with and is activated by the MAVS-TBK1 complex, and translocates into the nucleus to bind interferon promoters. Cooperative binding with ELF4 increases the binding affinity of interferon regulatory factors IRF3 and IRF7, which is mediated by EICE elements. Thus, in addition to identifying a regulator of innate immune signaling, we uncovered a role for EICE elements in interferon transactivation.


Subject(s)
DNA-Binding Proteins/immunology , Interferon-beta/immunology , Transcription Factors/immunology , West Nile Fever/immunology , West Nile virus/immunology , Animals , Cell Line , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Immunoblotting , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Protein Binding/immunology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/immunology , West Nile Fever/virology , West Nile virus/physiology
8.
Immunity ; 44(4): 913-923, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27096320

ABSTRACT

The participation of a specific subset of B cells and how they are regulated in cancer is unclear. Here, we demonstrate that the proportion of CD5(+) relative to interleukin-6 receptor α (IL-6Rα)-expressing B cells was greatly increased in tumors. CD5(+) B cells responded to IL-6 in the absence of IL-6Rα. IL-6 directly bound to CD5, leading to activation of the transcription factor STAT3 via gp130 and its downstream kinase JAK2. STAT3 upregulated CD5 expression, thereby forming a feed-forward loop in the B cells. In mouse tumor models, CD5(+) but not CD5(-) B cells promoted tumor growth. CD5(+) B cells also showed activation of STAT3 in multiple types of human tumor tissues. Thus, our findings demonstrate a critical role of CD5(+) B cells in promoting cancer.


Subject(s)
B-Lymphocytes/immunology , CD5 Antigens/metabolism , Interleukin-6/metabolism , Melanoma, Experimental/pathology , STAT3 Transcription Factor/immunology , Animals , CD5 Antigens/biosynthesis , Cell Line, Tumor , Cytokine Receptor gp130/metabolism , Humans , Interleukin-6/immunology , Janus Kinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Protein Binding , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , Transcriptional Activation/immunology
9.
Proc Natl Acad Sci U S A ; 119(36): e2206327119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037380

ABSTRACT

Cerebral malaria (CM) is a life-threatening form of Plasmodium falciparum infection caused by brain inflammation. Brain endothelium dysfunction is a hallmark of CM pathology, which is also associated with the activation of the type I interferon (IFN) inflammatory pathway. The molecular triggers and sensors eliciting brain type I IFN cellular responses during CM remain largely unknown. We herein identified the stimulator of interferon response cGAMP interactor 1 (STING1) as the key innate immune sensor that induces Ifnß1 transcription in the brain of mice infected with Plasmodium berghei ANKA (Pba). This STING1/IFNß-mediated response increases brain CXCL10 governing the extent of brain leukocyte infiltration and blood-brain barrier (BBB) breakdown, and determining CM lethality. The critical role of brain endothelial cells (BECs) in fueling type I IFN-driven brain inflammation was demonstrated in brain endothelial-specific IFNß-reporter and STING1-deficient Pba-infected mice, which were significantly protected from CM lethality. Moreover, extracellular particles (EPs) released from Pba-infected erythrocytes activated the STING1-dependent type I IFN response in BECs, a response requiring intracellular acidification. Fractionation of the EPs enabled us to identify a defined fraction carrying hemoglobin degradation remnants that activates STING1/IFNß in the brain endothelium, a process correlated with heme content. Notably, stimulation of STING1-deficient BECs with heme, docking experiments, and in vitro binding assays unveiled that heme is a putative STING1 ligand. This work shows that heme resultant from the parasite heterotrophic activity operates as an alarmin, triggering brain endothelial inflammatory responses via the STING1/IFNß/CXCL10 axis crucial to CM pathogenesis and lethality.


Subject(s)
Brain , Heme , Interferon-beta , Malaria, Cerebral , Membrane Proteins , Animals , Brain/parasitology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/parasitology , Endothelium/immunology , Endothelium/parasitology , Heme/metabolism , Interferon-beta/immunology , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Plasmodium berghei/metabolism , Transcriptional Activation/immunology
10.
J Immunol ; 208(2): 247-256, 2022 01 15.
Article in English | MEDLINE | ID: mdl-35017214

ABSTRACT

IFNs are comprised of three families of cytokines that confer protection against pathogen infection and uncontrolled cellular proliferation. The broad role IFNs play in innate and adaptive immune regulation has placed them under heavy scrutiny to position them as "friend" or "foe" across pathologies. Genetic lesions in genes involving IFN synthesis and signaling underscore the disparate outcomes of aberrant IFN signaling. Abrogation of the response leads to susceptibility to microbial infections whereas unabated IFN induction underlies a variety of inflammatory diseases and tumor immune evasion. Type I and III IFNs have overlapping roles in antiviral protection, yet the mechanisms by which they are induced and promote the expression of IFN-stimulated genes and inflammation can distinguish their biological functions. In this review, we examine the molecular factors that shape the shared and distinct roles of type I and III IFNs in immunity.


Subject(s)
Interferon Type I/immunology , Interferons/immunology , Virus Diseases/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Inflammation/immunology , Interferon Type I/metabolism , Interferon Type I/therapeutic use , Interferons/metabolism , Interferons/therapeutic use , Signal Transduction/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Interferon Lambda
11.
Nat Immunol ; 12(8): 709-14, 2011 Jun 19.
Article in English | MEDLINE | ID: mdl-21772279

ABSTRACT

Activation of NF-κB transcription factors by receptors of the innate or adaptive immune system is essential for host defense. However, after danger is eliminated, NF-κB signaling needs to be tightly downregulated for the maintenance of tissue homeostasis. This review highlights key negative regulatory principles that affect the amount, localization or conformational properties of NF-κB-activating proteins to attenuate the NF-κB response. These mechanisms are needed to prevent inflammation, autoimmune disease and oncogenesis.


Subject(s)
NF-kappa B/immunology , Animals , Down-Regulation/immunology , Feedback , Homeostasis/immunology , Humans , NF-kappa B/antagonists & inhibitors , Signal Transduction/immunology , Transcription, Genetic/immunology , Transcriptional Activation/immunology
12.
Nat Immunol ; 12(2): 160-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21186367

ABSTRACT

During immunoglobulin class-switch recombination (CSR), the cytidine deaminase AID induces double-strand breaks into transcribed, repetitive DNA elements called switch sequences. The mechanism that promotes the binding of AID specifically to switch regions remains to be elucidated. Here we used a proteomic screen with in vivo biotinylation of AID to identify the splicing regulator PTBP2 as a protein that interacts with AID. Knockdown of PTBP2 mediated by short hairpin RNA in B cells led to a decrease in binding of AID to transcribed switch regions, which resulted in considerable impairment of CSR. PTBP2 is thus an effector of CSR that promotes the binding of AID to switch-region DNA.


Subject(s)
Cytidine Deaminase/metabolism , DNA/metabolism , Nerve Tissue Proteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , DNA/genetics , Immunoglobulin Class Switching/genetics , Immunoglobulin Switch Region/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/immunology , Protein Binding/genetics , RNA, Small Interfering/genetics , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Transgenes/genetics
13.
Nat Immunol ; 12(2): 121-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21245903

ABSTRACT

The last 10 years have witnessed the identification of a new class of intracellular pattern-recognition molecules--the nucleotide-binding domain and leucine-rich repeat-containing family (NLR). Members of this family garnered interest as pattern-recognition receptors able to trigger inflammatory responses against pathogens. Many studies support a pathogen-recognition function for human NLR proteins and shed light on their role in the broader control of adaptive immunity and various disease states. Other evidence suggests that NLRs function in processes unrelated to pathogen detection. Here we discuss recent advances in our understanding of the biology of the human NLR proteins and their non-pathogen-recognition function in tissue homeostasis, apoptosis, graft-versus-host disease and early development.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Carrier Proteins/immunology , Graft vs Host Disease/immunology , Th1 Cells/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins , Autoimmune Diseases/genetics , Carrier Proteins/genetics , Embryonic Development/immunology , Genetic Predisposition to Disease , Humans , Lymphocyte Activation , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Ovulation/immunology , Polymorphism, Genetic , Signal Transduction/immunology , Transcriptional Activation/immunology , Transplantation Immunology
14.
Nat Immunol ; 12(2): 129-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21186366

ABSTRACT

The nuclear adaptor Ldb1 functions as a core component of multiprotein transcription complexes that regulate differentiation in diverse cell types. In the hematopoietic lineage, Ldb1 forms a complex with the non-DNA-binding adaptor Lmo2 and the transcription factors E2A, Scl and GATA-1 (or GATA-2). Here we demonstrate a critical and continuous requirement for Ldb1 in the maintenance of both fetal and adult mouse hematopoietic stem cells (HSCs). Deletion of Ldb1 in hematopoietic progenitors resulted in the downregulation of many transcripts required for HSC maintenance. Genome-wide profiling by chromatin immunoprecipitation followed by sequencing (ChIP-Seq) identified Ldb1 complex-binding sites at highly conserved regions in the promoters of genes involved in HSC maintenance. Our results identify a central role for Ldb1 in regulating the transcriptional program responsible for the maintenance of HSCs.


Subject(s)
Adult Stem Cells/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Fetal Development , Hematopoietic Stem Cells/metabolism , Adoptive Transfer , Adult Stem Cells/cytology , Adult Stem Cells/immunology , Adult Stem Cells/transplantation , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/transplantation , Female , Fetal Development/genetics , Fetal Development/immunology , Gene Expression Regulation, Developmental/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , High-Throughput Nucleotide Sequencing , LIM Domain Proteins , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy , Protein Binding , Regulatory Elements, Transcriptional/genetics , Regulatory Elements, Transcriptional/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
15.
Nat Immunol ; 12(11): 1119-26, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964606

ABSTRACT

Binding of antigen to the B cell antigen receptor (BCR) triggers both BCR signaling and endocytosis. How endocytosis regulates BCR signaling remains unknown. Here we report that BCR signaling was not extinguished by endocytosis of BCRs; instead, BCR signaling initiated at the plasma membrane continued as the BCR trafficked intracellularly with the sequential phosphorylation of kinases. Blocking the endocytosis of BCRs resulted in the recruitment of both proximal and downstream kinases to the plasma membrane, where mitogen-activated protein kinases (MAPKs) were hyperphosphorylated and the kinase Akt and its downstream target Foxo were hypophosphorylated, which led to the dysregulation of gene transcription controlled by these pathways. Thus, the cellular location of the BCR serves to compartmentalize kinase activation to regulate the outcome of signaling.


Subject(s)
B-Lymphocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Compartmentation/immunology , Cells, Cultured , Endocytosis/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Phosphorylation/immunology , Protein Transport/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Transcriptional Activation/immunology
16.
Immunity ; 40(6): 896-909, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24882217

ABSTRACT

Animal host defense against infection requires the expression of defense genes at the right place and the right time. Understanding such tight control of host defense requires the elucidation of the transcription factors involved. By using an unbiased approach in the model Caenorhabditis elegans, we discovered that HLH-30 (known as TFEB in mammals) is a key transcription factor for host defense. HLH-30 was activated shortly after Staphylococcus aureus infection, and drove the expression of close to 80% of the host response, including antimicrobial and autophagy genes that were essential for host tolerance of infection. TFEB was also rapidly activated in murine macrophages upon S. aureus infection and was required for proper transcriptional induction of several proinflammatory cytokines and chemokines. Thus, our data suggest that TFEB is a previously unappreciated, evolutionarily ancient transcription factor in the host response to infection.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Staphylococcal Infections/immunology , Animals , Autophagy/genetics , Autophagy/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans Proteins/genetics , Enterococcus faecalis/immunology , Immunity, Innate , Macrophages/immunology , Mice , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , RNA Interference , RNA, Small Interfering , Salmonella Infections/immunology , Salmonella enterica/immunology , Signal Transduction/immunology , Staphylococcus aureus/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
17.
Proc Natl Acad Sci U S A ; 117(47): 29823-29831, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33168738

ABSTRACT

Nuclear factor-ĸB (NF-ĸB) transcription factor is a family of essential regulators of the immune response and cell proliferation and transformation. A typical factor is a heterodimer made of either p50 or p52, which are limited processing products of either p105 or p100, respectively, and a member of the Rel family of proteins, typically p65. The transcriptional program of NF-ĸB is tightly regulated by the composition of the dimers. In our previous work, we demonstrated that the ubiquitin ligase KPC1 is involved in ubiquitination and proteasomal processing of p105 to generate p50. Its overexpression and the resulting high level of p50 stimulates transcription of a broad array of tumor suppressors. Here we demonstrate that additional mechanisms are involved in the p50-mediated tumor-suppressive effect. p50 down-regulates expression of a major immune checkpoint inhibitor, the programmed cell death-ligand 1 (PD-L1), both in cells and in tumors. Importantly, the suppression is abrogated by overexpression of p65. This highlights the importance of the cellular quantities of the two different subunits of NF-ĸB which determine the composition of the dimer. While the putative p50 homodimer is tumor-suppressive, the "canonical" p50p65 heterodimer is oncogenic. We found that an additional mechanism is involved in the tumor-suppressive phenomenon: p50 up-regulates expression of the proinflammatory chemokines CCL3, CCL4, and CCL5, which in turn recruit into the tumors active natural killer (NK) cells and macrophages. Overall, p50 acts as a strong tumor suppressor via multiple mechanisms, including overexpression of tumor suppressors and modulation of the tumor microenvironment by recruiting active immune cells.


Subject(s)
B7-H1 Antigen/metabolism , Gene Expression Regulation, Neoplastic/immunology , NF-kappa B p50 Subunit/metabolism , Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Adoptive Transfer , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Chemokines/immunology , Chemokines/metabolism , HEK293 Cells , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Macrophages/immunology , Macrophages/metabolism , Mice , Neoplasms/immunology , Neoplasms/pathology , Primary Cell Culture , Transcription Factor RelA/metabolism , Transcriptional Activation/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Ubiquitination/genetics , Ubiquitination/immunology , Up-Regulation , Xenograft Model Antitumor Assays
19.
Nat Immunol ; 11(5): 442-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20383150

ABSTRACT

The CD4 versus CD8 lineage specification of thymocytes is linked to coreceptor expression. The transcription factor MAZR has been identified as an important regulator of Cd8 expression. Here we show that variegated CD8 expression by loss of Cd8 enhancers was reverted in MAZR-deficient mice, which confirms that MAZR negatively regulates the Cd8 loci during the transition to the double-positive (DP) stage. Moreover, loss of MAZR led to partial redirection of major histocompatibility complex (MHC) class I-restricted thymocytes into CD4(+) helper-like T cells, which correlated with derepression of Th-POK, a central transcription factor for helper-lineage development. MAZR bound the silencer of the gene encoding Th-POK, which indicated direct regulation of this locus by MAZR. Thus, MAZR is part of the transcription factor network that regulates the CD8 lineage differentiation of DP thymocytes.


Subject(s)
CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Lineage , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Animals , Bone Marrow Transplantation , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cell Transdifferentiation/genetics , Cell Transdifferentiation/immunology , Cells, Cultured , Gene Regulatory Networks , H-2 Antigens/genetics , H-2 Antigens/metabolism , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Protein Binding/genetics , Protein Binding/immunology , Radiation Chimera , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Repressor Proteins/genetics , Repressor Proteins/immunology , Silencer Elements, Transcriptional/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transcriptional Activation/immunology
20.
Nat Immunol ; 11(5): 419-26, 2010 May.
Article in English | MEDLINE | ID: mdl-20364151

ABSTRACT

Pattern-recognition receptors (PRRs) elicit antiviral immune responses to human immunodeficiency virus type 1 (HIV-1). Here we show that HIV-1 required signaling by the PRRs Toll-like receptor 8 (TLR8) and DC-SIGN for replication in dendritic cells (DCs). HIV-1 activated the transcription factor NF-kappaB through TLR8 to initiate the transcription of integrated provirus by RNA polymerase II (RNAPII). However, DC-SIGN signaling was required for the generation of full-length viral transcripts. Binding of the HIV-1 envelope glycoprotein gp120 to DC-SIGN induced kinase Raf-1-dependent phosphorylation of the NF-kappaB subunit p65 at Ser276, which recruited the transcription-elongation factor pTEF-b to nascent transcripts. Transcription elongation and generation of full-length viral transcripts was dependent on pTEF-b-mediated phosphorylation of RNAPII at Ser2. Inhibition of either pathway abrogated replication and prevented HIV-1 transmission. Thus, HIV-1 subverts crucial components of the immune system for replication that might be targeted to prevent infection and dissemination.


Subject(s)
Dendritic Cells/metabolism , HIV Infections/immunology , HIV-1/physiology , Immunity, Innate , Toll-Like Receptor 8/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV-1/pathogenicity , Humans , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding/genetics , Protein Engineering , Proto-Oncogene Proteins c-raf/metabolism , RNA Polymerase II/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Second Messenger Systems/genetics , Second Messenger Systems/immunology , Sequence Deletion/genetics , Toll-Like Receptor 8/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL