Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Nat Immunol ; 15(6): 562-70, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24777531

ABSTRACT

Deubiquitinases (DUBs) are a new class of drug targets, although the physiological function of only few DUBs has been characterized. Here we identified the DUB USP15 as a crucial negative regulator of T cell activation. USP15 stabilized the E3 ubiquitin ligase MDM2, which in turn negatively regulated T cell activation by targeting the degradation of the transcription factor NFATc2. USP15 deficiency promoted T cell activation in vitro and enhanced T cell responses to bacterial infection and tumor challenge in vivo. USP15 also stabilized MDM2 in cancer cells and regulated p53 function and cancer-cell survival. Our results suggest that inhibition of USP15 may both induce tumor cell apoptosis and boost antitumor T cell responses.


Subject(s)
NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-mdm2/immunology , Th1 Cells/immunology , Ubiquitin-Specific Proteases/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Survival , HCT116 Cells , Humans , Leupeptins/pharmacology , Listeria monocytogenes/immunology , Listeriosis/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Escape , Tumor Suppressor Protein p53/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics , Ubiquitination/immunology
2.
Nat Immunol ; 12(12): 1221-9, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22057289

ABSTRACT

During infection, naive CD8(+) T cells differentiate into effector cells, which are armed to eliminate pathogens, and memory cells, which are poised to protect against reinfection. The transcriptional program that regulates terminal differentiation into short-lived effector-memory versus long-lived memory cells is not clearly defined. Through the use of mice expressing reporters for the DNA-binding inhibitors Id2 and Id3, we identified Id3(hi) precursors of long-lived memory cells before the peak of T cell population expansion or upregulation of cell-surface receptors that indicate memory potential. Deficiency in Id2 or Id3 resulted in loss of distinct CD8(+) effector and memory populations, which demonstrated unique roles for these inhibitors of E-protein transcription factors. Furthermore, cytokines altered the expression of Id2 and Id3 differently, which provides insight into how external cues influence gene expression.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation , Immunologic Memory/immunology , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/metabolism , T-Lymphocyte Subsets/immunology , Transcription, Genetic , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Infections/genetics , Infections/immunology , Infections/microbiology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Proteins/genetics , Lectins, C-Type , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets/cytology , Transcription, Genetic/drug effects
3.
Immunity ; 40(3): 342-54, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24656046

ABSTRACT

Production of type I interferons (IFN-I) is a crucial innate immune mechanism against viral infections. IFN-I induction is subject to negative regulation by both viral and cellular factors, but the underlying mechanism remains unclear. We report that the noncanonical NF-κB pathway was stimulated along with innate immune cell differentiation and viral infections and had a vital role in negatively regulating IFN-I induction. Genetic deficiencies in major components of the noncanonical NF-κB pathway caused IFN-I hyperinduction and rendered cells and mice substantially more resistant to viral infection. Noncanonical NF-κB suppressed signal-induced histone modifications at the Ifnb promoter, an action that involved attenuated recruitment of the transcription factor RelA and a histone demethylase, JMJD2A. These findings reveal an unexpected function of the noncanonical NF-κB pathway and highlight an important mechanism regulating antiviral innate immunity.


Subject(s)
Immunity, Innate , Interferon Type I/biosynthesis , NF-kappa B/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme Activation , Female , Gene Expression Regulation/drug effects , Hematopoietic Cell Growth Factors/pharmacology , Histone Demethylases/metabolism , Histones/metabolism , Immunity, Innate/drug effects , Interferon-beta/genetics , Interferon-beta/metabolism , Ligands , Mice , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptors/metabolism , Transcription Factor RelA/metabolism , Virus Diseases/genetics , NF-kappaB-Inducing Kinase
4.
Proc Natl Acad Sci U S A ; 115(10): E2311-E2319, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463696

ABSTRACT

Blood cell formation must be appropriately maintained throughout life to provide robust immune function, hemostasis, and oxygen delivery to tissues, and to prevent disorders that result from over- or underproduction of critical lineages. Persistent inflammation deregulates hematopoiesis by damaging hematopoietic stem and progenitor cells (HSPCs), leading to elevated myeloid cell output and eventual bone marrow failure. Nonetheless, antiinflammatory mechanisms that protect the hematopoietic system are understudied. The transcriptional regulator STAT3 has myriad roles in HSPC-derived populations and nonhematopoietic tissues, including a potent antiinflammatory function in differentiated myeloid cells. STAT3 antiinflammatory activity is facilitated by STAT3-mediated transcriptional repression of Ube2n, which encodes the E2 ubiquitin-conjugating enzyme Ubc13 involved in proinflammatory signaling. Here we demonstrate a crucial role for STAT3 antiinflammatory activity in preservation of HSPCs and lineage-balanced hematopoiesis. Conditional Stat3 removal from the hematopoietic system led to depletion of the bone marrow lineage- Sca-1+ c-Kit+ CD150+ CD48- HSPC subset (LSK CD150+ CD48- cells), myeloid-skewed hematopoiesis, and accrual of DNA damage in HSPCs. These responses were accompanied by intrinsic transcriptional alterations in HSPCs, including deregulation of inflammatory, survival and developmental pathways. Concomitant Ube2n/Ubc13 deletion from Stat3-deficient hematopoietic cells enabled lineage-balanced hematopoiesis, mitigated depletion of bone marrow LSK CD150+ CD48- cells, alleviated HSPC DNA damage, and corrected a majority of aberrant transcriptional responses. These results indicate an intrinsic protective role for STAT3 in the hematopoietic system, and suggest that this is mediated by STAT3-dependent restraint of excessive proinflammatory signaling via Ubc13 modulation.


Subject(s)
Blood Cells/immunology , Hematopoiesis , STAT3 Transcription Factor/immunology , Animals , Blood Cells/cytology , Cell Lineage , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/immunology , STAT3 Transcription Factor/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/immunology
5.
Gastroenterology ; 157(1): 163-178, 2019 07.
Article in English | MEDLINE | ID: mdl-30885780

ABSTRACT

BACKGROUND & AIMS: The peroxisome proliferator-activated receptor delta (PPARD) regulates cell metabolism, proliferation, and inflammation and has been associated with gastric and other cancers. Villin-positive epithelial cells are a small population of quiescent gastric progenitor cells. We expressed PPARD from a villin promoter to investigate the role of these cells and PPARD in development of gastric cancer. METHODS: We analyzed gastric tissues from mice that express the Ppard (PPARD1 and PPARD2 mice) from a villin promoter, and mice that did not carry this transgene (controls), by histology and immunohistochemistry. We performed cell lineage-tracing experiments and analyzed the microbiomes, chemokine and cytokine production, and immune cells and transcriptomes of stomachs of these mice. We also performed immunohistochemical analysis of PPARD levels in 2 sets of human gastric tissue microarrays. RESULTS: Thirty-eight percent of PPARD mice developed spontaneous, invasive gastric adenocarcinomas, with severe chronic inflammation. Levels of PPARD were increased in human gastric cancer tissues, compared with nontumor tissues, and associated with gastric cancer stage and grade. We found an inverse correlation between level of PPARD in tumor tissue and patient survival time. Gastric microbiomes from PPARD and control mice did not differ significantly. Lineage-tracing experiments identified villin-expressing gastric progenitor cells (VGPCs) as the origin of gastric tumors in PPARD mice. In these mice, PPARD up-regulated CCL20 and CXCL1, which increased infiltration of the gastric mucosa by immune cells. Immune cell production of inflammatory cytokines promoted chronic gastric inflammation and expansion and transformation of VGPCs, leading to tumorigenesis. We identified a positive-feedback loop between PPARD and interferon gamma signaling that sustained gastric inflammation to induce VGPC transformation and gastric carcinogenesis. CONCLUSIONS: We found PPARD overexpression in VPGCs to result in inflammation, dysplasia, and tumor formation. PPARD and VGPCs might be therapeutic targets for stomach cancer.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Cytokines/immunology , Gastric Mucosa/metabolism , Interferon-gamma/immunology , Receptors, Cytoplasmic and Nuclear/genetics , Stem Cells/metabolism , Stomach/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Carcinogenesis/immunology , Cell Lineage , Cell Transformation, Neoplastic/immunology , Chemokine CCL20/metabolism , Chemokine CXCL1/metabolism , Chemokines , Feedback, Physiological , Gene Expression Profiling , Inflammation , Mice , Microbiota/immunology , Microfilament Proteins/genetics , Stem Cells/immunology , Stomach/microbiology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology
6.
Immunol Rev ; 261(1): 84-101, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25123278

ABSTRACT

The term innate immunity typically refers to a quick but non-specific host defense response against invading pathogens. The innate immune system comprises particular immune cell populations, epithelial barriers, and numerous secretory mediators including cytokines, chemokines, and defense peptides. Innate immune cells are also now recognized to play important contributing roles in cancer and pathological inflammatory conditions. Innate immunity relies on rapid signal transduction elicited upon pathogen recognition via pattern recognition receptors (PRRs) and cell:cell communication conducted by soluble mediators, including cytokines. A majority of cytokines involved in innate immune signaling use a molecular cascade encompassing receptor-associated Jak protein tyrosine kinases and STAT (signal transducer and activator of transcription) transcriptional regulators. Here, we focus on roles for STAT proteins in three major innate immune subsets: neutrophils, macrophages, and dendritic cells (DCs). While knowledge in this area is only now emerging, understanding the molecular regulation of these cell types is necessary for developing new approaches to treat human disorders such as inflammatory conditions, autoimmunity, and cancer.


Subject(s)
Dendritic Cells/immunology , Immune System Diseases/immunology , Macrophages/immunology , Neoplasms/immunology , Neutrophils/immunology , STAT Transcription Factors/metabolism , Animals , Cell Communication/immunology , Cytokines/metabolism , Humans , Immunity, Innate , Janus Kinases/metabolism , Receptors, Pattern Recognition/metabolism , STAT Transcription Factors/genetics , Signal Transduction , Transcriptional Activation
7.
FASEB J ; 29(6): 2359-70, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25713055

ABSTRACT

The IL-6/signal transducer and activator of transcription 3 (STAT3) pathway is a critical signaling pathway for colitis-associated colorectal cancer (CAC). Peroxisome proliferator-activated receptor (PPAR)-δ, a lipid nuclear receptor, up-regulates IL-6. 15-Lipoxygenase-1 (15-LOX-1), which is crucial to production of lipid signaling mediators to terminate inflammation, down-regulates PPAR-δ. 15-LOX-1 effects on IL-6/STAT3 signaling and CAC tumorigenesis have not been determined. We report that intestinally targeted transgenic 15-LOX-1 expression in mice inhibited azoxymethane- and dextran sodium sulfate-induced CAC, IL-6 expression, STAT3 phosphorylation, and IL-6/STAT3 downstream target (Notch3 and MUC1) expression. 15-LOX-1 down-regulation was associated with IL-6 up-regulation in human colon cancer mucosa. Reexpression of 15-LOX-1 in human colon cancer cells suppressed IL-6 mRNA expression, STAT3 phosphorylation, IL-6 promoter activity, and PPAR-δ mRNA and protein expression. PPAR-δ overexpression in colonic epithelial cells promoted CAC tumorigenesis in mice and increased IL-6 expression and STAT3 phosphorylation, whereas concomitant 15-LOX-1 expression in colonic epithelial cells (15-LOX-1-PPAR-δ-Gut mice) suppressed these effects: the number of tumors per mouse (mean ± sem) was 4.22 ± 0.68 in wild-type littermates, 6.67 ± 0.83 in PPAR-δ-Gut mice (P = 0.026), and 2.25 ± 0.25 in 15-LOX-1-PPAR-δ-Gut mice (P = 0.0006). Identification of 15-LOX-1 suppression of PPAR-δ to inhibit IL-6/STAT3 signaling-driven CAC tumorigenesis provides mechanistic insights that can be used to molecularly target CAC.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Colitis/metabolism , Colonic Neoplasms/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Animals , Arachidonate 15-Lipoxygenase/genetics , Azoxymethane , Blotting, Western , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/genetics , Colitis/chemically induced , Colitis/genetics , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Dextran Sulfate , Gene Expression , HCT116 Cells , Humans , Immunohistochemistry , Interleukin-6/genetics , Mice, Transgenic , PPAR delta/genetics , PPAR delta/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
8.
Eur J Haematol ; 97(3): 261-70, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26660446

ABSTRACT

Heterozygous mutations in the transcriptional regulator GATA-2 associate with multilineage immunodeficiency, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML). The majority of these mutations localize in the zinc finger (ZnF) domains, which mediate GATA-2 DNA binding. Deregulated hematopoiesis with GATA-2 mutation frequently develops in adulthood, yet GATA-2 function in the bone marrow remains unresolved. To investigate this, we conditionally deleted the GATA-2 C-terminal ZnF (C-ZnF) coding sequences in adult mice. Upon Gata2 C-ZnF deletion, we observed rapid peripheral cytopenia, bone marrow failure, and decreased c-Kit expression on hematopoietic progenitors. Transplant studies indicated GATA-2 has a cell-autonomous role in bone marrow hematopoiesis. Moreover, myeloid lineage populations were particularly sensitive to Gata2 hemizygosity, while molecular assays indicated GATA-2 regulates c-Kit expression in multilineage progenitor cells. Enforced c-Kit expression in Gata2 C-ZnF-deficient hematopoietic progenitors enhanced myeloid colony activity, suggesting GATA-2 sustains myelopoiesis via a cell intrinsic role involving maintenance of c-Kit expression. Our results provide insight into mechanisms regulating hematopoiesis in bone marrow and may contribute to a better understanding of immunodeficiency and bone marrow failure associated with GATA-2 mutation.


Subject(s)
Anemia, Aplastic/genetics , Bone Marrow Diseases/genetics , Bone Marrow/pathology , GATA2 Transcription Factor/genetics , Hemoglobinuria, Paroxysmal/genetics , Protein Interaction Domains and Motifs/genetics , Proto-Oncogene Proteins c-kit/deficiency , Sequence Deletion , Zinc Fingers/genetics , Anemia, Aplastic/diagnosis , Anemia, Aplastic/metabolism , Anemia, Aplastic/mortality , Animals , Biomarkers , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Diseases/diagnosis , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/mortality , Bone Marrow Failure Disorders , Bone and Bones/pathology , Chromatin Immunoprecipitation , Decalcification, Pathologic/genetics , Disease Models, Animal , GATA2 Transcription Factor/chemistry , GATA2 Transcription Factor/metabolism , Gene Expression , Gene Expression Regulation , Gene Frequency , Genes, Reporter , Genotype , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hemoglobinuria, Paroxysmal/diagnosis , Hemoglobinuria, Paroxysmal/metabolism , Hemoglobinuria, Paroxysmal/mortality , High-Throughput Nucleotide Sequencing , Immunophenotyping , Mice , Mice, Knockout , Prognosis , Side-Population Cells
9.
Blood ; 120(22): 4363-73, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23033267

ABSTRACT

Cytokines and transcription factors play key roles in dendritic cell (DC) development, yet information about regulatory interactions between these signals remains limited. Here we show that the cytokines GM-CSF and Flt3L induce the transcriptional mediators Id2 and E2-2 and control DC lineage diversification by STAT-dependent pathways. We found that STAT5 is required for tissue CD103(+) DC generation and plasmacytoid DC (pDC) suppression in steady state or response to GM-CSF. STAT5 stimulates GM-CSF-dependent expression of Id2, which controls CD103(+) DC production and pDC inhibition. By contrast, pDCs, but not CD103(+) DCs, are dependent on STAT3. Consistently, STAT3 stimulates Flt3L-responsive expression of the pDC regulator Tcf4 (E2-2). These data suggest that STATs contribute to DC development by controlling transcription factors involved in lineage differentiation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Dendritic Cells/physiology , Inhibitor of Differentiation Protein 2/genetics , STAT3 Transcription Factor/physiology , STAT5 Transcription Factor/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Inhibitor of Differentiation Protein 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Transcription Factor 4
10.
J Biol Chem ; 287(14): 11234-9, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22318729

ABSTRACT

Recent work has identified a new subset of CD4(+) T cells named as Tfh cells that are localized in germinal centers and critical in germinal center formation. Tfh cell differentiation is regulated by IL-6 and IL-21, possibly via STAT3 factor, and B cell lymphoma 6 (Bcl6) is specifically expressed in Tfh cells and required for their lineage specification. In the current study, we characterized the role of STAT5 in Tfh cell development. We found that a constitutively active form of STAT5 effectively inhibited Tfh differentiation by suppressing the expression of Tfh-associated factors (CXC motif) receptor 5 (CXCR5), musculoaponeurotic fibrosarcoma (c-Maf), Bcl6, basic leucine zipper transcription factor ATF-like (Batf), and IL-21, and STAT5 deficiency greatly enhanced Tfh gene expression. Importantly, STAT5 regulated the expression of Tfh cell suppressor factor B lymphocyte-induced maturation protein 1 (Blimp-1); STAT5 deficiency impaired Blimp-1 expression and resulted in elevated expression of Tfh-specific genes. Similarly, inhibition of IL-2 potentiated Tfh generation, associated with dampened Blimp-1 expression; Blimp-1 overexpression inhibited Tfh gene expression in Stat5-deficient T cells, suggesting that the IL-2/STAT5 axis functions to regulate Blimp-1 expression. In vivo, deletion of STAT5 in CD4(+) T cells resulted in enhanced development of Tfh cells and germinal center B cells and led to an impairment of B cell tolerance in a well defined mouse tolerance model. Taken together, this study demonstrates that STAT5 controls Tfh differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , STAT5 Transcription Factor/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Mice , Positive Regulatory Domain I-Binding Factor 1 , STAT5 Transcription Factor/deficiency , Transcription Factors/genetics , Up-Regulation
11.
Blood ; 118(14): 3879-89, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21828128

ABSTRACT

Plasmacytoid dendritic cells (pDCs) reside in bone marrrow and lymphoid organs in homeostatic conditions and typically secrete abundant quantities of type I interferons (IFNs) on Toll-like receptor triggering. Recently, a pDC population was identified within Peyer patches (PPs) of the gut that is distinguished by its lack of IFN production; however, the relationship of PP pDCs to pDCs in other organs has been unclear. We report that PP pDCs are derived from common DC progenitors and accumulate in response to Fms-like tyrosine kinase 3 ligand, yet appear divergent in transcription factor profile and surface marker phenotype, including reduced E2-2 and CCR9 expression. Type I IFN signaling via STAT1 has a cell-autonomous role in accrual of PP pDCs in vivo. Moreover, IFN-α enhances pDC generation from DC progenitors by a STAT1-dependent mechanism. pDCs that have been developed in the presence of IFN-α resemble PP pDCs, produce inflammatory cytokines, stimulate Th17 cell generation, and fail to secrete IFN-α on Toll-like receptor engagement. These results indicate that IFN-α influences the development and function of pDCs by inducing emergence of an inflammatory (Th17-inducing) antigen-presenting subset, and simultaneously regulating accumulation of pDCs in the intestinal microenvironment.


Subject(s)
Dendritic Cells/immunology , Interferon-alpha/immunology , Peyer's Patches/cytology , STAT1 Transcription Factor/immunology , Animals , Cell Differentiation , Dendritic Cells/cytology , Mice , Mice, Inbred C57BL , Stem Cells/cytology , Stem Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , fms-Like Tyrosine Kinase 3/immunology
12.
J Exp Med ; 220(2)2023 02 06.
Article in English | MEDLINE | ID: mdl-36367776

ABSTRACT

Immune checkpoint blockade (ICB) has revolutionized cancer treatment, yet quality of life and continuation of therapy can be constrained by immune-related adverse events (irAEs). Limited understanding of irAE mechanisms hampers development of approaches to mitigate their damage. To address this, we examined whether mice gained sensitivity to anti-CTLA-4 (αCTLA-4)-mediated toxicity upon disruption of gut homeostatic immunity. We found αCTLA-4 drove increased inflammation and colonic tissue damage in mice with genetic predisposition to intestinal inflammation, acute gastrointestinal infection, transplantation with a dysbiotic fecal microbiome, or dextran sodium sulfate administration. We identified an immune signature of αCTLA-4-mediated irAEs, including colonic neutrophil accumulation and systemic interleukin-6 (IL-6) release. IL-6 blockade combined with antibiotic treatment reduced intestinal damage and improved αCTLA-4 therapeutic efficacy in inflammation-prone mice. Intestinal immune signatures were validated in biopsies from patients with ICB colitis. Our work provides new preclinical models of αCTLA-4 intestinal irAEs, mechanistic insights into irAE development, and potential approaches to enhance ICB efficacy while mitigating irAEs.


Subject(s)
Colitis , Interleukin-6 , Mice , Animals , Quality of Life , Colitis/pathology , Immunotherapy , Inflammation
13.
Blood ; 115(16): 3354-63, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20185584

ABSTRACT

Neutrophil mobilization, the release of neutrophils from the bone marrow reserve into circulating blood, is important to increase peripheral neutrophil amounts during bacterial infections. Granulocyte colony-stimulating factor (G-CSF) and chemokines, such as macrophage-inflammatory protein-2 (MIP-2; CXCL2), can induce neutrophil mobilization, but the mechanism(s) they use remain unclear. Signal transducers and activator of transcription 3 (STAT3) is the principal intracellular signaling molecule activated upon G-CSF ligation of its receptor. Using a murine model with conditional STAT3 deletion in bone marrow, we demonstrated previously that STAT3 regulates acute G-CSF-responsive neutrophil mobilization and MIP-2-dependent neutrophil chemotaxis. In this study, we show STAT3 is also necessary for MIP-2-elicited neutrophil mobilization. STAT3 appears to function by controlling extracellular signal-regulated kinase (ERK) activation, which is important for MIP-2-mediated chemotaxis. In addition, we demonstrate that G-CSF stimulates the expression of the MIP-2 receptor via STAT3-dependent transcriptional activation of Il8rb. G-CSF treatment also induces STAT3-dependent changes in bone marrow chemokine expression levels which may further affect neutrophil retention and release. Taken together, our study demonstrates that STAT3 regulates multiple aspects of chemokine and chemokine receptor expression and function within the bone marrow, indicating a central role in the neutrophil mobilization response.


Subject(s)
Chemokine CXCL2/metabolism , Chemotaxis, Leukocyte/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neutrophils/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Animals , Cell Separation , Chemokine CXCL2/immunology , Electrophoretic Mobility Shift Assay , Flow Cytometry , Gene Expression , Gene Expression Regulation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunoblotting , Immunoprecipitation , Mice , Mice, Transgenic , Neutrophils/immunology , Receptors, Interleukin-8 , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/immunology
14.
Blood ; 116(14): 2462-71, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20581311

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) mediates "emergency" granulopoiesis during infection, a process that is mimicked by clinical G-CSF use, yet we understand little about the intracellular signaling cascades that control demand-driven neutrophil production. Using a murine model with conditional deletion of signal transducer and activator of transcription 3 (STAT3) in bone marrow, we investigated the cellular and molecular mechanisms of STAT3 function in the emergency granulopoiesis response to G-CSF administration or infection with Listeria monocytogenes, a pathogen that is restrained by G-CSF signaling in vivo. Our results show that STAT3 deficiency renders hematopoietic progenitor cells and myeloid precursors refractory to the growth-promoting functions of G-CSF or L monocytogenes infection. STAT3 is necessary for accelerating granulocyte cell-cycle progression and maturation in response to G-CSF. STAT3 directly controls G-CSF-dependent expression of CCAAT-enhancer-binding protein ß (C/EBPß), a crucial factor in the emergency granulopoiesis response. Moreover, STAT3 and C/EBPß coregulate c-Myc through interactions with the c-myc promoter that control the duration of C/EBPα occupancy during demand-driven granulopoiesis. These results place STAT3 as an essential mediator of emergency granulopoiesis by its regulation of transcription factors that direct G-CSF-responsive myeloid progenitor expansion.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Leukopoiesis , STAT3 Transcription Factor/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Cell Cycle , Gene Expression Regulation , Granulocytes/metabolism , Granulocytes/microbiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Listeria monocytogenes/pathogenicity , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction
15.
Elife ; 102021 08 11.
Article in English | MEDLINE | ID: mdl-34378531

ABSTRACT

Maintenance of immune homeostasis involves a synergistic relationship between the host and the microbiome. Canonical interferon (IFN) signaling controls responses to acute microbial infection, through engagement of the STAT1 transcription factor. However, the contribution of tonic levels of IFN to immune homeostasis in the absence of acute infection remains largely unexplored. We report that STAT1 KO mice spontaneously developed an inflammatory disease marked by myeloid hyperplasia and splenic accumulation of hematopoietic stem cells. Moreover, these animals developed inflammatory bowel disease. Profiling gut bacteria revealed a profound dysbiosis in the absence of tonic IFN signaling, which triggered expansion of TH17 cells and loss of splenic Treg cells. Reduction of bacterial load by antibiotic treatment averted the TH17 bias and blocking IL17 signaling prevented myeloid expansion and splenic stem cell accumulation. Thus, tonic IFNs regulate gut microbial ecology, which is crucial for maintaining physiologic immune homeostasis and preventing inflammation.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome , Inflammation/genetics , Interferons/administration & dosage , Interleukin-17/genetics , STAT1 Transcription Factor/genetics , Animals , Female , Interleukin-17/metabolism , Mice , Mice, Knockout , STAT1 Transcription Factor/metabolism
16.
Cancer Immunol Res ; 8(6): 794-805, 2020 06.
Article in English | MEDLINE | ID: mdl-32213626

ABSTRACT

Clinical response rates after adoptive cell therapy (ACT) are highly correlated with in vivo persistence of the infused T cells. However, antigen-specific T cells found in tumor sites are often well-differentiated effector cells with limited persistence. Central memory CD8+ T cells, capable of self-renewal, represent desirable ACT products. We report here that exposure to a histone deacetylase inhibitor (HDACi) and IL21 could reprogram differentiated human CD8+ T cells into central memory-like T cells. Dedifferentiation of CD8+ T cells was initiated by increased H3 acetylation and chromatin accessibility at the CD28 promoter region. This led to IL21-mediated pSTAT3 binding to the CD28 region, and subsequent upregulation of surface CD28 and CD62L (markers of central memory T cells). The reprogrammed cells exhibited enhanced proliferation in response to both IL2 and IL15, and a stable memory-associated transcriptional signature (increased Lef1 and Tcf7). Our findings support the application of IL21 and HDACi for the in vitro generation of highly persistent T-cell populations that can augment the efficacy of adoptively transferred T cells.


Subject(s)
CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Histone Deacetylase Inhibitors/pharmacology , Immunologic Memory/immunology , Interleukins/metabolism , T-Lymphocytes/immunology , Adolescent , CD28 Antigens/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Humans , Immunologic Memory/drug effects , Immunotherapy, Adoptive , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
17.
J Immunother Cancer ; 8(1)2020 04.
Article in English | MEDLINE | ID: mdl-32273347

ABSTRACT

BACKGROUND: Type 1 conventional dendritic cells (cDC1s) possess efficient antigen presentation and cross-presentation activity, as well as potent T cell priming ability. Tissue-resident cDC1s (CD103+ cDC1s in mice, CD141+ cDC1s in humans) are linked with improved tumor control, yet the efficacy of immunotherapy using this population is understudied. METHODS: We generated murine CD103+ cDC1s in vitro and examined their expression of cDC1-related factors, antigen cross-presentation activity, and accumulation in tumor-draining lymph nodes (TdLNs). The antitumor efficacy of the in vitro-generated CD103+ cDC1s was studied in murine melanoma and osteosarcoma models. We evaluated tumor responses on vaccination with CD103+ cDC1s, compared these to vaccination with monocyte-derived DCs (MoDCs), tested CD103+ cDC1 vaccination with checkpoint blockade, and examined the antimetastatic activity of CD103+ cDC1s. RESULTS: In vitro-generated CD103+ cDC1s produced cDC1-associated factors such as interleukin-12p70 and CXCL10, and demonstrated antigen cross-presentation activity on stimulation with the toll-like receptor 3 agonist polyinosinic:polycytidylic acid (poly I:C). In vitro-generated CD103+ cDC1s also migrated to TdLNs following poly I:C treatment and intratumoral delivery. Vaccination with poly I:C-activated and tumor antigen-loaded CD103+ cDC1s enhanced tumor infiltration of tumor antigen-specific and interferon-γ+ CD8+ T cells, and suppressed melanoma and osteosarcoma growth. CD103+ cDC1s showed superior antitumor efficacy compared with MoDC vaccination, and led to complete regression of 100% of osteosarcoma tumors in combination with CTLA-4 antibody-mediated checkpoint blockade. In vitro-generated CD103+ cDC1s effectively protected mice from pulmonary melanoma and osteosarcoma metastases. CONCLUSIONS: Our data indicate an in vitro-generated CD103+ cDC1 vaccine elicits systemic and long-lasting tumor-specific T cell-mediated cytotoxicity, which restrains primary and metastatic tumor growth. The CD103+ cDC1 vaccine was superior to MoDCs and enhanced response to immune checkpoint blockade. These results indicate the potential for new immunotherapies based on use of cDC1s alone or in combination with checkpoint blockade.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Integrin alpha Chains/immunology , Lung Neoplasms/immunology , Melanoma, Experimental/immunology , Osteosarcoma/immunology , Sarcoma, Experimental/immunology , Vaccines/administration & dosage , Animals , Antigen Presentation/immunology , Antigens, CD/metabolism , Antigens, Neoplasm/immunology , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Cross-Priming , Dendritic Cells/transplantation , Immunotherapy , In Vitro Techniques , Integrin alpha Chains/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Osteosarcoma/pathology , Osteosarcoma/therapy , Sarcoma, Experimental/pathology , Sarcoma, Experimental/therapy , Tumor Cells, Cultured
18.
Cancers (Basel) ; 12(1)2020 Jan 04.
Article in English | MEDLINE | ID: mdl-31947933

ABSTRACT

Conventional dendritic cells (cDCs) are a critical immune population, composed of multiple subsets, and responsible for controlling adaptive immunity and tolerance. Although migratory type 1 cDCs (CD103+ cDC1s in mice) are necessary to mount CD8+ T cell-mediated anti-tumor immunity, whether and how tumors modulate CD103+ cDC1 function remain understudied. Signal Transducer and Activator of Transcription 3 (STAT3) mediates the intracellular signaling of tumor-associated immunosuppressive cytokines, such as interleukin (IL)-10; thus, we hypothesized that STAT3 restrained anti-tumor immune responses elicited by CD103+ cDC1s. Herein, we show that in vitro-derived STAT3-deficient (Stat3∆/∆) CD103+ cDC1s are refractory to the inhibitory effects of IL-10 on Toll-like receptor 3 (TLR3) agonist-induced maturation responses. In a tumor vaccination approach, we found Stat3∆/∆ CD103+ cDC1s restrained mammary gland tumor growth and increased mouse survival more effectively than STAT3-sufficient CD103+ cDC1s. In addition, vaccination with Stat3∆/∆ CD103+ cDC1s elicited increased amounts of tumor antigen-specific CD8+ T cells and IFN-γ+ CD4+ T cells in tumors and tumor-draining lymph nodes versus phosphate-buffered saline (PBS)-treated animals. Furthermore, IL-10 receptor-deficient CD103+ cDC1s controlled tumor growth to a similar degree as Stat3∆/∆ CD103+ cDC1s. Taken together, our data reveal an inhibitory role for STAT3 in CD103+ cDC1 maturation and regulation of anti-tumor immunity. Our results also suggest IL-10 is a key factor eliciting immunosuppressive STAT3 signaling in CD103+ cDC1s in breast cancer. Thus, inhibition of STAT3 in cDC1s may provide an important strategy to improve their efficacy in tumor vaccination approaches and cDC1-mediated control of anti-tumor immunity.

19.
Clin Immunol ; 130(1): 74-82, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18951849

ABSTRACT

We previously identified by linkage analysis a region on chromosome 1 (Eam1) that confers susceptibility to experimental autoimmune myocarditis (EAM). To evaluate the role of Eam1, we created a congenic mouse strain, carrying the susceptible Eam1 locus of A.SW on the resistant B10.S background (B10.A-Eam1 congenic) and analyzed three outcomes: 1) the incidence and severity of EAM, 2) the susceptibility of lymph node cells (LNCs) to Cy-enhanced cell death, and 3) susceptibility of lymphocytes to antigen-induced cell death. Incidence of myocarditis in B10.A-Eam1 congenic mice was comparable to A.SW mice, confirming that Eam1 plays an important role in disease development. Caspase 3, 8 and 9 activation in LNCs following Cy treatment and in CD4(+) T cells after immunization with myosin/CFA was significantly lower in A.SW than B10.S mice whereas B10.A-Eam1 congenic mice exhibited an intermediate phenotype. Our results show that Eam1 reduces lymphocyte apoptosis and increases susceptibility to EAM.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Chromosomes, Mammalian/genetics , Genetic Predisposition to Disease/genetics , Lymphocytes/cytology , Lymphocytes/metabolism , Myocarditis/immunology , Animals , Autoimmune Diseases/metabolism , Caspases/metabolism , Cell Death , Enzyme Activation , Immunization , Lymphocyte Activation/immunology , Male , Mice , Myocarditis/genetics , Myocarditis/metabolism , Myosins/immunology
20.
Mol Immunol ; 110: 24-39, 2019 06.
Article in English | MEDLINE | ID: mdl-29549977

ABSTRACT

Dendritic cells (DCs) are the principal antigen-presenting cells of the immune system and play key roles in controlling immune tolerance and activation. As such, DCs are chief mediators of tumor immunity. DCs can regulate tolerogenic immune responses that facilitate unchecked tumor growth. Importantly, however, DCs also mediate immune-stimulatory activity that restrains tumor progression. For instance, emerging evidence indicates the cDC1 subset has important functions in delivering tumor antigens to lymph nodes and inducing antigen-specific lymphocyte responses to tumors. Moreover, DCs control specific therapeutic responses in cancer including those resulting from immune checkpoint blockade. DC generation and function is influenced profoundly by cytokines, as well as their intracellular signaling proteins including STAT transcription factors. Regardless, our understanding of DC regulation in the cytokine-rich tumor microenvironment is still developing and must be better defined to advance cancer treatment. Here, we review literature focused on the molecular control of DCs, with a particular emphasis on cytokine- and STAT-mediated DC regulation. In addition, we highlight recent studies that delineate the importance of DCs in anti-tumor immunity and immune therapy, with the overall goal of improving knowledge of tumor-associated factors and intrinsic DC signaling cascades that influence DC function in cancer.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/physiology , Homeostasis , Inflammation , Neoplasms , Animals , Homeostasis/genetics , Homeostasis/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL