Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 124(3): 403-11, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24891320

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is considered a negative regulator of inflammation, as inhibition of STAT3 signaling enhances antitumor immunity. However, STAT3 activation is a key oncogenic pathway in natural killer (NK)-lineage large granular lymphomas, and we recently reported enhanced proliferation and function of human NK cells activated with IL-21, which signals primarily through STAT3. These IL-21-expanded NK cells also have increased NKG2D expression, which led us to focus our investigation on whether STAT3 regulates NKG2D. In this study, we show that modulation of STAT3 phosphorylation with cytokines and small-molecule inhibitors correlates with NKG2D expression on human NK cells, leading to altered NK-cell degranulation. Moreover, NKG2D expression on murine NK cells having conditional STAT3 ablation is lower than on NK cells from wild-type mice, and human NK cells carrying dominant-negative STAT3 mutations have decreased baseline NKG2D expression and blunted responses to IL-10 and IL-21. Lastly, we show binding of STAT3 to a predicted STAT3 binding site upstream of the NKG2D gene, which is enhanced by IL-10 and IL-21 and decreased by STAT3 inhibition. Taken together, these data show that NKG2D expression in NK cells is regulated at the transcriptional level by STAT3, resulting in a functional NK cell defect in patients with STAT3 mutations.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , STAT3 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites/genetics , DNA/genetics , DNA/metabolism , Humans , Interleukin-10/metabolism , Interleukin-15/metabolism , Interleukins/metabolism , Job Syndrome/genetics , Job Syndrome/immunology , Job Syndrome/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Phosphorylation , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , Transcription, Genetic , Tyrosine/metabolism
2.
J Leukoc Biol ; 92(6): 1215-25, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23024284

ABSTRACT

Neutrophil mobilization from the bone marrow is a critical aspect of the innate immune response, enabling a rapid deployment of phagocytes to infected or inflamed tissue. The cytokine G-CSF, which is induced rapidly during infection, elicits a swift and potent mobilizing response, yet its mechanisms of action remain poorly understood. Here, we studied the role of G-CSF and its principal signal transducer STAT3 in regulating expression of the neutrophil chemoattractant MIP-2. Our studies revealed Gr-1(hi) mature neutrophils as major sources of Cxcl2 (MIP-2) mRNA in bone marrow and G-CSF-responsive MIP-2 protein production. Induction of Cxcl2 was regulated directly by G-CSF-activated STAT3 via interaction at a STAT consensus element in the Cxcl2 promoter. G-CSF coordinately stimulated the association of STAT3, induction of the transcriptionally active H3K4me3 modification, and recruitment of RNA Pol II at the Cxcl2 proximal promoter, as well as the promoter region of Il8rb, encoding the MIP-2 receptor. These results suggest that the G-CSF-STAT3 pathway directly regulates transcriptional events that induce neutrophil mobilization.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Chemokine CXCL2/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , STAT3 Transcription Factor/metabolism , Animals , Bone Marrow Cells/immunology , Cell Line , Chemokine CXCL2/genetics , Chromatin Assembly and Disassembly , Gene Expression Regulation/drug effects , Histones/metabolism , Immunophenotyping , Mice , Mice, Knockout , Neutrophils/immunology , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Receptors, Cell Surface/metabolism , Receptors, Interleukin-8B/genetics , STAT3 Transcription Factor/genetics , Transcriptional Activation/drug effects
3.
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
4.
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
5.
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
6.
Clin Cancer Res ; 11(1): 113-22, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15671535

ABSTRACT

PURPOSE: Epithelial ovarian cancer (EOC) is characterized by early peritoneal involvement ultimately contributing to morbidity and mortality. To study the role of the peritoneum in fostering tumor invasion, we analyzed differences between the transcriptional repertoires of peritoneal tissue lacking detectable cancer in patients with EOC versus benign gynecologic disease. EXPERIMENTAL DESIGN: Specimens were collected at laparotomy from patients with benign disease (b) or malignant (m) ovarian pathology and comprised primary ovarian tumors, paired bilateral specimens from adjacent peritoneum and attached stroma (PE), subjacent stroma (ST), peritoneal washes, ascites, and peripheral blood mononuclear cells. Specimens were immediately frozen. RNA was amplified by in vitro transcription and cohybridized with reference RNA to a custom-made 17.5k cDNA microarray. RESULTS: Principal component analysis and unsupervised clustering did not segregate specimens from patients with benign or malignant pathology. Class comparison identified differences between benign and malignant PE and ST specimens deemed significant by permutation test (P = 0.027 and 0.012, respectively). A two-tailed Student's t test identified 402 (bPE versus mPE) and 663 (mST versus bST) genes differentially expressed at a significance level of P2 < or = 0.005 when all available paired samples from each patient were analyzed. The same comparison using one sample per patient reduced the pool of differentially expressed genes but retained permutation test significance for bST versus mST (P = 0.031) and borderline significance for bPE versus mPE (P = 0.056) differences. CONCLUSIONS: The presence of EOC may foster peritoneal implantation and growth of cancer cells by inducing factors that may represent molecular targets for disease control.


Subject(s)
Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/pathology , Stromal Cells/pathology , Computational Biology , DNA, Complementary/metabolism , Extracellular Matrix/metabolism , Female , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Ovary/pathology , Peritoneum/pathology , RNA/metabolism , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...