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










Publication year range
1.
PLoS Biol ; 17(11): e3000530, 2019 11.
Article in English | MEDLINE | ID: mdl-31730616

ABSTRACT

Type I interferon (IFN-I) is a family of multifunctional cytokines that modulate the innate and adaptive immunity and are used to treat mastocytosis. Although IFN-I is known to suppress mast cell function, including histamine release, the mechanisms behind its effects on mast cells have been poorly understood. We here investigated IFN-I's action on mast cells using interferon-α/ß receptor subunit 1 (Ifnar1)-deficient mice, which lack a functional IFN-I receptor complex, and revealed that IFN-I in the steady state is critical for mast cell homeostasis, the disruption of which is centrally involved in systemic anaphylaxis. Ifnar1-deficient mice showed exacerbated systemic anaphylaxis after sensitization, which was associated with increased histamine in the circulation, even though the mast cell numbers and high affinity immunoglobulin E receptor (FcεRI) expression levels were similar between Ifnar1-deficient and wild-type (WT) mice. Ifnar1-deficient mast cells showed increased secretory granule synthesis and exocytosis, which probably involved the increased transcription of Tfeb. Signal transducer and activator of transcription 1(Stat1) and Stat2 were unexpectedly insufficient to mediate these IFN-I functions, and instead, Stat3 played a critical role in a redundant manner with Stat1. Our findings revealed a novel regulation mechanism of mast cell homeostasis, in which IFN-I controls lysosome-related organelle biogenesis.


Subject(s)
Anaphylaxis/immunology , Interferon Type I/physiology , Mast Cells/immunology , Secretory Vesicles/metabolism , Animals , Cells, Cultured , Histamine/blood , Homeostasis , Mice , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/physiology , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/physiology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , Signal Transduction
2.
PLoS Pathog ; 15(4): e1007745, 2019 04.
Article in English | MEDLINE | ID: mdl-31009517

ABSTRACT

The mechanisms by which the gut luminal environment is disturbed by the immune system to foster pathogenic bacterial growth and survival remain incompletely understood. Here, we show that STAT2 dependent type I IFN signaling contributes to the inflammatory environment by disrupting hypoxia enabling the pathogenic S. Typhimurium to outgrow the microbiota. Stat2-/- mice infected with S. Typhimurium exhibited impaired type I IFN induced transcriptional responses in cecal tissue and reduced bacterial burden in the intestinal lumen compared to infected wild-type mice. Although inflammatory pathology was similar between wild-type and Stat2-/- mice, we observed decreased hypoxia in the gut tissue of Stat2-/- mice. Neutrophil numbers were similar in wild-type and Stat2-/- mice, yet Stat2-/- mice showed reduced levels of myeloperoxidase activity. In vitro, the neutrophils from Stat2-/- mice produced lower levels of superoxide anion upon stimulation with the bacterial ligand N-formylmethionyl-leucyl-phenylalanine (fMLP) in the presence of IFNα compared to neutrophils from wild-type mice, indicating that the neutrophils were less functional in Stat2-/- mice. Cytochrome bd-II oxidase-mediated respiration enhances S. Typhimurium fitness in wild-type mice, while in Stat2-/- deficiency, this respiratory pathway did not provide a fitness advantage. Furthermore, luminal expansion of S. Typhimurium in wild-type mice was blunted in Stat2-/- mice. Compared to wild-type mice which exhibited a significant perturbation in Bacteroidetes abundance, Stat2-/- mice exhibited significantly less perturbation and higher levels of Bacteroidetes upon S. Typhimurium infection. Our results highlight STAT2 dependent type I IFN mediated inflammation in the gut as a novel mechanism promoting luminal expansion of S. Typhimurium.


Subject(s)
Dysbiosis/immunology , Gastroenteritis/immunology , Inflammation/immunology , Interferon Type I/immunology , STAT2 Transcription Factor/physiology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Cells, Cultured , Dysbiosis/metabolism , Dysbiosis/pathology , Female , Gastroenteritis/metabolism , Gastroenteritis/microbiology , Gastroenteritis/pathology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interferon Type I/metabolism , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Neutrophils/pathology , STAT1 Transcription Factor/physiology , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella Infections/pathology
3.
J Infect Dis ; 218(suppl_5): S438-S447, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30192975

ABSTRACT

Marburg virus (MARV; family Filoviridae) causes sporadic outbreaks of Marburg hemorrhagic fever in sub-Saharan Africa with case fatality rates reaching 90%. Wild-type filoviruses, including MARV and the closely related Ebola virus, are unable to suppress the type I interferon response in rodents, and therefore require adaptation of the viruses to cause disease in immunocompetent animals. In the current study, we demonstrate that STAT2 knockout Syrian hamsters are susceptible to infection with different wild-type MARV variants. MARV Musoke causes a robust and systemic infection resulting in lethal disease. Histopathological findings share features similar to those observed in human patients and other animal models of filovirus infection. Reverse-transcription polymerase chain reaction analysis of host transcripts shows a dysregulation of the innate immune response. Our results demonstrate that the STAT2 knockout hamster represents a novel small animal model of severe MARV infection and disease without the requirement for virus adaptation.


Subject(s)
Marburg Virus Disease/etiology , STAT2 Transcription Factor/physiology , Animals , Cricetinae , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility , Female , Male , Marburg Virus Disease/immunology , Marburg Virus Disease/pathology
4.
PLoS One ; 12(5): e0176994, 2017.
Article in English | MEDLINE | ID: mdl-28472186

ABSTRACT

The JAK/STAT signaling pathway is suggested to play an important role in the pathogenesis of psoriasis, and recently JAK/STAT inhibitors have shown promising results in psoriasis treatment. The present study aimed to characterize the role of STAT2 in psoriasis. We demonstrated an increased expression of STAT2 and an increased level of phosphorylated/activated STAT2 in lesional compared with nonlesional psoriatic skin. Gene silencing of STAT2 by siRNA in human keratinocytes revealed that upon IFNα stimulation CXCL11 and CCL5 were the only two cytokines, among 102 analyzed, found to be regulated through a STAT2-dependent mechanism. Moreover, the regulation of CXCL11 and CCL5 depended on IRF9, but not on STAT1 and STAT6. The CXCL11 and CCL5 expression was increased in lesional compared with nonlesional psoriatic skin, and analysis demonstrated positive correlation between the expression of CXCL11 and IFNγ and between the expression of CCL5 and IFNγ in lesional psoriatic skin. In contrast, no correlation between the expression of CXCL11 and IL-17A and the expression of CCL5 and IL-17A in lesional psoriatic skin was found. Our data suggest that STAT2 plays a role in the psoriasis pathogenesis by regulating the expression of CXCL11 and CCL5, and thereby attracting IFNγ-producing immune cells to the skin.


Subject(s)
Chemokine CCL5/biosynthesis , Chemokine CXCL11/biosynthesis , Genetic Predisposition to Disease , Keratinocytes/metabolism , Psoriasis/genetics , STAT2 Transcription Factor/physiology , Adult , Chemokine CCL5/genetics , Chemokine CXCL11/genetics , Humans , Phosphorylation , Psoriasis/pathology , STAT2 Transcription Factor/genetics
5.
PLoS Biol ; 14(10): e2000117, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27780205

ABSTRACT

STAT2 is the quintessential transcription factor for type 1 interferons (IFNs), where it functions as a heterodimer with STAT1. However, the human and murine STAT2-deficient phenotypes suggest important additional and currently unidentified type 1 IFN-independent activities. Here, we show that STAT2 constitutively bound to STAT1, but not STAT3, via a conserved interface. While this interaction was irrelevant for type 1 interferon signaling and STAT1 activation, it precluded the nuclear translocation specifically of STAT1 in response to IFN-γ, interleukin-6 (IL-6), and IL-27. This is explained by the dimerization between activated STAT1 and unphosphorylated STAT2, whereby the semiphosphorylated dimers adopted a conformation incapable of importin-α binding. This, in turn, substantially attenuated cardinal IFN-γ responses, including MHC expression, senescence, and antiparasitic immunity, and shifted the transcriptional output of IL-27 from STAT1 to STAT3. Our results uncover STAT2 as a pervasive cytokine regulator due to its inhibition of STAT1 in multiple signaling pathways and provide an understanding of the type 1 interferon-independent activities of this protein.


Subject(s)
STAT1 Transcription Factor/antagonists & inhibitors , STAT2 Transcription Factor/physiology , Signal Transduction , Animals , Binding Sites , Cell Nucleus/metabolism , DNA/metabolism , Dimerization , Gene Expression/physiology , Humans , Interferon-gamma/metabolism , Interferon-gamma/physiology , Phosphorylation , Protein Binding , Protein Conformation , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism
6.
Domest Anim Endocrinol ; 52: 60-70, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25863197

ABSTRACT

The core of the leptin resistance hypothesis promulgated several years ago to explain obesity as a result of environmental causes consists of 2 tenets: the extinction of leptin-induced intracellular signaling downstream of leptin binding to the long form of the neuronal receptor LTRb in the hypothalamus and the impedance to leptin entry imposed at the blood-brain barrier (BBB). A recent comprehensive investigation concluded that a central leptin insufficiency associated with obesity can be attributed to a decreased efficiency of BBB leptin transport and not to leptin insensitivity within the hypothalamus. Interestingly, anorectic leptin's effects are counteracted in some individuals by a natural resistance associated with hyperleptinemia, which is related to changes in hypothalamic sensitivity to leptin (eg, due to malnutrition, obesity, or seasonal variations due to day-length-dependent reproduction changes). In sheep, it has been observed that the hypothalamus is resistant to leptin in some periods, which is related to the adaptation of these animals to annual changes in energy supply and demand. However, a broad range of ambiguities exists regarding the implications that the intracellular signaling of signal transducer and activator of transcription-2/suppressor of cytokine signaling 3 (STAT2/SOCS3) imparts central leptin resistance. Furthermore, several plausible alternative possibilities have been proposed, such as compensatory functional and anatomic reorganizations in the appetite regulating network, rearrangements in the afferent hormonal feedback signaling involved in weight homeostasis, and modifications in leptin transport to the hypothalamus across the BBB. Taken together, these observations suggest that the contention that impaired intracellular signaling downstream of leptin entry into the appetite regulating network expedites environmentally induced obesity remains unsubstantiated and requires further evidence. Furthermore, pregnancy decreases hypothalamic sensitivity to leptin (or other unknown mechanisms), and lactation can also alter the appetite-suppressing central activity of leptin. The objective of this review was to offer an approach to understanding (1) how information regarding nutritional status is transmitted to and interpreted within the hypothalamus in animals, with special attention on seasonally breeding animals and (2) whether central leptin resistance and/or leptin insufficiency in the hypothalamus favors the development of obesity.


Subject(s)
Brain/drug effects , Leptin/physiology , Seasons , Animals , Appetite Regulation , Biological Transport , Blood-Brain Barrier , Breeding , Drug Resistance , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Leptin/metabolism , Leptin/pharmacology , Mammary Glands, Animal/chemistry , Nutritional Status , Obesity , Photoperiod , Pregnancy , Prolactin/physiology , RNA, Messenger/analysis , Receptors, Leptin/metabolism , STAT2 Transcription Factor/physiology , Sheep , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/physiology
7.
Int J Cancer ; 136(1): 117-26, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-24895110

ABSTRACT

The role of STAT2 in mediating the antigrowth effects of type I interferon (IFN) is well-documented in vitro. Yet evidence of IFN-activated STAT2 as having tumor suppressor function in vivo and participation in antitumor immunity is lacking. Here we show in a syngeneic tumor transplantation model that STAT2 reduces tumor growth. Stat2(-/-) mice formed larger tumors compared to wild type (WT) mice. IFN-ß treatment of Stat2(-/-) mice did not cause tumor regression. Gene expression analysis revealed a small subset of immunomodulatory genes to be downregulated in tumors established in Stat2(-/-) mice. Additionally, we found tumor antigen cross-presentation by Stat2(-/-) dendritic cells to T cells to be impaired. Adoptive transfer of tumor antigen specific CD8(+) T cells primed by Stat2(-/-) dendritic cells into tumor-bearing Stat2(-/-) mice did not induce tumor regression with IFN-ß intervention. We observed that an increase in the number of CD4(+) and CD8(+) T cells in the draining lymph nodes of IFN-ß-treated tumor-bearing WT mice was absent in IFN-ß treated Stat2(-/-) mice. Thus our study provides evidence for further evaluation of STAT2 function in cancer patients receiving type I IFN based immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Interferon-beta/pharmacology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , STAT2 Transcription Factor/physiology , Tumor Burden/immunology , Adoptive Transfer , Animals , Cell Line, Tumor , Culture Media, Conditioned , Dendritic Cells , Down-Regulation , Gene Expression Regulation, Neoplastic/immunology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Transplantation , STAT1 Transcription Factor/metabolism , T-Lymphocytes/immunology
8.
Biochem Pharmacol ; 84(9): 1164-73, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22902832

ABSTRACT

Dysregulation of signal transducer and activator of transcription (STAT) signaling is usually associated with intricate immune diseases and rebuilding the balance of STAT1 and STAT3 signaling is being explored as a useful approach for the treatment of these diseases. However, few chemicals have been reported to rebuild the balance of these two signalings for immune hepatitis therapy. In the present study, we found that fusaruside, a new kind of cerebroside isolated from an endophytic fungus Fusarium sp. IFB-121 in Quercus variabilis, significantly ameliorated concanavalin A (Con A)-induced T-cell-mediated fulminant hepatitis in mice, which was closely associated with the improvement of histopathological parameters, inhibition of activation of liver CD4(+) T cells and NKT cells, regulation of balance of Th1/Th2/Th17/Treg cytokines and protection of hepatocyte from apoptosis. Moreover, T-cell proliferation and activation was also notably inhibited by fusaruside in vitro. Furthermore, the protective effect of fusaruside was attributable to a novel regulatory mechanism through down-regulating STAT1 activation and T-bet expression in liver CD4(+) T cells and up-regulating STAT3 activation and Bcl-X(L) expression in hepatocytes. In conclusion, fusaruside exhibited its capability against T-cell-mediated liver injury in vivo, through rebuilding the balance of STAT1 and STAT3 signalings. These results suggest that fusaruside is potentially useful for the treatment of T-cell-mediated human liver disorders.


Subject(s)
Cerebrosides/pharmacology , Hepatitis, Animal/drug therapy , Liver Failure, Acute/drug therapy , STAT1 Transcription Factor/physiology , STAT2 Transcription Factor/physiology , T-Lymphocytes/drug effects , Animals , CD4 Antigens/metabolism , Cell Proliferation/drug effects , Cerebrosides/isolation & purification , Concanavalin A , Cytokines/metabolism , Female , Fusarium/chemistry , Hepatitis, Animal/etiology , Hepatitis, Animal/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Failure, Acute/etiology , Liver Failure, Acute/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Signal Transduction , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , bcl-X Protein/metabolism
9.
Viruses ; 4(1): 62-82, 2012 01.
Article in English | MEDLINE | ID: mdl-22355452

ABSTRACT

The development of animal models of dengue virus (DENV) infection and disease has been challenging, as epidemic DENV does not naturally infect non-human species. Non-human primates (NHPs) can sustain viral replication in relevant cell types and develop a robust immune response, but they do not develop overt disease. In contrast, certain immunodeficient mouse models infected with mouse-adapted DENV strains show signs of severe disease similar to the 'vascular-leak' syndrome seen in severe dengue in humans. Humanized mouse models can sustain DENV replication and show some signs of disease, but further development is needed to validate the immune response. Classically, immunocompetent mice infected with DENV do not manifest disease or else develop paralysis when inoculated intracranially; however, a new model using high doses of DENV has recently been shown to develop hemorrhagic signs after infection. Overall, each model has its advantages and disadvantages and is differentially suited for studies of dengue pathogenesis and immunopathogenesis and/or pre-clinical testing of antiviral drugs and vaccines.


Subject(s)
Dengue , Models, Animal , Animals , Antiviral Agents/therapeutic use , Chimera , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dengue/drug therapy , Dengue/immunology , Dengue/physiopathology , Dengue/transmission , Dengue Vaccines , Dengue Virus/pathogenicity , Dengue Virus/physiology , Drug Evaluation, Preclinical , Hematopoietic Stem Cell Transplantation , Host Specificity , Humans , Immunity, Cellular , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Macaca mulatta , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/physiology , Species Specificity , Viremia/immunology
10.
J Gen Virol ; 93(Pt 4): 716-721, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22158879

ABSTRACT

Human cytomegalovirus (HCMV) immediate-early (IE) 1 protein associates with chromosomes in mitotic cells using its carboxyl-terminal 16 aa region. However, the role of this IE1 activity in viral growth has not been evaluated in the context of mutant virus infection. We produced a recombinant HCMV encoding mutant IE1 with the carboxyl-terminal chromosome-tethering domain (CTD) deleted. This IE1(ΔCTD) virus grew like the wild-type virus in fibroblasts, indicating that the CTD is not essential for viral replication in permissive cells. Unlike wild-type virus infections, PML and STAT2, which interact with IE1, did not accumulate at mitotic chromosomes in IE1(ΔCTD) virus-infected fibroblasts, demonstrating that their associations with chromosomes are IE1 CTD-dependent. IE1 SUMOylation did not affect IE1 association with chromosomes. Our results provide genetic evidence that the CTD is required for the associations of IE1, PML and STAT2 with mitotic chromosomes, but that these IE1-related activities are not essential for viral replication in fibroblasts.


Subject(s)
Chromatin/virology , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Immediate-Early Proteins/physiology , Chromatin/metabolism , Chromosomes, Human/metabolism , Chromosomes, Human/virology , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Mitosis/physiology , Recombination, Genetic , STAT2 Transcription Factor/metabolism , STAT2 Transcription Factor/physiology , SUMO-1 Protein/metabolism , SUMO-1 Protein/physiology , Virus Replication/genetics , Virus Replication/physiology
11.
PLoS Pathog ; 7(2): e1001297, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21379341

ABSTRACT

Dengue virus (DENV) is a mosquito-borne flavivirus, and symptoms of infection range from asymptomatic to the severe dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). High viral loads correlate with disease severity, and both type I & II interferons (IFNs) are crucial for controlling viral replication. We have previously reported that signal transducer and activator of transcription (STAT) 1-deficient mice are resistant to DENV-induced disease, but little is known about this STAT1-independent mechanism of protection. To determine the molecular basis of the STAT1-independent pathway, mice lacking STAT1, STAT2, or both STAT1 and STAT2 were infected with a virulent mouse-adapted strain of DENV2. In the first 72 hours of infection, the single-deficient mice lacking STAT1 or STAT2 possessed 50-100 fold higher levels of viral RNA than wild type mice in the serum, spleen, and other visceral tissues, but remained resistant to DENV-induced death. In contrast, the double-deficient mice exhibited the early death phenotype previously observed in type I and II IFN receptor knockout mice (AG129), indicating that STAT2 is the mediator of the STAT1-independent host defense mechanism. Further studies demonstrated that this STAT2-dependent STAT1-independent mechanism requires the type I IFN receptor, and contributes to the autocrine amplification of type I IFN expression. Examination of gene expression in the spleen and bone marrow-derived macrophages following DENV infection revealed STAT2-dependent pathways can induce the transcription of a subset of interferon stimulated genes even in the absence of STAT1. Collectively, these results help elucidate the nature of the poorly understood STAT1-independent host defense mechanism against viruses by identifying a functional type I IFN/STAT2 signaling pathway following DENV infection in vivo.


Subject(s)
Dengue Virus/pathogenicity , Dengue/immunology , Dengue/virology , Immunity, Innate , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/physiology , STAT2 Transcription Factor/physiology , Animals , Blotting, Western , Chromatin Immunoprecipitation , DNA, Viral/genetics , Dengue/genetics , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Macrophages , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , RNA, Viral/genetics , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Virus Replication
12.
Mol Cancer Res ; 8(1): 80-92, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20068068

ABSTRACT

Type I IFNs (IFN-alpha/beta) are pleitropic cytokines widely used in the treatment of certain malignancies, hepatitis B and C, and multiple sclerosis. IFN resistance is a challenging clinical problem to overcome. Hence, understanding the molecular mechanism by which IFN immunotherapy ceases to be effective is of translational importance. In this study, we report that continuous IFN-alpha stimulation of the human Jurkat variant H123 led to resistance to type I IFN-induced apoptosis due to a loss of signal transducers and activators of transcription 2 (STAT2) expression. The apoptotic effects of IFN-alpha were hampered as STAT2-deficient cells were defective in activating the mitochondrial-dependent death pathway and ISGF3-mediated gene activation. Reconstitution of STAT2 restored the apoptotic effects of IFN-alpha as measured by the loss of mitochondrial membrane potential, cytochrome c release from mitochondria, caspase activation, and ultimately cell death. Nuclear localization of STAT2 was a critical event as retention of tyrosine-phosphorylated STAT2 in the cytosol was not sufficient to activate apoptosis. Furthermore, silencing STAT2 gene expression in Saos2 and A375S.2 tumor cell lines significantly reduced the apoptotic capacity of IFN-alpha. Altogether, we show that STAT2 is a critical mediator in the activation of type I IFN-induced apoptosis. More importantly, defects in the expression or nuclear localization of STAT2 could lessen the efficacy of type I IFN immunotherapy.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Drug Resistance, Neoplasm/genetics , Interferon-alpha/pharmacology , STAT2 Transcription Factor/genetics , Cell Nucleus/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Deletion , Gene Silencing/physiology , Humans , Jurkat Cells , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Phosphorylation/drug effects , Phosphorylation/genetics , Protein-Tyrosine Kinases/metabolism , STAT2 Transcription Factor/metabolism , STAT2 Transcription Factor/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transfection , Tumor Cells, Cultured
13.
J Virol ; 83(24): 12854-70, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19812155

ABSTRACT

Our previous work has shown that efficient evasion from type I interferon responses by human cytomegalovirus (hCMV) requires expression of the 72-kDa immediate-early 1 (IE1) protein. It has been suggested that IE1 inhibits interferon signaling through intranuclear sequestration of the signal transducer and activator of transcription 2 (STAT2) protein. Here we show that physical association and subnuclear colocalization of IE1 and STAT2 depend on short acidic and serine/proline-rich low-complexity motifs in the carboxy-terminal region of the 491-amino-acid viral polypeptide. These motifs compose an essential core (amino acids 373 to 420) and an adjacent ancillary site (amino acids 421 to 445) for STAT2 interaction that are predicted to form part of a natively unstructured domain. The presence of presumably "disordered" carboxy-terminal domains enriched in low-complexity motifs is evolutionarily highly conserved across all examined mammalian IE1 orthologs, and the murine cytomegalovirus IE1 protein appears to interact with STAT2 just like the human counterpart. A recombinant hCMV specifically mutated in the IE1 core STAT2 binding site displays hypersensitivity to alpha interferon, delayed early viral protein accumulation, and attenuated growth in fibroblasts. However, replication of this mutant virus is specifically restored by knockdown of STAT2 expression. Interestingly, complex formation with STAT2 proved to be entirely separable from disruption of nuclear domain 10 (ND10), another key activity of IE1. Finally, our results demonstrate that IE1 counteracts the antiviral interferon response and promotes viral replication by at least two distinct mechanisms, one depending on sequestration of STAT2 and the other one likely involving ND10 interaction.


Subject(s)
Immediate-Early Proteins/chemistry , STAT2 Transcription Factor/chemistry , Amino Acid Sequence , Binding Sites , Cells, Cultured , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Humans , Immediate-Early Proteins/physiology , Interferon Type I/pharmacology , Molecular Sequence Data , STAT2 Transcription Factor/physiology , Virus Replication/drug effects
14.
J Neuroendocrinol ; 21(3): 200-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19207826

ABSTRACT

Adrenal medullary chromaffin cells are an integral part of the neuroendocrine system, playing an important role in the physiological adaptation to stress. In response to a wide variety of stimuli, including acetylcholine released from the splanchnic nerve, hormones such as angiotensin II or paracrine signals such as prostaglandins, chromaffin cells synthesise and secrete catecholamines and a number of biologically active peptides. This adrenal medullary output mediates a complex and diverse stress response. We report that chromaffin cells also respond both acutely and chronically to interferon (IFN)-alpha, thus providing a mechanism of interaction between the immune system and the stress response. Incubation of isolated bovine chromaffin cells maintained in culture, with IFN-alpha resulted in a rapid, transient activation of the extracellular signal-regulated protein kinase (ERK)1/2, which was maximal after 5 min. IFN-alpha mediated activation of ERK1/2 appeared to be responsible for the increased phosphorylation of tyrosine hydroxylase, the rate-limiting enzyme in catecholamine synthesis. This tyrosine hydroxylase phosphorylation was exclusively on serine 31, with no change in the phosphorylation of serine 19 or 40. This increase in the serine 31 phosphorylation of tyrosine hydroxylase was prevented by inhibition of protein kinase C or ERK1/2 activation. Incubation with IFN-alpha also resulted in a time- and concentration-dependent phosphorylation and nuclear translocation of signal transducer and activator of transcription proteins (STAT)1 and 2. This response was maximal after approximately 60 min. Prolonged treatment with IFN-alpha (12-48 h) resulted in increased expression of STAT1 and, to a lesser extent, STAT2. Thus, these findings demonstrate that adrenal medullary chromaffin cells are responsive to IFN-alpha and provide a possible cellular mechanism by which this immune-derived signal can potentially influence and integrate with the stress response.


Subject(s)
Adrenal Medulla/metabolism , Chromaffin Cells/metabolism , Interferon-alpha/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/metabolism , STAT1 Transcription Factor/physiology , STAT2 Transcription Factor/physiology , Tyrosine 3-Monooxygenase/metabolism , Active Transport, Cell Nucleus/drug effects , Adrenal Medulla/cytology , Adrenal Medulla/drug effects , Animals , Cattle , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Chromaffin Cells/drug effects , Interferon-alpha/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/physiology , Phosphorylation/drug effects , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Serine/chemistry , Serine/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tyrosine 3-Monooxygenase/chemistry
15.
J Immunol ; 182(5): 2849-58, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234180

ABSTRACT

Immediately after viral infection, innate responses including expression of IFN-alpha/beta and IFN-stimulated genes (ISGs) are elicited ubiquitously by recruitment of specific pathogen recognition receptors. The velocity to induce IFN-alpha/beta and ISGs in response to an infection is often decisive for virulence. Interestingly, in primary endothelial cells ISGs are induced later by hantaviruses pathogenic to humans than those considered to be nonpathogenic or of low virulence. Here we demonstrate that pathogenic Hantaan (HTNV) and putatively nonpathogenic Prospect Hill hantavirus (PHV) differentially activate innate responses in the established cell lines A549 and HuH7. STAT1alpha phosphorylation was detectable 3 h after PHV inoculation but not within the first 2 days after HTNV inoculation. The velocity to induce the ISGs MxA and ISG15 correlated inversely with amounts of virus produced. Moreover, expression of the inflammatory chemokine CCL5 was also induced differentially. Both hantaviruses induced innate responses via TRAF3 (TNF receptor-associated factor 3), and TLR3 was required for HTNV-induced expression of MxA, but not for the MxA induction triggered by PHV. Infection of RIG-I-deficient HuH7.5 cells revealed that RIG-I (retinoic acid receptor I) was not necessary for induction of innate responses by PHV. Taken together, these data suggest that HTNV and PHV elicit different signaling cascades that converge via TRAF3. Early induction of antiviral responses might contribute to efficient elimination of PHV. Subsequent to clearance of the infection, innate responses most likely cease; vice versa, retarded induction of antiviral responses could lead to increased HTNV replication and dissemination, which might cause a prolonged inflammatory response and might contribute to the in vivo virulence.


Subject(s)
Gene Expression Regulation, Viral/immunology , Hantaan virus/immunology , Immunity, Innate , Toll-Like Receptor 3/physiology , Virus Activation/immunology , Animals , Cell Line, Transformed , Cell Line, Tumor , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chlorocebus aethiops , Cytokines/physiology , DEAD Box Protein 58 , DEAD-box RNA Helicases/physiology , GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/genetics , Hantaan virus/pathogenicity , Humans , Interferon-alpha/physiology , Interferon-beta/physiology , Myxovirus Resistance Proteins , Receptors, Immunologic , STAT2 Transcription Factor/physiology , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/physiology , Ubiquitins/physiology , Vero Cells , Virus Replication/immunology
16.
J Biol Chem ; 283(49): 34029-36, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18835816

ABSTRACT

Skeletal muscle satellite cell-derived myoblasts are mainly responsible for postnatal muscle growth and injury-induced regeneration. However, the cellular signaling pathways that control proliferation and differentiation of myoblasts remain poorly defined. Recently, we found that JAK1/STAT1/STAT3 not only participate in myoblast proliferation but also actively prevent them from premature differentiation. Unexpectedly, we found that a related pathway consisting of JAK2, STAT2, and STAT3 is required for early myogenic differentiation. Interference of this pathway by either a small molecule inhibitor or small interfering RNA inhibits myogenic differentiation. Consistently, all three molecules are activated upon differentiation. The pro-differentiation effect of JAK2/STAT2/STAT3 is partially mediated by MyoD and MEF2. Interestingly, the expression of the IGF2 gene and the HGF gene is also regulated by JAK2/STAT2/STAT3, suggesting that this pathway could also promote differentiation by regulating signaling molecules known to be involved in myogenic differentiation. In summary, our current study reveals a novel role for the JAK2/STAT2/STAT3 pathway in myogenic differentiation.


Subject(s)
Gene Expression Regulation , Janus Kinase 2/metabolism , Myoblasts/metabolism , STAT2 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Differentiation , Cell Proliferation , Janus Kinase 2/physiology , Mice , Mice, Inbred C57BL , Models, Biological , Muscle Development , Plasmids/metabolism , RNA, Small Interfering/metabolism , STAT2 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Signal Transduction
17.
Dev Comp Immunol ; 32(5): 563-74, 2008.
Article in English | MEDLINE | ID: mdl-17981327

ABSTRACT

Lactobacillus acidophilus has been shown to exert immunostimulating activities in a number of species, including the chicken. To examine the molecular mechanisms of this phenomenon, we investigated spatial and temporal expression of immune system genes in chicken cecal tonsil and spleen mononuclear cells in response to structural constituents of L. acidophilus. Using a low-density chicken immune system microarray, we found that cecal tonsil cells responded more rapidly than spleen cells to the bacterial stimuli, with the most potent stimulus for cecal tonsil cells being DNA and for splenocytes being the bacterial cell wall components. We also discovered that in both splenocytes and cecal tonsil cells, STAT2 and STAT4 genes were highly induced. Given the close interactions between cecal tonsil cells and commensal bacteria, we further examined the involvement of STAT2 and STAT4 signaling pathways in cellular responses to bacterial DNA. Our results revealed that the expression of STAT2, STAT4, IL-18, MyD88, IFN-alpha, and IFN-gamma genes were up-regulated in cecal tonsil cells after treatment with L. acidophilus DNA.


Subject(s)
Chickens/immunology , Gene Expression Profiling , Lactobacillus acidophilus , Lymphocytes/metabolism , Probiotics/pharmacology , Animals , Cytokines/genetics , Cytokines/physiology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/physiology , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/physiology
18.
J Immunol ; 179(1): 463-71, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579067

ABSTRACT

MHC type II (MHC II) expression is tightly regulated in macrophages and potently induced by IFN-gamma (type II IFN). In contrast, type I IFNs (IFN-Is), which are far more widely expressed, fail to induce MHC II expression, even though both classes of IFNs direct target gene expression through Stat1. The unexpected finding that IFN-Is effectively induce MHC II expression in Stat2(-/-) macrophages provided an opportunity to explore this conundrum. The ensuing studies revealed that deletion of Stat2, which uniquely transduces signals for IFN-Is, leads to a loss in the IFN-I-dependent induction of suppressor of cytokine signaling-1. Impairment in the expression of this important negative regulator led to a striking prolongation in IFN-I-dependent Stat1 activation, as well as enhanced expression of the target gene, IFN-regulatory factor-1. The prolonged activity of these two transcription factors synergized to drive the transcription of CIITA, the master regulator of MHC II expression, analogous to the pattern observed in IFN-gamma-treated macrophages. Thus, IFN-I-dependent suppressor of cytokine signaling-1 expression plays an important role in distinguishing the biological response between type I and II IFNs in macrophages.


Subject(s)
Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/genetics , STAT2 Transcription Factor/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , Humans , Interferon-alpha/physiology , L Cells , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Promoter Regions, Genetic/immunology , STAT2 Transcription Factor/deficiency , STAT2 Transcription Factor/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/physiology , Trans-Activators/antagonists & inhibitors , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription, Genetic/immunology
19.
Scand J Immunol ; 64(3): 271-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16918696

ABSTRACT

Monocytic differentiation is coordinated through the ordered activation of multiple signalling pathways, controlling transcription of specific subsets of genes that regulate the development of the mature phenotype. To identify key transcription factors involved in this process, we used the human monoblastic U-937 cell line as a model of monocytic differentiation. U-937 cells can be differentiated by treatment with all-trans retinoic acid (ATRA) and 1,25alpha-dihydroxycholecalciferol (VitD3), resulting in G(0)/G(1)-arrested cells expressing monocytic surface markers. We have previously shown that ATRA-induced differentiation and cell cycle arrest specifically requires Stat1 activation, through phosphorylation of tyrosine 701 and serine 727. In this report, we used U-937 cells expressing phosphorylation-deficient mutants of Stat1 (Stat1Y701F and Stat1S727A) to determine myeloid-specific transcription factors that are activated downstream of Stat1 during induced monocytic differentiation. We demonstrate that ATRA-induced upregulation of Stat2, ICSBP/IRF8 and C/EBPepsilon, key transcription factors linked to myelomonocytic differentiation, is selectively impaired in cells expressing mutant Stat1. In contrast, ATRA-induced expression of PU.1, C/EBPalpha, C/EBPbeta and IRF-1 was unaffected. Taken together, our data suggest that ATRA-induced regulation of Stat2, ICSBP and C/EBPepsilon is dependent on active Stat1, and that a failure to correctly regulate these transcription factors is associated with the inhibition of monocytic differentiation.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Interferon Regulatory Factors/metabolism , Monocytes/physiology , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Cell Differentiation , Dihydroxycholecalciferols/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Phosphorylation , Proto-Oncogene Proteins/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/physiology , STAT2 Transcription Factor/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , U937 Cells , Up-Regulation
20.
J Leukoc Biol ; 79(6): 1286-94, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16624932

ABSTRACT

In human monocyte-derived dendritic cells (DC), infection with Mycobacterium tuberculosis and viruses or stimulation with Toll-like receptor type 3 and 4 agonists causes the release of type I interferon (IFN). Here, we describe that the IFN-beta released upon stimulation with lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C) is responsible for a rapid and sustained signal transducer and activator of transcription 1 and 2 activation and expression of IFN-stimulated genes, such as the transcription factor IFN regulatory factor 7 and the chemokine CXC chemokine ligand 10. The autocrine production of IFN-beta from LPS and poly I:C-matured DC (mDC) induced a temporary saturation of the response to type I IFN and a marked decline in the level of the two IFN receptor (IFNAR) subunits. It is interesting that we found that upon clearing of the released cytokines, LPS-stimulated DC reacquired full responsiveness to IFN-beta but only partial responsiveness to IFN-alpha, and their maturation process was unaffected. Monitoring of surface and total levels of the receptor subunits showed that maximal expression of IFNAR2 resumed within 24 h of clearing, and IFNAR1 expression remained low. Thus, mDC can modulate their sensitivity to two IFN subtypes through a differential regulation of the IFNAR subunits.


Subject(s)
Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Interferon Inducers/pharmacology , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Membrane Proteins/biosynthesis , Receptors, Interferon/biosynthesis , Antibodies, Monoclonal/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chemokine CXCL10 , Chemokines, CXC/physiology , Down-Regulation/drug effects , Endotoxins/pharmacology , Humans , Interferon alpha-2 , Membrane Proteins/genetics , Monocytes/cytology , Poly I-C/pharmacology , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Recombinant Proteins/pharmacology , STAT1 Transcription Factor/physiology , STAT2 Transcription Factor/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...