Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 177(1): 128-144, 2020 01.
Article in English | MEDLINE | ID: mdl-31465536

ABSTRACT

BACKGROUND AND PURPOSE: T helper cell 1 (Th1)-skewed neurotoxicity contributes to the poor outcome of stroke in rodents. Here, we have elucidated the mechanism of the Th1/Th2 shift in acute ischaemic stroke (AIS) patients at hyperacute phase and have looked for a miRNA-based therapeutic target. EXPERIMENTAL APPROACH: MiR-494 levels in blood from AIS patients and controls were measured by real-time PCR. C57BL/6J mice were subjected to transient middle cerebral artery occlusion, and cortical neurons were subjected to oxygen-glucose deprivation. Luciferase reporter system, chromatin immunoprecipitation sequencing (ChIP-Seq), and ChIP-PCR were used to uncover possible mechanisms. KEY RESULTS: In lymphocytes from AIS patients, there was a Th1/Th2 shift and histone deacetylase 2 (HDAC2) was markedly down-regulated. ChIP-seq showed that HDAC2 binding sites were enriched in regulation of Th1 cytokine production, and ChIP-PCR confirmed that HDAC2 binding was changed at the intron of STAT4 and the promoter of T-box transcription factor 21 (T-bet) in lymphocytes from AIS patients. MiR-494 was the most significantly increased miRNA in lymphocytes from AIS patients, and miR-494-3p directly targeted HDAC2. A strong association existed between miR-494 and Th1 cytokines, and neurological deficit as measured by the National Institute of Health Stroke Scale (NIHSS) in AIS patients. In vitro and in vivo experiments showed that antagomir-494 reduced Th1 shift-mediated neuronal and sensorimotor functional damage in the mouse model of ischaemic stroke, via the HDAC2-STAT4 pathway. CONCLUSION AND IMPLICATIONS: We demonstrated that miR-494 inhibition prevented Th1-skewed neurotoxicity through regulation of the HDAC2-STAT4 cascade.


Subject(s)
Histone Deacetylase 2/metabolism , Ischemic Stroke/metabolism , MicroRNAs/metabolism , STAT4 Transcription Factor/metabolism , Th1 Cells/metabolism , Aged , Animals , Antagomirs/pharmacology , Female , Histone Deacetylase 2/antagonists & inhibitors , Humans , Ischemic Stroke/pathology , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Middle Aged , STAT4 Transcription Factor/antagonists & inhibitors , Th1 Cells/drug effects , Th1 Cells/pathology
2.
Inflammation ; 42(4): 1179-1189, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30848408

ABSTRACT

Signal transducer and activator of transcription 4 (STAT4) has been implicated in the progression of myocarditis. The aim of the current study was to investigate the role by which STAT4 influences autoimmune myocarditis in an attempt to identify a theoretical therapeutic perspective for the condition. After successful establishment of an autoimmune myocarditis rat model, the expression patterns of STAT4, NF-κB pathway-related genes, Th1 inflammatory cytokines (IFN-γ and IL-2), and Th2 inflammatory cytokines (IL-6 and IL-10) were subsequently determined. The rats with autoimmune myocarditis were treated with oe-STAT4 or sh-STAT4 lentiviral vectors to evaluate the role of STAT4 in autoimmune myocarditis, or administrated with 1 mL 10 µmol/L of BAY11-7082 (the NF-κB pathway inhibitor) via tail vein to investigate the effect of the NF-κB pathway on autoimmune myocarditis. Finally, cell apoptosis was evaluated. The serum levels of IFN-γ and IL-2, extent of IκBα and P65 phosphorylation, and the expression of STAT4 were elevated, while the serum levels of IL-6 and IL-10 as well as the expression of IκBα were reduced among the rats with autoimmune myocarditis, which was accompanied by an increase in the apoptotic cells. More importantly, the silencing of STAT4 or the inhibition of the NF-κB pathway was detected to result in a decrease in the serum levels of IFN-γ and IL-2 and an elevation of the serum levels of IL-6 and IL-10, and inhibited myocardial cell apoptosis in rats with autoimmune myocarditis. Moreover, STAT4 silencing was also observed to decrease the extent of IκBα and P65 phosphorylation while acting to elevate the expression of IκBα. Taken together, silencing of STAT4 could hinder the progression of autoimmune myocarditis by balancing the expression of Th1/Th2 inflammatory cytokines via the NF-κB pathway, which may provide a novel target for experimental autoimmune myocarditis (EAM) treatment.


Subject(s)
Myocarditis/immunology , NF-kappa B/antagonists & inhibitors , STAT4 Transcription Factor/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Apoptosis , Autoimmune Diseases , Cytokines/blood , Disease Progression , Myocarditis/prevention & control , NF-kappa B/metabolism , Rats , STAT4 Transcription Factor/antagonists & inhibitors , STAT4 Transcription Factor/metabolism
3.
J Med Chem ; 56(11): 4521-36, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23668484

ABSTRACT

Herein we report our lead optimization effort to identify potent, selective, and orally bioavailable TYK2 inhibitors, starting with lead molecule 3. We used structure-based design to discover 2,6-dichloro-4-cyanophenyl and (1R,2R)-2-fluorocyclopropylamide modifications, each of which exhibited improved TYK2 potency and JAK1 and JAK2 selectivity relative to 3. Further optimization eventually led to compound 37 that showed good TYK2 enzyme and interleukin-12 (IL-12) cell potency, as well as acceptable cellular JAK1 and JAK2 selectivity and excellent oral exposure in mice. When tested in a mouse IL-12 PK/PD model, compound 37 showed statistically significant knockdown of cytokine interferon-γ (IFNγ), suggesting that selective inhibition of TYK2 kinase activity might be sufficient to block the IL-12 pathway in vivo.


Subject(s)
4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/chemical synthesis , Aminopyridines/chemical synthesis , Benzamides/chemical synthesis , TYK2 Kinase/antagonists & inhibitors , 4-Aminopyridine/pharmacokinetics , 4-Aminopyridine/pharmacology , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Benzamides/pharmacokinetics , Benzamides/pharmacology , Biological Availability , Crystallography, X-Ray , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-12/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Mice , Microsomes, Liver/metabolism , Models, Molecular , Protein Binding , Rats , STAT4 Transcription Factor/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
4.
EMBO Mol Med ; 4(10): 1112-25, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23027613

ABSTRACT

T lymphocytes exhibit pro-inflammatory or anti-inflammatory activities in obesity and diabetes, depending on their subtypes. Guanidine-rich immunosuppressive oligodeoxynucleotides (ODNs) effectively control Th1/Th2-cell counterbalance. This study reveals a non-toxic regulatory ODN (ODNR01) that inhibits Th1- and Th17-cell polarization by binding to STAT1/3/4 and blocking their phosphorylation without affecting Th2 and regulatory T cells. ODNR01 improves glucose tolerance and insulin sensitivity in both diet-induced obese (DIO) and genetically generated obese (ob/ob) mice. Mechanistic studies show that ODNR01 suppresses Th1- and Th17-cell differentiation in white adipose tissue, thereby reducing macrophage accumulation and M1 macrophage inflammatory molecule expression without affecting M2 macrophages. While ODNR01 shows no effect on diabetes in lymphocyte-free Rag1-deficient DIO mice, it enhances glucose tolerance and insulin sensitivity in CD4(+) T-cell-reconstituted Rag1-deficient DIO mice, suggesting its beneficial effect on insulin resistance is T-cell-dependent. Therefore, regulatory ODNR01 reduces obesity-associated insulin resistance through modulation of T-cell differentiation.


Subject(s)
Cell Differentiation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Mice, Obese , Oligodeoxyribonucleotides/administration & dosage , Th1 Cells/drug effects , Th17 Cells/drug effects , Animals , Disease Models, Animal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphorylation/drug effects , STAT1 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , STAT4 Transcription Factor/antagonists & inhibitors , Treatment Outcome
5.
J Immunol ; 189(2): 832-40, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22685315

ABSTRACT

A transcription factor network that includes STAT4, T-bet, and Runx3 promotes the differentiation of Th1 cells and inflammatory immune responses. How additional transcription factors regulate the function of Th1 cells has not been defined. In this study we show that the negative regulatory factor Twist1 decreases expression of T-bet, Runx3, and IL-12Rß2 as it inhibits IFN-γ production. Ectopic expression of Runx3, but not T-bet or IL-12Rß2, compensates for the effects of Twist1 on IFN-γ production, and Twist1 regulation of Ifng depends on complex formation with Runx3. Twist1 decreases Runx3 and T-bet binding at the Ifng locus, and it decreases chromatin looping within the Ifng locus. These data define an IL-12/STAT4-induced negative regulatory loop that impacts multiple components of the Th1 transcriptional network and provide further insight into regulation of Th1 differentiation.


Subject(s)
Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/physiology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Nuclear Proteins/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Twist-Related Protein 1/physiology , Animals , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/prevention & control , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , STAT4 Transcription Factor/antagonists & inhibitors , STAT4 Transcription Factor/deficiency , STAT4 Transcription Factor/physiology , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology , Twist-Related Protein 1/genetics
6.
Kidney Int ; 79(4): 452-63, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20980973

ABSTRACT

Polymorphisms in the transcription factor Stat4 gene have been implicated as risk factors for systemic lupus erythematosus. Although some polymorphisms have a strong association with autoantibodies and nephritis, their impact on pathophysiology is still unknown. To explore this further we used signal transducers and activators of transcription 4 (Stat4) knockout MRL/MpJ-Fas(lpr)/Fas(lpr) (MRL-Fas(lpr)) mice and found that they did not differ in survival or renal function from Stat4-intact MRL-Fas(lpr) mice. Circulating interleukin (IL)-18 levels, however, were elevated in Stat4-deficient compared to Stat4-intact mice, suggesting that this interleukin might contribute to the progression of lupus nephritis independent of Stat4. In a second approach, Stat4 antisense or missense oligonucleotides or vehicle were given to MRL-Fas(lpr) mice with advanced nephritis. Each of these treatments temporarily ameliorated disease, although IL-18 was increased in each setting. Based on these findings, studies using gene transfer to overexpress IL-18 in MRL-Fas(lpr) and IL-12p40/IL-23 knockout MRL-Fas(lpr) mice reveal a critical role for IL-18 in mediating disease. Thus, the Stat4 and IL-12 (an activator of Stat4)-independent factor, IL-18, can drive autoimmune lupus nephritis in MRL-Fas(lpr) mice. Temporarily blocking Stat4 during advanced nephritis ameliorates disease, suggesting a time-dependent compensatory proinflammatory mechanism.


Subject(s)
Interleukin-18/metabolism , Lupus Nephritis/etiology , STAT4 Transcription Factor/deficiency , Animals , Base Sequence , DNA Primers/genetics , Female , Gene Knockout Techniques , Gene Transfer Techniques , Interferon-gamma/biosynthesis , Interleukin-12/metabolism , Interleukin-18/antagonists & inhibitors , Interleukin-18/genetics , Interleukin-23/metabolism , Kidney/metabolism , Kidney/pathology , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, Knockout , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , STAT4 Transcription Factor/antagonists & inhibitors , STAT4 Transcription Factor/genetics
7.
J Immunol ; 185(6): 3593-601, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20693420

ABSTRACT

To deepen our knowledge of the natural host response to pathogens, our team undertook an in vivo screen of mutagenized 129S1 mice with Salmonella Typhimurium. One mutation affecting Salmonella susceptibility was mapped to a region of 1.3 Mb on chromosome 6 that contains 15 protein-coding genes. A missense mutation was identified in the Usp18 (ubiquitin-specific peptidase 18) gene. This mutation results in an increased inflammatory response (IL-6, type 1 IFN) to Salmonella and LPS challenge while paradoxically reducing IFN-gamma production during bacterial infection. Increased STAT1 phosphorylation correlated with impaired STAT4 phosphorylation, resulting in overwhelming IL-6 secretion but reduced IFN-gamma production during infection. The reduced IFN-gamma levels, along with the increased inflammation, rationalize the S. Typhimurium susceptibility in terms of increased bacterial load in target organs and cytokine-induced septic shock and death.


Subject(s)
Endopeptidases/genetics , Ethylnitrosourea/toxicity , Interferon-alpha/physiology , Interferon-beta/physiology , Interferon-gamma/antagonists & inhibitors , Mutation, Missense , STAT4 Transcription Factor/antagonists & inhibitors , Salmonella Infections, Animal/immunology , Signal Transduction/immunology , Animals , Endopeptidases/deficiency , Female , Genetic Predisposition to Disease , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon-gamma/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mutagens/toxicity , Mutation, Missense/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , STAT4 Transcription Factor/metabolism , STAT4 Transcription Factor/physiology , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Ubiquitin/immunology , Ubiquitin Thiolesterase
8.
J Immunol ; 178(6): 3427-36, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17339437

ABSTRACT

The transcription factor STAT4 mediates signals of various proinflammatory cytokines, such as IL-12, IL-15, and IL-23, that initiate and stabilize Th1 cytokine production. Although Th1 cytokine production has been suggested to play a major pathogenic role in rheumatoid arthritis, the role of STAT4 in this disease is poorly understood. In this study, we demonstrate a key functional role of STAT4 in murine collagen-induced arthritis (CIA). In initial studies we found that STAT4 expression is strongly induced in CD4(+) T cells and to a lesser extent in CD11b(+) APCs during CIA. To analyze the role of STAT4 for arthritis manifestation, we next investigated the outcome of interfering with STAT4 gene expression in CIA by using STAT4-deficient mice. Interestingly, STAT4-deficient mice developed significantly less severe arthritis than wild-type control mice and the T cells from such mice produced less IL-6, TNF, and IL-17. In addition, the targeting of STAT4 expression by a specific antisense phosphorothioate oligonucleotide directed at the translation start site suppressed STAT4 levels and signs of CIA even when applied during the onset of disease manifestation. These data suggest a key regulatory role of STAT4 in the pathogenesis and manifestation of murine collagen-induced arthritis. Furthermore, the targeting of STAT4 emerges as a novel approach to therapy for chronic arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Oligonucleotides, Antisense/pharmacology , STAT4 Transcription Factor/antagonists & inhibitors , Th1 Cells/immunology , Thionucleotides/pharmacology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , CD11b Antigen/immunology , Cells, Cultured , Codon, Initiator/antagonists & inhibitors , Codon, Initiator/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , STAT4 Transcription Factor/deficiency , STAT4 Transcription Factor/immunology , Th1 Cells/pathology
9.
J Immunol ; 177(3): 1679-88, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16849477

ABSTRACT

Dendritic cells (DCs) have been suggested to direct a type of Th differentiation through their cytokine profile, e.g., high IL-12/IL-23 for Th1 (named DC1/immunogenic DCs) and IL-10 for Th2 (DC2/tolerogenic DCs). Suppressor of cytokine signaling (SOCS)-3 is a potent inhibitor of Stat3 and Stat4 transduction pathways for IL-23 and IL-12, respectively. We thus hypothesize that an enhanced SOCS-3 expression in DCs may block the autocrine response of IL-12/IL-23 in these cells, causing them to become a DC2-type phenotype that will subsequently promote Th2 polarization of naive T cells. Indeed, in the present study we found that bone marrow-derived DCs transduced with SOCS-3 significantly inhibited IL-12-induced activation of Stat4 and IL-23-induced activation of Stat3. These SOCS-3-transduced DCs expressed a low level of MHC class II and CD86 on their surface, produced a high level of IL-10 but low levels of IL-12 and IFN-gamma, and expressed a low level of IL-23 p19 mRNA. Functionally, SOCS-3-transduced DCs drove naive myelin oligodendrocyte glycoprotein-specific T cells to a strong Th2 differentiation in vitro and in vivo. Injection of SOCS-3-transduced DCs significantly suppressed experimental autoimmune encephalomyelitis, a Th1 cell-mediated autoimmune disorder of the CNS and an animal model of multiple sclerosis. These results indicate that transduction of SOCS-3 in DCs is an effective approach to generating tolerogenic/DC2 cells that then skew immune response toward Th2, thus possessing therapeutic potential in Th1-dominant autoimmune disorders such as multiple sclerosis.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance/genetics , Immunophenotyping , Suppressor of Cytokine Signaling Proteins/genetics , Th2 Cells/immunology , Transduction, Genetic , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cells, Cultured , Coculture Techniques , Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Gene Expression Regulation/immunology , Interleukin-12/antagonists & inhibitors , Interleukin-12/physiology , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/antagonists & inhibitors , Interleukins/physiology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , STAT4 Transcription Factor/antagonists & inhibitors , STAT4 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/physiology , Th2 Cells/cytology
10.
Sci STKE ; 2005(304): pe49, 2005 Oct 04.
Article in English | MEDLINE | ID: mdl-16204702

ABSTRACT

The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway has evolved to serve highly specialized functions in the regulation of hematopoiesis, cell metabolism, and immune responses. The duration, strength, and specificity of cytokine signaling are controlled by several mechanisms, including the ubiquitin-proteasome pathway, which modulates the turnover of cytokine receptors and activated JAKs. The specificity of the ubiquitin pathway is achieved through various E3 ligase complexes that recognize and interact with distinct target proteins, often in a phosphorylation-dependent manner. Intriguing new information about the ubiquitin pathway came with the identification of an E3 ubiquitin ligase, SLIM, that specifically interacts with activated STAT1 and STAT4 and induces their ubiquitination and degradation. These findings, together with the evidence from paramyxoviruses about the role of ubiquitination as a highly specific STAT inhibition mechanism, highlight the role of E3 ubiquitin ligases as specificity determinants in the regulation of STAT activation, and open the field for investigation of additional E3s that target other STAT proteins.


Subject(s)
Cytokines/physiology , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction/physiology , Ubiquitin-Protein Ligases/physiology , Adaptor Proteins, Signal Transducing , Animals , Gene Expression Regulation/physiology , Humans , LIM Domain Proteins , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/physiology , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/physiology , STAT Transcription Factors/physiology , STAT4 Transcription Factor/antagonists & inhibitors , STAT4 Transcription Factor/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/chemistry , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...