Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 327
Filtrar
1.
J Exp Clin Cancer Res ; 43(1): 67, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429845

RESUMO

BACKGROUND: Docetaxel resistance represents a significant obstacle in the treatment of prostate cancer. The intricate interplay between cytokine signalling pathways and transcriptional control mechanisms in cancer cells contributes to chemotherapeutic resistance, yet the underlying molecular determinants remain only partially understood. This study elucidated a novel resistance mechanism mediated by the autocrine interaction of interleukin-11 (IL-11) and its receptor interleukin-11 receptor subunit alpha(IL-11RA), culminating in activation of the JAK1/STAT4 signalling axis and subsequent transcriptional upregulation of the oncogene c-MYC. METHODS: Single-cell secretion profiling of prostate cancer organoid was analyzed to determine cytokine production profiles associated with docetaxel resistance.Analysis of the expression pattern of downstream receptor IL-11RA and enrichment of signal pathway to clarify the potential autocrine mechanism of IL-11.Next, chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) was performed to detect the nuclear localization and DNA-binding patterns of phosphorylated STAT4 (pSTAT4). Coimmunoprecipitation and reporter assays were utilized to assess interaction between pSTAT4 and the cotranscription factor CREB-binding protein (CBP) as well as their role in c-MYC transcriptional activity. RESULTS: Autocrine secretion of IL-11 was markedly increased in docetaxel-resistant prostate cancer cells. IL-11 stimulation resulted in robust activation of JAK1/STAT4 signalling. Upon activation, pSTAT4 translocated to the nucleus and associated with CBP at the c-MYC promoter region, amplifying its transcriptional activity. Inhibition of the IL-11/IL-11RA interaction or disruption of the JAK1/STAT4 pathway significantly reduced pSTAT4 nuclear entry and its binding to CBP, leading to downregulation of c-MYC expression and restoration of docetaxel sensitivity. CONCLUSION: Our findings identify an autocrine loop of IL-11/IL-11RA that confers docetaxel resistance through the JAK1/STAT4 pathway. The pSTAT4-CBP interaction serves as a critical enhancer of c-MYC transcriptional activity in prostate cancer cells. Targeting this signalling axis presents a potential therapeutic strategy to overcome docetaxel resistance in advanced prostate cancer.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Interleucina-11 , Neoplasias da Próstata , Humanos , Masculino , Docetaxel/farmacologia , Regulação da Expressão Gênica , Interleucina-11/genética , Interleucina-11/metabolismo , Janus Quinase 1/genética , Janus Quinase 1/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais , Fator de Transcrição STAT4/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética
2.
Int J Mol Sci ; 25(6)2024 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-38542139

RESUMO

Our goal was to investigate the effects of epidermal growth factor (EGF) and interferons (IFNs) on signal transducer and activator of transcription STAT1 and STAT4 mRNA and active phosphorylated protein expression in Sjögren's syndrome cell culture models. iSGECs (immortalized salivary gland epithelial cells) and A253 cells were treated with EGF, IFN-alpha, -beta, -gamma, or mitogen-activated protein kinase p38 alpha (p38-MAPK) inhibitor for 0-24-48-72 h. STAT1 and STAT4 mRNA expression was quantified by qRT-PCR. Untreated and treated cells were compared using the delta-delta-CT method based on glyceraldehyde-3-phosphate dehydrogenase (GAPDH) normalized relative fold changes. phospho-tyrosine-701-STAT1 and phospho-serine-721-STAT4 were detected by Western blot analysis. STAT4 mRNA expression decreased 48 h after EGF treatment in A253 cells, immortalized salivary gland epithelial cells iSGECs nSS2 (sicca patient origin), and iSGECs pSS1 (anti-SSA negative Sjögren's Syndrome patient origin). EGF and p38-MAPK inhibitor decreased A253 STAT4 mRNA levels. EGF combined with IFN-gamma increased phospho-STAT4 and phospho-STAT1 after 72 h in all cell lines, suggesting additive effects for phospho-STAT4 and a major effect from IFN-gamma for phospho-STAT1. pSS1 and nSS2 cells responded differently to type I and type II interferons, confirming unique functional characteristics between iSGEC cell lines. EGF/Interferon related pathways might be targeted to regulate STAT1 and STAT4 expression in salivary gland epithelial cells. Further investigation is required learn how to better target the Janus kinases/signal transducer and activator of transcription proteins (JAK/STAT) pathway-mediated inflammatory response in Sjögren's syndrome.


Assuntos
Fator de Crescimento Epidérmico , Síndrome de Sjogren , Humanos , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Síndrome de Sjogren/tratamento farmacológico , Síndrome de Sjogren/genética , Interferon-alfa/farmacologia , Fatores Imunológicos , Técnicas de Cultura de Células , RNA Mensageiro/metabolismo , Suplementos Nutricionais , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fosforilação , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo
3.
Neurochem Int ; 175: 105683, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38341034

RESUMO

BACKGROUND: Oxidative stress and neuroinflammation are proven to play critical roles in the pathogenesis of Parkinson's disease (PD). As reported, patients with PD have lower level of STAT4 compared with healthy subjects. However, the biological functions and mechanisms of STAT4 in PD pathogenesis remain uncertain. This study aimed to investigate the roles and related mechanisms of STAT4 in PD development. METHODS: The intraperitoneal injection of MPTP (20 mg/kg) dissolved in physiological saline was performed to mimic PD-like conditions in vivo. MPP + solution was prepared for cell model of PD. Cell viability was measured by CCK-8. Griess reaction was conducted to measure NO concentrations. The mRNA and protein levels were evaluated by RT-qPCR and western blotting. ROS generation was assessed by DCFH-DA. The levels of inflammatory cytokines were measured by ELISA. Cell apoptosis was examined by flow cytometry and western blotting. Moreover, the SH-SY5Y cells were treated with conditioned medium from LPS-stimulated microglia and subjected to CCK-8 assays and ELISA. Mechanistically, CHIP assays and luciferase reporter assays were performed to verify the binding relationship between KISS1 and STAT4. For in vivo analysis, the histological changes of midbrain tissues of mice were determined by hematoxylin and eosin staining. The expression of tyrosine hydroxylase (TH) was detected by immunohistochemistry staining. Iba-1 positive microglial cells in the striatum were assessed by immunofluorescence staining. RESULTS: For in vitro analysis, STAT4 level was downregulated after MPP+ treatment, and STAT4 upregulation inhibited the oxidative damage, inflammation and apoptosis in SH-SY5Y cells. STAT4 bound at +215-228 region of KISS1, and KISS1 upregulation counteracted the protection of STAT4 upregulation against cell damage. Moreover, STAT4 upregulation inhibited cell viability loss and inflammation induced by conditioned medium from LPS-treated microglia, whereas KISS1 upregulation had the opposite effect. For in vivo analysis, the protective effects of STAT4 upregulation against inflammatory response, oxidative stress, dopaminergic neuronal loss and microglia activation were attenuated by KISS1 upregulation. Moreover, the inactivation of MAPK pathway caused by STAT4 upregulation was reversed by KISS1 upregulation, and MAPK inhibition attenuated the MPP+-induced inflammation, oxidative stress and apoptosis in SH-SY5Y cells. CONCLUSION: STAT4 inhibits KISS1 to attenuate the oxidative damage, inflammation and neuronal apoptosis in PD by inactivating the MAPK pathway.


Assuntos
Neuroblastoma , Doença de Parkinson , Animais , Humanos , Camundongos , Apoptose , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Kisspeptinas , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Doença de Parkinson/metabolismo , Sincalida/efeitos adversos , Sincalida/metabolismo , Fator de Transcrição STAT4/metabolismo
4.
J Cutan Pathol ; 51(3): 232-238, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37932931

RESUMO

BACKGROUND: Diagnosis of early mycosis fungoides (eMF) is challenging and often delayed as many of its clinical and histopathologic features may mimic various benign inflammatory dermatoses (BIDs). The products of the thymocyte selection-associated high mobility group box (TOX), twist family BHLH transcription factor 1 (TWIST1), signal transducer and activator of transcription 4 (STAT4), and special AT-rich sequence-binding protein 1 (SATB1) genes function as transcription factors and are involved in the pathogenesis of MF. OBJECTIVES: We aim to determine the diagnostic value of TOX, TWIST1, STAT4, and SATB1 protein expressions in eMF. METHODS: This non-randomized, controlled, prospective analytic study was conducted by performing immunohistochemistry staining with TOX, TWIST1, STAT4, and SATB1 polyclonal antibodies in lesional skin biopsies of eMF and BID patients. Nuclear staining of lymphocytes was compared between eMF and BIDs, and the capacity of these antibodies to predict eMF was determined. RESULTS: Immunostainings with anti-TWIST1 showed an increase in protein expression (p = 0.003) and showed a decrease with anti-SATB1 antibodies in eMF compared to BIDs (p = 0.005) while anti-TOX and anti-STAT4 antibodies did not exhibit significant differences (p = 0.384; p = 0.150). Receiver operating characteristic analysis showed that immunohistochemical evaluations of TWIST1 and SATB1 protein expressions can differentiate eMF (area under the curve [AUC]: 0.728, 95% confidence interval [CI]: 0.605-0.851, p = 0.002; AUC: 0.686, 95% CI: 0.565-0.807, p = 0.013). CONCLUSIONS: TWIST1 and SATB1 are potential diagnostic markers for the histologic diagnosis of eMF.


Assuntos
Proteínas de Ligação à Região de Interação com a Matriz , Micose Fungoide , Neoplasias Cutâneas , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Micose Fungoide/patologia , Proteínas Nucleares/metabolismo , Estudos Prospectivos , Neoplasias Cutâneas/patologia , Fator de Transcrição STAT4/metabolismo , Proteína 1 Relacionada a Twist/metabolismo
5.
J Physiol Biochem ; 80(1): 219-233, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38091230

RESUMO

This study aimed to investigate the role of ERG in the HLX/STAT4/Perforin signaling axis, impacting natural killer (NK) cell cytotoxicity and myocardial infarction (MI) progression. NK cell cytotoxicity was assessed via co-culture and 51Cr release assays. Datasets GSE34198 and GSE97320 identified common differentially expressed genes in MI. NK cell gene expression was analyzed in MI patients and healthy individuals using qRT-PCR and Western blotting. ERG's regulation of HLX and STAT4's regulation of perforin were studied through computational tools (MEM) and ChIP experiments. HLX's influence on STAT4 was explored with the MG132 proteasome inhibitor. Findings were validated in a mouse MI model.ERG, a commonly upregulated gene, was identified in NK cells from MI patients and mice. ERG upregulated HLX, leading to STAT4 proteasomal degradation and reduced Perforin expression. Consequently, NK cell cytotoxicity decreased, promoting MI progression. ERG mediates the HLX/STAT4/Perforin axis to inhibit NK cell cytotoxicity, fostering MI progression. These results provide vital insights into MI's molecular mechanisms.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais , Animais , Humanos , Camundongos , Proteínas de Homeodomínio , Células Matadoras Naturais/metabolismo , Perforina/genética , Perforina/metabolismo , Transdução de Sinais , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo , Fatores de Transcrição/metabolismo , Regulador Transcricional ERG/metabolismo
6.
Arthritis Res Ther ; 25(1): 221, 2023 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-37978415

RESUMO

OBJECTIVES: The induction direction of interferon (IFN)-α in T-cell phenotype and function varies depending on the activation state of the cell and the time of stimulation. To assess the effects of elevated IFN-α on regulatory T cells (Tregs) in systemic lupus erythematosus (SLE) patients, we investigated the differentiation of Th1-like Tregs under in-sequence and out-of-sequence conditions and the reversal effect of activating TIGIT on immune suppression. METHODS: Phenotypes and activation levels of Tregs from SLE patients and healthy controls were analyzed using flow cytometry. In vitro culture conditions based on the sequence of TCR activation and IFN-α stimulation simulated in-sequence or out-of-sequence effects. CD4+T cells and Tregs were cultured under the above conditions with or without TIGIT agonist. Expression of related characteristic markers and phosphorylation levels of AKT, mTOR, and STATs were detected using flow cytometry and ELISA. RESULTS: The frequency of Th1-like Tregs and activation levels of Tregs increased, but TIGIT expression in Tregs decreased in SLE patients. IFN-α promoted the conversation of Tregs to Th1-like Tregs while reducing immunosuppressive function under in-sequence conditions. The STAT4 pathway, but not the STAT1 pathway, was crucial for the IFN-α-mediated in-sequence effects. Reactivation of TIGIT reversed Th1 polarization of Tregs by suppressing AKT/mTOR and STAT4 signaling. CONCLUSIONS: Our findings suggest that IFN-α mediated in-sequence effects on Tregs may be responsible for the expansion of Th1-like Tregs in SLE. TIGIT can restore immune suppression damage in Tregs and represents a potential therapeutic target for SLE.


Assuntos
Lúpus Eritematoso Sistêmico , Proteínas Proto-Oncogênicas c-akt , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T Reguladores , Interferon-alfa/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Receptores Imunológicos/metabolismo , Fator de Transcrição STAT4/metabolismo
7.
Commun Biol ; 6(1): 967, 2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37783748

RESUMO

Neuroinflammation is associated with neurodegenerative diseases, including Alzheimer's and Parkinson's. The cytokine interleukin-12 activates signal transducer and activator of transcription 4 (Stat4), and consumption of a high-fat, high-cholesterol diet (HFD-C) and Stat4 activity are associated with inflammation, atherosclerosis, and a diabetic metabolic phenotype. In studies of in vitro hippocampal slices from control Stat4fl/flLdlr-/- mice fed a HFD-C diabetogenic diet, we show that Schaffer collateral-CA1 synapses exhibited larger reductions in activity-dependent, long-term potentiation (LTP) of synaptic transmission, compared to mice fed a standard diet. Glucose tolerance and insulin sensitivity shifts produced by HFD-C diet were reduced in Stat4ΔLysMLdlr-/- mice compared to Stat4fl/flLdlr-/- controls. Stat4ΔLysMLdlr-/- mice, which lack Stat4 under control of the LysMCre promoter, were resistant to HFD-C induced impairments in LTP. In contrast, Schaffer collateral-CA1 synapses in Stat4ΔLysMLdlr-/- mice fed the HFD-C diet showed larger LTP than control Stat4fl/flLdlr-/- mice. Expression of a number of neuroinflammatory and synaptic plasticity genes was reduced by HFD-C diet in control mice, and less affected by HFD-C diet in Stat4ΔLysMLdlr-/- mice. These data suggest that suppression of Stat4 activation may protect against effects of Western diet on cognition, type 2 diabetes, and reduce risk of Alzheimer's disease and other neurodegenerative disorders associated with neuroinflammation.


Assuntos
Diabetes Mellitus Tipo 2 , Fator de Transcrição STAT4 , Camundongos , Animais , Fator de Transcrição STAT4/metabolismo , Doenças Neuroinflamatórias , Plasticidade Neuronal , Colesterol/metabolismo , Células Mieloides/metabolismo
8.
J Immunol ; 211(10): 1469-1474, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37830760

RESUMO

NK cells represent a cellular component of the mammalian innate immune system, and they mount rapid responses against viral infection, including the secretion of the potent antiviral effector cytokine IFN-γ. Following mouse CMV infection, Bhlhe40 was the most highly induced transcription factor in NK cells among the basic helix-loop-helix family. Bhlhe40 upregulation in NK cells depended upon IL-12 and IL-18 signals, with the promoter of Bhlhe40 enriched for STAT4 and the permissive histone H3K4me3, and with STAT4-deficient NK cells showing an impairment of Bhlhe40 induction and diminished H3K4me3. Transcriptomic and protein analysis of Bhlhe40-deficient NK cells revealed a defect in IFN-γ production during mouse CMV infection, resulting in diminished protective immunity following viral challenge. Finally, we provide evidence that Bhlhe40 directly promotes IFN-γ by binding throughout the Ifng loci in activated NK cells. Thus, our study reveals how STAT4-mediated control of Bhlhe40 drives protective IFN-γ secretion by NK cells during viral infection.


Assuntos
Infecções por Citomegalovirus , Células Matadoras Naturais , Camundongos , Animais , Interferon gama , Citocinas/metabolismo , Interleucina-12/metabolismo , Infecções por Citomegalovirus/metabolismo , Fator de Transcrição STAT4/metabolismo , Mamíferos/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo
9.
Oncol Rep ; 50(1)2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37264954

RESUMO

Ovarian serous carcinoma (OC) is a common cause of mortality among gynecological malignancies. Although tumor­infiltrating CD8 T cells are associated with a favorable prognosis of OC, the underlying mechanisms are not clearly understood. The present study identified the key genes and potential molecular mechanisms associated with CD8 T­cell infiltration in OC. The score of CD8 T cells in The Cancer Genome Atlas dataset (376 samples from patients with OC) was estimated using the quanTIseq and MCP­counter algorithms. Thereafter, a protein­protein interaction network of differentially expressed genes was constructed and the hub genes were identified using cytoHubba in Cytoscape. The results revealed that signal transducer and activator of transcription 4 (STAT4) was strongly correlated with CD8 T­cell infiltration in OC. Furthermore, the prognostic value of STAT4 in OC was verified by Kaplan­Meier curve, and univariate and multivariate analyses. The biological functions of STAT4 were determined by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses, which revealed that STAT4 is closely related to cytokines in OC. Moreover, Spearman correlation analysis suggested that STAT4 was most positively correlated with CC chemokine ligand 5 (CCL5). CCL5 was revealed to be critical for orchestrating T­cell infiltration in tumors. Moreover, immunohistochemistry and reverse transcription­quantitative PCR showed that STAT4, CCL5 and CD8A (a marker for CD8 T cells) were closely related in OC. Moreover, in vitro analysis revealed that STAT4 knockdown led to a decrease in CCL5 expression and CD8 T­cell migration. Taken together, the present study suggested that STAT4 may regulate CD8 T­cell infiltration in OC tissues by inducing CCL5 secretion. Furthermore, STAT4 may be considered a promising prognostic biomarker for OC.


Assuntos
Carcinoma , Neoplasias Ovarianas , Humanos , Feminino , Quimiocinas CC/metabolismo , Ligantes , Prognóstico , Linfócitos T CD8-Positivos/metabolismo , Carcinoma/patologia , Neoplasias Ovarianas/patologia , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo
10.
J Transl Med ; 21(1): 326, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37194066

RESUMO

BACKGROUND: Renal tubulointerstitial fibrosis is the hallmark of various chronic kidney diseases. Symmetric dimethylarginine (SDMA) is an independent cardiovascular risk factor in patients with chronic kidney diseases, which is mostly excreted through renal tubules. However, the effect of SDMA on kidneys in a pathological condition is currently unknown. In this study, we investigated the role of SDMA in renal tubulointerstitial fibrosis and explored its underlying mechanisms. METHODS: Mouse unilateral ureteral obstruction (UUO) and unilateral ischemia-reperfusion injury (UIRI) models were established to study renal tubulointerstitial fibrosis. SDMA was injected into kidneys through ureter retrogradely. TGF-ß stimulated human renal epithelial (HK2) cells were used as an in vitro model and treated with SDMA. Signal transducer and activator of transcription-4 (STAT4) was inhibited by berbamine dihydrochloride or siRNA or overexpressed by plasmids in vitro. Masson staining and Western blotting were performed to evaluate renal fibrosis. Quantitative PCR was performed to validate findings derived from RNA sequencing analysis. RESULTS: We observed that SDMA (from 0.01 to 10 µM) dose-dependently inhibited the expression of pro-fibrotic markers in TGF-ß stimulated HK2 cells. Intrarenal administration of SDMA (2.5 µmol/kg or 25 µmol/kg) dose-dependently attenuated renal fibrosis in UUO kidneys. A significant increase in SDMA concentration (from 19.5 to 117.7 nmol/g, p < 0.001) in mouse kidneys was observed after renal injection which was assessed by LC-MS/MS. We further showed that intrarenal administration of SDMA attenuated renal fibrosis in UIRI induced mouse fibrotic kidneys. Through RNA sequencing analysis, we found that the expression of STAT4 was reduced by SDMA in UUO kidneys, which was further confirmed by quantitative PCR and Western blotting analysis in mouse fibrotic kidneys and renal cells. Inhibition of STAT4 by berbamine dihydrochloride (0.3 mg/ml or 3.3 mg/ml) or siRNA reduced the expression of pro-fibrotic markers in TGF-ß stimulated HK2 cells. Furthermore, blockage of STAT4 attenuated the anti-fibrotic effect of SDMA in TGF-ß stimulated HK2 cells. Conversely, overexpression of STAT4 reversed the anti-fibrotic effect of SDMA in TGF-ß stimulated HK2 cells. CONCLUSION: Taken together, our study indicates that renal SDMA ameliorates renal tubulointerstitial fibrosis through inhibition of STAT4.


Assuntos
Nefropatias , Insuficiência Renal Crônica , Obstrução Ureteral , Humanos , Camundongos , Animais , Cromatografia Líquida , Espectrometria de Massas em Tandem , Nefropatias/complicações , Rim/patologia , Obstrução Ureteral/complicações , Obstrução Ureteral/genética , Obstrução Ureteral/patologia , Insuficiência Renal Crônica/patologia , Fator de Crescimento Transformador beta/metabolismo , Fibrose , RNA Interferente Pequeno , Fator de Crescimento Transformador beta1/metabolismo , Fator de Transcrição STAT4/metabolismo
11.
J Immunol ; 210(11): 1667-1676, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37093664

RESUMO

Effector CD4 T cells are central to the development of autoimmune chronic inflammatory diseases, yet factors that mediate pathogenicity remain ill-defined. Single-nucleotide polymorphisms in the human STAT4 locus are associated with susceptibility to multiple autoimmune disorders, and Stat4 is linked to the pathogenic Th17 gene signature; however, Th17 cells differentiate independently of STAT4. Hence the interplay between STAT4 and CD4 T cell function, especially Th17 cells, during autoimmune disease is unclear. In this article, we demonstrate that CD4 T cell-intrinsic STAT4 expression is essential for the induction of autoimmune CNS inflammation in mice, in part by regulating the migration of CD4 T cells to the inflamed CNS. Moreover, unbiased transcriptional profiling revealed that STAT4 controls the expression of >200 genes in Th17 cells and is important for the upregulation of genes associated with IL-23-stimulated, pathogenic Th17 cells. Importantly, we show that Th17 cells specifically require STAT4 to evoke autoimmune inflammation, highlighting, to our knowledge, a novel function for STAT4 in Th17 pathogenicity.


Assuntos
Linfócitos T CD4-Positivos , Encefalomielite Autoimune Experimental , Humanos , Camundongos , Animais , Células Th17 , Células Th1 , Virulência , Inflamação , Diferenciação Celular , Fator de Transcrição STAT4/metabolismo
12.
J Biol Chem ; 299(5): 104703, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37059181

RESUMO

The conversion of signal transducer and activator of transcription (STAT) proteins from latent to active transcription factors is central to cytokine signaling. Triggered by their signal-induced tyrosine phosphorylation, it is the assembly of a range of cytokine-specific STAT homo- and heterodimers that marks a key step in the transition of hitherto latent proteins to transcription activators. In contrast, the constitutive self-assembly of latent STATs and how it relates to the functioning of activated STATs is understood less well. To provide a more complete picture, we developed a co-localization-based assay and tested all 28 possible combinations of the seven unphosphorylated STAT (U-STAT) proteins in living cells. We identified five U-STAT homodimers-STAT1, STAT3, STAT4, STAT5A, and STAT5B-and two heterodimers-STAT1:STAT2 and STAT5A:STAT5B-and performed semi-quantitative assessments of the forces and characterizations of binding interfaces that support them. One STAT protein-STAT6-was found to be monomeric. This comprehensive analysis of latent STAT self-assembly lays bare considerable structural and functional diversity in the ways that link STAT dimerization before and after activation.


Assuntos
Regulação da Expressão Gênica , Fatores de Transcrição STAT , Transativadores , Citocinas/metabolismo , Fosforilação , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transativadores/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Multimerização Proteica
13.
J Immunol ; 210(9): 1292-1304, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36961447

RESUMO

It is generally accepted that influenza A virus (IAV) infection promotes a Th1-like CD4 T cell response and that this effector program underlies its protective impact. Canonical Th1 polarization requires cytokine-mediated activation of the transcription factors STAT1 and STAT4 that synergize to maximize the induction of the "master regulator" Th1 transcription factor, T-bet. Here, we determine the individual requirements for these transcription factors in directing the Th1 imprint primed by influenza infection in mice by tracking virus-specific wild-type or T-bet-deficient CD4 T cells in which STAT1 or STAT4 is knocked out. We find that STAT1 is required to protect influenza-primed CD4 T cells from NK cell-mediated deletion and for their expression of hallmark Th1 attributes. STAT1 is also required to prevent type I IFN signals from inhibiting the induction of the Th17 master regulator, Rorγt, in Th17-prone T-bet-/- cells responding to IAV. In contrast, STAT4 expression does not appreciably impact the phenotypic or functional attributes of wild-type or T-bet-/- CD4 T cell responses. However, cytokine-mediated STAT4 activation in virus-specific CD4 T cells enhances their Th1 identity in a T-bet-dependent manner, indicating that influenza infection does not promote maximal Th1 induction. Finally, we show that the T-bet-dependent protective capacity of CD4 T cell effectors against IAV is optimized by engaging both STAT1 and STAT4 during Th1 priming, with important implications for vaccine strategies aiming to generate T cell immunity.


Assuntos
Linfócitos T CD4-Positivos , Influenza Humana , Camundongos , Animais , Humanos , Antivirais/metabolismo , Proteínas com Domínio T/metabolismo , Interferon gama/metabolismo , Fatores de Transcrição/metabolismo , Células Th1 , Fator de Transcrição STAT4/metabolismo , Diferenciação Celular , Fator de Transcrição STAT1/metabolismo
14.
J Orthop Res ; 41(4): 747-758, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35880357

RESUMO

Osteoarthritis (OA) is a chronic degenerative bone and joint disease common in middle-aged and elderly people. Currently, there is no satisfactory pharmacological treatment. Eugenol is a phenolic compound that has been shown to exert biological anti-inflammatory, antioxidant, and antiapoptotic effects in multiple systems and organs of the human body. However, its therapeutic effect on OA is unclear. This study examined the effect of eugenol on OA using an anterior cruciate ligament transection (ACLT) model in mice and its related signaling pathways in interleukin-1ß (IL-1ß)-stimulated human chondrocytes. A certain concentration of eugenol inhibited the decrease in cell viability induced by IL-1ß or carbonyl cyanide 3-chlorophenylhydrazone (CCCP). In vitro, eugenol effectively inhibited CCCP-induced chondrocyte apoptosis and mitochondrial membrane potential changes and inhibited the expressions of ADAMTS4 and MMP13 upregulated by IL-1ß. In vivo, ACLT induced destruction of the articular cartilage and subchondral bone of the mouse tibial plateau, while eugenol effectively protected the cartilage and subchondral bone from such damage. At the same time, eugenol reduced the ACLT-induced upregulation of ADAMTS4 and MMP13 and the downregulation of type II collagen (COLII) and aggrecan in the mouse knee cartilage. Eugenol also inhibited the increased expression of cartilage metabolism signaling molecules such as C-telopeptides of COLII (CTX-II) in ACLT-induced mouse serum. Consistent with the specific changes in the messenger RNA chip, eugenol inhibited the phosphorylation of JAK3 and STAT4 induced by IL-1ß. Together, these results suggest eugenol as an effective new drug for the prevention and treatment of OA.


Assuntos
Cartilagem Articular , Osteoartrite , Idoso , Pessoa de Meia-Idade , Humanos , Camundongos , Animais , Condrócitos/metabolismo , Cartilagem Articular/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Eugenol/metabolismo , Eugenol/farmacologia , Eugenol/uso terapêutico , Carbonil Cianeto m-Clorofenil Hidrazona , Osteoartrite/metabolismo , Transdução de Sinais , Interleucina-1beta/metabolismo , Modelos Animais de Doenças , Janus Quinase 3/metabolismo , Fator de Transcrição STAT4/metabolismo
15.
J Neuroinflammation ; 19(1): 253, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36217178

RESUMO

BACKGROUND: The immune system has been implicated in synaptic plasticity, inflammation, and the progression of Alzheimer's disease (AD). However, there were few studies on improving the niche microenvironment of neural stem cells (NSCs) in the brain of AD to promote adult hippocampal neurogenesis (AHN) by regulating the function of non-parenchymal immune cells. METHODS: The lymph nodes of amyloid precursor protein/presenilin 1 (APP/PS1) and 3xTg (APP/PS1/tau) mouse models of AD were treated with photobiomodulation therapy (PBMT) for 10 J/cm2 per day for 1 month (10 min for each day), T lymphocytes isolated from these two AD models were treated with PBMT for 2 J/cm2 (5 min for each time). The NSCs isolated from hippocampus of these two AD models at E14, and the cells were co-cultivated with PBMT-treated T lymphocyte conditioned medium for NSCs differentiation. RESULTS: Our results showed that PBMT treatment could promote AHN and reverse cognitive deficits in AD mouse model. The expression of interferon-γ (IFN-γ) and interleukin-10 (IL-10) was upregulated in the brain of these two AD models after PBMT treated, which was induced by the activation of Janus kinase 2 (JAK2)-mediated signal transducer and activator of transcription 4 (STAT4)/STAT5 signaling pathway in CD4+ T cells. In addition, elevated CD4+ T cell levels and upregulated transforming growth factor-ß1 (TGFß1)/insulin-like growth factors-1 (IGF-1)/brain-derived neurotrophic factor (BDNF) protein expression levels were also detected in the brain. More importantly, co-cultivated the PBMT-treated T lymphocyte conditioned medium with NSCs derived from these two AD models was shown to promote NSCs differentiation, which was reflected in the upregulation of both neuronal class-III ß-tubulin (Tuj1) and postsynaptic density protein 95 (PSD95), but the effects of PBMT was blocked by reactive oxygen species (ROS) scavenger or JAK2 inhibitor. CONCLUSION: Our research suggests that PBMT exerts a beneficial neurogenesis modulatory effect through activating the JAK2/STAT4/STAT5 signaling pathway to promote the expression of IFN-γ/IL-10 in non-parenchymal CD4+ T cells, induction of improvement of brain microenvironmental conditions and alleviation of cognitive deficits in APP/PS1 and 3xTg-AD mouse models.


Assuntos
Doença de Alzheimer , Terapia com Luz de Baixa Intensidade , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Cognição , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Janus Quinase 2/metabolismo , Camundongos , Camundongos Transgênicos , Neurogênese/fisiologia , Presenilina-1/genética , Presenilina-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT4/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Tubulina (Proteína)/metabolismo
16.
Scand J Immunol ; 96(2): e13177, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35484925

RESUMO

Chrysin (CHR) is a flavonoid with extensive pharmacological activity. The molecular formula of CHR is C15 H10 O4 . CHR is reported to have antioxidative, antitumour and antiviral functions. To evaluate its potential function as a vaccine adjuvant, we prepared a melanoma vaccine using a soluble protein extract of B16F10 melanoma cells as antigen and CHR as an adjuvant. The melanoma model was developed after two immunizations, and it was discovered that combining B16F10 soluble protein antigen-mixed CHR vaccine could inhibit tumour growth in the mouse model, and the overall survival rate was higher than that of the B16F10 antigen vaccine alone. In vivo and in vitro experiments were conducted to determine whether CHR functioned as an adjuvant by activating antigen-presenting cells (APCs). We discovered that CHR activated APCs both in vivo and in vitro and may enhance Th1 cell function by activating the IL12-STAT4 signal pathway, thereby enhancing the antitumour response of cytotoxic T lymphocytes (CTLs) in vivo. Next, to verify the critical role of CD8+ T cells in suppressing melanoma development, we transplanted CD8+ T cells from immunized mice to B16F10 tumour-bearing mice and discovered that the survival rate of tumour-bearing mice was significantly prolonged. In summary, our experimental results indicate that CHR can be used as a potential adjuvant to enhance antigen immunogenicity, inhibit B16F10 tumour growth in mice and improve tumour immune response.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer , Flavonoides , Melanoma Experimental , Animais , Linfócitos T CD8-Positivos , Modelos Animais de Doenças , Flavonoides/farmacologia , Imunidade , Interleucina-12/metabolismo , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT4/metabolismo , Transdução de Sinais
17.
Brain Res Bull ; 184: 68-75, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35367598

RESUMO

BACKGROUND: Heart failure (HF) and depression are common disorders that markedly compromise quality of life and impose a great financial burden on the society. Although increasing evidence has supported the closely linkage between the two disorders, the comorbidity mechanisms remain to be fully illuminated. We performed a bioinformatics network analysis to understand potential diagnostic biomarkers and therapeutic targets for HF comorbided with depression. METHODS: We downloaded the datasets of HF and depression from the Gene Expression Omnibus (GEO) database and constructed co-expression networks by Weighted Gene Co-Expression Network Analysis (WGCNA) to identify key modules. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed on the common genes existing in the HF and depression related modules. Then, we employed the STRING database to construct the protein-protein interaction (PPI) network and detected the hub genes in the network. Finally, we validated the expression difference of hub genes from additional datasets of HF and depression. RESULTS: Functional enrichment analysis indicated that platelet activation, chemokine signaling and focal adhesion were probably involved in HF comorbided with depression. PPI network construction indicated that HF comorbided with depression is likely related to 5 hub genes, including STAT4, CD83, CX3CR1, COL1A2, and SH2D1B. In validated datasets, STAT4 and COL1A2 were especially involved in the comorbidity of HF and depression. CONCLUSION: Our work indicated a total of 5 hub genes including STAT4, CD83, CX3CR1, COL1A2, and SH2D1B, in which STAT4 and COL1A2 especially underlie the comorbidity mechanisms of HF and depression. These shared pathways might provide new targets for further mechanistic studies of the pathogenesis and treatment of HF and depression.


Assuntos
Redes Reguladoras de Genes , Insuficiência Cardíaca , Biomarcadores/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Biologia Computacional , Depressão/diagnóstico , Depressão/genética , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/genética , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/epidemiologia , Insuficiência Cardíaca/genética , Humanos , Qualidade de Vida , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo
18.
Acta Histochem ; 124(4): 151872, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35367814

RESUMO

OBJECTIVE: This study aimed to investigate the protective role of the signal transducer and activator of transcription 4 (STAT4) in diabetic cardiomyopathy. MATERIALS AND METHODS: Male Sprague-Dawley (SD) rats (6-8 weeks old) were purchased from the Experimental Animal Center of Zhengzhou University. The rats were randomly divided into the control and diabetic cardiomyopathy groups. Rat models of diabetic cardiomyopathy were established by a high-sugar and high-fat diet combined with a peritoneal injection of streptozocin. Pathological changes in the heart were visualized using Hematoxylin-eosin (HE) staining and Masson's staining. Moreover, cell apoptosis was detected using terminal deoxyribonucleotidyl transferase (TdT)-mediated biotin-16-dUTP nick-end labeling (TUNEL) staining and Annexin V apoptosis detection kit. Furthermore, H9C2 cells were transfected with lentivirus overexpressing STAT4 and treated with high glucose. The CCK-8 assay was performed to determine cell viability. Finally, Western blotting was used to determine the expression of STAT4, Bax, and Bcl-2. RESULTS: The myocardial tissue of the diabetic cardiomyopathy models showed hypertrophy, myocardial fibrosis and collagen deposition. Furthermore, TUNEL staining showed increased apoptosis and decreased expression of STAT4 in the myocardial cells. Moreover, the myocardial tissues of the DCM models showed increased expression of Bax/Bcl-2 and a high percentage of Annexin V positive cells. The H9C2 cells showed decreased expression of STAT4 following high glucose treatment. However, the H9C2 cells overexpressing STAT4 showed decreased expression of Bax/Bcl-2 and reduced percentage of Annexin V positive cells. CONCLUSION: The DCM group had decreased myocardial expression of STAT4. Furthermore, overexpression of STAT4 was shown to reduce high glucose-induced apoptosis.


Assuntos
Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas , Animais , Anexina A5/efeitos adversos , Apoptose , Diabetes Mellitus Experimental/patologia , Cardiomiopatias Diabéticas/patologia , Glucose , Humanos , Masculino , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT4/metabolismo , Proteína X Associada a bcl-2
19.
Cell Death Dis ; 13(2): 130, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35136014

RESUMO

Although there are many studies on the relationship between genetic polymorphisms and the incidence of diseases, mechanisms are rarely known. We report the mechanism by which signal transducer and activator of transcription 4 (stat4) rs7574865 promotes the occurrence and progression of hepatocellular carcinoma (HCC). We found that the GG genotype at stat4 rs7574865 was a risk genotype, and STAT4 levels in serum and peritumoral tissue from HCC patients with the GG genotype were significantly higher than those found in TT or TG carriers. Furthermore, HCC patients with the GG genotype or elevated STAT4 levels had poor prognoses. In vitro experiments demonstrated that STAT4 silencing promoted apoptosis and inhibited the invasion and migration of HepG2 and L02 cells. Proteomic analysis of HCC peritumors identified 273 proteins related to STAT4, of which CYP2E1 activity and FGL2 content exhibited the highest positive correlation. The relationship between CYP2E1 and FGL2 was also confirmed in cyp2e1-/- mice and in CYP2E1 inhibitor-treated mice. In conclusion, this study elucidates the mechanism by which the stat4 rs7574865 polymorphism promotes the occurrence and progression of HCC via the Stat4/CYP2E1/FGL2 pathway.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Fator de Transcrição STAT4 , Animais , Carcinoma Hepatocelular/epidemiologia , Carcinoma Hepatocelular/genética , Estudos de Casos e Controles , Citocromo P-450 CYP2E1/genética , Citocromo P-450 CYP2E1/metabolismo , Fibrinogênio/metabolismo , Predisposição Genética para Doença , Genótipo , Humanos , Neoplasias Hepáticas/epidemiologia , Neoplasias Hepáticas/genética , Camundongos , Polimorfismo de Nucleotídeo Único , Proteômica , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo
20.
J Cell Mol Med ; 26(8): 2363-2376, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35224833

RESUMO

Bladder cancer (BLCA) is a common genitourinary cancer in patients, and tumour angiogenesis is indispensable for its occurrence and development. However, the indepth mechanism of tumour angiogenesis in BLCA remains elusive. According to recent studies, the tight junction protein family member occludin (OCLN) is expressed at high levels in BLCA tissues and correlates with a poor prognosis. Downregulation of OCLN inhibits tumour angiogenesis in BLCA cells and murine xenografts, whereas OCLN overexpression exerts the opposite effect. Mechanistically, the RT-qPCR analysis and Western blotting results showed that OCLN increased interleukin-8 (IL8) and p-signal transducer and activator of transcription 3 (STAT3) levels to promote BLCA angiogenesis. RNA sequencing analysis and dual-luciferase reporter assays indicated that OCLN regulated IL8 transcriptional activity via the transcription factor STAT4. In summary, our results provide new perspectives on OCLN, as this protein participates in the development of BLCA angiogenesis by activating the IL8/STAT3 pathway via STAT4 and may serve as a novel and unique therapeutic target.


Assuntos
Interleucina-8 , Ocludina , Fator de Transcrição STAT4 , Neoplasias da Bexiga Urinária , Animais , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Camundongos , Neovascularização Patológica/genética , Ocludina/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo , Neoplasias da Bexiga Urinária/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...