Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 910
Filtrar
1.
FASEB J ; 36(4): e22111, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35230716

RESUMEN

Epidermal tissues play vital roles in maintaining homeostasis and preventing the dysregulation of the cutaneous barrier. Sphingomyelin (SM), a sphingolipid synthesized by sphingomyelin synthase (SMS) 1 and 2, is involved in signal transduction via modulation of lipid-raft functions. Though the implications of SMS on inflammatory diseases have been reported, its role in dermatitis has not been clarified. In this study, we investigated the role of SM in the cutaneous barrier using a dermatitis model established by employing Sgms1 and 2 deficient mice. SM deficiency impaired the cutaneous inflammation and upregulated signal transducer and activator of transcription 3 (STAT3) phosphorylation in epithelial tissues. Furthermore, using mouse embryonic fibroblast cells, the sensitivity of STAT3 to Interleukin-6 stimulation was increased in Sgms-deficient cells. Using tofacitinib, a clinical JAK inhibitor, the study showed that SM deficiency might participate in STAT3 phosphorylation via JAK activation. Overall, these results demonstrate that SM is essential for maintaining the cutaneous barrier via the STAT3 pathway, suggesting SM could be a potential therapeutic target for dermatitis treatment.


Asunto(s)
Factor de Transcripción STAT3/fisiología , Piel/metabolismo , Esfingomielinas/fisiología , Animales , Células Cultivadas , Dermatitis/tratamiento farmacológico , Dermatitis/etiología , Humanos , Ratones , Ratones Endogámicos C57BL , Fosforilación , Transducción de Señal/fisiología , Esfingomielinas/uso terapéutico , Transferasas (Grupos de Otros Fosfatos Sustitutos)/fisiología
2.
J Cancer Res Clin Oncol ; 148(2): 309-319, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34661759

RESUMEN

BACKGROUND: Histone demethylase UTX has been reported to participate in the occurrence and development of many cancers in tissue-specific manners. However, the role of UTX in non-small cell lung cancer (NSCLC) and exactly what regulates the expression of UTX remains unclear. Here, we analyzed the role of UTX in NSCLC in association with the widely recognized tumor driver epidermal growth factor receptor (EGFR). METHODS: UTX levels in clinical samples were detected by immunohistochemistry staining, western blotting and real-time quantitative PCR. The expression of UTX in tumor tissue was correlated with the phosphorylation of EGFR. Cell proliferation and migration were evaluated by MTT and wound-healing assays. The impact of EGFR and its downstream pathways on UTX was explored with corresponding inhibitors, and examined by western blotting and real-time quantitative PCR. RESULTS: In this study, we found that the expression of UTX in cancer tissues of patients with NSCLC was significantly higher than that in paracancerous tissues, and positively associated with EGFR phosphorylation levels. In addition, in NSCLC cell lines, UTX can promote proliferation and migration, while inhibition of its enzyme activity suppressed cell growth. Moreover, UTX expression was significantly upregulated when EGFR signaling pathway was activated, and vice versa when EGFR pathway was inhibited by tyrosine kinase inhibitor. Further mechanistic studies suggested that the activation of EGFR activated its downstream JAK/STAT3 signaling pathway and promoted STAT3 phosphorylation; the phosphorylated STAT3 transcriptionally promoted the levels of UTX. CONCLUSIONS: These results suggest an "EGFR-STAT3-UTX" axis that plays an oncogenic role in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Histona Demetilasas/genética , Neoplasias Pulmonares/genética , Factor de Transcripción STAT3/fisiología , Células A549 , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Células Cultivadas , Receptores ErbB/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Fosforilación , Transducción de Señal/genética , Regulación hacia Arriba/genética
3.
Front Immunol ; 12: 663177, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867936

RESUMEN

Dominant-negative mutations associated with signal transducer and activator of transcription 3 (STAT3) signaling, which controls epithelial proliferation in various tissues, lead to atopic dermatitis in hyper IgE syndrome. This dermatitis is thought to be attributed to defects in STAT3 signaling in type 17 helper T cell specification. However, the role of STAT3 signaling in skin epithelial cells remains unclear. We found that STAT3 signaling in keratinocytes is required to maintain skin homeostasis by negatively controlling the expression of hair follicle-specific keratin genes. These expression patterns correlated with the onset of dermatitis, which was observed in specific pathogen-free conditions but not in germ-free conditions, suggesting the involvement of Toll-like receptor-mediated inflammatory responses. Thus, our study suggests that STAT3-dependent gene expression in keratinocytes plays a critical role in maintaining the homeostasis of skin, which is constantly exposed to microorganisms.


Asunto(s)
Folículo Piloso/fisiología , Factor de Transcripción STAT3/fisiología , Animales , Dermatitis Atópica/etiología , Dermatitis Atópica/genética , Dermatitis Atópica/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Folículo Piloso/inmunología , Homeostasis , Humanos , Queratinocitos/inmunología , Queratinocitos/fisiología , Queratinas/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal , Piel/inmunología , Piel/microbiología , Fenómenos Fisiológicos de la Piel , Células Th17/inmunología
4.
Front Immunol ; 12: 763065, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868008

RESUMEN

Neuropsychiatric lupus (NPSLE), the nervous system presentation of systemic lupus erythematosus (SLE), remains challenging to treat due to its unclear pathogenesis and lack of available targeted therapies. A potential contributor to disease progression is brain tertiary lymphoid structures (TLS); these ectopic lymphoid follicles that can develop tissue-targeted antibodies have recently been described in the MRL/lpr lupus mouse strain, a classic model for studying NPSLE. The brains of MRL/lpr mice show a significant increase of CXCL13, an important chemokine in lymphoid follicle formation and retention that may also play a role in the disease progression of NPSLE. The aim of the present study was to inhibit CXCL13 and examine the effect of this intervention on lymphoid formation and the development of neurobehavioral manifestations in lupus mice. Female MRL/lpr mice were injected with an anti-CXCL13 antibody, an IgG1 isotype-matched antibody, or PBS either three times a week for 12 weeks intraperitoneally (IP) starting at 6-8 weeks of age, or continuously intracerebroventricularly (ICV) with an osmotic pump over a two-week period starting at 15 weeks of age. Cognitive dysfunction and depression-like behavior were assessed at the end of treatment. When treatment was delivered IP, anti-CXCL13 treated mice showed significant improvement in cognitive function when compared to control treated mice. Depression-like behavior was attenuated as well. Furthermore, mice that received anti-CXCL13 by the ICV route showed similar beneficial effects. However, the extent of lymphocyte infiltration into the brain and the general composition of the aggregates were not substantively changed by anti-CXCL13 irrespective of the mode of administration. Nevertheless, analysis of brain gene expression in anti-CXCL13 treated mice showed significant differences in key immunological and neuro-inflammatory pathways that most likely explained the improvement in the behavioral phenotype. Our results indicate that CXCL13 affects the behavioral manifestations in the MRL/lpr strain and is important to the pathogenesis of murine NPSLE, suggesting it as a potential therapeutic target.


Asunto(s)
Quimiocina CXCL13/antagonistas & inhibidores , Vasculitis por Lupus del Sistema Nervioso Central/tratamiento farmacológico , Animales , Encéfalo/patología , Quimiocina CXCL13/fisiología , Cognición/efectos de los fármacos , Femenino , Inyecciones Intraventriculares , Vasculitis por Lupus del Sistema Nervioso Central/patología , Vasculitis por Lupus del Sistema Nervioso Central/psicología , Ratones , Ratones Endogámicos MRL lpr , Factor de Transcripción STAT3/fisiología
5.
Sci Rep ; 11(1): 21649, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34737337

RESUMEN

The E2 component of the mitochondrial pyruvate dehydrogenase complex (PDC) is the key autoantigen in primary biliary cholangitis (PBC) and STAT3 is an inflammatory modulator that participates in the pathogenesis of many liver diseases. This study investigated whether PDC-E2 interacts with STAT3 in human cholangiocytes (NHC) and hepatocytes (Hep-G2) under cholestatic conditions induced by glyco-chenodeoxycholic acid (GCDC). GCDC induced PDC-E2 expression in the cytoplasmic and nuclear fraction of NHC, whereas in Hep-G2 cells PDC-E2 expression was induced only in the cytoplasmic fraction. GCDC-treatment stimulated phosphorylation of STAT3 in the cytoplasmic fraction of NHC. siRNA-mediated gene silencing of PDC-E2 reduced the expression of pY-STAT3 in NHC but not in HepG2 cells. Immunoprecipitation and a proximity ligation assay clearly demonstrated that GCDC enhanced pY-STAT3 binding to PDC-E2 in the nuclear and cytoplasmic fraction of NHC cells. Staining with Mitotracker revealed mitochondrial co-localization of PDC-E2/pS-STAT3 complexes in NHC and Hep-G2 cells. In cirrhotic PBC livers the higher expression of both PDC-E2 and pY-STAT3 was observed. The immunoblot analysis demonstrated the occurrence of double bands of PDC-E2 protein in control livers, which was associated with a lower expression of pY-STAT3. Our data indicate the interaction between PDC-E2 and phosphorylated STAT3 under cholestatic conditions, which may play a role in the development of PBC.


Asunto(s)
Autoantígenos/metabolismo , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/metabolismo , Proteínas Mitocondriales/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Factor de Transcripción STAT3/metabolismo , Autoantígenos/fisiología , Conductos Biliares/patología , Línea Celular , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/fisiología , Células Epiteliales/metabolismo , Ácido Glicoquenodesoxicólico/farmacología , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Immunoblotting/métodos , Inmunoprecipitación/métodos , Hígado/patología , Cirrosis Hepática Biliar/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/fisiología , Complejo Piruvato Deshidrogenasa/fisiología , Factor de Transcripción STAT3/fisiología
6.
J Neuroimmunol ; 361: 577745, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34695768

RESUMEN

The symptoms of Fragile X syndrome (FXS) are driven in part by abnormal glial-mediated function. FXS astrocytes release elevated levels of immune-related factors interleukin-6 (IL-6) and tenascin C (TNC), and also demonstrate increased purinergic signaling, a pathway linked to signaling factor release. Here, in cortical astrocytes from the Fmr1 knockout (KO) FXS mouse model, purinergic agonism enhanced TNC secretion and STAT3 phosphorylation, two processes linked to elevated IL-6 secretion in FXS, while STAT3 knockdown and TLR4 antagonism normalized Fmr1 KO IL-6 release. We therefore suggest that purinergic signaling and immune regulatory pathways converge to drive FXS cortical pro-inflammatory responses.


Asunto(s)
Astrocitos/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Interleucina-6/metabolismo , Agonistas del Receptor Purinérgico P2Y/farmacología , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología , Tenascina/metabolismo , Uridina Trifosfato/farmacología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Masculino , Ratones , Ratones Noqueados , Fosforilación , Fosfotirosina/metabolismo , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Receptores de Interleucina-6/biosíntesis , Receptores de Interleucina-6/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Transducción de Señal/inmunología , Sulfonamidas/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo
7.
Mol Neurodegener ; 16(1): 64, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526055

RESUMEN

BACKGROUND: Human genetic association studies point to immune response and lipid metabolism, in addition to amyloid-beta (Aß) and tau, as major pathways in Alzheimer's disease (AD) etiology. Accumulating evidence suggests that chronic neuroinflammation, mainly mediated by microglia and astrocytes, plays a causative role in neurodegeneration in AD. Our group and others have reported early and dramatic losses of brain sulfatide in AD cases and animal models that are mediated by ApoE in an isoform-dependent manner and accelerated by Aß accumulation. To date, it remains unclear if changes in specific brain lipids are sufficient to drive AD-related pathology. METHODS: To study the consequences of CNS sulfatide deficiency and gain insights into the underlying mechanisms, we developed a novel mouse model of adult-onset myelin sulfatide deficiency, i.e., tamoxifen-inducible myelinating glia-specific cerebroside sulfotransferase (CST) conditional knockout mice (CSTfl/fl/Plp1-CreERT), took advantage of constitutive CST knockout mice (CST-/-), and generated CST/ApoE double knockout mice (CST-/-/ApoE-/-), and assessed these mice using a broad range of methodologies including lipidomics, RNA profiling, behavioral testing, PLX3397-mediated microglia depletion, mass spectrometry (MS) imaging, immunofluorescence, electron microscopy, and Western blot. RESULTS: We found that mild central nervous system (CNS) sulfatide losses within myelinating cells are sufficient to activate disease-associated microglia and astrocytes, and to increase the expression of AD risk genes (e.g., Apoe, Trem2, Cd33, and Mmp12), as well as previously established causal regulators of the immune/microglia network in late-onset AD (e.g., Tyrobp, Dock, and Fcerg1), leading to chronic AD-like neuroinflammation and mild cognitive impairment. Notably, neuroinflammation and mild cognitive impairment showed gender differences, being more pronounced in females than males. Subsequent mechanistic studies demonstrated that although CNS sulfatide losses led to ApoE upregulation, genetically-induced myelin sulfatide deficiency led to neuroinflammation independently of ApoE. These results, together with our previous studies (sulfatide deficiency in the context of AD is mediated by ApoE and accelerated by Aß accumulation) placed both Aß and ApoE upstream of sulfatide deficiency-induced neuroinflammation, and suggested a positive feedback loop where sulfatide losses may be amplified by increased ApoE expression. We also demonstrated that CNS sulfatide deficiency-induced astrogliosis and ApoE upregulation are not secondary to microgliosis, and that astrogliosis and microgliosis seem to be driven by activation of STAT3 and PU.1/Spi1 transcription factors, respectively. CONCLUSION: Our results strongly suggest that sulfatide deficiency is an important contributor and driver of neuroinflammation and mild cognitive impairment in AD pathology.


Asunto(s)
Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Trastornos de la Memoria/metabolismo , Vaina de Mielina/química , Enfermedades Neuroinflamatorias/metabolismo , Sulfoglicoesfingolípidos/metabolismo , Edad de Inicio , Enfermedad de Alzheimer/etiología , Aminopiridinas/toxicidad , Animales , Apolipoproteínas E/metabolismo , Química Encefálica , Sistema Nervioso Central/metabolismo , Disfunción Cognitiva/etiología , Perfilación de la Expresión Génica , Gliosis/metabolismo , Humanos , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Prueba del Laberinto Acuático de Morris , Neuroglía/enzimología , Neuroglía/fisiología , Enfermedades Neuroinflamatorias/etiología , Prueba de Campo Abierto , Proteínas Proto-Oncogénicas/fisiología , Pirroles/toxicidad , Factor de Transcripción STAT3/fisiología , Sulfoglicoesfingolípidos/análisis , Sulfotransferasas/deficiencia , Transactivadores/fisiología
8.
J BUON ; 26(4): 1635-1641, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34565029

RESUMEN

PURPOSE: The purpose of this study was to analyze the function of curcumin to suppress the proliferative and invasive abilities of papillary thyroid carcinoma (PTC) through inhibiting the JAK2/STAT3 pathway. METHODS: After treatment of different doses of curcumin in TPC-1 and SW1736 cells, changes in viability, clonality, cell cycle, apoptosis, wound healing and invasion were determined. Western blot analyses were performed to detect protein levels of apoptosis-associated genes, JAK2 and STAT3 in TPC-1 and SW1736 cells treated with different doses of curcumin. RESULTS: Curcumin treatment dose-dependently reduced viability, clonality and metastatic ability in TPC-1 and SW1736 cells. After treatment of 10 µM or 20 µM curcumin, PTC cells were blocked in G2/M phase, and their apoptotic rate increased. Curcumin treatment downregulated Bcl-2 and upregulated Bax in PTC cells. In addition, curcumin treatment downregulated p-JAK2 and p-STAT3 in TPC-1 and SW1736 cells. CONCLUSIONS: Curcumin treatment blocks PTC cells to proliferate and invade via inhibiting the JAK2/STAT3 pathway.


Asunto(s)
Curcumina/farmacología , Curcumina/uso terapéutico , Janus Quinasa 2/efectos de los fármacos , Janus Quinasa 2/fisiología , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/fisiología , Cáncer Papilar Tiroideo/tratamiento farmacológico , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/patología , Proliferación Celular/efectos de los fármacos , Humanos , Invasividad Neoplásica , Células Tumorales Cultivadas
9.
Front Immunol ; 12: 710977, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566964

RESUMEN

Growth differentiation factor 15 (GDF15) is involved in the occurrence and development of many diseases, and there are few studies on its relationship with sepsis. This article aims to explore the clinical value of GDF15 in sepsis and to preliminarily explore its prospective regulatory effect on macrophage inflammation and its functions. We recruited 320 subjects (132 cases in sepsis group, 93 cases in nonsepsis group, and 95 cases in control group), then detected the serum GDF15 levels and laboratory indicators, and further investigated the correlation between GDF15 and laboratory indicators, and also analyzed the clinical value of GDF15 in sepsis diagnosis, severity assessment, and prognosis. In vitro, we used LPS to stimulate THP-1 and RAW264.7 cells to establish the inflammatory model, and detected the expression of GDF15 in the culture medium and cells under the inflammatory state. After that, we added GDF15 recombinant protein (rGDF15) pretreatment to explore its prospective regulatory effect on macrophage inflammation and its functions. The results showed that the serum GDF15 levels were significantly increased in the sepsis group, which was correlated with laboratory indexes of organ damage, coagulation indexes, inflammatory factors, and SOFA score. GDF15 also has a high AUC in the diagnosis of sepsis, which can be further improved by combining with other indicators. The dynamic monitoring of GDF15 levels can play an important role in the judgment and prognosis of sepsis. In the inflammatory state, the expression of intracellular and extracellular GDF15 increased. GDF15 can reduce the levels of cytokines, inhibit M1 polarization induced by LPS, and promote M2 polarization. Moreover, GDF15 also enhances the phagocytosis and bactericidal function of macrophages. Finally, we observed a decreased level of the phosphorylation of JAK1/STAT3 signaling pathway and the nuclear translocation of NF-κB p65 with the pretreatment of rGDF15. In summary, our study found that GDF15 has good clinical application value in sepsis and plays a protective role in the development of sepsis by regulating the functions of macrophages and inhibiting the activation of JAK1/STAT3 pathway and nuclear translocation of NF-κB p65.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/fisiología , Sepsis/etiología , Adulto , Anciano , Animales , Femenino , Factor 15 de Diferenciación de Crecimiento/sangre , Humanos , Janus Quinasa 1/fisiología , Macrófagos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Pronóstico , Células RAW 264.7 , Factor de Transcripción STAT3/fisiología , Sepsis/sangre , Sepsis/diagnóstico , Sepsis/mortalidad , Índice de Severidad de la Enfermedad , Choque Séptico/diagnóstico , Transducción de Señal , Células THP-1 , Factor de Transcripción ReIA/metabolismo
10.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34507993

RESUMEN

Interleukin 6 (IL-6) is known to regulate the CD4 T cell function by inducing gene expression of a number of cytokines through activation of Stat3 transcription factor. Here, we reveal that IL-6 strengthens the mechanics of CD4 T cells. The presence of IL-6 during activation of mouse and human CD4 T cells enhances their motility (random walk and exploratory spread), resulting in an increase in travel distance and higher velocity. This is an intrinsic effect of IL-6 on CD4 T-cell fitness that involves an increase in mitochondrial Ca2+ Although Stat3 transcriptional activity is dispensable for this process, IL-6 uses mitochondrial Stat3 to enhance mitochondrial Ca2+-mediated motility of CD4 T cells. Thus, through a noncanonical pathway, IL-6 can improve competitive fitness of CD4 T cells by facilitating cell motility. These results could lead to alternative therapeutic strategies for inflammatory diseases in which IL-6 plays a pathogenic role.


Asunto(s)
Movimiento Celular/fisiología , Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Citocinas/metabolismo , Femenino , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Factor de Transcripción STAT3/fisiología , Transducción de Señal/efectos de los fármacos
11.
Front Immunol ; 12: 675542, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394075

RESUMEN

Autoreactive T cells play a crucial role in the pathogenesis of systemic lupus erythematosus (SLE). TGF-ß type I receptor (TGFßRI) is pivotal in determining T cell activation. Here, we showed that TGFßRI expression in naïve CD4+ T cells was decreased in SLE patients, especially in those with high disease activity. Moreover, IL-6 was found to downregulate TGFßRI expression through JAK/STAT3 pathway in SLE patients. In vitro, the JAK inhibitor tofacitinib inhibited SLE T cell activating by upregulating TGFßRI expression in a dose-dependent manner. In MRL/lpr mice, tofacitinib treatment ameliorated the clinical indicators and lupus nephritis, as evidenced by reduced plasma anti-dsDNA antibody levels, decreased proteinuria, and lower renal histopathological score. Consistently, tofacitinib enhanced TGFßRI expression and inhibited T cell activation in vivo. TGFßRI inhibitor SB431542 reversed the effects of tofacitinib on T cell activation. Thus, our results have indicated that tofacitinib can suppress T cell activation by upregulating TGFßRI expression, which provides a possible molecular mechanism underlying clinical efficacy of tofacitinib in treating SLE patients.


Asunto(s)
Lupus Eritematoso Sistémico/tratamiento farmacológico , Activación de Linfocitos/efectos de los fármacos , Piperidinas/farmacología , Pirimidinas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/fisiología , Linfocitos T/efectos de los fármacos , Adulto , Animales , Femenino , Humanos , Interleucina-6/fisiología , Quinasas Janus/fisiología , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Persona de Mediana Edad , Piperidinas/uso terapéutico , Pirimidinas/uso terapéutico , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Factor de Transcripción STAT3/fisiología , Linfocitos T/inmunología , Adulto Joven
12.
Mol Hum Reprod ; 27(8)2021 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-34240166

RESUMEN

Recurrent spontaneous abortion (RSA) is a common complication of early pregnancy. Dendritic cells (DCs) are thought to confer fetal-maternal immunotolerance and play a crucial role in ensuring a successful pregnancy. A decrease of plasmacytoid dendritic cells (pDCs) was found to be involved in RSA, but the underlying mechanisms of decreased pDC in RSA remain unclear. MicroRNAs (miRNAs) play critical roles in RSA as well as the development, differentiation and functional regulation of pDCs; however, the regulatory effect of miRNAs on pDC in RSA has not been fully investigated. Here we demonstrated that both the proportion of pDC and signal transducer and activator of transcription (STAT3)/transcription factor 4 (Tcf4/E2-2) expression decreased in the peripheral blood mononuclear cells and decidua of patients with RSA compared to those with normal pregnancy (NP), and there was a significantly positive correlation between pDC and STAT3 mRNA. MiRNA microarray assay and quantitative reverse transcription PCR results showed that miR-6875-5p expression was markedly increased in women with RSA and negatively correlated with mRNA expression level of STAT3. Up-regulated miR-6875-5p could sensitively discriminate patients with RSA from NP subjects. Overexpression of miR-6875-5p significantly down-regulated the mRNA expression of STAT3 and E2-2 as well as the protein and phosphorylation level of STAT3, while miR-6875-5p knockdown showed opposite results. Dual luciferase reporter verified that miR-6875-5p regulated STAT3 expression by directly binding to its 3'untranslated region. Overall, our results suggested that increased miR-6875-5p is involved in RSA by decreasing the differentiation of pDCs via inhibition of the STAT3/E2-2 signaling pathway. miR-6875-5p may be explored as a promising diagnostic marker and therapeutic target for RSA.


Asunto(s)
Aborto Habitual/inmunología , Células Dendríticas/citología , MicroARNs/metabolismo , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología , Proteína 2 Similar al Factor de Transcripción 7/fisiología , Regiones no Traducidas 3'/genética , Aborto Habitual/genética , Adulto , Diferenciación Celular , Decidua/metabolismo , Células Dendríticas/metabolismo , Femenino , Genes Reporteros , Células HEK293 , Humanos , Leucocitos Mononucleares/metabolismo , Embarazo
13.
Front Immunol ; 12: 675909, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34113349

RESUMEN

The lipopolysaccharides (LPSs) of Rhodobacter are reported to be TLR4 antagonists. Accordingly, the extract of Rhodobacter azotoformans (RAP99) is used as a health supplement for humans and animals in Japan to regulate immune responses in vivo. We previously analyzed the LPS structure of RAP99 (RAP99-LPS) and found it is different from that of E. coli-LPS but similar to lipid A from Rhodobacter sphaeroides (RSLA), a known antagonist of TLR4, with both having three C14 fatty acyl groups, two C10 fatty acyl groups, and two phosphates. Here we show that RAP99-LPS has an immune stimulatory activity and acts as a TLR4 agonist. Pretreatment of RAP99-LPS suppressed E. coli-LPS-mediated weight loss, suggesting it is an antagonist against E. coli-LPS like other LPS isolated from Rhodobacter. However, injections of RAP99-LPS caused splenomegaly and increased immune cell numbers in C57BL/6 mice but not in C3H/HeJ mice, suggesting that RAP99-LPS stimulates immune cells via TLR4. Consistently, RAP99-LPS suppressed the lung metastasis of B16F1 tumor cells and enhanced the expression of TLR3-mediated chemokines. These results suggest that RAP99-LPS is a TLR4 agonist that enhances the activation status of the immune system to promote anti-viral and anti-tumor activity in vivo.


Asunto(s)
Quimiocinas/genética , Lipopolisacáridos/farmacología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Rhodobacter/química , Receptor Toll-Like 3/fisiología , Receptor Toll-Like 4/agonistas , Animales , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Factor de Transcripción STAT3/fisiología
14.
J Zhejiang Univ Sci B ; 22(6): 492-503, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128372

RESUMEN

Dysregulated crosstalk between different signaling pathways contributes to tumor development, including resistance to cancer therapy. In the present study, we found that the mitogen-activated extracellular signal-regulated kinase (MEK) inhibitor trametinib failed to suppress the proliferation of PANC-1 and MGC803 cells by activating the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway, while the JAK2 inhibitor fedratinib failed to inhibit the growth of the PANC-1 cells upon stimulation of extracellular signal-regulated kinase (ERK) signaling. In particular, the most prominent enhancement of the anti-proliferative effect resulted from the concurrent blockage of the JAK2/STAT3 and ERK signaling pathways. Furthermore, the combination of the two inhibitors resulted in a reduced tumor burden in mice. Our evidence suggests novel crosstalk between JAK2/STAT3 and ERK signaling in gastric cancer (GC) and pancreatic ductal adenocarcinoma (PDAC) cells and provides a therapeutic strategy to overcome potential resistance in gastrointestinal cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Neoplasias Gastrointestinales/tratamiento farmacológico , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Neoplasias Gastrointestinales/patología , Humanos , Janus Quinasa 2/fisiología , Ratones , Ratones Endogámicos BALB C , Piridonas/uso terapéutico , Pirimidinonas/uso terapéutico , Pirrolidinas/uso terapéutico , Factor de Transcripción STAT3/fisiología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/uso terapéutico , Microambiente Tumoral
15.
Can J Physiol Pharmacol ; 99(9): 847-856, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34161158

RESUMEN

This study explored the potential effects of mild hyperhomocysteinemia (HHcy) on the blood-brain barrier (BBB) and neuroinflammation. Seven-week-old male wild-type C57BL/6 mice were fed normal mouse chow (the control group) or a methionine-enriched diet (the HHcy group) for 14 weeks. Mice in the HHcy group exhibited a slight increase in serum Hcy levels (13.56 ± 0.61 µmol/L). Activation of the ERK signaling pathway, up-regulation of matrix metalloproteinase-9 (MMP-9), and degradation of tight junction proteins (occludin and claudin-5) were observed in both the cerebral cortex and hippocampus of mice with mild HHcy. However, microglia were not activated and the levels of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) were not changed in either the cerebral cortex or hippocampus of mice with mild HHcy. Moreover, the signaling activity of STAT3 also did not differ significantly between the two groups. These findings demonstrate that the BBB is highly vulnerable to homocysteine insult. Even a slight increase in serum homocysteine levels up-regulates MMP-9 expression and disrupts the BBB integrity. Meanwhile, microglia activation or the STAT3 pathway might not contribute to the effects of mild HHcy on the brain.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Corteza Cerebral/inmunología , Hipocampo/inmunología , Hiperhomocisteinemia/fisiopatología , Enfermedades Neuroinflamatorias/etiología , Animales , Citocinas/análisis , Homocisteína/sangre , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Metaloproteinasa 9 de la Matriz/análisis , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología
16.
Sci Rep ; 11(1): 10495, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34006897

RESUMEN

IL-4 production is associated with low-avidity, poorly cytotoxic T cell induction that contributes to viral immune evasion and the failure of T cell-based vaccines. Yet, the precise mechanisms that regulate IL-4 signalling in T cells remain elusive. Mounting evidence indicates that cells can dynamically alter their IL-4/IL-13 receptor signature to modulate downstream immune outcomes upon pathogen encounter. Here, we describe how naïve (CD62L+CD44lo-mid) CD4 and CD8 T cells distinctly engage both STAT6 and STAT3 in response to IL-4. We further show that IL-4R⍺ expression is both time- and IL-4 concentration-dependent. Remarkably, our findings reveal that STAT3 inhibition can ablate IL-4R⍺ and affect transcriptional expression of other Stat and Jak family members. By extension, the loss of STAT3 lead to aberrant STAT6 phosphorylation, revealing an inter-regulatory relationship between the two transcription factors. Moreover, IL-4 stimulation down-regulated TGF-ß1 and IFN-γR1 expression on naïve T cells, possibly signifying the broad regulatory implications of IL-4 in conditioning lineage commitment decisions during early infection. Surprisingly, naïve T cells were unresponsive to IL-13 stimulation, unlike dendritic cells. Collectively, these findings could be exploited to inform more efficacious vaccines, as well as design treatments against IL-4/IL-13-associated disease conditions.


Asunto(s)
Interleucina-4/metabolismo , Factor de Transcripción STAT3/fisiología , Transducción de Señal , Linfocitos T/metabolismo , Animales , Biomarcadores/metabolismo , Femenino , Ratones , Ratones Endogámicos BALB C , Fosforilación , Receptores de Interleucina-4/metabolismo , Factor de Transcripción STAT6/metabolismo
17.
Int Urol Nephrol ; 53(6): 1247-1254, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33942213

RESUMEN

AIM: High glucose (HG) induces the production of transforming growth factor (TGF)-ß and reactive oxygen species, which further activates JAK/STAT signaling and promotes the synthesis of matrix proteins, contributes to the pathophysiological processes of diabetic nephropathy. This study aims to investigate the protection role of vitamin D (VD) in the kidney in high glucose condition. METHODS: Rat glomerular mesangial cells were cultured in high glucose medium, with or without VD or VD receptor (VDR) siRNAs treatment. The levels of TGF-ß and fibronectin were detected by qRT-PCR, immunoblotting and enzyme-linked immunosorbent assay (ELISA). The levels of phosphorylated JAK2, STAT1 and STAT3, and JAK/STAT signaling downstream genes were examined by immunoblotting and qRT-PCR. RESULTS: In rat glomerular mesangial cells, VD treatment can repress the tyrosine phosphorylation of JAK2, STAT1 and STAT3. VD inhibited TGF-ß and fibronectin expression which was rescued by vitamin d receptor (VDR) siRNA and STATs inhibitor perficitinib. The JAK/STAT signaling downstream protein coding genes including SOCS1, SOCS3 and type IV collagen were repressed by VD. Meanwhile, the expression of non-coding RNAs such as miR-181a, miR-181b, was repressed by VD, and the expression of miR-34a and Let-7b was upregulated by VD. CONCLUSION: Vitamin D (VD) treatment inhibits the function of HG on fibronectin production through regulating JAK/STAT pathway. These results provide direct evidences that VD protects glomerular mesangial cells from high glucose-induced injury through repressing JAK/STAT signaling, which has the potential for clinical DN treatment.


Asunto(s)
Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/fisiología , Células Mesangiales/efectos de los fármacos , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/fisiología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/fisiología , Transducción de Señal/efectos de los fármacos , Vitamina D/farmacología , Vitaminas/farmacología , Animales , Células Cultivadas , Glucosa/metabolismo , Masculino , Células Mesangiales/fisiología , Ratas , Ratas Sprague-Dawley
18.
J Invest Dermatol ; 141(11): 2668-2678.e6, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33992648

RESUMEN

Exacerbated IL-22 activity induces tissue inflammation and immune disorders such as psoriasis. However, because IL-22 is also essential for tissue repair and defense at barrier interfaces, targeting IL-22 activity to treat psoriasis bears the risk of deleterious effects at mucosal sites such as the gut. We previously showed in vitro that IL-22 signaling relies on IL-22 receptor alpha (IL-22Rα) Y-dependent and -independent pathways. The second depends on the C-terminal Y-less region of IL-22Rα and leads to a massive signal transducer and activator of transcription 3 (STAT3) activation. Because STAT3 activation is associated with the development of psoriasis, we hypothesized that the specific inhibition of the noncanonical STAT3 activation by the Y-less region of IL-22Rα could reduce psoriasis-like disease while leaving intact its tissue defense functions in the gut. We show that mice expressing a C-terminally truncated version of IL-22Rα (ΔCtermut/mut mice) are protected from the development of psoriasis-like dermatitis lesions induced by imiquimod to a lesser extent than Il22ra-/- mice. In contrast, only Il22ra-/- mice lose weight after Citrobacter rodentium infection. Altogether, our data suggest that specific targeting of the noncanonical STAT3 activation by IL-22 could serve to treat psoriasis-like skin inflammation without affecting IL-22‒dependent tissue repair or barrier defense at other sites.


Asunto(s)
Imiquimod/toxicidad , Psoriasis/inducido químicamente , Receptores de Interleucina/fisiología , Factor de Transcripción STAT3/fisiología , Animales , Citrobacter rodentium , Infecciones por Enterobacteriaceae/inmunología , Interleucinas/farmacología , Ratones , Ratones Endogámicos C57BL , Interleucina-22
19.
Mediators Inflamm ; 2021: 8856326, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33867859

RESUMEN

Non-small-cell lung cancer (NSCLC) remains the most common malignancy with the highest morbidity and mortality worldwide. In our previous study, we found that a classic traditional Chinese medicine (TCM) formula Ze-Qi-Tang (ZQT), which has been used in the treatment of respiratory diseases for thousands of years, could directly inhibit the growth of human NSCLC cells via the p53 signaling pathway. In this study, we explored the immunomodulatory functions of ZQT. We found that ZQT significantly prolonged the survival of orthotopic lung cancer model mice by modulating the tumor microenvironment (TME). ZQT remarkably reduced the number of MDSCs (especially G-MDSCs) and inhibited their immunosuppressive activity by inducing apoptosis in these cells via the STAT3/S100A9/Bcl-2/caspase-3 signaling pathway. When G-MDSCs were depleted, the survival promotion effect of ZQT and its inhibitory effect on lung luminescence signal disappeared in tumor-bearing mice. This is the first study to illustrate the immunomodulatory effect of ZQT in NSCLC and the underlying molecular mechanism.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Granulocitos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Medicina Tradicional China , Células Supresoras de Origen Mieloide/efectos de los fármacos , Animales , Calgranulina B/fisiología , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Caspasa 3/fisiología , Línea Celular Tumoral , Medicamentos Herbarios Chinos/uso terapéutico , Granulocitos/patología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/patología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Factor de Transcripción STAT3/fisiología , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral
20.
Front Immunol ; 12: 630196, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897686

RESUMEN

Objective: Kawasaki disease (KD) is the most common cause of acquired pediatric heart disease in the developed world. 10% of KD patients are resistant to front-line therapy, and no interventions exist to address secondary complications such as myocardial fibrosis. We sought to identify proteins and pathways associated with disease and anti-IL-1 treatment in a mouse model of KD. Methods: Vasculitis was induced via Lactobacillus casei cell wall extract (LCWE) injection in 5-week-old male mice. Groups of mice were injected with LCWE alone, LCWE and IL-1 receptor antagonist anakinra, or saline for controls. Upper heart tissue was assessed by quantitative mass spectrometry analysis. Expression and activation of STAT3 was assessed by immunohistochemistry, immunofluorescence and Western blot, and IL-6 expression by RNA-seq and ELISA. A STAT3 small molecular inhibitor and anti-IL-6R antibody were used to evaluate the role of STAT3 and IL-6 in disease development. Results: STAT3 was highly expressed and phosphorylated in cardiac tissue of LCWE-injected mice, and reduced following anakinra treatment. Il6 and Stat3 gene expression was enhanced in abdominal aorta of LCWE-injected mice and reduced with Anakinra treatment. IL-6 serum levels were enhanced in LCWE-injected mice and normalized by anakinra. However, neither inhibition of STAT3 nor blockade of IL-6 altered disease development. Conclusion: Proteomic analysis of cardiac tissues demonstrates differential protein expression between KD-like, control and anakinra treated cardiac tissue. STAT3 and IL-6 were highly upregulated with LCWE and normalized by anakinra treatment. However, both STAT3 and IL-6 were dispensable for disease development indicating they may be bystanders of inflammation.


Asunto(s)
Interleucina-6/fisiología , Síndrome Mucocutáneo Linfonodular/etiología , Factor de Transcripción STAT3/fisiología , Proteína Amiloide A Sérica/antagonistas & inhibidores , Animales , Pared Celular , Modelos Animales de Enfermedad , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-6/antagonistas & inhibidores , Interleucina-6/sangre , Lacticaseibacillus casei , Masculino , Ratones , Ratones Endogámicos C57BL , Síndrome Mucocutáneo Linfonodular/tratamiento farmacológico , Miocardio/metabolismo , Proteómica , Factor de Transcripción STAT3/análisis , Factor de Transcripción STAT3/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...