Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 339
Filtrar
1.
Mol Med Rep ; 30(5)2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39239743

RESUMEN

The regulation of cardiac function by the nuclear transcription factor signal transducer and activator of transcription 4 (STAT4) has been recently recognized. Nevertheless, the role and mechanisms of action of STAT4 in myocardial ischemia­reperfusion (I/R) injury remain unknown. Consequently, the present study constructed a rat model of I/R by ligation of the left anterior descending coronary artery. Following sacrifice, the rat hearts were excised and analyzed to investigated the effects of STAT4 on I/R­induced myocardial injury. Western blotting demonstrated that expression of STAT4 decreased significantly in the rat model of cardiac I/R and in H9C2 cells that were subjected to hypoxia and reoxygenation (H/R). The overexpression of STAT4 in H9C2 cells reduced cell damage and apoptosis induced by H/R. Furthermore, both in vivo and in vitro, the level of PI3K decreased significantly. Although the AKT protein expression levels were not altered, the AKT phosphorylation levels decreased significantly. STAT4 overexpression enhanced the expression of PI3K and AKT in the H9C2 cells. On the whole, the present study demonstrated that STAT4 alleviated I/R­induced myocardial injury through the PI3K/AKT signaling pathway.


Asunto(s)
Apoptosis , Daño por Reperfusión Miocárdica , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Factor de Transcripción STAT4 , Transducción de Señal , Animales , Factor de Transcripción STAT4/metabolismo , Factor de Transcripción STAT4/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Ratas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación
2.
Proc Natl Acad Sci U S A ; 121(32): e2400153121, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39088391

RESUMEN

Although many cytokine pathways are important for dendritic cell (DC) development, it is less clear what cytokine signals promote the function of mature dendritic cells. The signal transducer and activator of transcription 4 (STAT4) promotes protective immunity and autoimmunity downstream of proinflammatory cytokines including IL-12 and IL-23. In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), Stat4-/- mice are resistant to the development of inflammation and paralysis. To define whether STAT4 is required for intrinsic signaling in mature DC function, we used conditional mutant mice in the EAE model. Deficiency of STAT4 in CD11c-expressing cells resulted in decreased T cell priming and inflammation in the central nervous system. EAE susceptibility was recovered following adoptive transfer of wild-type bone marrow-derived DCs to mice with STAT4-deficient DCs, but not adoptive transfer of STAT4- or IL-23R-deficient DCs. Single-cell RNA-sequencing (RNA-seq) identified STAT4-dependent genes in DC subsets that paralleled a signature in MS patient DCs. Together, these data define an IL-23-STAT4 pathway in DCs that is key to DC function during inflammatory disease.


Asunto(s)
Células Dendríticas , Encefalomielitis Autoinmune Experimental , Interleucina-23 , Factor de Transcripción STAT4 , Transducción de Señal , Animales , Factor de Transcripción STAT4/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interleucina-23/metabolismo , Interleucina-23/inmunología , Ratones , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Ratones Noqueados , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/inmunología , Inflamación/metabolismo , Inflamación/inmunología , Traslado Adoptivo , Ratones Endogámicos C57BL , Humanos , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
Int J Mol Sci ; 25(14)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39063055

RESUMEN

Obesity is a major risk factor for the development of life-threatening malignant ventricular tachyarrhythmias (VT) and sudden cardiac death (SCD). Risks may be highest for patients with high levels of the proinflammatory cytokine interleukin (IL)-6. We used our guinea pig model of high-fat diet (HFD)-induced arrhythmias that exhibit a heightened proinflammatory-like pathology, which is also observed in human obesity arrhythmias, as well as immunofluorescence and confocal microscopy approaches to evaluate the pathological IL-6 trans-signaling function and explore the underlying mechanisms. Using blind-stick and electrocardiogram (ECG) techniques, we tested the hypothesis that heightened IL-6 trans-signaling would exhibit increased ventricular arrhythmia/SCD incidence and underlying arrhythmia substrates. Remarkably, compared to low-fat diet (LFD)-fed controls, HFD promoted phosphorylation of the IL-6 signal transducer and activator of transcription 4 (STAT4), leading to its activation and enhanced nuclear translocation of pSTAT4/STAT4 compared to LFD controls and pSTAT3/STAT3 nuclear expression. Overactivation of IL-6 trans-signaling in guinea pigs prolonged the QT interval, which resulted in greater susceptibility to arrhythmias/SCD with isoproterenol challenge, as also observed with the downstream Janus kinase (JAK) 2 activator. These findings may have potentially profound implications for more effective arrhythmia therapy in the vulnerable obese patient population.


Asunto(s)
Arritmias Cardíacas , Dieta Alta en Grasa , Interleucina-6 , Factor de Transcripción STAT4 , Transducción de Señal , Animales , Cobayas , Dieta Alta en Grasa/efectos adversos , Interleucina-6/metabolismo , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/etiología , Factor de Transcripción STAT4/metabolismo , Masculino , Obesidad/metabolismo , Fosforilación , Modelos Animales de Enfermedad
4.
Toxicol Appl Pharmacol ; 488: 116980, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38823456

RESUMEN

Multiple sclerosis (MS) is a class of autoimmune diseases mainly caused by the immune system attacking the myelin sheath of the axons in the nervous system. Although the pathogenesis of MS is complex, studies have shown that dendritic cells (DCs) play a vital role in the pathogenesis of MS. Quercetin (QU) has a unique advantage in clinical application, especially for treating autoimmune diseases. However, the mechanism of QU in the treatment of experimental autoimmune encephalomyelitis (EAE) remains unclear. In this study, we explore the potential role of QU in EAE. Finally, we find that QU has anti-inflammatory activities and neural protective effects in EAE. The experimental results suggest that the cellular basis for QU's function is to inhibit the activation of DCs while modulating the Th17 cell differentiation in the co-culture system. Further, QU may target STAT4 to inhibit its activation in DCs. This work will be of great significance for the future development and utilization of QU.


Asunto(s)
Células Dendríticas , Encefalomielitis Autoinmune Experimental , Ratones Endogámicos C57BL , Quercetina , Factor de Transcripción STAT4 , Células Th17 , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Animales , Quercetina/farmacología , Factor de Transcripción STAT4/metabolismo , Femenino , Ratones , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Diferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , Antiinflamatorios/farmacología
5.
Gene ; 927: 148719, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38917875

RESUMEN

Renal cell carcinoma (RCC) represents a significant portion of genitourinary cancers, marked by challenging prognosis and high metastasis rates. Immunotherapy has been applied in managing advanced renal cell carcinoma, but the therapeutic outcomes are unsatisfactory. In this study, we order to construct a Janus kinase/signal transduction and activator transcriptional (JAK/STAT)-related signature linked to kidney patient outcomes for better predicting the efficacy to immune checkpoint inhibitors (ICIs) and to provide guidance for effective combination therapy. We screened 25 differentially expressed genes (DEGs) that exhibited high expression in RCC samples and were enriched in the JAK-STAT signaling pathway. Among these genes, 11 key genes were identified and correlated with the expectation of Kidney Clear Cell Carcinoma (KIRC) patients and all these genes was significantly elevated in RCC tumor tissues and cancer cells compared to para-cancer tissues and normal renal cells. Utilizing these 11 genes, we divided RCC patients into high-risk and low-risk groups. We found a clear correlation between the clinicopathologic factors of KIRC patients and the JAK-STAT-related risk score. And the IHC results shown that the JAK3 and STAT4 expression of tumor was significantly higher than normal tissue in RCC patients, the level of JAK3 and STAT4 was positively related to the T stage of RCC patients. In addition, high-risk patients had a poorer prognosis and greater protumor immune cell infiltration, and benefitted less from immunotherapy than did low-risk patients. Furthermore, the JAK-STAT-related risk score can predict disease-free survival (DFS) in RCC patients according to the nomogram, which constructed in combination with other clinical features such as age, TNM-staging and stage. Our study demonstrated the JAK-STAT signaling pathway's important regulatory function in RCC tumor immunity. This insight not only enhances our ability to accurately predict the survival rate of RCC patients, but also underscores a potential therapeutic alternative for RCC, involving the combined targeting of the JAK-STAT pathway and immune checkpoints.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Renales , Regulación Neoplásica de la Expresión Génica , Inmunoterapia , Neoplasias Renales , Transducción de Señal , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Pronóstico , Inmunoterapia/métodos , Femenino , Masculino , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo , Janus Quinasa 3/genética , Janus Quinasa 3/metabolismo , Quinasas Janus/metabolismo , Quinasas Janus/genética , Persona de Mediana Edad , Factores de Transcripción STAT/metabolismo , Factores de Transcripción STAT/genética , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Transcriptoma , Perfilación de la Expresión Génica
6.
Int J Med Sci ; 21(6): 1176-1186, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38774752

RESUMEN

Background: To uncover the potential significance of JAK-STAT-SOCS1 axis in penile cancer, our study was the pioneer in exploring the altered expression processes of JAK-STAT-SOCS1 axis in tumorigenesis, malignant progression and lymphatic metastasis of penile cancer. Methods: In current study, the comprehensive analysis of JAK-STAT-SOCS1 axis in penile cancer was analyzed via multiple analysis approaches based on GSE196978 data, single-cell data (6 cancer samples) and bulk RNA data (7 cancer samples and 7 metastasis lymph nodes). Results: Our study observed an altered molecular expression of JAK-STAT-SOCS1 axis during three different stages of penile cancer, from tumorigenesis to malignant progression to lymphatic metastasis. STAT4 was an important dominant molecule in penile cancer, which mediated the immunosuppressive tumor microenvironment by driving the apoptosis of cytotoxic T cell and was also a valuable biomarker of immune checkpoint inhibitor treatment response. Conclusions: Our findings revealed that the complexity of JAK-STAT-SOCS1 axis and the predominant role of STAT4 in penile cancer, which can mediate tumorigenesis, malignant progression, and lymphatic metastasis. This insight provided valuable information for developing precise treatment strategies for patients with penile cancer.


Asunto(s)
Progresión de la Enfermedad , Quinasas Janus , Metástasis Linfática , Neoplasias del Pene , Factor de Transcripción STAT4 , Proteína 1 Supresora de la Señalización de Citocinas , Humanos , Masculino , Neoplasias del Pene/patología , Neoplasias del Pene/genética , Neoplasias del Pene/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Metástasis Linfática/patología , Metástasis Linfática/genética , Quinasas Janus/metabolismo , Factor de Transcripción STAT4/metabolismo , Factor de Transcripción STAT4/genética , Regulación Neoplásica de la Expresión Génica , Carcinogénesis/genética , Carcinogénesis/patología , Transducción de Señal , Microambiente Tumoral/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología
7.
J Anim Sci ; 1022024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38713167

RESUMEN

Follicular fluid meiosis-activating sterol (FF-MAS) is a small molecule compound found in FF, named for its ability to induce oocyte resumption of meiosis. Granulosa cells (GCs) within the follicle are typically located in a hypoxic environment under physiologic conditions due to limited vascular distribution. Previous research suggests that hypoxia-induced cell cycle arrest and apoptosis in GCs may be crucial triggering factors in porcine follicular atresia. However, the impact of FF-MAS on GCs within follicles has not been explored so far. In this study, we uncovered a novel role of FF-MAS in facilitating GC survival under hypoxic conditions by inhibiting STAT4 expression. We found that STAT4 expression was upregulated in porcine GCs exposed to 1% O2. Both gain and loss of function assays confirmed that STAT4 was required for cell apoptosis under hypoxia conditions, and that the GC apoptosis caused by hypoxia was markedly attenuated following FF-MAS treatment through inhibition of STAT4 expression. Correlation analysis in vivo revealed that GC apoptosis was associated with increased STAT4 expression, while the FF-MAS content in follicular fluid was negatively correlated with STAT4 mRNA levels and cell apoptosis. These findings elucidate a novel role of FF-MAS-mediated protection of GCs by inhibiting STAT4 expression under hypoxia, which might contribute to the mechanistic understanding of follicular development.


Granulosa cells (GCs) influence follicle growth and development, with their proliferation and differentiation promoting follicle development and ovulation, while their programmed cell death and degeneration trigger follicular atresia. In this study, to investigate the effect of FF-MAS on GCs of follicles, we performed gene expression profiling in the domestic pig (Sus scrofa). We discovered STAT4 is required for GC apoptosis under hypoxia conditions both in vitro and in vivo and FF-MAS prevents porcine ovarian granulosa cells from hypoxia-induced apoptosis via inhibiting STAT4 expression.


Asunto(s)
Apoptosis , Líquido Folicular , Células de la Granulosa , Meiosis , Factor de Transcripción STAT4 , Animales , Células de la Granulosa/efectos de los fármacos , Femenino , Apoptosis/efectos de los fármacos , Porcinos , Líquido Folicular/química , Meiosis/efectos de los fármacos , Factor de Transcripción STAT4/metabolismo , Factor de Transcripción STAT4/genética , Esteroles , Hipoxia/veterinaria
8.
Tuberculosis (Edinb) ; 147: 102517, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38733881

RESUMEN

The extensive inability of the BCG vaccine to produce long-term immune protection has not only accelerated the disease burden but also progressed towards the onset of drug resistance. In our previous study, we have reported the promising effects of Bergenin (Berg) in imparting significant protection as an adjunct immunomodulator against tuberculosis (TB). In congruence with our investigations, we delineated the impact of Berg on T cells, wherein it enhanced adaptive memory responses by modulating key transcription factors, STAT4 and Akt. We translated this finding into the vaccine model of TB and observed a notable reduction in the burden of Mycobacterium tuberculosis (M.tb) in BCG-Berg co-immunized mice as compared to BCG vaccination. Moreover, Berg, along with BCG, also aided in a heightened proinflammatory response milieu that corroborates the host protective immune response against TB. Furthermore, this response aligns with the escalated central and resident memory responses by modulating the Akt-Foxo-Stat4 axis, which plays a crucial role in enhancing the vaccine efficacy of BCG. These findings showcase the utilization of immunomodulator Berg as an immunoprophylactic agent to upgrade immunological memory, making it a more effective defender against TB.


Asunto(s)
Inmunidad Adaptativa , Vacuna BCG , Benzopiranos , Memoria Inmunológica , Ratones Endogámicos C57BL , Mycobacterium tuberculosis , Proteínas Proto-Oncogénicas c-akt , Factor de Transcripción STAT4 , Transducción de Señal , Animales , Vacuna BCG/inmunología , Vacuna BCG/farmacología , Memoria Inmunológica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Mycobacterium tuberculosis/inmunología , Benzopiranos/farmacología , Factor de Transcripción STAT4/metabolismo , Inmunidad Adaptativa/efectos de los fármacos , Femenino , Tuberculosis/inmunología , Tuberculosis/microbiología , Interacciones Huésped-Patógeno , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Ratones
9.
Free Radic Biol Med ; 218: 1-15, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38574973

RESUMEN

Sjogren's syndrome (SS) is an autoimmune disease characterized by dysfunction of exocrine glands, such as salivary glands. However, the molecular mechanism of salivary secretion dysfunction in SS is still unclear. Given the significance of glutathione peroxidase 4 (GPX4) in cellular redox homeostasis, we hypothesized that dysregulation of GPX4 may play a pivotal role in the pathogenesis of salivary secretion dysfunction observed in SS. The salivary gland of SS patients and the SS mouse model exhibited reduced expression of the ferroptosis inhibitor GPX4 and the important protein aquaporin 5 (AQP5), which is involved in salivary secretion. GPX4 overexpression upregulated and GPX4 knockdown downregulated AQP5 expression in salivary gland epithelial cells (SGECs) and salivary secretion. Bioinformatics analysis of GSE databases from SS patients' salivary glands revealed STAT4 as a key intermediary regulator between GPX4 and AQP5. A higher level of nuclear pSTAT4 was observed in the salivary gland of the SS mouse model. GPX4 overexpression inhibited and GPX4 knockdown promoted STAT4 phosphorylation and nuclear translocation in SGECs. CHIP assay confirmed the binding of pSTAT4 within the promoter of AQP5 inhibiting AQP5 transcription. GPX4 downregulation accumulates intracellular lipid ROS in SGECs. Lipid ROS inhibitor ferrostatin-1 treatment during in vitro and in vivo studies confirmed that lipid ROS activates STAT4 phosphorylation and nuclear translocation in SGECs. In summary, the downregulated GPX4 in SGECs contributes to salivary secretion dysfunction in SS via the lipid ROS/pSTAT4/AQP5 axis. This study unraveled novel targets to revitalize the salivary secretion function in SS patients.


Asunto(s)
Acuaporina 5 , Células Epiteliales , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Especies Reactivas de Oxígeno , Factor de Transcripción STAT4 , Glándulas Salivales , Síndrome de Sjögren , Síndrome de Sjögren/metabolismo , Síndrome de Sjögren/genética , Síndrome de Sjögren/patología , Animales , Humanos , Ratones , Glándulas Salivales/metabolismo , Glándulas Salivales/patología , Acuaporina 5/metabolismo , Acuaporina 5/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT4/metabolismo , Factor de Transcripción STAT4/genética , Modelos Animales de Enfermedad , Femenino , Regulación hacia Abajo , Masculino , Transducción de Señal , Regulación de la Expresión Génica , Ferroptosis/genética , Saliva/metabolismo , Persona de Mediana Edad
10.
Biomed Pharmacother ; 174: 116537, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38579402

RESUMEN

Chronic Lymphocytic Leukemia (CLL) patients have a defective expression of the proapoptotic protein p66Shc and of its transcriptional factor STAT4, which evoke molecular abnormalities, impairing apoptosis and worsening disease prognosis and severity. p66Shc expression is epigenetically controlled and transcriptionally modulated by STAT4; epigenetic modifiers are deregulated in CLL cells and specific histone deacetylases (HDACs) like HDAC1, are overexpressed. Reactivation of STAT4/p66Shc expression may represent an attractive and challenging strategy to reverse CLL apoptosis defects. New selective class I HDAC inhibitors (HDACis, 6a-g) were developed with increased potency over existing agents and preferentially interfering with the CLL-relevant isoform HDAC1, to unveil the role of class I HDACs in the upregulation of STAT4 expression, which upregulates p66Shc expression and hence normalizes CLL cell apoptosis. 6c (chlopynostat) was identified as a potent HDAC1i with a superior profile over entinostat. 6c induces marked apoptosis of CLL cells compared with SAHA, which was associated with an upregulation of STAT4/p66Shc protein expression. The role of HDAC1, but not HDAC3, in the epigenetic upregulation of STAT4/p66Shc was demonstrated for the first time in CLL cells and was validated in siRNA-induced HDAC1/HDAC3 knock-down EBV-B cells. To sum up, HDAC1 inhibition is necessary to reactivate STAT4/p66Shc expression in patients with CLL. 6c is one of the most potent HDAC1is known to date and represents a novel pharmacological tool for reversing the impairment of the STAT4/p66Shc apoptotic machinery.


Asunto(s)
Apoptosis , Linfocitos B , Inhibidores de Histona Desacetilasas , Leucemia Linfocítica Crónica de Células B , Factor de Transcripción STAT4 , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Leucemia Linfocítica Crónica de Células B/metabolismo , Apoptosis/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Factor de Transcripción STAT4/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 1/antagonistas & inhibidores , Benzamidas/farmacología , Masculino , Anciano , Femenino , Persona de Mediana Edad
11.
Phytomedicine ; 128: 155558, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38547614

RESUMEN

BACKGROUND: The experimental autoimmune myocarditis (EAM) model is valuable for investigating myocarditis pathogenesis. M1-type macrophages and CD4+T cells exert key pathogenic effects on EAM initiation and progression. Baicalein (5,6,7-trihydroxyflavone, C15H10O5, BAI), which is derived from the Scutellaria baicalensis root, is a primary bioactive compound with potent anti-inflammatory and antioxidant properties. BAI exerts good therapeutic effects against various autoimmune diseases; however, its effect in EAM has not been thoroughly researched. PURPOSE: This study aimed to explore the possible inhibitory effect of BAI on M1 macrophage polarisation and CD4+T cell differentiation into Th1 cells via modulation of the JAK-STAT1/4 signalling pathway, which reduces the secretion of pro-inflammatory factors, namely, TNF-α and IFN-γ, and consequently inhibits TNF-α- and IFN-γ-triggered apoptosis in cardiomyocytes of the EAM model mice. STUDY DESIGN AND METHODS: Flow cytometry, immunofluorescence, real-time quantitative polymerase chain reaction (q-PCR), and western blotting were performed to determine whether BAI alleviated M1/Th1-secreted TNF-α- and IFN-γ-induced myocyte death in the EAM model mice through the inhibition of the JAK-STAT1/4 signalling pathway. RESULTS: These results indicate that BAI intervention in mice resulted in mild inflammatory infiltrates. BAI inhibited JAK-STAT1 signalling in macrophages both in vivo and in vitro, which attenuated macrophage polarisation to the M1 type and reduced TNF-α secretion. Additionally, BAI significantly inhibited the differentiation of CD4+T cells to Th1 cells and IFN-γ secretion both in vivo and in vitro by modulating the JAK-STAT1/4 signalling pathway. This ultimately led to decreased TNF-α and IFN-γ levels in cardiac tissues and reduced myocardial cell apoptosis. CONCLUSION: This study demonstrates that BAI alleviates M1/Th1-secreted TNF-α- and IFN-γ-induced cardiomyocyte death in EAM mice by inhibiting the JAK-STAT1/4 signalling pathway.


Asunto(s)
Apoptosis , Modelos Animales de Enfermedad , Flavanonas , Interferón gamma , Quinasas Janus , Miocarditis , Miocitos Cardíacos , Factor de Transcripción STAT1 , Transducción de Señal , Factor de Necrosis Tumoral alfa , Animales , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Quinasas Janus/metabolismo , Ratones , Flavanonas/farmacología , Masculino , Interferón gamma/metabolismo , Apoptosis/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Miocarditis/tratamiento farmacológico , Factor de Transcripción STAT4/metabolismo , Enfermedades Autoinmunes/tratamiento farmacológico , Ratones Endogámicos BALB C , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Scutellaria baicalensis/química , Células TH1/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos
12.
Tissue Eng Regen Med ; 21(4): 595-608, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38466361

RESUMEN

BACKGROUND: The purpose of this study was to investigate the specific effects of signal transducer and activator of transcription 4 (STAT4)-induced long intergenic nonprotein coding RNA 1278 (LINC01278) on the growth of non-small cell lung cancer (NSCLC) cells involved in the microRNA (miR)-877-5p/activated transcription factor 4 (ATF4) axis. METHODS: NSCLC tumor tissue and adjacent normal tissue were collected. Human normal lung epithelial cell BEAS-2B and human NSCLC cell lines (H1299, H1975, A549, H2228) were collected. The expression levels of STAT4, LINC01278, miR-877-5p, and ATF4 were detected. A549 cells were screened for subsequent experiments. The proliferation ability of cells was detected by colony formation experiment. Cell apoptosis was tested by flow cytometry. Scratch test and transwell assay were used to detect the migration and invasion ability of cells. Biological function of LINC01278 in NSCLC was confirmed by xenograft experiments. RESULTS: Low expression miR-877-5p and high expression of STAT4, LINC01278 and ATF4 were detected in NSCLC. Silenced LINC01278 in A549 cell depressed cell proliferation, migration and invasion, but facilitated cell apoptosis. LINC01278 was positively correlated with STAT4 and could directly bind to miR-877-5p. Upregulating miR-877-5p suppressed NSCLC cell progression, while downregulating miR-877-5p had the opposite effect. Upregulating miR-877-5p abrogated the effects of silenced LINC01278 on NSCLC cell progression. MiR-877-5p targeted ATF4. ATF4 upregulation could partly restore the carcinogenic effect of LINC01278 in vitro and in vivo. CONCLUSION: Our data supports that STAT4-induced upregulation of LINC01278 promotes NSCLC progression by modulating the miR-877-5p/ATF4 axis, suggesting a novel direction for NSCLC treatment.


Asunto(s)
Factor de Transcripción Activador 4 , Carcinoma de Pulmón de Células no Pequeñas , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Factor de Transcripción STAT4 , Regulación hacia Arriba , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Animales , Factor de Transcripción STAT4/metabolismo , Factor de Transcripción STAT4/genética , Invasividad Neoplásica , Línea Celular Tumoral , Células A549 , Ratones Desnudos , Ratones , Apoptosis , Femenino , Masculino , Movimiento Celular , Ratones Endogámicos BALB C
13.
Int J Mol Sci ; 25(6)2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38542139

RESUMEN

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.


Asunto(s)
Factor de Crecimiento Epidérmico , Síndrome de Sjögren , Humanos , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/genética , Interferón-alfa/farmacología , Factores Inmunológicos , Técnicas de Cultivo de Célula , ARN Mensajero/metabolismo , Suplementos Dietéticos , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Fosforilación , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo
14.
J Exp Clin Cancer Res ; 43(1): 67, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38429845

RESUMEN

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.


Asunto(s)
Resistencia a Antineoplásicos , Interleucina-11 , Neoplasias de la Próstata , Humanos , Masculino , Docetaxel/farmacología , Regulación de la Expresión Génica , Interleucina-11/genética , Interleucina-11/metabolismo , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Factor de Transcripción STAT4/metabolismo , Resistencia a Antineoplásicos/genética
15.
Am J Physiol Cell Physiol ; 326(5): C1494-C1504, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38406824

RESUMEN

Primary Sjögren's syndrome (pSS) is characterized by its autoimmune nature. This study investigates the role of the IFNγ SNP rs2069705 in modulating the susceptibility to pSS. Differential expression of IFNγ and BAFF was analyzed using the GEO database's mRNA microarray GSE84844. Genotyping of the IFNγ SNP rs2069705 was conducted via the dbSNP website. The JASPAR tool was used for predicting transcription factor bindings. Techniques such as dual-luciferase reporter assays, Chromatin immunoprecipitation, and analysis of a pSS mouse model were applied to study gene and protein interactions. A notable increase in the mutation frequency of IFNγ SNP rs2069705 was observed in MNCs from the exocrine glands of pSS mouse models. Bioinformatics analysis revealed elevated levels of IFNγ and BAFF in pSS samples. The model exhibited an increase in both CD20+ B cells and cells expressing IFNγ and BAFF. Knocking down IFNγ resulted in lowered BAFF expression and less lymphocyte infiltration, with BAFF overexpression reversing this suppression. Activation of the Janus kinase (JAK)/STAT1 pathway was found to enhance transcription in the BAFF promoter region, highlighting IFNγ's involvement in pSS. In addition, rs2069705 was shown to boost IFNγ transcription by promoting interaction between its promoter and STAT4. SNP rs2069705 in the IFNγ gene emerges as a pivotal element in pSS susceptibility, primarily by augmenting IFNγ transcription, activating the JAK/STAT1 pathway, and leading to B-lymphocyte infiltration in the exocrine glands.NEW & NOTEWORTHY The research employed a combination of bioinformatics analysis, genotyping, and experimental models, providing a multifaceted approach to understanding the complex interactions in pSS. We have uncovered that the rs2069705 SNP significantly affects the transcription of IFNγ, leading to altered immune responses and B-lymphocyte activity in pSS.


Asunto(s)
Linfocitos B , Interferón gamma , Polimorfismo de Nucleótido Simple , Síndrome de Sjögren , Activación Transcripcional , Animales , Femenino , Humanos , Ratones , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Interferón gamma/genética , Interferón gamma/metabolismo , Quinasas Janus/metabolismo , Quinasas Janus/genética , Polimorfismo de Nucleótido Simple/genética , Transducción de Señal , Síndrome de Sjögren/genética , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/metabolismo , Síndrome de Sjögren/patología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo
16.
Neurochem Int ; 175: 105683, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38341034

RESUMEN

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.


Asunto(s)
Neuroblastoma , Enfermedad de Parkinson , Animales , Humanos , Ratones , Apoptosis , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Kisspeptinas , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , Sincalida/efectos adversos , Sincalida/metabolismo , Factor de Transcripción STAT4/metabolismo
17.
J Cutan Pathol ; 51(3): 232-238, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37932931

RESUMEN

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.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz , Micosis Fungoide , Neoplasias Cutáneas , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Micosis Fungoide/patología , Proteínas Nucleares/metabolismo , Estudios Prospectivos , Neoplasias Cutáneas/patología , Factor de Transcripción STAT4/metabolismo , Proteína 1 Relacionada con Twist/metabolismo
18.
J Physiol Biochem ; 80(1): 219-233, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38091230

RESUMEN

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.


Asunto(s)
Citotoxicidad Inmunológica , Células Asesinas Naturales , Animales , Humanos , Ratones , Proteínas de Homeodominio , Células Asesinas Naturales/metabolismo , Perforina/genética , Perforina/metabolismo , Transducción de Señal , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo , Factores de Transcripción/metabolismo , Regulador Transcripcional ERG/metabolismo
19.
Mol Neurobiol ; 61(4): 2336-2356, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37875707

RESUMEN

Our previous study has proved that the Klotho up-regulation participated in cerebral ischemic preconditioning (CIP)-induced brain ischemic tolerance. However, the exact neuroprotective mechanism of Klotho in CIP remains unclear. We explored the hypothesis that STAT4-mediated Klotho up-regulation contributes to the CIP-induced brain ischemic tolerance via inhibiting neuronal pyroptosis. Firstly, the expressions of pyroptosis-associated proteins (i.e., NLRP3, GSDMD, pro-caspase-1, and cleaved caspase-1) in hippocampal CA1 region were determined during the process of brain ischemic tolerance. We found the expression of pyroptosis-associated proteins was significantly up-regulated in the ischemic insult (II) group, and showed no significant changes in the CIP group. The expression level of each pyroptosis-associated proteins was lower in the CIP + II group than that in the II group. Inhibition of Klotho expression increased the expression of pyroptosis-associated proteins in the CIP + II group and blocked the CIP-induced brain ischemic tolerance. Injection of Klotho protein decreased the expression of pyroptosis-associated proteins in the II group, and protected neurons from ischemic injury. Secondly, the transcription factor STAT4 of Klotho was identified by bioinformatic analysis. Double luciferase reporter gene assay and chromatin immunoprecipitation assay showed STAT4 can bind to the site between nt - 881 and - 868 on the Klotho promoter region and positively regulates Klotho expression. Moreover, we found CIP significantly enhanced the expression of STAT4. Knockdown STAT4 suppressed Klotho up-regulation after CIP and blocked the CIP-induced brain ischemic tolerance. Collectively, it can be concluded that STAT4-mediated the up-regulation of Klotho contributed to the brain ischemic tolerance induced by CIP via inhibiting pyroptosis.


Asunto(s)
Isquemia Encefálica , Precondicionamiento Isquémico , Ratas , Animales , Ratas Wistar , Regulación hacia Arriba , Piroptosis , Factor de Transcripción STAT4/metabolismo , Isquemia Encefálica/metabolismo , Región CA1 Hipocampal/metabolismo , Neuronas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
20.
Arthritis Res Ther ; 25(1): 221, 2023 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-37978415

RESUMEN

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.


Asunto(s)
Lupus Eritematoso Sistémico , Proteínas Proto-Oncogénicas c-akt , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos T Reguladores , Interferón-alfa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Receptores Inmunológicos/metabolismo , Factor de Transcripción STAT4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA