Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38877848

RESUMEN

BACKGROUND: Allergic rhinitis (AR) represents a significant global health concern that can give rise to numerous diseases and result in labor productivity. T regulatory (Treg) cells are pivotal players in the pathogenesis of AR, and their deficiencies are closely related to Prostaglandin E2 (PGE2). However, the downstream mechanisms of this relationship remain poorly understood. OBJECTIVE: This study aims to investigate the inhibitory mechanisms through which PGE2 impacts the differentiation of Treg cells. METHODS: We compared the differentiation of Treg cells from naïve CD4+ T cells of AR patients and healthy controls, with or without the presence of PGE2 by flow cytometry. Intracellular cAMP concentration, mRNA and protein levels of cyclic-AMP dependent protein kinase A (PKA), as well as their downstream target, Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ) were examined in Treg cells from AR and healthy donors. AR mouse model was established by pollen administration. RESULTS: PGE2 suppressed the differentiation of Treg cells from human naïve CD4+ T cells through the EP4 receptor. Furthermore, in AR patients and AR mouse, the expression of EP4 receptor were observed enhanced. The PGE2-EP4 signal was carried out by activating cAMP-PKA signaling pathway. Subsequently, phospholated PKA would suppress PPAR-γ expression. Treatment of Pioglitazone, a PPAR-γ agonist, was demonstrated to rescue the differentiation of Treg and help alleviate inflammation in the AR mouse model. CONCLUSION: In AR disease, the PGE2-EP4 signaling exerts an inhibitory effect on Treg differentiation by influencing the cAMP-PKA pathway and its downstream target PPAR-γ.

2.
Mol Oncol ; 17(12): 2694-2708, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37691307

RESUMEN

E3 SUMO-protein ligase CBX4 (CBX4), a key component of polycomb-repressive complexes 1 (PRC1), has been reported to regulate a variety of genes implicated in tumor growth, metastasis, and angiogenesis. However, its role in T-cell-mediated antitumor immunity remains elusive. To shed light on this issue, we generated mice with T-cell-specific deletion of Cbx4. Tumor growth was increased in the knockout mice. Additionally, their tumor-infiltrating lymphocytes exhibited impaired tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) production, with an elevated programmed cell death protein 1 (PD-1) level. In fact, dysregulated Pdcd1 expression was observed in all major subsets of peripheral T cells from the knockout mice, which was accompanied by a functional defect in response to T-cell receptor (TCR) stimulation. In support of a direct link between CBX4 and PD-1, Cbx4 overexpression resulted in the downregulation of Pdcd1 expression. Epigenetic analyses indicated that Cbx4 deficiency leads to diminished accumulation of inhibitory histone modifications at conserved region (CR)-C and CR-B sites of the Pdcd1 promoter, namely mono-ubiquitinated histone H2A at lysine 119 (H2AK119ub1) and trimethylated histone H3 at lysine 27 (H3K27me3). Moreover, inhibition of either the E3 ligase activity of polycomb-repressive complexes 1 (PRC1) or the methyltransferase activity of polycomb-repressive complexes 2 (PRC2) restores Pdcd1 expression in Cbx4-transfected cells. Cumulatively, this study reveals a novel function of CBX4 in the regulation of T-cell function and expands our understanding of the epigenetic control of Pdcd1 expression.


Asunto(s)
Neoplasias , Receptor de Muerte Celular Programada 1 , Animales , Ratones , Receptor de Muerte Celular Programada 1/genética , Lisina , Linfocitos T/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Ligasas/genética , Ligasas/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Neoplasias/genética , Ratones Noqueados
3.
Proc Natl Acad Sci U S A ; 120(19): e2212613120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126714

RESUMEN

Oxidative stress is a key feature in both chronic inflammation and cancer. P38 regulated/activated protein kinase (PRAK) deficiency can cause functional disorders in neutrophils and macrophages under high oxidative stress, but the precise mechanisms by which PRAK regulates reactive oxygen species (ROS) elimination and its potential impact on CD4+ T helper subset function are unclear. The present study reveals that the PRAK-NF-E2-related factor 2(NRF2) axis is essential for maintaining the intracellular redox homeostasis of T helper 17(Th17) cells, thereby promoting Th17 cell differentiation and antitumor effects. Through mechanistic analysis, we identify NRF2 as a novel protein substrate of PRAK and find that PRAK enhances the stability of the NRF2 protein through phosphorylation NRF2 Serine(S) 558 independent of protein ubiquitination. High accumulation of cellular ROS caused by loss of PRAK disrupts both glycolysis and PKM2-dependent phosphorylation of STAT3, which subsequently impairs the differentiation of Th17 cells. As a result, Prak knockout (KO) mice display significant resistance to experimental autoimmune encephalomyelitis (EAE) but impaired antitumor immunity in a MC38 tumor model. This work reveals that the PRAK-NRF2-mediated antioxidant pathway is a metabolic checkpoint that controls Th17-cell glycolysis and differentiation. Targeting PRAK is a promising strategy for maintaining an active ROS scavenging system and may lead to potent Th17 cell antitumor immunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Proteínas Quinasas , Animales , Ratones , Diferenciación Celular , Glucólisis , Homeostasis , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Proteínas Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Th17/metabolismo
4.
Front Public Health ; 10: 937844, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36530737

RESUMEN

Background: The COVID-19 pandemic has disrupted the diagnosis, treatment, and care for tuberculosis (TB). Delays in seeking TB care may result in increased community transmission and unfavorable treatment outcomes. We sought to understand the influence of the COVID-19 pandemic on the proportion of patients with TB who delayed seeking the diagnosis and care for TB and explore the reasons for their postponement. Methods: We surveyed a representative sample of outpatients treated for pulmonary TB from June to November 2020 using an anonymous standardized questionnaire. Multivariable logistic regression was used to calculate adjusted odds ratios (aOR) and 95% confidence intervals (CIs) of factors associated with the postponement of TB care. We used routinely collected surveillance data to assess trends of TB reports before and after the emergence of COVID-19 (2017-2019 vs. 2020-2022) in Tianjin, China. Results: Among 358 participants who were diagnosed with pulmonary TB during the COVID-19 response, 61 (17%) postponed seeking TB diagnosis due to COVID-19, with 39 (64%) citing fear as the primary reason. Female sex (aOR:2.0; 95% CI: 1.1-3.7), previous antituberculosis treatment (aOR:3.2; 95%CI: 1.4-7.6), and TB diagnosis during the first-level response (aOR = 3.2, 1.7-6.2) were associated with the postponement. Among all 518 participants receiving antituberculosis treatment, 57 (11%) had postponed their regular healthcare visits due to COVID-19, 175 (34%) received no treatment supervision, and 32 (6%) experienced treatment interruption. Compared to 2017-2019, reported pulmonary TB declined by 36.8% during the first-level response to COVID-19, 23.5% during the second-level response, 14% during the third-level response in 2020, and 4.3% in 2021. Conclusion: The COVID-19 response reduced the number of people who sought and received diagnosis, treatment, and care for TB in Tianjin, China. Integrative programs to ensure access and continuity of TB services should be considered and dual testing for SARS-CoV-2 and M. tuberculosis may facilitate finding cases.


Asunto(s)
COVID-19 , Tuberculosis Pulmonar , Tuberculosis , Humanos , Femenino , Pandemias , COVID-19/diagnóstico , COVID-19/epidemiología , SARS-CoV-2 , Tuberculosis Pulmonar/diagnóstico , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/epidemiología , Tuberculosis/diagnóstico , Tuberculosis/tratamiento farmacológico , Tuberculosis/epidemiología , China/epidemiología , Antituberculosos/uso terapéutico
5.
J Immunol ; 208(8): 1912-1923, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35379745

RESUMEN

The mechanism regulating the life span of short-lived plasma cells (SLPCs) remains poorly understood. Here we demonstrated that the EP4-mediated activation of AKT by PGE2 was required for the proper control of inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) hyperactivation and hence the endoplasmic reticulum (ER) homeostasis in IgM-producing SLPCs. Disruption of the PGE2-EP4-AKT signaling pathway resulted in IRE1α-induced activation of JNK, leading to accelerated death of SLPCs. Consequently, Ptger4-deficient mice (C57BL/6) exhibited a markedly impaired IgM response to T-independent Ags and increased susceptibility to Streptococcus pneumoniae infection. This study reveals a highly selective impact of the PGE2-EP4 signal on the humoral immunity and provides a link between ER stress response and the life span of SLPCs.


Asunto(s)
Supervivencia Celular , Dinoprostona , Estrés del Retículo Endoplásmico , Endorribonucleasas , Células Plasmáticas , Proteínas Serina-Treonina Quinasas , Animales , Supervivencia Celular/inmunología , Dinoprostona/inmunología , Estrés del Retículo Endoplásmico/inmunología , Endorribonucleasas/inmunología , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos C57BL , Células Plasmáticas/inmunología , Prostaglandinas/inmunología , Prostaglandinas E/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología
6.
Patient Prefer Adherence ; 16: 321-329, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35173417

RESUMEN

PURPOSE: Poor tuberculosis (TB) medication adherence increases the risk of treatment failure and development of drug-resistant TB, while universal implementation of directly observed therapy (DOT) is not feasible in China. EHealth technologies were reported to be promising patient-centered tools for improving adherence. However, only pilot studies have assessed patients' experiences, and the results were discrepant. PATIENTS AND METHODS: This prospective-cohort study was conducted among TB patients at the outpatient department from 3 March 2019 to 30 May 2020 in Tianjin, China. Data were downloaded from the Tuberculosis Doctor App and TB Information Management System (TBIMS) and merged them by the TBIMS notification number. Logistic regression analysis was used to analyze the factors associated with regular drug-intake. Odds ratios and 95% confidence intervals were estimated with and without adjustment for age, gender, ethnicity and occupation. RESULTS: Revisit examination was more regularly and frequently in APP group than non-APP group. In APP group, 33.28% patients were regular drug-intake. The whole drug-intake rate was 84.84%. Tuberculosis pleurisy (aOR: 0.42, 95CI%=0.26-0.69) and retreated patients (aOR: 0.40, 95CI% =0.27-0.59) were more likely to have poor medication compliance. Local residents tend to have better medication compliance (aOR: 1.80, 95CI% =1.16-2.79). CONCLUSION: APP could improve TB patients' revisit examination adherence. Medication adherence was poor in tuberculosis pleuritis and retreated patients, while local residents tend to have better medication adherence. To make full use of the mobile application in TB patient management, more incentive measures should be adopted for patients and doctors, respectively.

7.
Front Immunol ; 12: 755512, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804040

RESUMEN

Non-coding RNAs represent a class of important regulators in immune response. Previously, LINC02605 was identified as a candidate regulator in innate immune response by lncRNA microarray assays. In this study, we systematically analyzed the functions and the acting mechanisms of LINC02605 in antiviral innate immune response. LINC02605 was up-regulated by RNA virus, DNA virus, and type I IFNs in NF-κB and Jak-stat dependent manner. Overexpression of LINC02605 promotes RNA virus-induced type I interferon production and inhibited viral replication. Consistently, knockdown of LINC02605 resulted in reduced antiviral immune response and increased viral replication. Mechanistically, LINC02605 released the inhibition of hsa-miR-107 on the expression of phosphatase and tensin homolog (PTEN). By microRNA mimics and inhibitors, hsa-miR-107 was demonstrated to not only inhibit PTEN's expression but also negatively regulate the antiviral immune response. Knockdown of LINC02605 led to the reduction of PTEN expression both in mRNA and protein levels. Overexpression of LINC02605 had an opposite impact. Moreover, LINC02605 attenuated the serine 97 phosphorylation level of interferon regulatory factor 3 (IRF3) by promoting PTEN expression. Nucleoplasmic fragmentation assay showed that knocking down LINC02605 inhibited the nuclear translocation of IRF3, rendering the host cells more susceptible to viral invasion, while overexpression showed opposite effects. Therefore, LINC02605 is an induced lncRNA by viral infection and plays a positive feedback in antiviral immune response through modulating the nuclear translocation of IRF3.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/metabolismo , ARN Largo no Codificante/inmunología , Transporte Activo de Núcleo Celular/inmunología , Línea Celular , Humanos , Factor 3 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , MicroARNs/inmunología , Virosis/inmunología
8.
Nat Commun ; 12(1): 1736, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741957

RESUMEN

Metastasis is the leading cause of cancer-related death. Despite the recent advancements in cancer treatment, there is currently no approved therapy for metastasis. The present study reveals a potent and selective activity of PRAK in the regulation of tumor metastasis. While showing no apparent effect on the growth of primary breast cancers or subcutaneously inoculated tumor lines, Prak deficiency abrogates lung metastases in PyMT mice or mice receiving intravenous injection of tumor cells. Consistently, PRAK expression is closely associated with metastatic risk in human cancers. Further analysis indicates that loss of function of PRAK leads to a pronounced inhibition of HIF-1α protein synthesis, possibly due to reduced mTORC1 activities. Notably, pharmacological inactivation of PRAK with a clinically relevant inhibitor recapitulates the anti-metastatic effect of Prak depletion, highlighting the therapeutic potential of targeting PRAK in the control of metastasis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Neoplasias/terapia , Proteínas Serina-Treonina Quinasas/genética
9.
Cell Immunol ; 351: 104065, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32089259

RESUMEN

Many aspects remain elusive of the mechanisms governing T cell quiescence. Here we show that E protein activity helps to establish a quiescent program in naïve T cells. Decreased E protein activity, as the consequence of enforced expression of an Id1 transgene, led to the accumulation of CD4+CD44hi T cells. The naïve CD4+ T cells from this transgenic strain mounted a vigorous proliferative response upon TCR stimulation, as a result of direct inhibition of E protein activity. Transcriptome analyses demonstrated that Id1-tg naïve CD4+ T cells exhibited a transcriptional profile characteristic of activated CD4+ T cells, with particular enrichment in the gene set related to PI3K-AKT signaling. Western blot analysis confirmed low but constitutive activation of this pathway. Moreover, the Id1-tg CD4+ T cells displayed enhanced formation of TCR microcluster. Taken together, these data support that downregulation of E protein activity facilitates the exit of naïve T cells from quiescence.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Proteína 1 Inhibidora de la Diferenciación/inmunología , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología
10.
J Immunol ; 203(7): 1943-1951, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31451677

RESUMEN

Human UBL4A/GdX, encoding an ubiquitin-like protein, was shown in this study to be upregulated by viral infection and IFN stimulation. Then the functions of UBL4A in antiviral immune response were characterized. Overexpression of UBL4A promoted RNA virus-induced ISRE or IFN-ß or NF-κB activation, leading to enhanced type I IFN transcription and reduced virus replication. Consistently, knockdown of UBL4A resulted in reduced type I IFN transcription and enhanced virus replication. Additionally, overexpression of UBL4A promoted virus-induced phosphorylation of TBK1, IRF3, and IKKα/ß. Knockdown of UBL4A inhibited virus-induced phosphorylation of TBK1, IRF3, and IKKα/ß. Coimmunoprecipitation showed that UBL4A interacted with TRAF6, and this interaction was enhanced upon viral infection. Ubiquitination assays showed that UBL4A promoted the K63-linked ubiquitination of TRAF6. Therefore, we reveal a novel positive feedback regulation of UBL4A in innate immune response combating virus invasion by enhancing the K63-linked ubiquitination of TRAF6.


Asunto(s)
Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/inmunología , Macrófagos Peritoneales/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Ubiquitinación/inmunología , Ubiquitinas/inmunología , Animales , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Factor 6 Asociado a Receptor de TNF/genética , Ubiquitinación/genética , Ubiquitinas/genética , Virus/genética , Virus/inmunología
11.
Aging Dis ; 10(3): 497-509, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31164995

RESUMEN

Although autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus, are frequently associated with premature aging of the thymus, a direct link is missing between autoimmunity and thymic atrophy. Here we monitored the progression of thymic involution in Aire-deficient mice, in which defective negative selection causes spontaneous and progressive development of autoimmunity. In young and middle-aged mice, Aire deficiency appeared to be protective as supported by the reduced ß-gal+ epithelial cells and the enhanced thymic output. However, once the autoimmune phenotype was fully developed in aged Aire-deficient mice, their thymuses underwent accelerated involution. In comparison to the age-matched wildtype littermates, old Aire-deficient mice showed lower numbers of total thymocytes and recent thymic emigrants but more ß-gal+ thymic epithelial cells. This phenomenon may partly be attributable to the increased number of activated Th1 cells homing to the thymus. This speculation was further supported by the enhanced thymic aging following repeated challenges with complete Freund's adjuvant immunization. Taken together, the present study highlights a unique mechanism by which autoimmunity facilitates the senescence of thymic epithelial cells through returning Th1 cells.

12.
FASEB J ; 33(9): 10140-10151, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31238017

RESUMEN

Exposure to space environment induces alterations in glucose and lipid metabolism that contribute to muscular atrophy, bone loss, and cardiovascular disorders. Intestinal microbiota is also changed, but its impact on spaceflight-related metabolic disorder is not clear. We investigated the relationship between glucose metabolic changes and gut dysbiosis in a hind limb-unloading (HU) mouse model, a well-accepted ground-based spaceflight analog. Impaired body weight gain, glucose intolerance, and peripheral insulin resistance were found in 2-4-wk HU mice. Reduced abundance of gut Bifidobacterium spp. and Akkermansia muciniphila was observed within 3 d of HU. The ground-based control (Ctrl) mice that were cohoused with HU mice showed similar patterns of dysbiosis and metabolic changes. Compared with the Ctrls, higher levels of plasma LPS-binding protein and altered transcription of Tnfa and glucose metabolism-related genes in the liver were observed in HU mice. The supplementation of Bifidobacterium spp. suppressed endotoxemia and liver inflammation and improved glucose tolerance in HU mice. The results indicate a close relationship between dysbiosis and altered glucose metabolism in the HU model and also emphasize the importance of evaluating intestinal microbiota in astronauts and its effect on glucose metabolism.-Wang, Y., Zhao, W., Shi, J., Wang, J., Hao, J., Pang, X., Huang, X., Chen, X., Li, Y., Jin, R., Ge, Q. Intestinal microbiota contributes to altered glucose metabolism in simulated microgravity mouse model.


Asunto(s)
Disbiosis/metabolismo , Microbioma Gastrointestinal/fisiología , Intolerancia a la Glucosa/etiología , Glucosa/metabolismo , Ingravidez , Proteínas de Fase Aguda , Akkermansia , Animales , Bifidobacterium/aislamiento & purificación , Proteínas Portadoras/sangre , Corticosterona/sangre , Endotoxemia/prevención & control , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Inclinación de Cabeza , Hepatitis/prevención & control , Vivienda para Animales , Resistencia a la Insulina , Hígado/metabolismo , Masculino , Glicoproteínas de Membrana/sangre , Ratones , Ratones Endogámicos C57BL , Norepinefrina/sangre , Probióticos , Distribución Aleatoria , Organismos Libres de Patógenos Específicos , Verrucomicrobia/aislamiento & purificación , Ingravidez/efectos adversos
13.
Int J Med Sci ; 15(14): 1702-1712, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30588194

RESUMEN

Cancer-testis antigen MAGEA3, being restrictedly expressed in testis and various kinds of tumors, has long been considered as an ideal target for immunotherapy. In this study, we report that MAGEA3 interacts with STAT1 and regulates the expression of tyrosine phosphorylated STAT1 (pY-STAT1) in tumor cells. We show that pY-STAT1 is significantly up-regulated when MAGEA3 is silenced by MAGEA3-specific siRNA. RNA sequencing analysis identified 274 STAT1-related genes to be significantly altered in expression level in MAGEA3 knockdown cells. Further analysis of these differentially expressed genes with GO enrichment and KEGG pathway revealed that they are mainly enriched in plasma membrane, extracellular region and MHC class I protein complex, and involved in the interferon signaling pathways, immune response, antigen presentation and cell chemotaxis. The differentially expressed genes associated with chemokines, antigen presentation and vasculogenic mimicry formation were validated by biological experiments. Matrigel matrix-based tube formation assay showed that silencing MAGEA3 in tumor cells impairs tumor vasculogenic mimicry formation. These data indicate that MAGEA3 expression in tumor cells is associated with immune cells infiltration into tumor microenvironment and anti-tumor immune responses, implying that it may play an important role in tumor immune escape. Our findings reveal the potential impact of MAGEA3 on the immunosuppressive tumor microenvironment and will provide promising strategies for improving the efficacy of MAGEA3-targeted immunotherapy.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/inmunología , Factor de Transcripción STAT1/metabolismo , Escape del Tumor , Microambiente Tumoral/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/inmunología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias/patología , Fosforilación , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT1/inmunología , Transducción de Señal/inmunología , Tirosina/metabolismo , Regulación hacia Arriba
14.
FEBS Lett ; 592(14): 2444-2457, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29931672

RESUMEN

Parainfluenza virus infection is a common respiratory illness in children. Although lncRNAs are novel regulators of virus-induced innate immunity, a systemic attempt to characterize the differential expression of lncRNAs upon parainfluenza virus infection is lacking. In this report, we identify 207 lncRNAs and 166 mRNAs differentially expressed in SeV-infected HEK293T cells by microarray. The functional annotation analysis reveals that differentially regulated transcripts are predominantly involved in the host antiviral response pathway. The lncRNAs with the potential to regulate SeV-induced antiviral response are identified by building the lncRNA-mRNA coexpression network. Furthermore, silencing lncRNA ENST00000565297 results in reduced type I IFN signaling upon SeV infection. These catalogs may facilitate future analysis of the functions of lncRNAs in innate immunity and related diseases.


Asunto(s)
Inmunidad Innata/genética , Infecciones por Paramyxoviridae/genética , ARN Largo no Codificante/fisiología , Niño , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Paramyxoviridae/inmunología , ARN Largo no Codificante/genética , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/inmunología , Virus Sendai/inmunología , Virus Sendai/patogenicidad , Transcriptoma
15.
FASEB J ; 32(10): 5238-5249, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29688809

RESUMEN

Ubiquitination and deubiquitination are important post-translational regulatory mechanisms responsible for fine tuning the antiviral signaling. In this study, we identified a deubiquitinase, the ubiquitin-specific peptidase 7/herpes virus associated ubiquitin-specific protease (USP7/HAUSP) as an important negative modulator of virus-induced signaling. Overexpression of USP7 suppressed Sendai virus and polyinosinic-polycytidylic acid and poly(deoxyadenylic-deoxythymidylic)-induced ISRE and IFN-ß activation, and enhanced virus replication. Knockdown or knockout of endogenous USP7 expression had the opposite effect. Coimmunoprecipitation assays showed that USP7 physically interacted with tripartite motif (TRIM)27. This interaction was enhanced after SeV infection. In addition, TNF receptor-associated factor family member-associated NF-kappa-B-binding kinase (TBK)-1 was pulled down in the TRIM27-USP7 complex. Overexpression of USP7 promoted the ubiquitination and degradation of TBK1 through promoting the stability of TRIM27. Knockout of endogenous USP7 led to enhanced TRIM27 degradation and reduced TBK1 ubiquitination and degradation, resulting in enhanced type I IFN signaling. Our findings suggest that USP7 acts as a negative regulator in antiviral signaling by stabilizing TRIM27 and promoting the degradation of TBK1.-Cai, J., Chen, H.-Y., Peng, S.-J., Meng, J.-L., Wang, Y., Zhou, Y., Qian, X.-P., Sun, X.-Y., Pang, X.-W., Zhang, Y., Zhang, J. USP7-TRIM27 axis negatively modulates antiviral type I IFN signaling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Interferón Tipo I/metabolismo , Proteínas Nucleares/metabolismo , Infecciones por Respirovirus/metabolismo , Virus Sendai/metabolismo , Transducción de Señal , Peptidasa Específica de Ubiquitina 7/metabolismo , Proteínas de Unión al ADN/genética , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/genética , Proteínas Nucleares/genética , Proteolisis , Infecciones por Respirovirus/genética , Virus Sendai/genética , Peptidasa Específica de Ubiquitina 7/genética , Ubiquitinación
16.
J Cancer ; 7(6): 656-63, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27076846

RESUMEN

Cancer/testis antigen HCA587/MAGE-C2 has been considered as a tumor specific target for immunotherapy. It has been reported that HCA587/MAGE-C2 plays an active role in tumorigenesis by promoting the growth and survival of tumor cells. However, the regulation of HCA587/MAGE-C2 expression in cancer cells remains largely unknown. MicroRNAs (miRNAs), a large family of gene regulators, have been shown to negatively regulate the expression of important cancer-related genes and contribute to the initiation and development of cancers. In this study, we conducted searches of miRNAs that regulate HCA587/MAGE-C2 expression. We combined bioinformatics tools with biological validation assays to demonstrate that HCA587/MAGE-C2 is a direct target of microRNA-874 (miR-874). Furthermore, we investigated the expression levels of miR-874 in human hepatocellular carcinoma tissues and paired adjacent normal tissues by stem-loop reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The results revealed a significant downregulation of miR-874 expression in tumor tissues compared to adjacent normal tissues. Finally, we demonstrated that overexpression of miR-874, as well as HCA587/MAGE-C2 silencing, resulted in suppression of tumor cell proliferation and invasion. Moreover, the inhibition effects of miR-874 on cell proliferation and invasion were reversed by co-expression of HCA587/MAGE-C2 in A375 cells. Taken together, our data demonstrated that HCA587/MAGE-C2 is a direct target of miR-874, and miR-874 may function as a tumor suppressive miRNA, at least in part, by negatively regulating HCA587/MAGE-C2 expression in cancer cells.

17.
FASEB J ; 29(8): 3263-73, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25877215

RESUMEN

The immune systems can be altered by spaceflight in many aspects, but microgravity-related mucosal immune changes and its clinical significance have not been well studied. The purpose of this study was to investigate whether simulated microgravity influences the intestinal homeostasis and increases the susceptibility to colon inflammation. The hindlimb unloading (HU) mouse model was used to simulate the microgravity condition. Three percent dextran sulfate sodium (DSS) was given to mice to induce colitis. Compared to ground control (Ctrl) mice, the HU ones revealed an impaired intestinal homeostasis and increased susceptibility to DSS-induced colitis. This includes an early-onset, 4-fold expansion of segmented filamentous bacteria (SFB), more than 2-fold decrease in regulatory T (Treg) cell numbers and IL-10 production, ∼2-fold increase in colonic IL-1ß expression, 2-fold increase in circulating neutrophils, and colonic neutrophil infiltration. The application of antibiotics ameliorated the Treg and IL-10 reductions but did not significantly dampen neutrophilia and elevated expression of colonic IL-1ß. These results indicate that the intestinal microflora and innate immune system both respond to simulated microgravity and together, contribute to the proinflammatory shift in the gut microenvironment. The data also emphasize the necessity for evaluating the susceptibility to inflammatory bowel diseases (IBDs) in distant space travels.


Asunto(s)
Colitis/etiología , Susceptibilidad a Enfermedades/etiología , Homeostasis/fisiología , Intestinos/fisiología , Animales , Colitis/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Femenino , Inflamación/etiología , Inflamación/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Simulación de Ingravidez/métodos
18.
Immunol Cell Biol ; 93(8): 744-52, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25753394

RESUMEN

A key process in the development of T lymphocyte in the thymus is T-cell receptor (TCR) selection. It is controlled by complex signaling pathways that contain redox-sensitive molecules. However, the redox status early after TCR selection and how redox regulators promote the survival of post-selected DP thymocytes has not been directly addressed. The present study demonstrated that the transition from pre- to post-selected double-positive (DP) stages was accompanied with an increase of reactive oxygen species (ROS) and a transient surge in the expression of a variety of redox regulators. Among them, the thioredoxin (Trx)1/thioredoxin reductase (TrxR)1 system was found to be critically involved in the regulation of cell survival of DP thymocytes, especially that of post-selected CD69(+) subset, as its inhibition caused a specific reduction of these cells both in vitro and in vivo, most likely owing to increased apoptosis. Suppression of the glutathione-dependent redox system, on the other hand, showed no obvious impact. Biochemically, treatment of DP thymcoytes with TrxR1 inhibitor alone or in conjunction with anti-CD3 resulted in enhanced phosphorylation of redox-sensitive ASK-1, JNK and p38 MAPK, and upregulated expression of Bim. Taken together, the data presented here suggest that the timely upregulation of Trx1/TrxR1 and the active control of intracellular redox status is critical for the survival of thymocytes during and short after positive selection.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Tiorredoxina Reductasa 1/metabolismo , Tiorredoxinas/metabolismo , Timocitos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Expresión Génica , Ratones , Modelos Biológicos , Oxidación-Reducción , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Timocitos/citología , Timocitos/inmunología , Técnicas de Cultivo de Tejidos
19.
J Autoimmun ; 56: 87-97, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25468259

RESUMEN

Mechanisms of peripheral tolerance play a critical role in preventing T cells that escape from negative selection in the thymus from initiating autoimmune reactions. To investigate the site of peripheral tolerance induction, we examined migration and activation of recent thymic emigrants (RTEs) in liver, spleen, lymph node and peripheral blood. We show that a fraction of RTE precursors were retained in the liver independent of the secondary lymphoid organs. Compared to RTEs from the lymph nodes, RTEs from the liver proliferated more and many exhibited an activated phenotype with the capability of producing IL-10 upon activation. Liver RTEs also responded poorly to interleukin (IL)-7 and were more prone to apoptosis. Following transfer into RAG(-/-) recipients, liver RTEs induced more severe inflammation and T cell infiltration in the lung and colon. The extrathymic expression of MHC and Aire is required for the acquisition of tolerogenic phenotype of newly generated thymic emigrants in the liver. These results suggest that the liver is the first checkpoint in the periphery to filter, retain, and enforce tolerance to autoreactive CD4(+) thymic emigrants that escape from negative selection.


Asunto(s)
Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Movimiento Celular/inmunología , Tolerancia Inmunológica , Hígado/inmunología , Subgrupos de Linfocitos T/inmunología , Timocitos/inmunología , Animales , Antígenos de Superficie/metabolismo , Autoinmunidad/genética , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/inmunología , Antígenos de Histocompatibilidad/inmunología , Tolerancia Inmunológica/genética , Interleucina-10/metabolismo , Interleucina-7/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Fenotipo , Subgrupos de Linfocitos T/metabolismo , Timocitos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína AIRE
20.
J Gastroenterol ; 49(3): 419-26, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23604623

RESUMEN

BACKGROUND: To explore the potential application of placenta-specific PLAC1/Cancer Placenta (CP) 1 antigen for immunotherapy in CRC patients, further identification of the cytotoxic T lymphocyte epitopes from this antigen is necessary. METHODS: We assessed the protein expression of PLAC1/CP1 using a tissue chip and immunochemistry staining in CRC samples. Simultaneously, we predicted four PLAC1/CP1-derived HLA-A*0201-restricted peptides by using reverse immunology methods. Peptide-specific CD8(+) T cell responses were assessed by an IFN-γ release ELISPOT assay. Effector CD8(+) T cells lyse HLA-A*0201 CRC cell line SW620 was detected in a granzyme-B release ELISPOT cytotoxicity assay. RESULTS: Our results indicated that PLAC1/CP1 was highly expressed in 56.7 % (55/97) of adenocarcinomas. PLAC1/CP1 protein expression was associated with CRC tumor differentiation, the tumor/node/metastasis stage, and lymph node metastasis. Two of four peptides showed high affinities in an HLA-A2 binding assay. In 66.7 % (6/9) of peripheral blood mononuclear cells of CRC samples with PLAC1/CP1 protein-positive expression, these two peptides, PLAC1/CP1 p41-50 (FMLNNDVCV) and PLAC1/CP1 p69-77 (HAYQFTYRV), were immunogenic in the induction of peptide-specific CD8(+) T cell responses as assessed by an IFN-γ release ELISPOT assay. Furthermore, the generated effector CD8(+) T cells could specifically lyse the PLAC1/CP1 HLA-A*0201 CRC cell line SW620 in a granzyme-B release ELISPOT cytotoxicity assay. CONCLUSIONS: These results show that the PLAC1/CP1 antigen is a possible prognostic marker of CRC and that PLAC1/CP1 p41-50 and PLAC1/CP1 p69-77 are novel HLA-A*0201-restricted CD8(+) T cell epitopes and potential targets for peptide-based immunotherapy in CRC patients.


Asunto(s)
Neoplasias Colorrectales/inmunología , Antígeno HLA-A2/inmunología , Inmunoterapia/métodos , Proteínas Gestacionales/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Ensayo de Immunospot Ligado a Enzimas , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...