Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Immunol Lett ; 259: 30-36, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37247788

RESUMEN

BACKGROUND: Systemic lupus erythematosus (SLE) is one of the most common autoimmune diseases in China. At present, there are hundreds of autoantibodies in SLE patients; however, only a dozen of the autoantibodies can be routinely detected, and the available diagnostic antibodies are not sufficient for diagnosis or differential diagnosis of SLE patients with atypical clinical manifestations or other autoimmune diseases. Therefore, it is necessary to find new diagnostic markers to improve the diagnostic effect of SLE. METHODS: The displayed random peptide library and peptide microarray were combined to identify SLE-related epitope peptides. A case-control design was used. The IgG antibodies in the sera from SLE patients, healthy controls, and other autoimmune disease controls underwent a reaction with the phage-display random peptide library, respectively. Selected epitope peptides were used to construct a peptide chip. A total of 644 serum samples (including 296 SLE patients, 168 disease controls, and 180 healthy controls) were used for further screening and verification. Peptides with an area under the curve (AUC) > 0.650 were further verified by ELISA. Finally, 500 serum samples (including 200 SLE patients, 150 disease controls, and 150 healthy controls) were used to verify and evaluate the diagnostic and differential diagnostic efficacy of the selected peptides. RESULTS: After the previous screening, five epitope peptides (SLE_P19, SLE_P20, SLE_P27, SLE_P28, and SLE_P29) may have potential as SLE diagnostic markers. Additionally, SLE_P27 was superior to the other four peptides in the diagnosis and differential diagnosis of SLE and rheumatoid arthritis (RA). The AUC of SLE_P27 was 0.938, the sensitivity was 76.00%, the specificity was 92.70%, the positive likelihood ratio was 10.411, the negative likelihood ratio was 0.259, and the accuracy was 84.40%. The diagnostic efficacy of SLE can be increased by combining the five selected peptides with the anti-double stranded DNA antibody (anti-dsDNA) and anti-Smith antibodies (anti-Sm). CONCLUSIONS: In this study, we identified five peptides that may serve as potential biomarkers for SLE diagnosis using the strategy of combining the displayed random peptide library with the peptide microarray. The combination of selected peptides and existing autoantibodies can significantly improve the diagnostic efficiency. These specific peptides are expected to be new diagnostic markers for SLE.


Asunto(s)
Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Humanos , Epítopos , Biblioteca de Péptidos , Péptidos , Autoanticuerpos
2.
FASEB J ; 35(5): e21535, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33817835

RESUMEN

Thymic epithelial cells (TECs) are indispensable for T cell development, T cell receptor (TCR) repertoire selection, and specific lineage differentiation. Medullary thymic epithelial cells (mTECs), which account for the majority of TECs in adults, are critical for thymocyte selection and self-tolerance. CD74 is a nonpolymorphic transmembrane glycoprotein of major histocompatibility complex class II (MHCII) that is expressed in TECs. However, the exact role of CD74 in regulating the development of mTEC is poorly defined. In this research, we found that loss of CD74 resulted in a significant diminution in the medulla, a selective reduction in the cell number of mature mTECs expressing CD80 molecules, which eventually led to impaired thymic CD4+ T cell development. Moreover, RNA-sequence analysis showed that CD74 deficiency obviously downregulated the canonical nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway in mTECs. Our results suggest that CD74 positively controls mTEC cellularity and maturation partially by activating the canonical NF-κB signaling pathway.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/fisiología , Diferenciación Celular , Células Epiteliales/patología , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/fisiología , Activación de Linfocitos/inmunología , FN-kappa B/metabolismo , Timo/patología , Animales , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Transducción de Señal , Timo/inmunología , Timo/metabolismo
3.
Front Immunol ; 11: 628464, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329618

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2019.03099.].

4.
J Biol Chem ; 294(37): 13740-13754, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31346035

RESUMEN

Seminal amyloid fibrils are made up of naturally occurring peptide fragments and are key targets for the development of combination microbicides or antiviral drugs. Previously, we reported that the polysulfonic compound ADS-J1 is a potential candidate microbicide that not only inhibits HIV-1 entry, but also seminal fibrils. However, the carcinogenic azo moieties in ADS-J1 preclude its clinical application. Here, we screened several ADS-J1-like analogs and found that the antiparasitic drug suramin most potently inhibited seminal amyloid fibrils. Using various biochemical methods, including Congo red staining, CD analysis, transmission EM, viral infection assays, surface plasmon resonance imaging, and molecular dynamics simulations, we investigated suramin's inhibitory effects and its putative mechanism of action. We found that by forming a multivalent interaction, suramin binds to proteolytic peptides and mature fibrils, thereby inhibiting seminal fibril formation and blocking fibril-mediated enhancement of viral infection. Of note, suramin exhibited potent anti-HIV activities, and combining suramin with several antiretroviral drugs produced synergistic effects against HIV-1 in semen. Suramin also displayed a good safety profile for vaginal application. Moreover, suramin inhibited the semen-derived enhancer of viral infection (SEVI)/semen-mediated enhancement of HIV-1 transcytosis through genital epithelial cells and the subsequent infection of target cells. Collectively, suramin has great potential for further development as a combination microbicide to reduce the spread of the AIDS pandemic by targeting both viral and host factors involved in HIV-1 sexual transmission.


Asunto(s)
Amiloide/efectos de los fármacos , Semen/efectos de los fármacos , Suramina/farmacología , Adulto , Animales , Fármacos Anti-VIH/farmacología , Antirretrovirales/farmacología , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Voluntarios Sanos , Humanos , Masculino , Fragmentos de Péptidos/metabolismo , Péptidos/metabolismo , Conejos , Semen/metabolismo , Suramina/metabolismo
5.
EBioMedicine ; 45: 58-69, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31202814

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs) have been identified as regulators of a number of developmental and tumorigenic processes. However, the functions of most lncRNAs in glioma remain unknown and the mechanisms governing the proliferation of tumor cells remain poorly defined. METHODS: Both in vitro and in vivo assays were performed to investigate the roles of lncRNAs in the pathophysiology of gliomas. lncRNA arrays were used to identify differentially expressed lncRNAs. Subcutaneous tumor formation and a brain orthotopic tumor model in nude mice were used to investigate the functions of lncRNAs in vivo. The in vitro functions of lncRNAs were analyzed by fluorescence-activated cell sorting, colony formation, and western blot analyses. RNA fluorescence in situ hybridization and immunoprecipitation were used to explore the underlying mechanisms. FINDINGS: Here, we describe the newly discovered noncoding RNA RP11-732M18.3, which is highly overexpressed in glioma cells and interacts with 14-3-3ß/α to promote glioma growth, acting as an oncogene. Overexpression of lncRNA RP11-732 M18.3 was associated with the proliferation of glioma cells and tumor growth in vitro and in vivo. Remarkably, lncRNA RP11-732M18.3 promoted cell proliferation and G1/S cell cycle transition. lncRNA RP11-732M18.3 is predominately localized in the cytoplasm. Mechanistically, the interaction of lncRNA RP11-732M18.3 with 14-3-3ß/α increases the degradation of the p21 protein. lncRNA RP11-732M18.3 promoted the recruitment of ubiquitin-conjugating enzyme E2 E1 to 14-3-3ß/α and the binding of 14-3-3ß/α with ubiquitin-conjugating enzyme E2 E1 (UBE2E1) promoted the degradation of p21. INTERPRETATION: Overall these data demonstrated that lncRNA RP11-732M18.3 regulates glioma growth through a newly described lncRNA-protein interaction mechanism. The inhibition of lncRNA RP11-732M18.3 could provide a novel therapeutic target for glioma treatment.


Asunto(s)
Proteínas 14-3-3/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Glioma/tratamiento farmacológico , ARN Largo no Codificante/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , Xenoinjertos , Humanos , Hibridación Fluorescente in Situ , Ratones , Terapia Molecular Dirigida , Unión Proteica/genética , Proteolisis , Enzimas Ubiquitina-Conjugadoras/genética
6.
Clin Sci (Lond) ; 133(7): 821-838, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30872359

RESUMEN

Intestinal dysbiosis is implicated in Systemic Lupus Erythematosus (SLE). However, the evidence of gut microbiome changes in SLE is limited, and the association of changed gut microbiome with the activity of SLE, as well as its functional relevance with SLE still remains unknown. Here, we sequenced 16S rRNA amplicon on fecal samples from 40 SLE patients (19 active patients, 21 remissive patients), 20 disease controls (Rheumatoid Arthritis (RA) patients), and 22 healthy controls (HCs), and investigated the association of functional categories with taxonomic composition by Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). We demonstrated SLE patients, particularly the active patients, had significant dysbiosis in gut microbiota with reduced bacterial diversity and biased community constitutions. Amongst the disordered microbiota, the genera Streptococcus, Campylobacter, Veillonella, the species anginosus and dispar, were positively correlated with lupus activity, while the genus Bifidobacterium was negatively associated with the disease activity. PICRUSt analysis showed metabolic pathways were different between SLE and HCs, and also between active and remissive SLE patients. Moreover, we revealed that a random forest model could distinguish SLE from RA and HCs (area under the curve (AUC) = 0.792), and another random forest model could well predict the activity of SLE patients (AUC = 0.811). In summary, SLE patients, especially the active patients, show an apparent dysbiosis in gut microbiota and its related metabolic pathways. Amongst the disordered microflora, four genera and two species are associated with lupus activity. Furthermore, the random forest models are able to diagnose SLE and predict disease activity.


Asunto(s)
Bacterias/crecimiento & desarrollo , Microbioma Gastrointestinal , Intestinos/microbiología , Lupus Eritematoso Sistémico/microbiología , Adulto , Bacterias/genética , Estudios de Casos y Controles , Disbiosis , Heces/microbiología , Femenino , Interacciones Huésped-Patógeno , Humanos , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/terapia , Masculino , Persona de Mediana Edad , Inducción de Remisión , Ribotipificación , Adulto Joven
7.
Front Immunol ; 10: 3099, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32082299

RESUMEN

The thymus is the primary lymphoid organ responsible for the generation and maturation of T cells. Thymic epithelial cells (TECs) account for the majority of thymic stromal components. They are further divided into cortical and medullary TECs based on their localization within the thymus and are involved in positive and negative selection, respectively. Establishment of self-tolerance in the thymus depends on promiscuous gene expression (pGE) of tissue-restricted antigens (TRAs) by TECs. Such pGE is co-controlled by the autoimmune regulator (Aire) and forebrain embryonic zinc fingerlike protein 2 (Fezf2). Over the past two decades, research has found that TECs contribute greatly to thymopoiesis and T cell development. In turn, signals from T cells regulate the differentiation and maturation of TECs. Several signaling pathways essential for the development and maturation of TECs have been discovered. New technology and animal models have provided important observations on TEC differentiation, development, and thymopoiesis. In this review, we will discuss recent advances in classification, development, and maintenance of TECs and mechanisms that control TEC functions during thymic involution and central tolerance.


Asunto(s)
Diferenciación Celular , Células Epiteliales/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Timocitos/citología , Timocitos/metabolismo , Timo/citología , Timo/fisiología , Animales , Biomarcadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunofenotipificación , Linfopoyesis , FN-kappa B/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
8.
J Clin Invest ; 129(3): 1115-1128, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30589415

RESUMEN

Noncoding RNAs are emerging as important players in gene regulation and disease pathogeneses. Here, we show that a previously uncharacterized long noncoding RNA, nexilin F-actin binding protein antisense RNA 1 (NEXN-AS1), modulates the expression of the actin-binding protein NEXN and that NEXN exerts a protective role against atherosclerosis. An expression microarray analysis showed that the expression of both NEXN-AS1 and NEXN was reduced in human atherosclerotic plaques. In vitro experiments revealed that NEXN-AS1 interacted with the chromatin remodeler BAZ1A and the 5' flanking region of the NEXN gene and that it also upregulated NEXN expression. Augmentation of NEXN-AS1 expression inhibited TLR4 oligomerization and NF-κB activity, downregulated the expression of adhesion molecules and inflammatory cytokines by endothelial cells, and suppressed monocyte adhesion to endothelial cells. These inhibitory effects of NEXN-AS1 were abolished by knockdown of NEXN. In vivo experiments using ApoE-knockout mice fed a Western high-fat diet demonstrated that NEXN deficiency promoted atherosclerosis and increased macrophage abundance in atherosclerotic lesions, with heightened expression of adhesion molecules and inflammatory cytokines, whereas augmented NEXN expression deterred atherosclerosis. Patients with coronary artery disease were found to have lower blood NEXN levels than healthy individuals. These results indicate that NEXN-AS1 and NEXN represent potential therapeutic targets in atherosclerosis-related diseases.


Asunto(s)
Aterosclerosis/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteínas de Microfilamentos/biosíntesis , Placa Aterosclerótica/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Ratones , Ratones Noqueados para ApoE , Proteínas de Microfilamentos/genética , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , ARN Largo no Codificante/genética , Células THP-1
9.
Exp Ther Med ; 16(4): 3728-3734, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30233732

RESUMEN

Regulatory T cells (Tregs) serve an important role in the pathogenesis of rheumatoid arthritis (RA) by regulating autoimmunity and inflammation. Humans and mice contain inducible T-cell costimulator-positive (ICOS+) Tregs, although their role in RA is unclear. A total of 33 patients with RA and 17 normal control (NC) subjects were examined. The proportion of ICOS+ Tregs in the peripheral blood and intracellular cytokine levels in these cells were assessed using flow cytometry. The percentage of ICOS+ Tregs increased in the cohort of patients with RA compared with the NCs. Such increases were much larger in patients with inactive RA compared with patients with active RA. Additionally, ICOS+ Tregs expressed multiple suppressive cytokines, including interleukin (IL)-10, transforming growth factor-ß and IL-35, but expressed low levels of IL-17. Importantly, the expression of suppressive cytokines in ICOS+ Tregs from patients with active RA decreased, but IL-17 expression noticeably increased compared with patients with inactive RA. The present findings suggested that ICOS+ Tregs may perform inflammatory and inhibitory functions, and abnormal ICOS+ Tregs numbers and functions may contribute to the pathogenesis of RA.

10.
Int J Oncol ; 53(3): 973-986, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30015880

RESUMEN

Glioma is the most prevalent and fatal primary tumor of the central nervous system in adults, while the development of effective therapeutic strategies in clinical practice remain a challenge. Nucleotide-binding domain leucine-rich family pyrin-containing 3 (NLRP3) has been reported to be associated with tumorigenesis and progression; however, its expression and function in human glioma remain unclear. The present study was designed to explore the biological role and potential mechanism of NLRP3 in human glioma. The results demonstrated that overexpression of NLRP3, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), caspase­1 and interleukin (IL)­1ß protein in human glioma tissues were significantly correlated with higher World Health Organization grades. The in vitro biological experiments demonstrated that NLRP3 downregulation significantly inhibited the proliferation, migration and invasion, and promoted the apoptosis of SHG44 and A172 glioma cell lines. Furthermore, western blot assays revealed that the downregulation of NLRP3 significantly reduced the expression of ASC, caspase­1 and IL­1ß protein. Furthermore, NLRP3 knockdown caused the inhibition of epithelial-mesenchymal transition (EMT), and inhibited the phosphorylation of AKT serine/threonine kinase (AKT) and phosphorylation of phosphatase and tensin homolog (PTEN). Consistently, the upregulation of NLRP3 significantly increased the expression of ASC, caspase­1, IL­1ß and phosphorylated-PTEN, promoted proliferation, migration, invasion and EMT, inhibited apoptosis, and activated the AKT signaling pathway. The data of the present study indicate that NLRP3 affects human glioma progression and metastasis through multiple pathways, including EMT and PTEN/AKT signaling pathway regulation, enhanced inflammasome activation, and undefined inflammasome-independent mechanisms. Understanding the biological effects of NLRP3 in human glioma and the underlying mechanisms may offer novel insights for the development of glioma clinical therapeutic strategies.


Asunto(s)
Neoplasias del Sistema Nervioso Central/patología , Transición Epitelial-Mesenquimal/genética , Glioma/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal/genética , Adolescente , Adulto , Anciano , Apoptosis/genética , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Niño , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Masculino , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Clasificación del Tumor , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba , Adulto Joven
11.
J Cell Mol Med ; 22(1): 497-510, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28922548

RESUMEN

Current glioma therapies allow in situ delivery of cytotoxic drugs to the tumour; however, gliomas show early recurrence due to their highly proliferative character. Long non-coding (lnc)RNAs play critical roles in tumorigenesis by controlling cell proliferation and cycling. However, the mechanism of action of lncRNAs in glioma development remains unclear. Here, we report that the lncRNA PLAC2 induces cell cycle arrest by targeting ribosomal protein (RP)L36 in glioma. RPL36 promoted cell proliferation and G1/S cell cycle progression. Mass spectrometry analysis revealed that signal transducer and activator of transcription (STAT)1 interacted with both lncRNA PLAC2 and the RPL36 promoter. We also found that the nucleus PLAC2 bind with STAT1 and interact with RPL36 promoters but the cytoplasmic lncRNA PLAC2 inhibited STAT1 nuclear transfer, thereby decreasing RP36 expression, inhibiting cell proliferation and inducing cell cycle arrest. These results provide evidence for a novel cell cycle regulatory network in glioma comprising the lncRNA PLAC2 along with STAT1 and RPL36 that can serve as a therapeutic target for glioma treatment.


Asunto(s)
Ciclo Celular/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , ARN Largo no Codificante/genética , Proteínas Ribosómicas/genética , Factor de Transcripción STAT1/metabolismo , Adulto , Animales , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Fase G1/genética , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , ARN Largo no Codificante/metabolismo , Proteínas Ribosómicas/metabolismo , Fase S/genética
12.
Mol Med Rep ; 17(2): 3062-3068, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29207171

RESUMEN

Ginsenoside Rh2 (G­Rh2), the main bioactive component in American ginseng, is known to exert a wide variety of biological activities. Accumulating evidence suggests that G­Rh2 inhibits cell proliferation and induces apoptosis of tumor cells. However, the possible mechanism through which G­Rh2 exerts its action on malignant glioma cells have not been completely elucidated. The findings of the present study demonstrated that G­Rh2 decreased the viability of glioma cells in a dose­ and time­dependent manner, and induced cell cycle arrest. G­Rh2­induced cell cycle arrest was accompanied by the downregulation of cyclin­dependent kinase 4 and Cyclin E. In addition, G­Rh2 markedly reduced the expression of total­ RAC­α serine/threonine­protein kinase (Akt) and the levels of phosphorylated­Akt. These findings provide mechanistic details of how G­Rh2 acts on glioma cells and suggest that G­Rh2 may function as a potential anti­cancer drug for glioma treatment.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ginsenósidos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Humanos , Fosforilación
13.
Mol Med Rep ; 14(6): 5288-5296, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27779670

RESUMEN

Early reports suggest that nuclear factor IA (NFIA) is important in the pathogenesis of glioma. Our previous study demonstrated that the long non­coding RNA (lncRNA), RP5­833A20.1, suppressed the expression of NFIA in THP­1 macrophage-derived foam cells. However, the effect and possible mechanism of RP5­833A20.1 on glioma remains to be fully elucidated, and whether the NFIA-dependent pathway is involved in its progression has not been investigated. In the present study, the mechanisms by which RP5­833A20.1 regulates the expression of NFIA in glioma were investigated. The expression levels of RP5­833A20.1 and NFIA were determined in U251 cells and clinical samples using reverse transcription­quantitative polymerase chain reaction (PCR) analysis. The effects of RP5­833A20.1 on cell proliferation, invasion, cell cycle and apoptosis were evaluated using in vitro assays. The potential changes in protein expression were investigated using western blot analysis. The methylation status of the CpG island in the NFIA promoter was determined using bisulfite PCR (BSP) sequencing. It was found that the expression of RP5­833A20.1 was downregulated, whereas the expression of NFIA was upregulated in glioma tissues, compared with corresponding adjacent nontumor tissues from 20 patients with glioma. The overexpression of RP5­833A20.1 inhibited proliferation and cell cycle progression, and induced apoptosis in the U251 cells. The mRNA and protein levels of NFIA were markedly inhibited by overexpression of RP5­833A20.1 in the U251 cells. The overexpression of RP5­833A20.1 increased the expression of microRNA­382­5p in the U251 cells. The BSP assay revealed that the overexpression of RP5­833A20.1 enhanced the methylation level of the NFIA promoter. These results demonstrated that RP5­833A20.1 inhibited tumor cell proliferation, induced apoptosis and inhibited cell­cycle progression by suppressing the expression of NFIA in U251 cells. Collectively, these results indicated RP5­833A20.1 as a novel therapeutic target for glioma.


Asunto(s)
Ciclo Celular/genética , Factores de Transcripción NFI/genética , Interferencia de ARN , ARN Largo no Codificante/genética , Adulto , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Metilación de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética
14.
Sci Rep ; 6: 34939, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27731345

RESUMEN

T-cells play an important role in promoting mucosal immunity against pathogens, but the mechanistic basis for their homeostasis in the intestine is still poorly understood. We report here that T-cell-specific deletion of mTOR results in dramatically decreased CD4 and CD8 T-cell numbers in the lamina propria of both small and large intestines under both steady-state and inflammatory conditions. These defects result in defective host resistance against a murine enteropathogen, Citrobacter rodentium, leading to the death of the animals. We further demonstrated that mTOR deficiency reduces the generation of gut-homing effector T-cells in both mesenteric lymph nodes and Peyer's patches without obviously affecting expression of gut-homing molecules on those effector T-cells. Using mice with T-cell-specific ablation of Raptor/mTORC1 or Rictor/mTORC2, we revealed that both mTORC1 and, to a lesser extent, mTORC2 contribute to both CD4 and CD8 T-cell accumulation in the gastrointestinal (GI) tract. Additionally, mTORC1 but not mTORC2 plays an important role regulating the proliferative renewal of both CD4 and CD8 T-cells in the intestines. Our data thus reveal that mTOR is crucial for T-cell accumulation in the GI tract and for establishing local adaptive immunity against pathogens.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae/inmunología , Linfocitos T/citología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Proliferación Celular , Microbioma Gastrointestinal , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Homeostasis , Inflamación , Intestinos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa , Ganglios Linfáticos Agregados/inmunología , Proteína Reguladora Asociada a mTOR/metabolismo , Linfocitos T/microbiología
15.
J Lipid Res ; 57(8): 1398-411, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27281478

RESUMEN

Accumulated evidence shows that vanin-1 (VNN1) plays a key part in glucose metabolism. We explored the effect of VNN1 on cholesterol metabolism, inflammation, apoptosis in vitro, and progression of atherosclerotic plaques in apoE(-/-) mice. Oxidized LDL (Ox-LDL) significantly induced VNN1 expression through an ERK1/2/cyclooxygenase-2/PPARα signaling pathway. VNN1 significantly increased cellular cholesterol content and decreased apoAI and HDL-cholesterol (HDL-C)-mediated efflux by 25.16% and 23.13%, respectively, in THP-1 macrophage-derived foam cells (P < 0.05). In addition, VNN1 attenuated Ox-LDL-induced apoptosis through upregulation of expression of p53 by 59.15% and downregulation of expression of B-cell lymphoma-2 127.13% in THP-1 macrophage (P < 0.05). In vivo, apoE(-/-) mice were divided randomly into two groups and transduced with lentivirus (LV)-Mock or LV-VNN1 for 12 weeks. VNN1-treated mice showed increased liver lipid content and plasma levels of TG (124.48%), LDL-cholesterol (119.64%), TNF-α (148.74%), interleukin (IL)-1ß (131.81%), and IL-6 (156.51%), whereas plasma levels of HDL-C (25.75%) were decreased significantly (P < 0.05). Consistent with these data, development of atherosclerotic lesions was increased significantly upon infection of apoE(-/-) mice with LV-VNN1. These observations suggest that VNN1 may be a promising therapeutic candidate against atherosclerosis.


Asunto(s)
Amidohidrolasas/fisiología , Aterosclerosis/enzimología , Dieta Alta en Grasa/efectos adversos , Animales , Apolipoproteínas E/genética , Apoptosis , Aterosclerosis/etiología , Células CACO-2 , Ésteres del Colesterol/metabolismo , Proteínas Ligadas a GPI/fisiología , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Metabolismo de los Lípidos , Lipoproteínas LDL/fisiología , Hígado/metabolismo , Receptores X del Hígado/metabolismo , Macrófagos/enzimología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo
16.
Arch Biochem Biophys ; 604: 27-35, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27267730

RESUMEN

BACKGROUND: Atherosclerosis is a chronic inflammatory disease and represents the leading cause of morbidity and mortality throughout the world. Accumulating evidences have showed that Dihydrocapsaicin (DHC) has been found to exert multiple pharmacological and physiological effects. Nevertheless, the effects and possible mechanism of DHC on proinflammatory response remain largely unexplained. METHODS AND RESULTS: We found that DHC markedly upregulated NFIA and suppressed NF-κB expression in THP-1 macrophages. Up-regulation of proinflammatory cytokines induced by LPS including TNF-α, IL-1ß and IL-6 were markedly suppressed by DHC treatment. We also observed that protein level of NFIA was significantly increased while NF-κB and proinflammatory cytokines were decreased by DHC treatment in apoE(-/-) mice. Lentivirus-mediated overexpression of NFIA suppressed NF-κB and proinflammatory cytokines expression both in THP-1 macrophages and plaque tissues of apoE-/- mice. Moreover, treatment with lentivirus-mediated overexpression of NFIA made the down-regulation of DHC on NF-κB and proinflammatory cytokines expression notably accentuated in THP-1 macrophages and apoE(-/-) mice. In addition, treatment with siRNA targeting NF-κB accentuated the suppression of proinflammatory cytokines by lentivirus-mediated overexpression of NFIA. CONCLUSION: These observations demonstrated that DHC can significantly decrease proinflammatory cytokines through enhancing NFIA and inhibiting NF-κB expression and thus DHC may be a promising candidate as an anti-inflammatory drug for atherosclerosis as well as other disorders.


Asunto(s)
Capsaicina/análogos & derivados , Citocinas/metabolismo , Regulación de la Expresión Génica , FN-kappa B/metabolismo , Factores de Transcripción NFI/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antiinflamatorios/química , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Capsaicina/química , Perfilación de la Expresión Génica , Humanos , Inflamación , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , ARN Interferente Pequeño/metabolismo
17.
J Immunol ; 197(1): 141-50, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27233961

RESUMEN

Thymic epithelial cells (TECs) play important roles in T cell generation. Mechanisms that control TEC development and function are still not well defined. The mammalian or mechanistic target of rapamycin complex (mTORC)2 signals to regulate cell survival, nutrient uptake, and metabolism. We report in the present study that mice with TEC-specific ablation of Rictor, a critical and unique adaptor molecule in mTORC2, display thymic atrophy, which accompanies decreased TEC numbers in the medulla. Moreover, generation of multiple T cell lineages, including conventional TCRαß T cells, regulatory T cells, invariant NKT cells, and TCRγδ T cells, was reduced in TEC-specific Rictor-deficient mice. Our data demonstrate that mTORC2 in TECs is important for normal thymopoiesis and efficient T cell generation.


Asunto(s)
Células Epiteliales/fisiología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Linfopoyesis , Complejos Multiproteicos/metabolismo , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Timo/fisiología , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/genética , Proteína Asociada al mTOR Insensible a la Rapamicina , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Serina-Treonina Quinasas TOR/genética
18.
PLoS One ; 11(3): e0152641, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27022746

RESUMEN

Promiscuous expression of tissue restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs) is crucial for negative selection of self-reactive T cells to establish central tolerance. Intercellular transfer of self-peptide-MHC complexes from mTECs to thymic dendritic cells (DCs) allows DCs to acquire TRAs, which in turn contributes to negative selection and regulatory T cell generation. However, mTECs are unlikely to express all TRAs, such as immunoglobulins generated only in B cells after somatic recombination, hyper-mutation, or class-switches. We report here that both mTECs and cortical TECs can efficiently acquire not only cell surface but also intracellular proteins from thymocytes. This reveals a previously unappreciated intercellular sharing of molecules from thymocytes to TECs, which may broaden the TRA inventory in mTECs for establishing a full spectrum of central tolerance.


Asunto(s)
Células Epiteliales/metabolismo , Espacio Extracelular/metabolismo , Proteínas/metabolismo , Timocitos/metabolismo , Timo/citología , Animales , Hematopoyesis , Proteínas Luminiscentes/metabolismo , Complejo Mayor de Histocompatibilidad , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/metabolismo
19.
PLoS Biol ; 14(2): e1002370, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26889835

RESUMEN

Thymus is crucial for generation of a diverse repertoire of T cells essential for adaptive immunity. Although thymic epithelial cells (TECs) are crucial for thymopoiesis and T cell generation, how TEC development and function are controlled is poorly understood. We report here that mTOR complex 1 (mTORC1) in TECs plays critical roles in thymopoiesis and thymus function. Acute deletion of mTORC1 in adult mice caused severe thymic involution. TEC-specific deficiency of mTORC1 (mTORC1KO) impaired TEC maturation and function such as decreased expression of thymotropic chemokines, decreased medullary TEC to cortical TEC ratios, and altered thymic architecture, leading to severe thymic atrophy, reduced recruitment of early thymic progenitors, and impaired development of virtually all T-cell lineages. Strikingly, temporal control of IL-17-producing γδT (γδT17) cell differentiation and TCRVγ/δ recombination in fetal thymus is lost in mTORC1KO thymus, leading to elevated γδT17 differentiation and rearranging of fetal specific TCRVγ/δ in adulthood. Thus, mTORC1 is central for TEC development/function and establishment of thymic environment for proper T cell development, and modulating mTORC1 activity can be a strategy for preventing thymic involution/atrophy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Complejos Multiproteicos/metabolismo , Linfocitos T/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Timo/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Quimiocinas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Endogámicos C57BL , Proteína Reguladora Asociada a mTOR , Timo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA