Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
EMBO Rep ; 24(4): e56932, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36862324

RESUMEN

Obesity is associated with metabolic disorders and chronic inflammation. However, the obesity-associated metabolic contribution to inflammatory induction remains elusive. Here, we show that, compared with lean mice, CD4+ T cells from obese mice exhibit elevated basal levels of fatty acid ß-oxidation (FAO), which promote T cell glycolysis and thus hyperactivation, leading to enhanced induction of inflammation. Mechanistically, the FAO rate-limiting enzyme carnitine palmitoyltransferase 1a (Cpt1a) stabilizes the mitochondrial E3 ubiquitin ligase Goliath, which mediates deubiquitination of calcineurin and thus enhances activation of NF-AT signaling, thereby promoting glycolysis and hyperactivation of CD4+ T cells in obesity. We also report the specific GOLIATH inhibitor DC-Gonib32, which blocks this FAO-glycolysis metabolic axis in CD4+ T cells of obese mice and reduces the induction of inflammation. Overall, these findings establish a role of a Goliath-bridged FAO-glycolysis axis in mediating CD4+ T cell hyperactivation and thus inflammation in obese mice.


Asunto(s)
Ácidos Grasos , Inflamación , Animales , Ratones , Ratones Obesos , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Glucólisis , Ubiquitina-Proteína Ligasas/metabolismo , Oxidación-Reducción
2.
Cell Commun Signal ; 22(1): 143, 2024 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383403

RESUMEN

Phase separation is a cellular phenomenon where macromolecules aggregate or segregate, giving rise to biomolecular condensates resembling "droplets" and forming distinct, membrane-free compartments. This process is pervasive in biological cells, contributing to various essential cellular functions. However, when phase separation goes awry, leading to abnormal molecular aggregation, it can become a driving factor in the development of diseases, including tumor. Recent investigations have unveiled the intricate connection between dysregulated phase separation and tumor pathogenesis, highlighting its potential as a novel therapeutic target. This article provides an overview of recent phase separation research, with a particular emphasis on its role in tumor, its therapeutic implications, and outlines avenues for further exploration in this intriguing field.


Asunto(s)
Condensados Biomoleculares , Neoplasias , Humanos , Separación de Fases
3.
PLoS Biol ; 18(10): e3000837, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33017390

RESUMEN

Amyloid-ß (Aß) accumulation in the brain is a hallmark of Alzheimer's disease (AD) pathology. However, the molecular mechanism controlling microglial Aß phagocytosis is poorly understood. Here we found that the E3 ubiquitin ligase Pellino 1 (Peli1) is induced in the microglia of AD-like five familial AD (5×FAD) mice, whose phagocytic efficiency for Aß was then impaired, and therefore Peli1 depletion suppressed the Aß deposition in the brains of 5×FAD mice. Mechanistic characterizations indicated that Peli1 directly targeted CCAAT/enhancer-binding protein (C/EBP)ß, a major transcription factor responsible for the transcription of scavenger receptor CD36. Peli1 functioned as a direct E3 ubiquitin ligase of C/EBPß and mediated its ubiquitination-induced degradation. Consequently, loss of Peli1 increased the protein levels of C/EBPß and the expression of CD36 and thus, promoted the phagocytic ability in microglial cells. Together, our findings established Peli1 as a critical regulator of microglial phagocytosis and highlighted the therapeutic potential by targeting Peli1 for the treatment of microglia-mediated neurological diseases.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Microglía/citología , Microglía/metabolismo , Proteínas Nucleares/metabolismo , Fagocitosis , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Ratones Transgénicos , Proteínas Nucleares/deficiencia , Transcripción Genética , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitinación
4.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 52(2): 139-147, 2023 Apr 25.
Artículo en Inglés, Zh | MEDLINE | ID: mdl-37283097

RESUMEN

OBJECTIVES: To construct a prognosis risk model based on long noncoding RNAs (lncRNAs) related to cuproptosis and to evaluate its application in assessing prognosis risk of bladder cancer patients. METHODS: RNA sequence data and clinical data of bladder cancer patients were downloaded from the Cancer Genome Atlas database. The correlation between lncRNAs related to cuproptosis and bladder cancer prognosis was analyzed with Pearson correlation analysis, univariate Cox regression, Lasso regression, and multivariate Cox regression. Then a cuproptosis-related lncRNA prognostic risk scoring equation was constructed. Patients were divided into high-risk and low-risk groups based on the median risk score, and the immune cell abundance between the two groups were compared. The accuracy of the risk scoring equation was evaluated using Kaplan-Meier survival curves, and the application of the risk scoring equation in predicting 1, 3 and 5-year survival rates was evaluated using receiver operating characteristic (ROC) curves. Univariate and multivariate Cox regression were used to screen for prognostic factors related to bladder cancer patients, and a prognostic risk assessment nomogram was constructed, the accuracy of which was evaluated with calibration curves. RESULTS: A prognostic risk scoring equation for bladder cancer patients was constructed based on nine cuproptosis-related lncRNAs. Immune infiltration analysis showed that the abundances of M0 macrophages, M1 macrophages, M2 macrophages, resting mast cells and neutrophils in the high-risk group were significantly higher than those in the low-risk group, while the abundances of CD8+ T cells, helper T cells, regulatory T cells and plasma cells in the low-risk group were significantly higher than those in the high-risk group (all P<0.05). Kaplan-Meier survival curve analysis showed that the total survival and progression-free survival of the low-risk group were longer than those of the high-risk group (both P<0.01). Univariate and multivariate Cox analysis showed that the risk score, age and tumor stage were independent factors for patient prognosis. The ROC curve analysis showed that the area under the curve (AUC) of the risk score in predicting 1, 3 and 5-year survival was 0.716, 0.697 and 0.717, respectively. When combined with age and tumor stage, the AUC for predicting 1-year prognosis increased to 0.725. The prognostic risk assessment nomogram for bladder cancer patients constructed based on patient age, tumor stage, and risk score had a prediction value that was consistent with the actual value. CONCLUSIONS: A bladder cancer patient prognosis risk assessment model based on cuproptosis-related lncRNA has been successfully constructed in this study. The model can predict the prognosis of bladder cancer patients and their immune infiltration status, which may also provide a reference for tumor immunotherapy.


Asunto(s)
Apoptosis , ARN Largo no Codificante , Neoplasias de la Vejiga Urinaria , Humanos , Linfocitos T CD8-positivos , Pronóstico , ARN Largo no Codificante/genética , Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/genética , Cobre
5.
J Transl Med ; 20(1): 20, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991623

RESUMEN

BACKGROUND: The aberrant expression of E3 ubiquitin ligase Pellino-1 (PELI1) contributes to several human cancer development and progression. However, its expression patterns and functional importance in papillary thyroid cancer (PTC) remains unknown. METHODS: PELI1 expression profiles in PTC tissues were obtained and analyzed through the starBase v3.0 analysis. Real-time PCR, Immunohistochemical assays (IHC) and Western blot were used to investigate the mRNA and protein levels of PELI1 in PTC. The effects of PELI1 on PTC cell progression were evaluated through CCK-8, colony formation, Transwell, and Wound healing assay in vitro, and a PTC xenograft mouse model in vivo. The downstream target signal of PELI1 in PTC was analyzed by using Kyoto encyclopedia of genes and genomes (KEGG), and bioinformatics tools were used to identify potential miRNAs targeting PELI1. Human umbilical cord mesenchymal stem cells were modified by miR-30c-5p and the miR-30c-5p containing extracellular vesicles were collected (miR-30c-5p-EVs) by ultra-high-speed centrifugation method. Then, the effects of miR-30c-5p-EVs on PELI1 expression and PTC progression were evaluated both in vitro and in vivo. RESULTS: Both mRNA and protein expression of PELI1 were widely increased in PTC tissues, and overexpression of PELI1 was positively correlated with bigger tumor size and lymph node metastases. PELI1 promoted PTC cell proliferation and migration in vitro. While, PELI1 silencing significantly suppressed PTC growth in vivo accompanied with reduced expression of Ki-67 and matrix metallopeptidase 2 (MMP-2). Mechanistically, PI3K-AKT pathway was identified as the downstream target of PELI1, and mediated the functional influence of PELI1 in PTC cells. Moreover, we found that the expression of miR-30c-5p was inversely correlated with PELI1 in PTC samples and further confirmed that miR-30c-5p was a tumor-suppressive miRNA that directly targeted PELI1 to inhibit PTC cell proliferation and migration. Furthermore, we showed that miR-30c-5p-EVs could effectively downregulate PELI1 expression and suppress the PTC cell growth in vitro and in vivo. CONCLUSION: This study not only supported the first evidence that miR-30c-5p loss-induced PELI1 accumulation facilitated cell proliferation and migration by activating the PI3K-AKT pathway in PTC but also provided novel insights into PTC therapy based on miR-carrying-hUCMSC-EVs.


Asunto(s)
MicroARNs/genética , Neoplasias de la Tiroides , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
Int J Med Sci ; 19(4): 631-639, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35582426

RESUMEN

To elucidate the effect of Schistosoma japonicum peptide (SJMHE1) on pyroptosis in thyroid follicular epithelial cells (TFCs) induced by excessive iodine and the potential mechanism, the effects of SJMHE1 were investigated in NaI-treated Nthy-ori 3-1 cells; and the involvement of the ROS/MAPK/NF-κB signaling pathways in these effects was evaluated by employing CCK-8 assays, flow cytometry, ELISA, and Western blotting experiments. We found that SJMHE1 significantly reduced NLRP3, N-terminus of gasdermin D (GSDMD-N) and cleaved caspase-1 (C-caspase-1) expression, and decreased IL-1ß secretion in TFCs. SJMHE1 also markedly reduced reactive oxygen species (ROS) production, and decreased the phosphorylation levels of MAPK and NF-κB pathway members. Moreover, blocking of the Toll-like receptor 2 significantly impaired SJMHE1-mediated protection from excessive iodine-induced pyroptosis in TFCs. Therefore, our results suggested a protective role of SJMHE1 in excessive iodine-induced pyroptosis in TFCs, which might be attributed to its suppression for ROS/MAPK/NF-κB signaling pathway by binding of SJMHE1 with TLR2.


Asunto(s)
Yodo , Piroptosis , Caspasa 1 , Células Epiteliales/metabolismo , Humanos , Yodo/farmacología , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Glándula Tiroides , Receptor Toll-Like 2/metabolismo
7.
BMC Cancer ; 21(1): 877, 2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34332557

RESUMEN

BACKGROUND: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), such as gefitinib, have achieved good efficacy in EGFR mutation-positive non-small-cell lung cancer (NSCLC) patients, but eventual drug resistance is inevitable. Thus, new TKI-based combination therapies should be urgently explored to extend the overall survival time of these patients. CD8 + CD56+ natural killer T (NKT) cells are a natural and unique subset of lymphocytes in humans that present characteristics of T and NK cells and exert cytotoxicity on tumour cells in a granzyme B-dependent manner. The aim of this trial was to explore the efficacy and safety of CD8 + CD56+ NKT cell immunotherapy combined with gefitinib in patients with advanced EGFR-mutated NSCLC. METHODS: The study was designed as a prospective, randomized, controlled, open-label, phase I/II trial that includes 30 patients with EGFR mutation-positive stage III/IV NSCLC. All patients will be randomized in blocks at a 1:1 ratio and treated with gefitinib 250 mg/day monotherapy or combination therapy with allogeneic CD8 + CD56+ NKT cell infusions twice per month for 12 cycles or until disease progression occurs. The effectiveness of this treatment will be evaluated based on by progression-free survival (PFS), the time to progression (TTP), overall response rate (ORR), disease control rate (DCR) and overall survival (OS). The safety of the trail is being assessed based on adverse events (AEs). Recruitment and data collection, which started in December 2017, are ongoing. DISCUSSION: Although immunotherapy, including programmed death-1/programmed death-1 ligand (PD-1/PD-L1) immunotherapy, has been used for NSCLC treatment with or without EGFR-TKIs, its clear efficacy still has not been shown. Assessing the safety and therapeutic potential of allogeneic CD8 + CD56+ NKT killer cells in combination with EGFR-TKIs in NSCLC will be of great interest. TRIAL REGISTRATION: This trial (Phase I/II Trails of NKT Cell in Combination With Gefitinib For Non Small Cell Lung Cancer) was registered on 21 November 2017 with www.chictr.org.cn , ChiCTR-IIR-17013471 .


Asunto(s)
Traslado Adoptivo , Carcinoma de Pulmón de Células no Pequeñas/terapia , Gefitinib/uso terapéutico , Neoplasias Pulmonares/terapia , Mutación , Células T Asesinas Naturales/inmunología , Traslado Adoptivo/efectos adversos , Traslado Adoptivo/métodos , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/etiología , Terapia Combinada , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Gefitinib/administración & dosificación , Gefitinib/efectos adversos , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/etiología , Terapia Molecular Dirigida , Células T Asesinas Naturales/metabolismo , Resultado del Tratamiento
8.
J Cell Mol Med ; 24(13): 7460-7469, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32436312

RESUMEN

Peripheral nerve injury and regeneration are complex processes and involve multiple molecular and signalling components. However, the involvement of long non-coding RNA (lncRNA) in this process is not fully clarified. In this study, we evaluated the expression of the lncRNA maternally expressed gene 3 (MEG3) in rats after sciatic nerve transection and explored its potential mechanisms. The expression of lncRNA MEG3 was up-regulated following sciatic nerve injury and observed in Schwann cells (SCs). The down-regulation of lncRNA MEG3 in SCs enhanced the proliferation and migration of SCs via the PTEN/PI3K/AKT pathway. The silencing of lncRNA MEG3 promoted the migration of SCs and axon outgrowth in rats after sciatic nerve transection and facilitated rat nerve regeneration and functional recovery. Our findings indicated that lncRNA MEG3 may be involved in nerve injury and injured nerve regeneration in rats with sciatic nerve defects by regulating the proliferation and migration of SCs. This gene may provide a potential therapeutic target for improving peripheral nerve injury.


Asunto(s)
Movimiento Celular/genética , Regulación hacia Abajo/genética , Regeneración Nerviosa/genética , ARN Largo no Codificante/metabolismo , Células de Schwann/patología , Nervio Ciático/lesiones , Nervio Ciático/fisiopatología , Animales , Axones/metabolismo , Proliferación Celular/genética , Masculino , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transporte de ARN/genética , ARN Largo no Codificante/genética , Ratas Sprague-Dawley , Recuperación de la Función , Células de Schwann/metabolismo , Transducción de Señal , Regulación hacia Arriba/genética
9.
J Cell Mol Med ; 23(4): 2822-2835, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30772948

RESUMEN

Peripheral nerve injury results in limited nerve regeneration and severe functional impairment. Mesenchymal stem cells (MSCs) are a remarkable tool for peripheral nerve regeneration. The involvement of human umbilical cord MSC-derived extracellular vesicles (hUCMSC-EVs) in peripheral nerve regeneration, however, remains unknown. In this study, we evaluated functional recovery and nerve regeneration in rats that received hUCMSC-EV treatment after nerve transection. We observed that hUCMSC-EV treatment promoted the recovery of motor function and the regeneration of axons; increased the sciatic functional index; resulted in the generation of numerous axons and of several Schwann cells that surrounded individual axons; and attenuated the atrophy of the gastrocnemius muscle. hUCMSC-EVs aggregated to rat nerve defects, down-regulated interleukin (IL)-6 and IL-1ß, up-regulated IL-10 and modulated inflammation in the injured nerve. These effects likely contributed to the promotion of nerve regeneration. Our findings indicate that hUCMSC-EVs can improve functional recovery and nerve regeneration by providing a favourable microenvironment for nerve regeneration. Thus, hUCMSC-EVs have considerable potential for application in the treatment of peripheral nerve injury.


Asunto(s)
Vesículas Extracelulares/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/terapia , Nervio Ciático/lesiones , Cordón Umbilical/citología , Animales , Células Cultivadas , Humanos , Masculino , Músculo Esquelético/lesiones , Atrofia Muscular/etiología , Atrofia Muscular/patología , Atrofia Muscular/terapia , Traumatismos de los Nervios Periféricos/etiología , Traumatismos de los Nervios Periféricos/patología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Nervio Ciático/cirugía
10.
Endocr J ; 65(9): 893-901, 2018 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-29877208

RESUMEN

Hashimoto's thyroiditis (HT) is considered a T helper-type 1 (Th1) cytokine-dominant autoimmune thyroid disease. Caveolin-1 (Cav-1), a part of the thyroxisome multiprotein complex, is localized at the apical pole of thyrocytes and is indispensable for synthesis of thyroid hormones and modulation of oxidative stress in order to avoid cell damage and apoptosis. Reduced autophagy induces thyroid follicular cells (TFC) apoptosis by activating reactive oxygen species (ROS) in HT patients. Nevertheless, whether Cav-1 has roles in the regulation of autophagy remains largely unclear. In this study, we examined Th1 cytokines and Cav-1 expression in HT thyroid tissues, determined the effects of interleukin-1beta (IL-1ß) and interferon-gamma (IFN-γ) on Cav-1 and autophagy activity in TFC, and investigated the association between Cav-1 and autophagy activity in vitro. Our results indicate that higher levels of IL-1ß and IFN-γ and lower levels of Cav-1 were expressed in thyroid tissues of HT patients than in those of normal controls. Cav-1 mRNA and protein levels were significantly decreased in TFC exposed to IL-1ß and IFN-γ, accompanied by decreased expression of autophagy-related protein LC3B-II. Interestingly, small interfering RNA (siRNA)-mediated Cav-1 knockdown in TFC reduced LC3B-II protein expression. Taken together, these results suggest that lack of Cav-1 expression inhibited autophagy activity in TFC exposed to Th1 cytokines (IL-1ß and IFN-γ), which might be a novel pathogenetic mechanism of HT.


Asunto(s)
Autofagia/genética , Caveolina 1/fisiología , Enfermedad de Hashimoto/genética , Células Epiteliales Tiroideas/fisiología , Autofagia/efectos de los fármacos , Estudios de Casos y Controles , Caveolina 1/genética , Células Cultivadas , Técnicas de Silenciamiento del Gen , Enfermedad de Hashimoto/metabolismo , Humanos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Células TH1/metabolismo , Células TH1/fisiología , Células Epiteliales Tiroideas/efectos de los fármacos , Células Epiteliales Tiroideas/metabolismo
11.
Acta Biochim Biophys Sin (Shanghai) ; 50(9): 862-868, 2018 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-30032173

RESUMEN

Noncanonical NF-κB pathway is essential for the B cell activation and antibody production, which centralize the critical role of B cells in regulating the pathogenesis of systemic lupus erythematosus (SLE). We have previously demonstrated that Pellino1 (Peli1) negatively regulates noncanonical NF-κB activation and lupus autoimmunity. Here, we showed that poly IC is a potent inducer of Peli1 protein in mouse splenic B cells in dose- and time-dependent manners, and poly IC-induced Peli1 protein dramatically suppressed the activation of noncanonical NF-κB pathway. In addition, poly IC-pretreated B cells failed to induce lupus-like disease in BM12 CD4+ T cell-immunized mice. Accordingly, the induction of antibody-producing plasma cells and germinal center B cells, as well as the production of autoantibodies were significantly impaired in immunized µMT mice that were transferred with poly IC-pretreated B cells. Our findings demonstrate that poly IC-induced Peli1 negatively regulates the noncanonical NF-κB pathway in the context of restraining the pathogenesis of lupus-like disease.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Lupus Eritematoso Sistémico/prevención & control , Proteínas Nucleares/inmunología , Poli I-C/farmacología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Autoinmunidad/genética , Autoinmunidad/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Línea Celular Tumoral , Humanos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/inmunología , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
12.
J Cell Mol Med ; 21(3): 475-486, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27677654

RESUMEN

Helminth-derived products have recently been shown to prevent the development of inflammatory diseases in mouse models. However, most identified immunomodulators from helminthes are mixtures or macromolecules with potentially immunogenic side effects. We previously identified an immunomodulatory peptide called SJMHE1 from the HSP60 protein of Schistosoma japonicum. In this study, we assessed the ability of SJMHE1 to affect murine splenocytes and human peripheral blood mononuclear cells (PBMCs) stimulated by toll-like receptor (TLR) ligands in vitro and its treatment effect on mice with collagen-induced arthritis (CIA). We show that SJMHE1 not only modulates the cytokine production of murine macrophage (MΦ) and dendritic cell but also affects cytokine production upon coculturing with allogeneic CD4+ T cell. SJMHE1 potently inhibits the cytokine response to TLR ligands lipopolysaccharide (LPS), CpG oligodeoxynucleotides (CpG) or resiquimod (R848) from mouse splenocytes, and human PBMCs stimulated by LPS. Furthermore, SJMHE1 suppressed clinical signs of CIA in mice and blocked joint erosion progression. This effect was mediated by downregulation of key cytokines involved in the pathogenesis of CIA, such as interferon-γ (IFN-γ), tumour necrosis factor-α (TNF-α), interleukin (IL)-6, IL-17, and IL-22 and up-regulation of the inhibitory cytokine IL-10, Tgf-ß1 mRNA, and CD4+ CD25+ Foxp3+ Tregs. This study provides new evidence that the peptide from S. japonicum, which is the 'safe' selective generation of small molecule peptide that has evolved during host-parasite interactions, is of great value in the search for novel anti-inflammatory agents and therapeutic targets for autoimmune diseases.


Asunto(s)
Artritis Experimental/inducido químicamente , Artritis Experimental/tratamiento farmacológico , Colágeno/farmacología , Citocinas/metabolismo , Péptidos/farmacología , Schistosoma japonicum/metabolismo , Receptores Toll-Like/metabolismo , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Linfocitos T CD4-Positivos/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Proteínas del Helminto/farmacología , Interacciones Huésped-Parásitos/efectos de los fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Regulación hacia Arriba/efectos de los fármacos
13.
J Autoimmun ; 75: 50-57, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27448770

RESUMEN

The incidence of the autoimmune thyroid disease Hashimoto thyroiditis (HT) has increased in recent years, and increasing evidence supports the contribution of excess iodine intake to thyroid disease. In this study, we examined the status of autophagy and apoptosis in thyroid tissues obtained from patients with HT, and we determined the effects of excessive iodine on the autophagy and apoptosis of thyroid follicular cells (TFCs) in an attempt to elucidate the effects of excess iodine on HT development. Our results showed decreases in the autophagy-related protein LC3B-II, and increases in caspase-3 were observed in thyroid tissues from HT patients. Interestingly, the suppression of autophagy activity in TFCs was induced by excess iodine in vitro, and this process is mediated through transforming growth factor-ß1 downregulation and activation of the Akt/mTOR signaling pathway. In addition, excess iodine induced autophagy suppression and enhanced reactive oxygen species (ROS) production and apoptosis of TFCs, which could be rescued by the activation of autophagy. Taken together, our results demonstrated that excess iodine contributed to autophagy suppression and apoptosis of TFCs, which could be important factors predisposing to increased risk of HT development.


Asunto(s)
Apoptosis/inmunología , Autofagia/inmunología , Células Epiteliales/inmunología , Enfermedad de Hashimoto/inmunología , Yodo/inmunología , Acetilcisteína/farmacología , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/metabolismo , Caspasa 3/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Enfermedad de Hashimoto/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Yodo/metabolismo , Yodo/farmacología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Sirolimus/farmacología , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/patología , Factor de Crecimiento Transformador beta1/farmacología
14.
Endocr J ; 63(7): 655-62, 2016 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-27237400

RESUMEN

The neurohypophysial hormone, oxytocin, is involved in the regulation of energy metabolism. Adiponectin (APN) is an adipose tissue-specific serum protein that inversely associates with metabolic syndrome (MetS). High-molecular-weight adiponectin (HMW APN) is considered the active form. In the present study, we aimed to determine the relationships of oxytocin and HMW APN to MetS and investigate whether or not the combination of oxytocin and HMW APN is associated with further metabolic abnormalities compared to each of them alone. A total of 170 subjects (75 with MetS and 95 non-MetS) were enrolled. Anthropometric parameters, oral glucose tolerance test (OGTT), blood lipids, hs-CRP, oxytocin and HMW APN levels were measured. Compared with non-MetS subjects, serum oxytocin and HMW APN levels were significantly lower in subjects with MetS (P<0.01). We then classified the subjects into three groups: high oxytocin and high HMW APN levels (high score group), low oxytocin and low HMW APN levels (low score group) and others. Participants in low score group showed the worst metabolic profiles and were more likely to have MetS compared to the other two group. In Spearman rank correlation coefficient, the classification by the combination of oxytocin and HMW APN was significantly correlated with a larger number of metabolic risk factors compared with classification by each of them alone. Individuals with low circulating oxytocin levels coupled with low HMW APN levels were at significantly increased risk of MetS. The combination of both markers would be useful for identifying MetS high risk patients.


Asunto(s)
Adiponectina/sangre , Síndrome Metabólico/sangre , Oxitocina/sangre , Adiponectina/química , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Síndrome Metabólico/epidemiología , Persona de Mediana Edad , Peso Molecular , Factores de Riesgo
15.
Endocr J ; 63(4): 359-65, 2016 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-26806193

RESUMEN

Fibroblast growth factor 1 (FGF1) has been recently characterized as a potent insulin sensitizer that regulates adipose tissue remodeling, but the physiological role of FGF1 remains unclear. This study measured serum FGF1 levels for the first time in patients with newly diagnosed type 2 diabetes mellitus (T2DM), and further explored the correlations between FGF1 levels and various metabolic parameters in T2DM. Serum FGF1 levels were determined using ELISA in age-, sex- and BMI- matched subjects with normal glucose tolerance (NGT) (n=80) and newly diagnosed T2DM (n=80). Oral glucose tolerance test (OGTT), glycosylated hemoglobin (HbA1C), blood lipids, and insulin secretion were also measured. Insulin resistance and pancreatic ß-cell function were assessed by homeostasis model assessment of insulin resistance (HOMA-IR) and homeostasis model assessment of beta cell function (HOMA-ß), respectively. Serum FGF1 levels were significantly higher in T2DM patients than in normal glucose tolerance subjects (74.52 [55.91∼101.34] vs. 60.31 [48.99∼83.91] pg/mL; P<0.05). In addition, serum FGF1 level positively correlated with body mass index (BMI), waist circumference (WC), waist to hip ratio (WHR), fasting plasma glucose (FPG), 2-h post-OGTT glucose (2h PG), and HbA1C (all P values <0.05) in T2DM subjects. Multivariate regression analyses showed that BMI and HbA1C were the independent factors influencing serum FGF1 levels. Logistic regression analyses demonstrated that serum FGF1 was significantly associated with type 2 diabetes (P<0.01). Circulating concentrations of FGF1 are significantly increased in T2DM patients. Our results suggest that FGF1 may play a role in the pathogenesis of T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Factor 1 de Crecimiento de Fibroblastos/sangre , Adulto , Glucemia/metabolismo , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/diagnóstico , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo , Regulación hacia Arriba
16.
Cell Immunol ; 289(1-2): 91-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24747824

RESUMEN

The INK4b-ARF-INK4a gene cluster encodes three tumor suppressors: p15(INK4b), p14(ARF), and p16(INK4a). Antisense non-coding RNA in the INK4 locus (ANRIL) is transcribed in the opposite direction from this gene cluster. Recent studies suggest that ANRIL represses the expression of p15(INK4b), p14(ARF), and p16(INK4a); however, the underlying mechanism is unclear. In this study, the expressions of ANRIL in human esophageal squamous cell carcinoma (ESCC) tissues and matched adjacent non-tumor tissues were examined by quantitative real-time polymerase chain reaction. Compared with matched adjacent non-tumor tissues, the expression levels of ANRIL in ESCC tissues were significantly increased. Furthermore, inhibition of ANRIL was found to increase the expression of p15(INK4b) and transforming growth factor ß1 (TGFß1) and depletion of ANRIL in ESCC cell lines may inhibit cellular proliferation. Thus, our findings suggest a significant role of ANRIL in the occurrence and development of ESCC through TGFß1 signaling pathways.


Asunto(s)
Carcinoma de Células Escamosas/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Neoplasias Esofágicas/genética , ARN Largo no Codificante/genética , Factor de Crecimiento Transformador beta1/genética , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/biosíntesis , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago , Humanos , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/biosíntesis , Transducción de Señal/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Proteínas Supresoras de Tumor/genética
17.
Endocr J ; 61(5): 513-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24621778

RESUMEN

Sitagliptin was used as monotherapy or in combination with metformin, thiazolidinedione or sulfonylurea. It is not clear whether effects are enhanced or unique when in combination with transient continuous subcutaneous insulin infusion (CSII) therapy. The aim of this study was to assess the safety and efficacy of sitagliptin in combination with transient CSII therapy in patients with newly diagnosed type 2 diabetes. Eighty patients with newly diagnosed type 2 diabetes from July 2011 to May 2013 were recruited into the study. These patients were randomly divided into a CSII monotherapy group (group A, n = 40) or a sitagliptin in combination with CSII therapy group (group B, n = 40) and received insulin intensive therapy. Treatments were maintained for 2 weeks. 75g oral glucose tolerance test (OGTT) was performed before and after treatments, and the levels of glucose, insulin and C-peptide were examined. The results indicated that, compared with CSII therapy group, the level of plasma glucose significantly decreased, the levels of insulin and C-peptide strikingly increased and homeostasis model assessment for beta-cell function (HOMA-ß) and Insulinogenic index (Ins index) were improved in the group of sitagliptin in combination with CSII therapy. Above all, the incidence of hypoglycemia was lower, insulin doses were less and the rate of recovery to normal glucose tolerance (NGT) or impaired glucose tolerance (IGT) determined by 75gOGTT was higher in the latter. So, Sitagliptin in combination with CSII therapy can be a new safe and effective therapy in patients with newly diagnosed type 2diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/administración & dosificación , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Pirazinas/administración & dosificación , Triazoles/administración & dosificación , Adulto , Anciano , Diabetes Mellitus Tipo 2/diagnóstico , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Quimioterapia Combinada , Femenino , Humanos , Hipoglucemiantes/efectos adversos , Infusiones Subcutáneas , Insulina/efectos adversos , Sistemas de Infusión de Insulina/efectos adversos , Masculino , Persona de Mediana Edad , Pirazinas/efectos adversos , Fosfato de Sitagliptina , Resultado del Tratamiento , Triazoles/efectos adversos
18.
Int J Mol Sci ; 15(12): 21674-86, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25429429

RESUMEN

Hashimoto's thyroiditis (HT) is an organ-specific immune disease characterized by the presence of lymphocytic infiltration and serum autoantibodies. Previous studies have confirmed the critical role of Th17 cells in the pathopoiesis of HT patients. Additionally, regulatory T cells (Treg) display a dysregulatory function in autoimmune disease. The purpose of this study is to investigate the alteration of Th17 and Treg cells in HT patients and explore contributing factors. We found there was an increased ratio of Th17/Treg in HT patients and a positive correlation with autoantibodies (anti-TgAb). In addition, there was an increased level of GITRL, which has been demonstrated to be correlated with the increassement of Th17 cells in the serum and thyroid glands of HT patients; the upregulated serum level of GITRL has a positive correlation with the percentage of Th17 cells in HT patients. In summary, an increase in GITRL may impair the balance of Th17/Treg, and contribute to the pathopoiesis of Hashimoto's thyroiditis.


Asunto(s)
Enfermedad de Hashimoto/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Factores de Necrosis Tumoral/metabolismo , Femenino , Enfermedad de Hashimoto/sangre , Enfermedad de Hashimoto/genética , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Masculino , Persona de Mediana Edad , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tiroglobulina/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/patología , Factores de Necrosis Tumoral/genética
19.
J Immunol Res ; 2024: 3604935, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38774604

RESUMEN

Objective: Immunotherapy has proven effective in treating advanced gastric cancer (AGC), yet its benefits are limited to a subset of patients. Our aim is to swiftly identify prognostic biomarkers using cytokines to improve the precision of clinical guidance and decision-making for PD-1 inhibitor-based cancer immunotherapy in AGC. Materials and Methods: The retrospective study compared 36 patients with AGC who received combined anti-PD-1 immunotherapy and chemotherapy (immunochemotherapy) with a control group of 20 patients who received chemotherapy alone. The concentrations of TNF-α, IL-1ß, IL-2R, IL-6, IL-8, IL-10, and IL-17 in the serum were assessed using chemiluminescence immunoassay at three distinct time intervals following the commencement of immunochemotherapy. Results: When compared to controls, patients undergoing immunochemotherapy demonstrated a generalized rise in cytokine levels after the start of treatment. However, patients who benefited from immunochemotherapy showed a decrease in IL-6 or IL-8 concentrations throughout treatment (with varied trends observed for IL-1ß, IL-2R, IL-10, IL-17, and TNF-α) was evident in patients benefiting from immunochemotherapy but not in those who did not benefit. Among these markers, the combination of IL-6, IL-8, and CEA showed optimal predictive performance for short-term efficacy of immunochemotherapy in AGC patients. Conclusion: Reductions in IL-6/IL-8 levels observed during immunochemotherapy correlated with increased responsiveness to treatment effectiveness. These easily accessible blood-based biomarkers are predictive and rapid and may play a crucial role in identifying individuals likely to derive benefits from PD-1 blockade immunotherapy.


Asunto(s)
Biomarcadores de Tumor , Inhibidores de Puntos de Control Inmunológico , Interleucina-6 , Interleucina-8 , Receptor de Muerte Celular Programada 1 , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/terapia , Neoplasias Gástricas/inmunología , Femenino , Masculino , Persona de Mediana Edad , Anciano , Biomarcadores de Tumor/sangre , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Interleucina-6/sangre , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Interleucina-8/sangre , Estudios Retrospectivos , Resultado del Tratamiento , Adulto , Pronóstico , Inmunoterapia/métodos , Estadificación de Neoplasias , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
20.
Discov Oncol ; 15(1): 231, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884851

RESUMEN

OBJECTIVE: To investigate the potential of group I pepsinogen (PG I) and group II pepsinogen (PG II) as diagnostic markers for recurrence in gastric cancer (GC) patients post-total gastrectomy. METHODS: Ninety-six patients who underwent total gastrectomy for GC between June 2022 and June 2023 were included in this study. Clinical data, serum samples, and ascites samples were collected. Patients were categorized based on recurrence status at the time of sample collection and the primary tumor site. PG I and PG II levels were determined using a chemiluminescent immunoassay, and their clinical utility following total gastrectomy for GC was evaluated via receiver operating characteristic (ROC) curve analysis. RESULTS: This study included 96 GC patients who underwent total gastrectomy, 55 of whom experienced postoperative recurrence (57.29%). The levels of serum PG I (27.86 (27.04, 30.97) vs. 26.05 (24.16, 27.09) ng/mL; P < 0.0001) and PG II (1.95 (1.23, 3.05) vs. 0.63 (0.47, 0.90) ng/mL; P < 0.0001) were significantly greater in the recurrent group compared to the non-recurrent group. The secretion of PG I and/or PG II by metastatic cancer cells correlated with the primary lesion site. When the cut-off value for serum PG I was 26.93 ng/mL, the area under the curve (AUC) for PG I was 0.77. When the cut-off value for serum PG II was 0.96 ng/mL, the AUC reached 0.90. The combined AUC was 0.97. CONCLUSION: These findings suggest that serum PG I and PG II are valuable biomarkers for identifying GC patients with biochemical recurrence post-total gastrectomy.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda