Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS Pathog ; 13(7): e1006503, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28732079

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are expanded in tumor microenvironments, including that of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC). The link between MDSC expansion and EBV infection in NPC is unclear. Here, we show that EBV latent membrane protein 1 (LMP1) promotes MDSC expansion in the tumor microenvironment by promoting extra-mitochondrial glycolysis in malignant cells, which is a scenario for immune escape initially suggested by the frequent, concomitant detection of abundant LMP1, glucose transporter 1 (GLUT1) and CD33+ MDSCs in tumor sections. The full process has been reconstituted in vitro. LMP1 promotes the expression of multiple glycolytic genes, including GLUT1. This metabolic reprogramming results in increased expression of the Nod-like receptor family protein 3 (NLRP3) inflammasome, COX-2 and P-p65 and, consequently, increased production of IL-1ß, IL-6 and GM-CSF. Finally, these changes in the environment of malignant cells result in enhanced NPC-derived MDSC induction. One key step is the physical interaction of LMP1 with GLUT1 to stabilize the GLUT1 protein by blocking its K48-ubiquitination and p62-dependent autolysosomal degradation. This work indicates that LMP1-mediated glycolysis regulates IL-1ß, IL-6 and GM-CSF production through the NLRP3 inflammasome, COX-2 and P-p65 signaling pathways to enhance tumor-associated MDSC expansion, which leads to tumor immunosuppression in NPC.


Asunto(s)
Carcinoma/fisiopatología , Infecciones por Virus de Epstein-Barr/fisiopatología , Herpesvirus Humano 4/metabolismo , Células Supresoras de Origen Mieloide/citología , Neoplasias Nasofaríngeas/fisiopatología , Proteínas de la Matriz Viral/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/virología , Línea Celular Tumoral , Proliferación Celular , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Regulación Neoplásica de la Expresión Génica , Glucólisis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virología , Transducción de Señal , Proteínas de la Matriz Viral/genética
2.
Immunology ; 155(2): 263-272, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29770434

RESUMEN

The expansion of myeloid-derived suppressor cells (MDSCs) correlates with tumorigenesis in colorectal cancer (CRC). Here, we found a significant association between CD33+ MDSC number and Yes-associated protein 1 (YAP1) and phosphatase and tensin homologue (PTEN) levels in CRC patients (P < 0·05). Moreover, the CD33+ MDSCs, YAP1 and PTEN were identified as predictors for the prognosis of CRC patients (P < 0·05). Notably, CD33+ MDSCs were an independent survival predictor for CRC patients through a Cox model analysis. In vitro data determined that the expression levels of YAP1 and PTEN in CRC-derived cell lines were associated with CRC-derived MDSC induction, and the blockade of YAP1 and PTEN decreased CRC-derived MDSC induction. A mechanistic analysis revealed that YAP1 promoted CRC-derived MDSC induction by suppressing PTEN expression to up-regulate COX-2, P-AKT and P-p65 in CRC-derived cells, leading to secretion of the cytokine granulocyte-macrophage colony-stimulating factor. Our findings establish a novel mechanism of pro-tumorigenic MDSC induction mediated by ectopic YAP1 and PTEN expression in CRC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Biomarcadores de Tumor , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Fosfohidrolasa PTEN/genética , Fosfoproteínas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Proliferación Celular , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Fosfohidrolasa PTEN/metabolismo , Fosfoproteínas/metabolismo , Pronóstico , Factores de Transcripción , Microambiente Tumoral , Proteínas Señalizadoras YAP , Adulto Joven
3.
J Pathol ; 240(3): 329-340, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27538493

RESUMEN

Recent studies have shown that extracellular microRNAs are not only potential biomarkers but are also involved in cell interactions to regulate the intercommunication between cancer cells and their microenvironments in various types of malignancies. In this study, we isolated exosomes from nasopharyngeal carcinoma (NPC) cell lines and patient sera (T-EXOs), or control NP69 cells and healthy donor sera (HD-EXOs). We found that miR-24-3p was markedly enriched in T-EXOs as compared with HD-EXOs; the serum exosomal miR-24-3p level was correlated with worse disease-free survival of patients (p < 0.05). Knockdown of exosomal miR-24-3p (miR-24-3p-sponge-T-EXOs) by a sponge RNA targeting miR-24-3p restored the T-EXO-mediated (control-sponge-T-EXO) inhibition of T-cell proliferation and Th1 and Th17 differentiation, and the induction of regulatory T cells (Tregs). Mechanistic analyses revealed that administration of exosomal miR-24-3p increased P-ERK, P-STAT1 and P-STAT3 expression while decreasing P-STAT5 expression during T-cell proliferation and differentiation. Moreover, by in vivo and in vitro assessments, we found FGF11 to be a direct target of miR-24-3p. However, both miR-24-3p-sponge-T-EXOs and T-EXOs (control-sponge-T-EXOs) impeded proliferation and Th1 and Th17 differentiation, but induced Treg differentiation, of lenti-shFGF11-transfected T cells. The levels of phosphorylated ERK and STAT proteins were different in lenti-ScshRNA-transfected T cells and lenti-shFGF11-transfected T cells following administration of miR-24-3p-sponge-T-EXO. Interestingly, tumour FGF11 expression was positively correlated with the number of CD4+ and CD8+ T cells in vivo, and predicted favourable patient DFS (p < 0.05). Additionally, hypoxia increased cellular and exosomal miR-24-3p levels and enhanced the inhibitory effect of T-EXO on T-cell proliferation and differentiation. Collectively, our findings suggest that exosomal miR-24-3p is involved in tumour pathogenesis by mediating T-cell suppression via repression of FGF11, and may serve as a potential prognostic biomarker in NPC. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Carcinoma/genética , Exosomas/genética , Factores de Crecimiento de Fibroblastos/genética , MicroARNs/genética , Neoplasias Nasofaríngeas/genética , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinoma/diagnóstico , Carcinoma/inmunología , Carcinoma/patología , Comunicación Celular , Diferenciación Celular , Proliferación Celular , Supervivencia sin Enfermedad , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/inmunología , Neoplasias Nasofaríngeas/patología , Pronóstico , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
4.
Eur J Cancer ; 191: 112965, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37540921

RESUMEN

PURPOSE: The safety and objective clinical responses were observed in the phase I study using adjuvant autologous tumour-infiltrating lymphocytes (TILs) following concurrent chemoradiotherapy (CCRT) in nasopharyngeal carcinoma (NPC) patients. METHODS AND MATERIALS: One hundred fifty-six patients with stage III-IVb and pretreatment Epstein-Barr virus DNA levels of ≥4000 copies/ml were randomly assigned to receive CCRT combined with TIL infusion (n = 78) or CCRT alone (n = 78). All patients received CCRT and patients assigned to the TIL group received TIL infusion within 1 week after CCRT. The primary endpoint was investigator-assessed progression-free survival (PFS) at 3 years. RESULTS: After a median follow-up of 62.3 months, no significant difference was observed in the 3-year PFS rate between the CCRT plus TIL infusion group and CCRT alone group (75.6% versus 74.4%, hazard ratios, 1.08; 95% confidence intervals, 0.62-1.89). TIL infusion was safe without grade 3 or 4 adverse events and all the high-grade adverse effects were associated with myelosuppression caused by CCRT. Exploratory analysis showed that a potential survival benefit was observed with TILs in patients with lower levels of circulating CD8+TIM3+ cells, serum IL-8 or PD-L1. The infused TIL products in patients with favourable outcomes were associated with increased transcription of interferon-γ and a series of inflammatory related genes and a lower exhausted score. CONCLUSION: The primary objective of prolonging PFS with CCRT plus TILs in high-risk NPC patients was not met. These findings may provide evidence for the design of future trials investigating the combination of TILs plus immune checkpoint inhibitors based on CCRT in high-risk NPC patients. TRIAL REGISTRATION NUMBER: NCT02421640.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Neoplasias Nasofaríngeas , Humanos , Adyuvantes Inmunológicos , Quimioradioterapia/métodos , Supervivencia sin Enfermedad , ADN , Infecciones por Virus de Epstein-Barr/complicaciones , Herpesvirus Humano 4 , Linfocitos Infiltrantes de Tumor , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/patología
5.
Sci Immunol ; 8(87): eabq2424, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37738362

RESUMEN

Metabolic fitness of T cells is essential for their vitality, which is largely dependent on the behavior of the mitochondria. The nature of mitochondrial behavior in tumor-infiltrating T cells remains poorly understood. In this study, we show that mitofusin-2 (MFN2) expression is positively correlated with the prognosis of multiple cancers. Genetic ablation of Mfn2 in CD8+ T cells dampens mitochondrial metabolism and function and promotes tumor progression. In tumor-infiltrating CD8+ T cells, MFN2 enhances mitochondria-endoplasmic reticulum (ER) contact by interacting with ER-embedded Ca2+-ATPase SERCA2, facilitating the mitochondrial Ca2+ influx required for efficient mitochondrial metabolism. MFN2 stimulates the ER Ca2+ retrieval activity of SERCA2, thereby preventing excessive mitochondrial Ca2+ accumulation and apoptosis. Elevating mitochondria-ER contact by increasing MFN2 in CD8+ T cells improves the efficacy of cancer immunotherapy. Thus, we reveal a tethering-and-buffering mechanism of organelle cross-talk that regulates the metabolic fitness of tumor-infiltrating CD8+ T cells and highlights the therapeutic potential of enhancing MFN2 expression to optimize T cell function.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Apoptosis , Retículo Endoplásmico , GTP Fosfohidrolasas , Mitocondrias , Proteínas Mitocondriales
6.
Cell Host Microbe ; 31(11): 1882-1897.e10, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37848029

RESUMEN

Epstein-Barr virus (EBV) is a global public health concern, as it is known to cause multiple diseases while also being etiologically associated with a wide range of epithelial and lymphoid malignancies. Currently, there is no available prophylactic vaccine against EBV. gB is the EBV fusion protein that mediates viral membrane fusion and participates in host recognition, making it critical for EBV infection in both B cells and epithelial cells. Here, we present a gB nanoparticle, gB-I53-50 NP, that displays multiple copies of gB. Compared with the gB trimer, gB-I53-50 NP shows improved structural integrity and stability, as well as enhanced immunogenicity in mice and non-human primate (NHP) preclinical models. Immunization and passive transfer demonstrate a robust and durable protective antibody response that protects humanized mice against lethal EBV challenge. This vaccine candidate demonstrates significant potential in preventing EBV infection, providing a possible platform for developing prophylactic vaccines for EBV.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Vacunas , Cricetinae , Animales , Ratones , Herpesvirus Humano 4 , Infecciones por Virus de Epstein-Barr/prevención & control , Formación de Anticuerpos , Células CHO , Anticuerpos Neutralizantes , Anticuerpos Antivirales
7.
Front Immunol ; 11: 1906, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973789

RESUMEN

T cell exhaustion is an obstacle to immunotherapy for solid tumors. An understanding of the mechanism by which T cells develop this phenotype in solid tumors is needed. Here, hypoxia, a feature of the tumor microenvironment, causes T cell exhaustion (TExh) by inducing a mitochondrial defect. Upon exposure to hypoxia, activated T cells with a TExh phenotype are characterized by mitochondrial fragmentation, decreased ATP production, and decreased mitochondrial oxidative phosphorylation activity. The TExh phenotype is correlated with the downregulation of the mitochondrial fusion protein mitofusin 1 (MFN1) and upregulation of miR-24. Overexpression of miR-24 alters the transcription of many metabolism-related genes including its target genes MYC and fibroblast growth factor 11 (FGF11). Downregulation of MYC and FGF11 induces TExh differentiation, reduced ATP production and a loss of the mitochondrial mass in T cell receptor (TCR)-stimulated T cells. In addition, we determined that MYC regulates the transcription of FGF11 and MFN1. In nasopharyngeal carcinoma (NPC) tissues, the T cells exhibit an increased frequency of exhaustion and loss of mitochondrial mass. In addition, inhibition of miR-24 signaling decreases NPC xenograft growth in nude mice. Our findings reveal a mechanism for T cell exhaustion in the tumor environment and provide potential strategies that target mitochondrial metabolism for cancer immunotherapy.


Asunto(s)
Linfocitos Infiltrantes de Tumor/metabolismo , Mitocondrias/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Linfocitos T/metabolismo , Microambiente Tumoral , Animales , Estudios de Casos y Controles , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias/genética , Mitocondrias/inmunología , Mitocondrias/patología , Dinámicas Mitocondriales , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/inmunología , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/inmunología , Neoplasias Nasofaríngeas/patología , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/patología , Hipoxia Tumoral
8.
Cell Res ; 30(11): 950-965, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32901110

RESUMEN

Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-associated malignancy with a complex tumor ecosystem. How the interplay between tumor cells, EBV, and the microenvironment contributes to NPC progression and immune evasion remains unclear. Here we performed single-cell RNA sequencing on ~104,000 cells from 19 EBV+ NPCs and 7 nonmalignant nasopharyngeal biopsies, simultaneously profiling the transcriptomes of malignant cells, EBV, stromal and immune cells. Overall, we identified global upregulation of interferon responses in the multicellular ecosystem of NPC. Notably, an epithelial-immune dual feature of malignant cells was discovered and associated with poor prognosis. Functional experiments revealed that tumor cells with this dual feature exhibited a higher capacity for tumorigenesis. Further characterization of the cellular components of the tumor microenvironment (TME) and their interactions with tumor cells revealed that the dual feature of tumor cells was positively correlated with the expression of co-inhibitory receptors on CD8+ tumor-infiltrating T cells. In addition, tumor cells with the dual feature were found to repress IFN-γ production by T cells, demonstrating their capacity for immune suppression. Our results provide new insights into the multicellular ecosystem of NPC and offer important clinical implications.


Asunto(s)
Perfilación de la Expresión Génica , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Análisis de la Célula Individual , Microambiente Tumoral/genética , Virosis/genética , Animales , Agregación Celular , Comunicación Celular , Femenino , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunomodulación , Interferones/metabolismo , Ligandos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Ratones Desnudos , Células Mieloides/metabolismo , Neoplasias Nasofaríngeas/inmunología , Neoplasias Nasofaríngeas/virología , Procesos Estocásticos , Células del Estroma/metabolismo , Linfocitos T/inmunología
9.
Cell Death Dis ; 10(2): 50, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30718502

RESUMEN

Regulatory T cells (Tregs) represent an important contributor to cancer immune escape, but the molecular mechanism responsible for Treg expansion in tumors is heterogeneous and unclear. Here, we investigated the role of S1P1, a receptor of the bioactive lipid sphingosine 1-phosphate (S1P), in regulating the crosstalk between tumor cells and tumor-associated Tregs in bladder cancer (BC). We found that the frequency of CD4+Foxp3+ Tregs was increased in circulating and tumor-infiltrating lymphocytes from BC patients. S1P1 expression was upregulated in BC tissues compared with tumor-adjacent tissues and was positively correlated with the density of tumor-infiltrated Foxp3+ Tregs. Both S1P1 and Treg predicted poor overall survival in BC patients. The in vitro data paralleled the in vivo data and suggested that the activation or overexpression of S1P1 in BC cells promoted the generation of BC-induced (i)Tregs from CD4+CD25-cells, and the generation of these cells was reversed by treatment with anti-IL-10 or anti-TGF-ß. Moreover, S1P1 promoted Treg migration mediated by BC cells. Mechanistically, S1P1 activated the TGF-ß signaling pathway, leading to the secretion of TGF-ß and IL-10 from BC cells. In total, our findings suggest that S1P1 induces tumor-derived Treg expansion in a cell-specific manner and serves as a potent prognostic biomarker and therapeutic target in BC.


Asunto(s)
Receptores de Esfingosina-1-Fosfato/inmunología , Linfocitos T Reguladores/inmunología , Neoplasias de la Vejiga Urinaria/inmunología , Animales , Línea Celular Tumoral , Femenino , Humanos , Masculino , Receptores de Esfingosina-1-Fosfato/biosíntesis , Análisis de Supervivencia , Neoplasias de la Vejiga Urinaria/genética
10.
Cell Death Differ ; 26(11): 2314-2328, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30816302

RESUMEN

Stimulator of interferon genes (STING), a major adaptor protein in antiviral innate immune signaling, is considered as one of the most important regulators of antiviral and antitumor immunity. Although STING agonists are now intensively studied in clinical trials as a new class of adjuvants to boost cancer immunotherapy, the tumor-intrinsic role of the STING pathway in shaping the tumor microenvironment remains controversial. Here, we discovered that STING plays a vital role in regulation of myeloid-derived suppressor cell (MDSC) differentiation and antitumor immunity in Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC). Mechanistic analyses reveal that STING represses NPC-derived MDSC induction by enhancing SOCS1 expression in both tumor cells and MDSCs. SOCS1 physically interacts with STAT3 through its SH2 domain to prevent STAT3 phosphorylation and dimerization, resulting in reduced MDSC induction via inhibition of GM-CSF and IL-6 production. Notably, reduced tumoral STING expression was found to be significantly associated with a poor prognosis for NPC patients. Our findings reveal a novel mechanism linking STING to tumor microenvironmental cytokine production and MDSC induction.


Asunto(s)
Proteínas de la Membrana/metabolismo , Células Supresoras de Origen Mieloide/citología , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/patología , Microambiente Tumoral/fisiología , Sistemas CRISPR-Cas , Diferenciación Celular/inmunología , Línea Celular Tumoral , Dimerización , Infecciones por Virus de Epstein-Barr/patología , Técnicas de Inactivación de Genes , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Células HEK293 , Herpesvirus Humano 4/inmunología , Humanos , Interleucina-6/antagonistas & inhibidores , Proteínas de la Membrana/genética , Células Supresoras de Origen Mieloide/inmunología , Carcinoma Nasofaríngeo/inmunología , Carcinoma Nasofaríngeo/virología , Neoplasias Nasofaríngeas/inmunología , Neoplasias Nasofaríngeas/virología , Fosforilación/fisiología , Pronóstico , Factor de Transcripción STAT3/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Microambiente Tumoral/efectos de los fármacos
11.
Clin Exp Pharmacol Physiol ; 35(2): 211-6, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17941891

RESUMEN

1. The present study was conducted to investigate whether hydroxysafflor yellow A (HSYA) has a protective effect against heart injury after ischaemia-reperfusion and to determine the possible mechanism involved. 2. Hearts isolated from male Sprague-Dawley rats were perfused on a Langendorff apparatus and subjected to 30 min global ischaemia, followed by 120 min reperfusion. Infarct size and the level of lactate dehydrogenase (LDH) in the coronary effluent were determined. In mitochondria from isolated perfused hearts, Ca(2+)-induced swelling was observed. In isolated ventricular myocytes, depolarization of the mitochondrial membrane was determined by tetramethyl-rhodamine ethyl ester (TMRE) fluorescence. Furthermore, levels of phosphorylated endothelial nitric oxide synthase (eNOS) protein were measured by western blot. 3. Pretreatment with HSYA for 5 min before ischaemia reduced infarct size and the release of LDH. Administration of 20 micromol/L atractyloside, an opener of the mitochondrial permeability transition pore, and 10 micromol/L N(G)-nitro-L-arginine methyl ester (L-NAME), an inhibitor of NOS, attenuated the protective effects of HSYA. In mitochondria isolated from hearts pretreated with 0.1 mmol/L HSYA for 5 min, a significant inhibition of Ca(2+)-induced swelling was observed and this inhibition was attenuated by l-NAME. In isolated ventricular myocytes, pretreatment with HSYA prevented ischaemia-induced cell death and depolarization of the mitochondrial membrane, whereas atractyloside or l-NAME attenuated the effects of HSYA. Levels of phosphorylated eNOS protein were significantly enhanced in the HSYA-treated group. 4. The findings of the present study indicate that HSYA protects the myocardium against ischaemia-reperfusion injury by inhibiting mitochondrial permeability transition pore opening. The effect of HSYA on mitochondrial permeability transition pore opening may be mediated through enhanced nitric oxide production by eNOS activation.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Chalcona/análogos & derivados , Mitocondrias Cardíacas/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Quinonas/farmacología , Animales , Atractilósido/farmacología , Fármacos Cardiovasculares/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Chalcona/farmacología , Chalcona/uso terapéutico , Inhibidores Enzimáticos/farmacología , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Dilatación Mitocondrial , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III , Perfusión , Fosforilación , Quinonas/uso terapéutico , Ratas , Ratas Sprague-Dawley
13.
FEMS Microbiol Lett ; 362(6)2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25667230

RESUMEN

Yeast Cia2p is a component of the cytosolic Fe/S protein assembly (CIA) machinery. Initial studies of the CIA machinery were performed in yeast, but the precise role of Cia2p in this eukaryote is still unknown. We report that CIA2 deficiency results in impaired oxidative stress response, as evidenced by increased sensitivity to the oxidant cumene hydroperoxide (CHP), impaired activities of superoxide dismutases and aconitase and decreased replicative lifespan in the mutants. Moreover, intracellular reactive oxygen species levels were significantly increased in CIA2-deficient cells after treatment with CHP. We also show that CIA2-deficient cells display an increased resistance to tunicamycin-induced endoplasmic reticulum (ER) stress, as evidenced by the upregulated splicing of the mRNA of HAC1, which encodes a functional transcription factor that regulates the transcription of unfolded protein response (UPR) target genes, suggesting enhanced intracellular UPR activity. Furthermore, the transcription of several canonical UPR target genes is strongly induced in CIA2-deficient cells as compared with wild-type controls. Taken together, these results suggest the involvement of Cia2p in oxidative and ER stress responses in yeast.


Asunto(s)
Estrés del Retículo Endoplásmico , Estrés Oxidativo , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/metabolismo , Respuesta de Proteína Desplegada , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Derivados del Benceno/farmacología , Citoplasma/genética , Citoplasma/fisiología , Mutación , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal , Factores de Transcripción , Tunicamicina/farmacología , Respuesta de Proteína Desplegada/genética , Regulación hacia Arriba
14.
Mech Ageing Dev ; 138: 53-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24486555

RESUMEN

Saccharomyces cerevisiae Nar1p is an essential Fe/S protein that exhibits striking similarity to bacterial iron-only hydrogenases. Nar1p is required for the maturation of cytosolic and nuclear, but not of mitochondrial Fe/S proteins, and plays a role in modulating sensitivity to oxygen in both yeast and Caenorhabditis elegans through unknown mechanisms. Here we report that Nar1 deficiency results in shortened lifespan and sensitivity to paraquat that is rescued by increased expression of mitochondrial superoxide dismutase. These data suggest that Nar1p promotes protection against oxidative stress and define a new role for Nar1p in promoting replicative lifespan.


Asunto(s)
Hidrogenasas , Proteínas Hierro-Azufre , Paraquat , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , Animales , Citosol/metabolismo , Herbicidas/metabolismo , Herbicidas/farmacología , Hidrogenasas/metabolismo , Proteínas Hierro-Azufre/deficiencia , Proteínas Hierro-Azufre/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Paraquat/metabolismo , Paraquat/farmacología , Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/ultraestructura , Superóxido Dismutasa/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA