Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell Host Microbe ; 31(11): 1882-1897.e10, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37848029

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections , Vaccines , Cricetinae , Animals , Mice , Herpesvirus 4, Human , Epstein-Barr Virus Infections/prevention & control , Antibody Formation , CHO Cells , Antibodies, Neutralizing , Antibodies, Viral
2.
Sci Immunol ; 8(87): eabq2424, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37738362

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Apoptosis , Endoplasmic Reticulum , GTP Phosphohydrolases , Mitochondria , Mitochondrial Proteins
3.
Eur J Cancer ; 191: 112965, 2023 09.
Article in English | MEDLINE | ID: mdl-37540921

ABSTRACT

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.


Subject(s)
Epstein-Barr Virus Infections , Nasopharyngeal Neoplasms , Humans , Adjuvants, Immunologic , Chemoradiotherapy/methods , Disease-Free Survival , DNA , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human , Lymphocytes, Tumor-Infiltrating , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/pathology
4.
Cell Res ; 30(11): 950-965, 2020 11.
Article in English | MEDLINE | ID: mdl-32901110

ABSTRACT

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.


Subject(s)
Gene Expression Profiling , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Single-Cell Analysis , Tumor Microenvironment/genetics , Virus Diseases/genetics , Animals , Cell Aggregation , Cell Communication , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunomodulation , Interferons/metabolism , Ligands , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Nude , Myeloid Cells/metabolism , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/virology , Stochastic Processes , Stromal Cells/metabolism , T-Lymphocytes/immunology
5.
Front Immunol ; 11: 1906, 2020.
Article in English | MEDLINE | ID: mdl-32973789

ABSTRACT

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.


Subject(s)
Lymphocytes, Tumor-Infiltrating/metabolism , Mitochondria/metabolism , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , T-Lymphocytes/metabolism , Tumor Microenvironment , Animals , Case-Control Studies , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/immunology , Mitochondria/pathology , Mitochondrial Dynamics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Hypoxia
6.
Cell Death Differ ; 26(11): 2314-2328, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30816302

ABSTRACT

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.


Subject(s)
Membrane Proteins/metabolism , Myeloid-Derived Suppressor Cells/cytology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Tumor Microenvironment/physiology , CRISPR-Cas Systems , Cell Differentiation/immunology , Cell Line, Tumor , Dimerization , Epstein-Barr Virus Infections/pathology , Gene Knockout Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , HEK293 Cells , Herpesvirus 4, Human/immunology , Humans , Interleukin-6/antagonists & inhibitors , Membrane Proteins/genetics , Myeloid-Derived Suppressor Cells/immunology , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/virology , Phosphorylation/physiology , Prognosis , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism , Tumor Microenvironment/drug effects
7.
Cell Death Dis ; 10(2): 50, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30718502

ABSTRACT

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.


Subject(s)
Sphingosine-1-Phosphate Receptors/immunology , T-Lymphocytes, Regulatory/immunology , Urinary Bladder Neoplasms/immunology , Animals , Cell Line, Tumor , Female , Humans , Male , Sphingosine-1-Phosphate Receptors/biosynthesis , Survival Analysis , Urinary Bladder Neoplasms/genetics
8.
Immunology ; 155(2): 263-272, 2018 10.
Article in English | MEDLINE | ID: mdl-29770434

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , PTEN Phosphohydrolase/genetics , Phosphoproteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers , Cell Proliferation , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , PTEN Phosphohydrolase/metabolism , Phosphoproteins/metabolism , Prognosis , Transcription Factors , Tumor Microenvironment , YAP-Signaling Proteins , Young Adult
9.
PLoS Pathog ; 13(7): e1006503, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28732079

ABSTRACT

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.


Subject(s)
Carcinoma/physiopathology , Epstein-Barr Virus Infections/physiopathology , Herpesvirus 4, Human/metabolism , Myeloid-Derived Suppressor Cells/cytology , Nasopharyngeal Neoplasms/physiopathology , Viral Matrix Proteins/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/virology , Cell Line, Tumor , Cell Proliferation , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic , Glycolysis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Herpesvirus 4, Human/genetics , Host-Pathogen Interactions , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , Signal Transduction , Viral Matrix Proteins/genetics
10.
J Pathol ; 240(3): 329-340, 2016 11.
Article in English | MEDLINE | ID: mdl-27538493

ABSTRACT

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.


Subject(s)
Carcinoma/genetics , Exosomes/genetics , Fibroblast Growth Factors/genetics , MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma/diagnosis , Carcinoma/immunology , Carcinoma/pathology , Cell Communication , Cell Differentiation , Cell Proliferation , Disease-Free Survival , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Prognosis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
11.
FEMS Microbiol Lett ; 362(6)2015 Mar.
Article in English | MEDLINE | ID: mdl-25667230

ABSTRACT

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.


Subject(s)
Endoplasmic Reticulum Stress , Oxidative Stress , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/metabolism , Unfolded Protein Response , Basic-Leucine Zipper Transcription Factors/genetics , Benzene Derivatives/pharmacology , Cytoplasm/genetics , Cytoplasm/physiology , Mutation , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/physiology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Transcription Factors , Tunicamycin/pharmacology , Unfolded Protein Response/genetics , Up-Regulation
12.
Mech Ageing Dev ; 138: 53-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24486555

ABSTRACT

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.


Subject(s)
Hydrogenase , Iron-Sulfur Proteins , Paraquat , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Animals , Cytosol/metabolism , Herbicides/metabolism , Herbicides/pharmacology , Hydrogenase/metabolism , Iron-Sulfur Proteins/deficiency , Iron-Sulfur Proteins/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Paraquat/metabolism , Paraquat/pharmacology , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae/ultrastructure , Superoxide Dismutase/metabolism , Time Factors
14.
Clin Exp Pharmacol Physiol ; 35(2): 211-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17941891

ABSTRACT

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.


Subject(s)
Cardiovascular Agents/pharmacology , Chalcone/analogs & derivatives , Mitochondria, Heart/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Quinones/pharmacology , Animals , Atractyloside/pharmacology , Cardiovascular Agents/therapeutic use , Cell Survival/drug effects , Chalcone/pharmacology , Chalcone/therapeutic use , Enzyme Inhibitors/pharmacology , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III , Perfusion , Phosphorylation , Quinones/therapeutic use , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...