Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Metabolites ; 11(8)2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-34436459

RESUMO

Alterations in metabolism following radiotherapy affect therapeutic efficacy, although the mechanism underlying such alterations is unclear. A new imaging technique-named dynamic nuclear polarization (DNP) carbon-13 magnetic resonance imaging (MRI)-probes the glycolytic flux in a real-time, dynamic manner. The [1-13C]pyruvate is transported by the monocarboxylate transporter (MCT) into cells and converted into [1-13C]lactate by lactate dehydrogenase (LDH). To capture the early glycolytic alterations in the irradiated cancer and immune cells, we designed a preliminary DNP 13C-MRI study by using hyperpolarized [1-13C]pyruvate to study human FaDu squamous carcinoma cells, HMC3 microglial cells, and THP-1 monocytes before and after irradiation. The pyruvate-to-lactate conversion rate (kPL [Pyr.]) calculated by kinetic modeling was used to evaluate the metabolic alterations. Western blotting was performed to assess the expressions of LDHA, LDHB, MCT1, and MCT4 proteins. Following irradiation, the pyruvate-to-lactate conversion rates on DNP 13C-MRI were significantly decreased in the FaDu and the HMC3 cells but increased in the THP-1 cells. Western blot analysis confirmed the similar trends in LDHA and LDHB expression levels. In conclusion, DNP 13C-MRI non-invasively captured the different glycolytic alterations among cancer and immune systems in response to irradiation, implying its potential for clinical use in the future.

2.
J Clin Med ; 8(6)2019 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-31248127

RESUMO

Insulin resistance and metabolic derangement are present in patients with type 2 diabetes mellitus (T2DM). However, the metabolomic signature of T2DM in cerebrospinal fluid (CSF) has not been investigated thus far. In this prospective metabolomic study, fasting CSF and plasma samples from 40 T2DM patients to 36 control subjects undergoing elective surgery with spinal anesthesia were analyzed by 1H nuclear magnetic resonance (NMR) spectroscopy. NMR spectra of CSF and plasma metabolites were analyzed and correlated with the presence of T2DM and diabetic microangiopathy (retinopathy, nephropathy, and neuropathy) using an area under the curve (AUC) estimation. CSF metabolomic profiles in T2DM patients vs. controls revealed significantly increased levels of alanine, leucine, valine, tyrosine, lactate, pyruvate, and decreased levels of histidine. In addition, a combination of alanine, histidine, leucine, pyruvate, tyrosine, and valine in CSF showed a superior correlation with the presence of T2DM (AUC:0.951), diabetic retinopathy (AUC:0.858), nephropathy (AUC:0.811), and neuropathy (AUC:0.691). Similar correlations also appeared in plasma profiling. These metabolic alterations in CSF suggest decreasing aerobic metabolism and increasing anaerobic glycolysis in cerebral circulation of patients with T2DM. In conclusion, our results provide clues for the metabolic derangements in diabetic central neuropathy among T2DM patients; however, their clinical significance requires further exploration.

3.
Oxid Med Cell Longev ; 2019: 5913635, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30755786

RESUMO

We have previously shown that GSH depletion alters global metabolism of cells. In the present study, we applied a metabolomic approach for studying the early changes in metabolism in hydrogen peroxide- (H2O2-) treated hepatoma cells which were destined to die. Levels of fructose 1,6-bisphosphate and an unusual metabolite, sedoheptulose 1,7-bisphosphate (S-1,7-BP), were elevated in hepatoma Hep G2 cells. Deficiency in G6PD activity significantly reduced S-1,7-BP formation, suggesting that S-1,7-BP is formed in the pentose phosphate pathway as a response to oxidative stress. Additionally, H2O2 treatment significantly increased the level of nicotinamide adenine dinucleotide phosphate (NADP+) and reduced the levels of ATP and NAD+. Severe depletion of ATP and NAD+ in H2O2-treated Hep G2 cells was associated with cell death. Inhibition of PARP-mediated NAD+ depletion partially protected cells from death. Comparison of metabolite profiles of G6PD-deficient cells and their normal counterparts revealed that changes in GSH and GSSG per se do not cause cell death. These findings suggest that the failure of hepatoma cells to maintain energy metabolism in the midst of oxidative stress may cause cell death.


Assuntos
Carcinoma Hepatocelular/metabolismo , Heptoses/metabolismo , Peróxido de Hidrogênio/metabolismo , Neoplasias Hepáticas/metabolismo , Humanos , Estresse Oxidativo
4.
Molecules ; 23(11)2018 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-30400243

RESUMO

Non-alcoholic fatty liver disease (NAFLD) as a global health problem has clinical manifestations ranging from simple non-alcoholic fatty liver (NAFL) to non-alcoholic steatohepatitis (NASH), cirrhosis, and cancer. The role of different types of fatty acids in driving the early progression of NAFL to NASH is not understood. Lipid overload causing lipotoxicity and inflammation has been considered as an essential pathogenic factor. To correlate the lipid profiles with cellular lipotoxicity, we utilized palmitic acid (C16:0)- and especially unprecedented palmitoleic acid (C16:1)-induced lipid overload HepG2 cell models coupled with lipidomic technology involving labeling with stable isotopes. C16:0 induced inflammation and cell death, whereas C16:1 induced significant lipid droplet accumulation. Moreover, inhibition of de novo sphingolipid synthesis by myriocin (Myr) aggravated C16:0 induced lipoapoptosis. Lipid profiles are different in C16:0 and C16:1-treated cells. Stable isotope-labeled lipidomics elucidates the roles of specific fatty acids that affect lipid metabolism and cause lipotoxicity or lipid droplet formation. It indicates that not only saturation or monounsaturation of fatty acids plays a role in hepatic lipotoxicity but also Myr inhibition exasperates lipoapoptosis through ceramide in-direct pathway. Using the techniques presented in this study, we can potentially investigate the mechanism of lipid metabolism and the heterogeneous development of NAFLD.


Assuntos
Marcação por Isótopo , Metabolismo dos Lipídeos , Metaboloma , Metabolômica , Ácidos Graxos/metabolismo , Ácidos Graxos Monoinsaturados/metabolismo , Células Hep G2 , Humanos , Marcação por Isótopo/métodos , Metabolômica/métodos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ácido Palmítico/metabolismo , Esfingolipídeos/biossíntese
5.
World J Gastroenterol ; 24(33): 3760-3769, 2018 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-30197481

RESUMO

AIM: To explore the correlation of metabolomics profiles of gastric cancer (GC) with its chromosomal instability (CIN) status. METHODS: Nineteen GC patients were classified as CIN and non-CIN type by The Cancer Genome Atlas Research Group system, based on 409 oncogenes and tumor suppressor genes sequenced. The aqueous metabolites of the GC tumor and its surrounding adjacent healthy tissues were identified through liquid chromatography-mass spectrometry. Groups were compared by defining variable importance in projection score of > 1.2, a fold change value or its reciprocal of > 1.2, and a P value of < 0.05 as a significant difference. RESULTS: In total, twelve men and seven women were enrolled, with a median age of 66 years (range, 47-87 years). The numbers of gene alterations in the CIN GC group were significantly higher than those in the non-CIN GC (32-218 vs 2-17; P < 0.0005). Compared with the adjacent healthy tissues, GC tumors demonstrated significantly higher aspartic acid, citicoline, glutamic acid, oxidized glutathione, succinyladenosine, and uridine diphosphate-N-acetylglucosamine levels, but significantly lower butyrylcarnitine, glutathione hydroxyhexanoycarnitine, inosinic acid, isovalerylcarnitine, and threonine levels (all P < 0.05). CIN tumors contained significantly higher phosphocholine and uridine 5'-monophosphate levels but significantly lower beta-citryl-L-glutamic acid levels than did non-CIN tumors (all P < 0.05). CIN GC tumors demonstrated additional altered pathways involving alanine, aspartate, and glutamate metabolism, glyoxylate and dicarboxylate metabolism, histidine metabolism, and phenylalanine, tyrosine, and tryptophan biosynthesis. CONCLUSION: Metabolomic profiles of GC tumors and the adjacent healthy tissue are distinct, and the CIN status is associated with downstream metabolic alterations in GC.


Assuntos
Instabilidade Cromossômica , Genes Supressores de Tumor , Redes e Vias Metabólicas/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Metabolômica/métodos , Pessoa de Meia-Idade , Mutação , Estudos Prospectivos , Estômago/patologia , Neoplasias Gástricas/patologia
6.
Redox Biol ; 12: 916-928, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28478381

RESUMO

To study the physiological roles of NADH and NADPH homeostasis in cancer, we studied the effect of NNT knockdown on physiology of SK-Hep1 cells. NNT knockdown cells show limited abilities to maintain NAD+ and NADPH levels and have reduced proliferation and tumorigenicity. There is an increased dependence of energy production on oxidative phosphorylation. Studies with stable isotope tracers have revealed that under the new steady-state metabolic condition, the fluxes of TCA and glycolysis decrease while that of reductive carboxylation increases. Increased [α-ketoglutarate]/[succinate] ratio in NNT-deficient cells results in decrease in HIF-1α level and expression of HIF-1α regulated genes. Reduction in NADPH level leads to repression of HDAC1 activity and an increase in p53 acetylation. These findings suggest that NNT is essential to homeostasis of NADH and NADPH pools, anomalies of which affect HIF-1α- and HDAC1-dependent pathways, and hence retrograde response of mitochondria.


Assuntos
Mitocôndrias/metabolismo , NADP Trans-Hidrogenase Específica para A ou B/genética , NADP/metabolismo , NAD/metabolismo , Neoplasias/enzimologia , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Silenciamento de Genes , Glicólise , Histona Desacetilase 1/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Proteínas Mitocondriais/genética , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/metabolismo , Fosforilação Oxidativa
7.
Sci Rep ; 7(1): 935, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28428548

RESUMO

No effective drug is currently available for treatment of enterovirus 71 (EV71) infection. Schizonepeta tenuifolia Briq. (ST) has been used as a herbal constituent of traditional Chinese medicine. We studied whether the aqueous extract of Schizonepeta tenuifolia Briq (STE) has antiviral activity. STE inhibited replication of EV71, as evident by its ability to diminish plaque formation and cytopathic effect induced by EV71, and to inhibit the synthesis of viral RNA and protein. Moreover, daily single-dose STE treatment significantly improved the survival of EV71-infected mice, and ameliorated the symptoms. Mechanistically, STE exerts multiple effects on enteroviral infection. Treatment with STE reduced viral attachment and entry; the cleavage of eukaryotic translation initiation factor 4 G (eIF4G) by EV71 protease, 2Apro; virus-induced reactive oxygen species (ROS) formation; and relocation of heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) from the nucleus to the cytoplasm. It was accompanied by a decline in EV71-associated hyperphosphorylation of p38 kinase and EPS15. It is plausible that STE may inhibit ROS-induced p38 kinase activation, and subsequent hnRNP A1 relocation and EPS15-mediated membrane trafficking in infected cells. These findings suggest that STE possesses anti-EV71 activities, and may serve as health food or candidate antiviral drug for protection against EV71.


Assuntos
Antivirais/uso terapêutico , Enterovirus Humano A/efeitos dos fármacos , Infecções por Enterovirus/tratamento farmacológico , Lamiaceae/química , Extratos Vegetais/uso terapêutico , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Enterovirus Humano A/fisiologia , Infecções por Enterovirus/virologia , Fator de Iniciação Eucariótico 4G/metabolismo , Ribonucleoproteína Nuclear Heterogênea A1/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos ICR , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Células Vero , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Biochem Pharmacol ; 117: 20-34, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27501919

RESUMO

Dehydroepiandrosterone (DHEA)-induced growth arrest of hepatoma cells is associated with metabolic disturbance. Our previous study has suggested that DHEA may cause cellular energy drain. It is possible that mitochondrial dysfunction may be mechanistically implicated in DHEA-induced changes in cellular phenotype. Treatment of SK-Hep-1 cells with DHEA caused significant reduction in proliferation, colony formation, and growth in semi-solid medium. Such changes in cellular phenotype were associated with mitochondrial depolarization, increase in mitochondrial mass, and decrease in respiratory activity. Level of reactive oxygen species (ROS) increased in DHEA-treated cells. To explore the mechanistic aspect of DHEA-induced mitochondrial dysfunction, we employed SILAC approach to study the changes in the mitoproteome of SK-Hep-1 cells after DHEA treatment. Respiratory chain complex proteins such as NDUFB8 and SDHB were differentially expressed. Of mitochondrial proteins with altered expression, FAST kinase domain-containing protein 2 (FASTKD2) showed significantly reduced expression. Exogenous expression of FASTKD2 in SK-Hep-1 cells increased their resistance to growth-inhibitory effect of DHEA, though it alone did not affect cell growth. FASTKD2 expression partially reversed the effect of DHEA on mitochondria, and reduced DHEA-induced ROS generation. Our results suggest that DHEA induces changes in mitochondrial proteins and respiratory activity, and contributes to growth arrest. FASTKD2 may be an important regulator of mitochondrial physiology, and represent a downstream target for DHEA.


Assuntos
Antineoplásicos Hormonais/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Desidroepiandrosterona/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Proteínas Mitocondriais/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/patologia , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteômica/métodos , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Succinato Desidrogenase/antagonistas & inibidores , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo
9.
Viruses ; 7(12): 6689-706, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26694452

RESUMO

Glucose-6-phosphate dehydrogenase (G6PD)-deficient cells are highly susceptible to viral infection. This study examined the mechanism underlying this phenomenon by measuring the expression of antiviral genes-tumor necrosis factor alpha (TNF-α) and GTPase myxovirus resistance 1 (MX1)-in G6PD-knockdown cells upon human coronavirus 229E (HCoV-229E) and enterovirus 71 (EV71) infection. Molecular analysis revealed that the promoter activities of TNF-α and MX1 were downregulated in G6PD-knockdown cells, and that the IκB degradation and DNA binding activity of NF-κB were decreased. The HSCARG protein, a nicotinamide adenine dinucleotide phosphate (NADPH) sensor and negative regulator of NF-κB, was upregulated in G6PD-knockdown cells with decreased NADPH/NADP⁺ ratio. Treatment of G6PD-knockdown cells with siRNA against HSCARG enhanced the DNA binding activity of NF-κB and the expression of TNF-α and MX1, but suppressed the expression of viral genes; however, the overexpression of HSCARG inhibited the antiviral response. Exogenous G6PD or IDH1 expression inhibited the expression of HSCARG, resulting in increased expression of TNF-α and MX1 and reduced viral gene expression upon virus infection. Our findings suggest that the increased susceptibility of the G6PD-knockdown cells to viral infection was due to impaired NF-κB signaling and antiviral response mediated by HSCARG.


Assuntos
Coronavirus Humano 229E/imunologia , Enterovirus Humano A/imunologia , Glucosefosfato Desidrogenase/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/virologia , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Resistência a Myxovirus/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Antioxid Redox Signal ; 22(9): 744-59, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25556665

RESUMO

AIMS: Glucose 6-phosphate dehydrogenase (G6PD) is essential for maintenance of nicotinamide dinucleotide hydrogen phosphate (NADPH) levels and redox homeostasis. A number of drugs, such as antimalarial drugs, act to induce reactive oxygen species and hemolytic crisis in G6PD-deficient patients. We used diamide (DIA) to mimic drug-induced oxidative stress and studied how these drugs affect cellular metabolism using a metabolomic approach. RESULTS: There are a few differences in metabolome between red blood cells (RBCs) from normal and G6PD-deficient individuals. DIA causes modest changes in normal RBC metabolism. In contrast, there are significant changes in various biochemical pathways, namely glutathione (GSH) metabolism, purine metabolism, and glycolysis, in G6PD-deficient cells. GSH depletion is concomitant with a shift in energy metabolism. Adenosine monophosphate (AMP) and adenosine diphosphate (ADP) accumulation activates AMP protein kinase (AMPK) and increases entry of glucose into glycolysis. However, inhibition of pyruvate kinase (PK) reduces the efficacy of energy production. Metabolic changes and protein oxidation occurs to a greater extent in G6PD-deficient RBCs than in normal cells, leading to severe irreversible loss of deformability of the former. INNOVATION AND CONCLUSION: Normal and G6PD-deficient RBCs differ in their responses to oxidants. Normal cells have adequate NADPH regeneration for maintenance of GSH pool. In contrast, G6PD-deficient cells are unable to regenerate enough NADPH under a stressful situation, and switch to biosynthetic pathway for GSH supply. Rapid GSH exhaustion causes energy crisis and futile AMPK activation. Our findings suggest that drug-induced oxidative stress differentially affects metabolism and metabolite signaling in normal and G6PD-deficient cells. It also provides an insight into the pathophysiology of acute hemolytic anemia in G6PD-deficient patients.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Eritrócitos/metabolismo , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Deficiência de Glucosefosfato Desidrogenase/patologia , Glutationa/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Anemia Hemolítica/metabolismo , Diamida/farmacologia , Eritrócitos/efeitos dos fármacos , Humanos , Metionina/metabolismo , NADP/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Via de Pentose Fosfato , Piruvato Quinase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Reagentes de Sulfidrila/farmacologia
11.
PLoS One ; 9(11): e113234, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25401329

RESUMO

Redox homeostasis is an important host factor determining the outcome of infectious disease. Enterovirus 71 (EV71) infection has become an important endemic disease in Southeast Asia and China. We have previously shown that oxidative stress promotes viral replication, and progeny virus induces oxidative stress in host cells. The detailed mechanism for reactive oxygen species (ROS) generation in infected cells remains elusive. In the current study, we demonstrate that mitochondria were a major ROS source in EV71-infected cells. Mitochondria in productively infected cells underwent morphologic changes and exhibited functional anomalies, such as a decrease in mitochondrial electrochemical potential ΔΨ(m) and an increase in oligomycin-insensitive oxygen consumption. Respiratory control ratio of mitochondria from infected cells was significantly lower than that of normal cells. The total adenine nucleotide pool and ATP content of EV71-infected cells significantly diminished. However, there appeared to be a compensatory increase in mitochondrial mass. Treatment with mito-TEMPO reduced eIF2α phosphorylation and viral replication, suggesting that mitochondrial ROS act to promote viral replication. It is plausible that EV71 infection induces mitochondrial ROS generation, which is essential to viral replication, at the sacrifice of efficient energy production, and that infected cells up-regulate biogenesis of mitochondria to compensate for their functional defect.


Assuntos
Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/virologia , Glioblastoma/virologia , Mitocôndrias/patologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Replicação Viral , Antioxidantes/farmacologia , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , China , Óxidos N-Cíclicos/farmacologia , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Microscopia Eletrônica de Transmissão , Mitocôndrias/metabolismo , Oxirredução , Consumo de Oxigênio , Células Tumorais Cultivadas
12.
PLoS One ; 8(11): e79566, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24223971

RESUMO

Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme in the pentose phosphate pathway and provides reducing energy to all cells by maintaining redox balance. The most common clinical manifestations in patients with G6PD deficiency are neonatal jaundice and acute hemolytic anemia. The effects of microbial infection in patients with G6PD deficiency primarily relate to the hemolytic anemia caused by Plasmodium or viral infections and the subsequent medication that is required. We are interested in studying the impact of bacterial infection in G6PD-deficient cells. G6PD knock down A549 lung carcinoma cells, together with the common pathogen Staphylococcus aureus, were employed in our cell infection model. Here, we demonstrate that a lower cell viability was observed among G6PD-deficient cells when compared to scramble controls upon bacterial infection using the MTT assay. A significant increase in the intracellular ROS was detected among S. aureus-infected G6PD-deficient cells by observing dichlorofluorescein (DCF) intensity within cells under a fluorescence microscope and quantifying this signal using flow cytometry. The impairment of ROS removal is predicted to enhance apoptotic activity in G6PD-deficient cells, and this enhanced apoptosis was observed by annexin V/PI staining under a confocal fluorescence microscope and quantified by flow cytometry. A higher expression level of the intrinsic apoptotic initiator caspase-9, as well as the downstream effector caspase-3, was detected by Western blotting analysis of G6PD-deficient cells following bacterial infection. In conclusion, we propose that bacterial infection, perhaps the secreted S. aureus α-hemolysin in this case, promotes the accumulation of intracellular ROS in G6PD-deficient cells. This would trigger a stronger apoptotic activity through the intrinsic pathway thereby reducing cell viability when compared to wild type cells.


Assuntos
Células Epiteliais/enzimologia , Células Epiteliais/microbiologia , Glucosefosfato Desidrogenase/metabolismo , Staphylococcus aureus/fisiologia , Apoptose , Linhagem Celular Tumoral , Farmacorresistência Bacteriana , Células Epiteliais/citologia , Glucosefosfato Desidrogenase/efeitos dos fármacos , Proteínas Hemolisinas/metabolismo , Humanos , Espaço Intracelular/metabolismo , Necrose , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia
13.
Free Radic Biol Med ; 54: 71-84, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23142419

RESUMO

Glucose-6-phosphate dehydrogenase (G6PD) is crucial to NADPH generation and redox homeostasis. We have recently shown that G6PD deficiency predisposes cells to oxidant-induced cell death, and it is associated with the impairment of glutathione regeneration. It remains unclear what other metabolic pathways are affected by G6PD deficiency and whether the altered metabolism disturbs cellular redox homeostasis and underlies increased susceptibility to oxidants. In this study, we examined the effects of diamide on global metabolite profiles of SK-Hep1-derived SK-i-Gi and SK-i-Sc cells, which could inducibly express short hairpin RNA (shRNA) against G6PD (Gi) and control shRNA (Sc), respectively. There was no significant difference in their metabolite profiles under uninduced conditions. Doxycycline (Dox) addition resulted in over 70% decrease in G6PD activity in SK-i-Gi cells. This was accompanied by relatively minor changes in the metabolome of SK-i-Gi cells. Upon further diamide treatment, the metabolite profiles of both SK-i-Gi and SK-i-Sc cells changed in a time-dependent manner. A number of metabolic pathways, including those involved in energy metabolism and metabolism of amino acids and glutathione, were affected. However, the changes in the metabolite profile of Dox-treated SK-i-Gi cells were distinct from those of control cells (i.e., Dox-treated SK-i-Sc, SK-i-Gi, and SK-i-Sc cells). Cellular glutathione was depleted, whereas its disulfide form increased significantly in diamide, Dox-treated SK-i-Gi cells. Metabolites related to energy metabolism, such as AMP, ADP, and acetylcarnitine, increased to a greater extent in these cells than in diamide-treated control cells. In contrast, NAD and glutathione dropped to lower levels in SK-i-Gi cells than in control cells. The NAD(+) depletion in SK-i-Gi cells was accompanied by a significant increase in NAD kinase activity. Targeted analyses revealed that NADP(+) and NADPH increased significantly in diamide, Dox-treated SK-i-Gi cells compared with similarly treated control cells. Our results suggest that diamide induces oxidation and depletion of glutathione in SK-i-Gi cells under conditions of G6PD shRNA induction and subsequently induces conversion of NAD(+) to NADP(+) through enhanced NAD kinase activity. This may represent a compensatory mechanism to restore cellular NADPH reserve in G6PD-deficient cells. It is accompanied by alteration in pathways of cellular energy metabolism, such as glycolysis and ß-oxidation.


Assuntos
Carcinoma Hepatocelular/enzimologia , Glucosefosfato Desidrogenase/metabolismo , Neoplasias Hepáticas/enzimologia , Metaboloma , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Diamida/metabolismo , Metabolismo Energético/genética , Técnicas de Silenciamento de Genes , Glucosefosfato Desidrogenase/genética , Glutationa/metabolismo , Humanos , Neoplasias Hepáticas/genética , Metaboloma/genética , NADP/metabolismo , Oxirredução , Estresse Oxidativo/genética , RNA Interferente Pequeno/genética
14.
Biochem Pharmacol ; 82(11): 1549-61, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21843511

RESUMO

DHEA is known to have chemopreventive and antiproliferative activities, and was initially thought to be mediated by inhibition of G6PD. Our previous study has shown that DHEA may act through interference with energy metabolism. To study the effect of pharmacological dose of DHEA on cellular metabolism, and to further delineate the mechanism underlying its antiproliferative effect, we applied a metabolomic approach to globally profile the changes in metabolites in SK-Hep1 cells underexpressing G6PD (Sk-Gi) and control cells (Sk-Sc) after DHEA treatment. RRLC-TOF-MS was used to identify metabolites, and tandem mass spectrometry was used to confirm their identity. DHEA induced changes in glutathione metabolism, lipid metabolism, s-adenosylmethionine (SAM) metabolism, as well as lysine metabolism. Elevation in level of glutathione disulfide, together with a concomitant decrease in level of reduced glutathione, was indicative of increased oxidative stress. Depletion of carnitine and its acyl derivatives reflected decline in fatty acid catabolism. These changes were associated with mitochondrial malfunction and reduction in cellular ATP content. Cardiolipin (CL) and phosphatidylcholine (PC) levels decreased significantly, suggesting that alterations in lipid composition are causally related to decline in mitochondrial function after DHEA treatment. The decline in cellular SAM content was accompanied by decreased expression of methionine adenosyltransferase genes MAT2A and MAT2B. SAM supplementation partially rescued cells from DHEA-induced growth stagnation. Our findings suggest that DHEA causes perturbation of multiple pathways in cellular metabolism. Decreased SAM production, and cardiolipin depletion and the resulting mitochondrial dysfunction underlie the antiproliferative effect of DHEA.


Assuntos
Anticarcinógenos/farmacologia , Antineoplásicos/farmacologia , Desidroepiandrosterona/farmacologia , Metaboloma , Carcinoma Hepatocelular , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida , Glucosefosfato Desidrogenase/metabolismo , Humanos , Neoplasias Hepáticas , Metionina Adenosiltransferase/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilcolinas/metabolismo , Fosforilcolina/metabolismo , S-Adenosilmetionina/farmacologia
15.
Free Radic Biol Med ; 49(3): 361-73, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20420899

RESUMO

Glucose-6-phosphate dehydrogenase (G6PD) plays a key role in the regeneration of NADPH and maintenance of cellular redox balance. In the present study, we investigate the effect of G6PD deficiency on H(2)O(2)-elicited signaling in HepG2 cells. H(2)O(2) was found to inhibit cellular protein tyrosine phosphatase (PTP) activity, resulting in activation of MAPKs. MKP-1 expression increased in the late phase of H(2)O(2) signaling. Using RNAi technology, we found that G6PD knockdown enhanced the inhibitory effect of H(2)O(2) on PTPs and led to sustained MAPK activation. This was accompanied by delayed expression and inhibition of MKP-1. Using a pharmacological inhibitor and siRNA, we demonstrate that MKP-1 acts as a regulator of MAPK activation in H(2)O(2) signaling. The prolonged MAPK activation in G6PD-knockdown cells was associated with an increased susceptibility to H(2)O(2)-induced apoptosis and growth retardation. Treatment with p38 and JNK inhibitors or N-acetylcysteine ameliorated such cellular effect, while triptolide and MKP-1-siRNA did the opposite. Glucose oxidase treatment had similar effects as addition of H(2)O(2). Taken together, these findings suggest that G6PD knockdown enhances the magnitude and duration of H(2)O(2)-induced MAPK signaling through inhibition of cellular PTPs, and the resultant anomalous signaling may lead to cell demise.


Assuntos
Apoptose/efeitos dos fármacos , Glucosefosfato Desidrogenase/genética , Peróxido de Hidrogênio/farmacologia , Acetilcisteína/farmacologia , Antracenos/farmacologia , Fosfatase 1 de Especificidade Dupla/antagonistas & inibidores , Fosfatase 1 de Especificidade Dupla/metabolismo , Flavonoides/farmacologia , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Imidazóis/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Piridinas/farmacologia
16.
Free Radic Biol Med ; 47(5): 529-35, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19497363

RESUMO

Glucose-6-phosphate dehydrogenase (G6PD) has been recently found to play growth-regulatory roles in nucleated cells. To identify any other physiologic roles of G6PD, we generated G6PD-knockdown Hep G2 cells and investigated their susceptibility to oxidants. Hep G2 cells expressing shRNA against G6PD (Gi) were more susceptible to diamide-induced cytotoxicity than control cells expressing scrambled control shRNA (Sc). The level of reactive oxygen species in the Gi cells substantially exceeded that in Sc cells. This was accompanied by increased membrane peroxidation and the appearance of high-molecular-weight aggregates of membrane-associated cytoskeletal proteins in Gi cells. G6PD knockdown was associated with an impaired ability to regenerate glutathione. Diamide caused a considerable decrease in cellular glutathione level and a concomitant increase in glutathione disulfide in Gi cells. Consistent with this finding, N-acetylcysteine mitigated diamide-induced oxidative stress and cell death. Our findings suggest that G6PD confers protection against oxidant-induced cytotoxicity through effective glutathione regeneration.


Assuntos
Diamida/toxicidade , Glucosefosfato Desidrogenase/genética , Glutationa/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Antioxidantes/farmacologia , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Citoproteção/genética , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/genética , Eficiência/fisiologia , Técnicas de Silenciamento de Genes , Glucosefosfato Desidrogenase/antagonistas & inibidores , Glucosefosfato Desidrogenase/metabolismo , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/genética , Estresse Oxidativo/genética , RNA Interferente Pequeno/farmacologia , Radiossensibilizantes/toxicidade
17.
J Agric Food Chem ; 57(14): 6140-7, 2009 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-19537794

RESUMO

Oxidative stress is known to be a determinant of a host's susceptibility to pathogens. Natural compounds with antioxidant activity may provide a preventive measure against infection. Tea polyphenols were evaluated for their ability to inhibit enterovirus 71 (EV71) replication in Vero cell culture. Among the polyphenolic compounds tested, epigallocatechin gallate (EGCG) and gallocatechin gallate (GCG) potently inhibited replication of EV71. EGCG and GCG reduced the titer of infectious progeny virus by 95%. Quantitative RT-PCR analysis also revealed that EGCG suppressed replication of genomic RNA. It was accompanied by an increased cytoprotective effect. EGCG and GCG caused 5-fold increase in the viability of EV71-infected cells. The viral inhibitory effect correlated well with the antioxidant capacity of polyphenol. Mechanistically, EV71 infection led to increased oxidative stress, as shown by increased dichlorofluorescein and MitoSOX Red fluorescence. Upon EGCG treatment, reactive oxygen species (ROS) generation was significantly reduced. Consistent with this, EV71 replication was enhanced in glucose-6-phosphate dehydrogenase deficient cells, and such enhancement was largely reversed by EGCG. These findings suggest that EGCG may suppress viral replication via modulation of cellular redox milieu.


Assuntos
Antioxidantes/farmacologia , Antivirais/farmacologia , Catequina/análogos & derivados , Enterovirus Humano A/efeitos dos fármacos , Animais , Catequina/farmacologia , Linhagem Celular , Chlorocebus aethiops , Enterovirus Humano A/genética , Enterovirus Humano A/fisiologia , Fibroblastos/virologia , Doença de Depósito de Glicogênio Tipo I , Humanos , Masculino , Estresse Oxidativo/efeitos dos fármacos , RNA Viral/biossíntese , Espécies Reativas de Oxigênio/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Vero , Replicação Viral/efeitos dos fármacos
18.
Int J Oncol ; 33(5): 969-77, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18949359

RESUMO

DHEA is known to have anti-proliferative effect. The mechanism is not completely understood. We investigated the mechanism underlying DHEA-induced growth arrest of hepatoma cells. Growth inhibition was associated with increased G6PD activity, and insensitive to reversal by mevalonate. Thus, DHEA does not act via inhibition of G6PD and HMGR. Instead, growth stagnation was accompanied by reduced expression of nucleus-encoded mitochondrial genes; morphological and functional alterations of mitochondria; and depletion of intracellular ATP. Conversely, pyruvate supplementation alleviated DHEA-induced growth inhibition. It is likely that DHEA suppresses cell growth by altering mitochondrial gene expression, morphology and functions.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proliferação de Células , DNA Mitocondrial/metabolismo , Desidroepiandrosterona/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/patologia , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Metabolismo Energético , Glucosefosfato Desidrogenase/metabolismo , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Neoplasias Hepáticas/genética , Ácido Pirúvico/metabolismo , Fatores de Tempo
19.
J Gen Virol ; 89(Pt 9): 2080-2089, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18753216

RESUMO

Variations in the cellular microenvironment affect the host's susceptibility to pathogens. Using glucose-6-phosphate dehydrogenase (G6PD)-deficient fibroblasts as a model, this study demonstrated that the cellular redox status affects infectivity as well as the outcome of enterovirus 71 (EV71) infection. Compared with their normal counterparts, G6PD-deficient cells supported EV71 replication more efficiently and showed greater cytopathic effect and loss of viability. Mechanistically, viral infection led to increased oxidative stress, as indicated by increased dichlorofluorescein fluorescence and a diminished ratio of glutathione (GSH) to its disulfide form (GSSG), with the effect being greater in G6PD-deficient cells. Exogenous expression of active G6PD in the deficient cells, which increased the intracellular GSH:GSSG ratio, suppressed the generation of viral progeny. Consistent with this, treatment with N-acetylcysteine offered resistance to EV71 propagation and a cytoprotective effect on the infected cells. These findings support the notion that G6PD status, and thus redox balance, is an important determinant of enteroviral infection.


Assuntos
Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/enzimologia , Infecções por Enterovirus/etiologia , Glucosefosfato Desidrogenase/metabolismo , Antioxidantes/farmacologia , Sequência de Bases , Células Cultivadas , Efeito Citopatogênico Viral , DNA Viral/genética , Enterovirus Humano A/genética , Enterovirus Humano A/fisiologia , Infecções por Enterovirus/prevenção & controle , Infecções por Enterovirus/virologia , Deficiência de Glucosefosfato Desidrogenase/complicações , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Deficiência de Glucosefosfato Desidrogenase/virologia , Glutationa , Dissulfeto de Glutationa/metabolismo , Humanos , Estresse Oxidativo , Ensaio de Placa Viral , Virulência , Replicação Viral
20.
J Agric Food Chem ; 54(5): 1638-45, 2006 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-16506813

RESUMO

Glucose-6-phosphate dehydrogenase (G6PD) plays important roles in the maintenance of cellular redox balance. It was not until recently that the importance of G6PD in regulation of cellular growth and apoptosis emerged. In the present study, we found that G6PD-deficient fibroblasts were more susceptible to peroxynitrite-induced cytotoxicity. Treatment with peroxynitrite generator 3-morpholinosydnonimine (SIN-1) hydrochloride caused apoptosis in human fibroblast in a dose-dependent manner. This was preceded by a decrease in the intracellular level of glutathione (GSH) as well as accumulation of p53. The extent of apoptosis and glutathione depletion were greater in G6PD-deficient fibroblasts than in the normal counterpart. Pretreatment with green tea polyphenol epigallocatechin-3-gallate (EGCG) effectively blocked peroxynitrite-induced glutathione depletion, p53 accumulation, and apoptosis in both normal and G6PD-deficient cells. EGCG, administered to cells alone or as pretreatment, caused activation of Akt. The protective effect was abolished by phosphatidylinositol 3-kinase (PI3K) inhibitors, wortmannin, and LY294002. Our findings suggest that G6PD deficiency enhances the toxicity of peroxynitrite and that EGCG initiates cell survival signaling via the PI3K/akt pathway.


Assuntos
Camellia sinensis/química , Catequina/análogos & derivados , Sobrevivência Celular/efeitos dos fármacos , Glucosefosfato Desidrogenase/fisiologia , Ácido Peroxinitroso/farmacologia , Apoptose/efeitos dos fármacos , Catequina/farmacologia , Linhagem Celular , Fibroblastos/enzimologia , Deficiência de Glucosefosfato Desidrogenase , Glutationa/metabolismo , Humanos , Molsidomina/análogos & derivados , Molsidomina/farmacologia , Oxirredução , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA