Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.472
Filtrar
1.
Int J Mol Sci ; 24(15)2023 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-37569345

RESUMO

In previous work, we showed that cancer cells do not depend on glycolysis for ATP production, but they do on fatty acid oxidation. However, we found some cancer cells induced cell death after glucose deprivation along with a decrease of ATP production. We investigated the different response of glucose deprivation with two types of cancer cells including glucose insensitive cancer cells (GIC) which do not change ATP levels, and glucose sensitive cancer cells (GSC) which decrease ATP production in 24 h. Glucose deprivation-induced cell death in GSC by more than twofold after 12 h and by up to tenfold after 24 h accompanied by decreased ATP production to compare to the control (cultured in glucose). Glucose deprivation decreased the levels of metabolic intermediates of the pentose phosphate pathway (PPP) and the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) in both GSC and GIC. However, glucose deprivation increased reactive oxygen species (ROS) only in GSC, suggesting that GIC have a higher tolerance for decreased NADPH than GSC. The twofold higher ratio of reduced/oxidized glutathione (GSH/GSSG) in GIS than in GSC correlates closely with the twofold lower ROS levels under glucose starvation conditions. Treatment with N-acetylcysteine (NAC) as a precursor to the biologic antioxidant glutathione restored ATP production by 70% and reversed cell death caused by glucose deprivation in GSC. The present findings suggest that glucose deprivation-induced cancer cell death is not caused by decreased ATP levels, but rather triggered by a failure of ROS regulation by the antioxidant system. Conclusion is clear that glucose deprivation-induced cell death is independent from ATP depletion-induced cell death.


Assuntos
Trifosfato de Adenosina , Glucose , Neoplasias , Espécies Reativas de Oxigênio , Glucose/deficiência , Trifosfato de Adenosina/metabolismo , Via de Pentose Fosfato , Espécies Reativas de Oxigênio/metabolismo , NADP/metabolismo , Glutationa/metabolismo , Acetilcisteína/metabolismo , Acetilcisteína/farmacologia , Células PC-3 , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Morte Celular
2.
Nature ; 618(7963): 151-158, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37198494

RESUMO

Pancreatic ductal adenocarcinoma (PDA) is a lethal disease notoriously resistant to therapy1,2. This is mediated in part by a complex tumour microenvironment3, low vascularity4, and metabolic aberrations5,6. Although altered metabolism drives tumour progression, the spectrum of metabolites used as nutrients by PDA remains largely unknown. Here we identified uridine as a fuel for PDA in glucose-deprived conditions by assessing how more than 175 metabolites impacted metabolic activity in 21 pancreatic cell lines under nutrient restriction. Uridine utilization strongly correlated with the expression of uridine phosphorylase 1 (UPP1), which we demonstrate liberates uridine-derived ribose to fuel central carbon metabolism and thereby support redox balance, survival and proliferation in glucose-restricted PDA cells. In PDA, UPP1 is regulated by KRAS-MAPK signalling and is augmented by nutrient restriction. Consistently, tumours expressed high UPP1 compared with non-tumoural tissues, and UPP1 expression correlated with poor survival in cohorts of patients with PDA. Uridine is available in the tumour microenvironment, and we demonstrated that uridine-derived ribose is actively catabolized in tumours. Finally, UPP1 deletion restricted the ability of PDA cells to use uridine and blunted tumour growth in immunocompetent mouse models. Our data identify uridine utilization as an important compensatory metabolic process in nutrient-deprived PDA cells, suggesting a novel metabolic axis for PDA therapy.


Assuntos
Glucose , Neoplasias Pancreáticas , Ribose , Microambiente Tumoral , Uridina , Animais , Camundongos , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Ribose/metabolismo , Uridina/química , Glucose/deficiência , Divisão Celular , Linhagem Celular Tumoral , Sistema de Sinalização das MAP Quinases , Uridina Fosforilase/deficiência , Uridina Fosforilase/genética , Uridina Fosforilase/metabolismo , Humanos
3.
Nat Commun ; 13(1): 572, 2022 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-35102143

RESUMO

Substrate inhibition of enzymes can be a major obstacle to the production of valuable chemicals in engineered microorganisms. Here, we show substrate inhibition of lycopene cyclase as the main limitation in carotenoid biosynthesis in Yarrowia lipolytica. To overcome this bottleneck, we exploit two independent approaches. Structure-guided protein engineering yields a variant, Y27R, characterized by complete loss of substrate inhibition without reduction of enzymatic activity. Alternatively, establishing a geranylgeranyl pyrophosphate synthase-mediated flux flow restrictor also prevents the onset of substrate inhibition by diverting metabolic flux away from the inhibitory metabolite while maintaining sufficient flux towards product formation. Both approaches result in high levels of near-exclusive ß-carotene production. Ultimately, we construct strains capable of producing 39.5 g/L ß-carotene at a productivity of 0.165 g/L/h in bioreactor fermentations (a 1441-fold improvement over the initial strain). Our findings provide effective approaches for removing substrate inhibition in engineering pathways for efficient synthesis of natural products.


Assuntos
Licopeno/metabolismo , Yarrowia/metabolismo , Acetilcoenzima A/metabolismo , Reatores Biológicos , Carbono/metabolismo , Citosol/metabolismo , Farnesiltranstransferase/metabolismo , Fermentação , Glucose/deficiência , Liases Intramoleculares/metabolismo , Metabolismo dos Lipídeos , Lipídeos/biossíntese , Licopeno/química , Análise do Fluxo Metabólico , Engenharia de Proteínas , Especificidade por Substrato , Terpenos/metabolismo
4.
Sci Rep ; 12(1): 1710, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35110605

RESUMO

The study is aimed at elucidating the effect of selenium nanoparticles (SeNPs) on the death of cells in the primary culture of mouse cerebral cortex during oxygen and glucose deprivation (OGD). A primary cell culture of the cerebral cortex containing neurons and astrocytes was subjected to OGD and reoxygenation to simulate cerebral ischemia-like conditions in vitro. To evaluate the neuroprotective effect of SeNPs, cortical astrocytes and neurons were incubated for 24 h with SeNPs, and then subjected to 2-h OGD, followed by 24-h reoxygenation. Vitality tests, fluorescence microscopy, and real-time PCR have shown that incubation of primary cultured neurons and astrocytes with SeNPs at concentrations of 2.5-10 µg/ml under physiological conditions has its own characteristics depending on the type of cells (astrocytes or neurons) and leads to a dose-dependent increase in apoptosis. At low concentration SeNPs (0.5 µg/ml), on the contrary, almost completely suppressed the processes of basic necrosis and apoptosis. Both high (5 µg/ml) and low (0.5 µg/ml) concentrations of SeNPs, added for 24 h to the cells of cerebral cortex, led to an increase in the expression level of genes Bcl-2, Bcl-xL, Socs3, while the expression of Bax was suppressed. Incubation of the cells with 0.5 µg/ml SeNPs led to a decrease in the expression of SelK and SelT. On the contrary, 5 µg/ml SeNPs caused an increase in the expression of SelK, SelN, SelT, SelP. In the ischemic model, after OGD/R, there was a significant death of brain cells by the type of necrosis and apoptosis. OGD/R also led to an increase in mRNA expression of the Bax, SelK, SelN, and SelT genes and suppression of the Bcl-2, Bcl-xL, Socs3, SelP genes. Pre-incubation of cell cultures with 0.5 and 2.5 µg/ml SeNPs led to almost complete inhibition of OGD/R-induced necrosis and greatly reduced apoptosis. Simultaneously with these processes we observed suppression of caspase-3 activation. We hypothesize that the mechanisms of the protective action of SeNPs involve the activation of signaling cascades recruiting nuclear factors Nrf2 and SOCS3/STAT3, as well as the activation of adaptive pathways of ESR signaling of stress arising during OGD and involving selenoproteins SelK and SelT, proteins of the Bcl-2 family ultimately leading to inactivation of caspase-3 and inhibition of apoptosis. Thus, our results demonstrate that SeNPs can act as neuroprotective agents in the treatment of ischemic brain injuries.


Assuntos
Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Córtex Cerebral/efeitos dos fármacos , Glucose/deficiência , Nanopartículas , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Compostos de Selênio/farmacologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Hipóxia Celular , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Técnicas de Cocultura , Feminino , Masculino , Camundongos , Necrose , Neurônios/metabolismo , Neurônios/patologia , Cultura Primária de Células , Selenoproteínas/genética , Selenoproteínas/metabolismo
5.
Int Immunopharmacol ; 104: 108464, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35021128

RESUMO

BACKGROUND: Emerging evidence has shown that circular RNAs (circRNAs) are involved in the pathogenesis of ischemic stroke (IS). Nonetheless, the function of circ_0000647 was not reported. METHODS: Oxygen-glucose deprivation and reperfusion (OGD/R)-treated SK-N-SH cells were used to mimic cerebral ischemia/reperfusion (I/R) conditions. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were used to measure the levels of circ_0000647, microRNA-126-5p (miR-126-5p) and TNF receptor associated factor 3 (TRAF3). Cell Counting Kit-8 (CCK-8) assay, 5'-ethynyl-2'-deoxyuridine (EDU) assay and flow cytometry analysis were employed to assess cell proliferation and apoptosis. Enzyme-linked immunosorbent assay (ELISA) was conducted for the concentrations of IL-6 and TNF-α. Oxidative stress was assessed by determining malondialdehyde (MDA) level and superoxide dismutase (SOD) activity. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were adopted to estimate the relationships of circ_0000647, miR-126-5p and TRAF3. The morphology and size of exosomes were observed via transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) analysis. RESULTS: Circ_0000647 was elevated in OGD/R-treated SK-N-SH cells. OGD/R treatment suppressed the proliferation and promoted the apoptosis, inflammation and oxidative stress in SK-N-SH cells, while circ_0000647 knockdown reversed the effects. Circ_0000647 could sponge miR-126-5p, which directly targeted TRAF3. MiR-126-5p overexpression alleviated OGD/R-induced SK-N-SH cell damage and miR-126-5p inhibition reversed the effect of circ_0000647 knockdown on OGD/R-induced SK-N-SH cell damage. Moreover, TRAF3 elevation abated miR-126-5p-mediated effect on SK-N-SH cell injury. In addition, exosomal circ_0000647 level was increased in OGD/R-stimulated SK-N-SH cells. CONCLUSION: Circ_0000647 interference relieved OGD/R-induced SK-N-SH cell damage by altering miR-126-5p/TRAF3 axis.


Assuntos
MicroRNAs , RNA Circular , Traumatismo por Reperfusão/genética , Fator 3 Associado a Receptor de TNF/genética , Hipóxia Celular/genética , Linhagem Celular Tumoral , Glucose/deficiência , Humanos , Interleucina-6/metabolismo , Modelos Biológicos , Oxigênio , Traumatismo por Reperfusão/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
Int Immunopharmacol ; 104: 108443, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35021129

RESUMO

AIM: Hepatic ischemia/reperfusion (I/R) injury is a significant pathological process that contributes to high morbidity and mortality rates, although the underlying mechanism is unknown. Recent studies have shown that transient receptor potential melastatin 2 (TRPM2) plays a critical role in organ I/R injury, but the exact mechanism is elusive. This study investigates the role and mechanism of TPRM2 in hepatic I/R injury and oxygen-glucosedeprivation/reoxygenation (OGD/R) induced hepatocyte injury. METHODS: We evaluated the effects of TRPM2 on hepatic I/R injury using a knockout mouse model of hepatic I/R. In a model of OGD/R in hepatocytes, we investigated the mechanism of TPRM2 in it using the autophagy agonist and inhibitor and an NLRP3 inhibitor. RESULTS: We discovered that knockout of TRPM2 protected against hepatic I/R accompanied by autophagy activation and NLRP3 inflammasome pathway inhibition. Furthermore, increasing autophagy attenuated OGD/R-induced cell injury and knockdown of TRPM2 alleviated the injury by activating autophagy. Additionally, we detected the expression of NLRP3 inflammasome pathway in the OGD/R-induced hepatocytes which had been treated with the autophagy agonist and inhibitor, and found that autophagy negatively regulated the NLRP3 inflammasome pathway. Moreover, we discovered that the administration of NLRP3-inhibitor INF39 increased cell viability and caused a decline in cell death in the OGD/R-treated hepatocytes. CONCLUSIONS: Downregulation of TRPM2 protected the liver against I/R injury and OGD/R induced injury, mediated by autophagy activation and inhibition of the NLRP3 inflammasome pathway, whereas autophagy negatively regulated the NLRP3 inflammasome pathway in this process.


Assuntos
Hepatopatias/genética , Traumatismo por Reperfusão/genética , Canais de Cátion TRPM/genética , Animais , Autofagia , Hipóxia Celular/genética , Linhagem Celular , Regulação para Baixo , Glucose/deficiência , Hepatócitos , Humanos , Inflamassomos/metabolismo , Hepatopatias/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais
7.
Mol Cell ; 82(1): 60-74.e5, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34995509

RESUMO

Acetyl-CoA is a key intermediate situated at the intersection of many metabolic pathways. The reliance of histone acetylation on acetyl-CoA enables the coordination of gene expression with metabolic state. Abundant acetyl-CoA has been linked to the activation of genes involved in cell growth or tumorigenesis through histone acetylation. However, the role of histone acetylation in transcription under low levels of acetyl-CoA remains poorly understood. Here, we use a yeast starvation model to observe the dramatic alteration in the global occupancy of histone acetylation following carbon starvation; the location of histone acetylation marks shifts from growth-promoting genes to gluconeogenic and fat metabolism genes. This reallocation is mediated by both the histone deacetylase Rpd3p and the acetyltransferase Gcn5p, a component of the SAGA transcriptional coactivator. Our findings reveal an unexpected switch in the specificity of histone acetylation to promote pathways that generate acetyl-CoA for oxidation when acetyl-CoA is limiting.


Assuntos
Gluconeogênese , Glucose/deficiência , Histonas/metabolismo , Metabolismo dos Lipídeos , Processamento de Proteína Pós-Traducional , Saccharomyces cerevisiae/metabolismo , Acetilcoenzima A/metabolismo , Acetilação , Regulação Fúngica da Expressão Gênica , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Metabolismo dos Lipídeos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transativadores/genética , Transativadores/metabolismo
8.
Sci Rep ; 12(1): 1230, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35075260

RESUMO

Human gingival fibroblasts (HGnFs) maintain periodontal tissue homeostasis through active proliferation and migration. Clinically, it is considered that the wound-healing ability of the gingival tissue is maintained even in environments with insufficient supply of nutrients, such as glucose, immediately after periodontal surgery. However, the effects of such glucose-deficient environments on HGnFs remain unclear. This study aimed to investigate the effects of low-glucose environment on HGnFs homeostasis. We evaluated gingival wound healing by examining cell proliferation and migration and collagen synthesis in HGnFs cultured in 100, 50, 25, and 0 mg/dL glucose in vitro. The cellular stress levels were determined by measuring the lactate dehydrogenase (LDH) and reactive oxygen species (ROS) levels. The glucose metabolism of HGnFs in the low-glucose concentrations was studied by measuring glucose transporter type 1 (GLUT1) mRNA expression, glucose uptake assays, lactate and ATP productions. Molecular effects were examined with a focus on the LKB1-AMPK signaling pathway. Autophagy activity in glucose-deprived HGnFs was evaluated by measuring the levels of autophagy-related proteins. Low glucose levels increased cellular stress levels, autophagy activity, and enhanced glucose metabolism through the LKB1-AMPK signaling pathway, providing more ATPs to promote wound healing. Our results regarding glucose transfer suggest the rapid healing of gingival wounds.


Assuntos
Autofagia , Fibroblastos/fisiologia , Gengiva/fisiologia , Glucose/deficiência , Cicatrização , Quinases Proteína-Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Células Cultivadas , Glicólise , Humanos
9.
Neurosci Lett ; 770: 136381, 2022 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-34906568

RESUMO

BACKGROUND: The maintenance of human brain microvascular endothelial cell (HBMEC) function is crucial to improve the outcomes of ischemic stroke (IS). Emerging evidence shows that circular RNAs (circRNAs) are involved in IS progression. This study aimed to investigate the role of circRNA FUN14 domain containing 1 (circFUNDC1) in oxygen-glucose deprivation (OGD)-treated HBMECs. METHODS: The expression of circFUNDC1, miR-375 and phosphatase and tensin homolog (PTEN) mRNA was detected by quantitative real-time PCR (qPCR). Cell viability, apoptosis, migration and angiogenesis were determined by CCK-8 assay, flow cytometry assay, transwell assay and tube formation assay. The protein level of PTEN was detected by western blot. The relationship between miR-375 and circFUNDC1 or PTEN was confirmed by pull-down assay, dual-luciferase reporter assay and RIP assay. Exosomes were identified by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). RESULTS: CircFUNDC1 expression was increased in peripheral blood of IS patients and OGD-treated HBMECs. CircFUNDC1 knockdown alleviated OGD-induced cell apoptosis and promoted OGD-blocked cell viability, migration and angiogenesis of HBMECs. MiR-375 was a target of circFUNDC1, and miR-375 restoration played similar effects with circFUNDC1 knockdown. The inhibition of miR-375 reversed the effects of circFUNDC1 knockdown. In addition, PTEN was a downstream target of miR-375, and PTEN overexpression abolished the effects of miR-375 restoration. The expression of circFUNDC1 was elevated in serum-derived exosomes of IS patients, and circFUNDC1 harbored diagnostic values. CONCLUSION: CircFUNDC1 knockdown alleviates OGD-induced HBMECs injuries by inhibiting PTEN via enriching miR-375.


Assuntos
Células Endoteliais/metabolismo , AVC Isquêmico/metabolismo , Proteínas de Membrana/genética , MicroRNAs/metabolismo , Proteínas Mitocondriais/genética , PTEN Fosfo-Hidrolase/genética , RNA Circular/metabolismo , Idoso , Encéfalo/irrigação sanguínea , Hipóxia Celular , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Exossomos/metabolismo , Feminino , Glucose/deficiência , Humanos , AVC Isquêmico/genética , Masculino , MicroRNAs/genética , Microvasos/citologia , Microvasos/metabolismo , Pessoa de Meia-Idade , Oxigênio/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , RNA Circular/genética
10.
Microvasc Res ; 140: 104302, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34919942

RESUMO

PURPOSE: Myocardial ischemia/reperfusion injury (MI/RI) is a major problem in the clinical treatment of ischemic cardiomyopathy, and its specific underlying mechanisms are complicated and still unclear. A number of studies have indicated that the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxidase-1(HO-1) signaling pathway might serve as an important target for the management of MI/RI. Catalpol is a kind of iridoid glucoside that has been found to exhibit diverse anti-inflammatory and antioxidant properties. This study was aimed at investigating the role of Catalpol in targeting MI/RI and its related mechanisms in an oxygen-glucose deprivation/reoxygenation (OGD/R) model in vitro and a preclinical ischemia/reperfusion (I/R) model. METHODS: This study using both in vitro and in vivo models investigated the possible role and underlying mechanisms used by Catalpol for modulating of MI/RI. The potential effects of Catalpol on the viability of cardiomyocytes were measured by cell counting kit-8 (CCK-8) assays. The phenotypes of myocardial injury, oxidative stress and inflammation markers were measured by western blot, immunofluorescence, enzyme-linked immunosorbent assay (ELISA) etc. Nrf2/HO-1 signaling pathway was detected by immunofluorescence and western blot analysis. RESULTS: We found that Catalpol significantly suppressed the process of MI/RI and protected OGD/R-treated cardiomyocytes by inhibiting the various markers of inflammation and suppressing oxidative stress. Additionally, mechanistically it was also demonstrated that Catalpol could effectively activate Nrf2/HO-1 signaling pathway to suppress the damage caused by inflammation and oxidative stress in MI/RI. CONCLUSION: In summary, the findings suggest that Catalpol exerted significant cardioprotective effects following myocardial ischemia, possibly through the activation of the Nrf2/HO-1 signaling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Heme Oxigenase-1/metabolismo , Glucosídeos Iridoides/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Hipóxia Celular , Linhagem Celular , Modelos Animais de Doenças , Glucose/deficiência , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais
11.
Microvasc Res ; 140: 104297, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34890690

RESUMO

Angiogenesis caused by acute vascular occlusion occurs in various ischemic diseases. The in vitro tube formation assay by endothelial cells is a rapid, quantitative method for drug discovery on angiogenesis. Tube formation assay on Matrigel has been widely used to identify the angiogenesis, however, there are some problems to limit its application. In this study, we found for the first time that sodium dithionite (SD) could induce endothelial cell tube formation without Matrigel under hypoxia condition. To further verify our findings, the angiogenesis related proteins and mRNA at different time points after tube formation were measured both in primary human large-vessel endothelial cell (HUVECs) and murine microvascular endothelial cell line (Bend.3). In conclusion, compared with traditional tube formation on Matrigel, the novel model exhibits the following advantages: (1) Combination oxygen glucose deprivation with sodium dithionite (OGD-SD) model is operated more easily than traditional tube formation. (2) OGD-SD can be used for not only cell imaging, but also immunofluorescence, protein extraction and gene analysis. (3) OGD-SD is more applicable to acute hypoxia model of endothelial cell in vitro. (4) OGD-SD may be more suitable to identify molecular mechanism of compound that intervenes processes of pro-tube formation, tube formation and tube disconnection.


Assuntos
Células Endoteliais/patologia , Células Endoteliais da Veia Umbilical Humana/patologia , Neovascularização Patológica , Neovascularização Fisiológica , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Bioensaio , Hipóxia Celular , Linhagem Celular , Movimento Celular , Ditionita/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Glucose/deficiência , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Transdução de Sinais
12.
Cancer Sci ; 113(1): 132-144, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34668620

RESUMO

In solid tumors, cancer cells have devised multiple approaches to survival and proliferate in response to glucose starvation that is often observed in solid tumor microenvironments. However, the precise mechanisms are far less known. Herein, we report that glucose deprivation activates 90-kDa ribosomal S6 kinase (p90 RSK), a highly conserved Ser/Thr kinase, and activated p90 RSK promotes cancer cell survival. Mechanistically, activated p90 RSK by glucose deprivation phosphorylates checkpoint kinase 1 (CHK1), a key transducer in checkpoint signaling pathways, at Ser280 and triggers CHK1 ubiquitination mediated by SCFß-TrCP ubiquitin ligase and proteasomal degradation, subsequently suppressing cancer cell apoptosis induced by glucose deprivation. Importantly, we identified an inverse correlation between p90 RSK activity and CHK1 levels within the solid tumor mass, with lower levels of CHK1 and higher activity of p90 RSK in the center of the tumor where low glucose concentrations are often observed. Thus, our study indicates that p90 RSK promotes CHK1 phosphorylation at Ser280 and its subsequent degradation, which allows cancer cells to escape from checkpoint signals under the stress of glucose deprivation, leading to cell survival and thus contributing to tumorigenesis.


Assuntos
Quinase 1 do Ponto de Checagem/metabolismo , Glucose/deficiência , Neoplasias/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Quinase 1 do Ponto de Checagem/química , Ativação Enzimática , Células HEK293 , Humanos , Camundongos , Fosforilação , Proteólise/efeitos dos fármacos , Pteridinas/farmacologia , Ubiquitinação/efeitos dos fármacos
13.
J Neurochem ; 160(3): 392-411, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34837397

RESUMO

TBC1Domain Family Member 25 (TBC1D25) is a protein that contains a TBC/RAB-GTPase activating protein (GAP) domain, which was shown to participate in autophagy in previous studies. However, the role of TBC1D25 in cerebral ischemia-reperfusion (I/R) injury remains unknown. In this study, we found that the mRNA and protein expression levels of TBC1D25 decreased in mouse brain after I/R injury and primary cortical neurons treated with oxygen and glucose deprivation/reoxygenation (OGD/R). Then TBC1D25 knockout (KO) mice were applied to demonstrate that TBC1D25 ablation aggravated cerebral I/R-induced neuronal loss and infarct size. In addition, neuronal apoptosis and inflammation were significantly potentiated in the TBC1D25-KO group. In in vitro OGD/R model, TBC1D25 knockdown can attenuate neuronal cell viability and aggravate the process of inflammation and apoptosis. Conversely, over-expression of TBC1D25 in primary neurons ameliorated the aforementioned processes. Mechanistically, RNA-sequencing (RNA-seq) analysis revealed mitogen-activated protein kinase (MAPK) signaling pathway was the most significant pathway that contributed to TBC1D25-mediated brain I/R injury process. Through experimental verification, TBC1D25 deficiency increased the phosphorylation of the transforming growth factor-ß-activated kinase 1 (TAK1)-c-Jun N-terminal kinase (JNK)/p38 axis in neurons during the brain I/R injury. Furthermore, we found that TAK1 blockade abrogated the apoptosis and inflammatory response produced by TBC1D25 knockdown in vitro. In conclusion, this study is the first to demonstrate the functional significance of TBC1D25 in the pathophysiology of brain I/R injury, and the protective mechanism of TBC1D25 is dependent on the TAK1-JNK/p38 pathway.


Assuntos
Isquemia Encefálica/genética , Proteínas Ativadoras de GTPase/genética , Proteínas Quinases JNK Ativadas por Mitógeno/genética , MAP Quinase Quinase Quinases/genética , Traumatismo por Reperfusão/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Animais , Apoptose , Isquemia Encefálica/fisiopatologia , Proteínas Ativadoras de GTPase/deficiência , Glucose/deficiência , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/fisiopatologia , Inflamação/genética , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , RNA-Seq , Traumatismo por Reperfusão/fisiopatologia
14.
Stroke ; 53(2): e37-e41, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34743535

RESUMO

BACKGROUND AND PURPOSE: Neuroprotective strategies for stroke remain inadequate. Nanoliposomes comprised of phosphatidylcholine, cholesterol, and monosialogangliosides (nanoliposomes) induced an antioxidant protective response in endothelial cells exposed to amyloid insults. We tested the hypotheses that nanoliposomes will preserve human neuroblastoma (SH-SY5Y) and human brain microvascular endothelial cells viability following oxygen-glucose deprivation (OGD)-reoxygenation and will reduce injury in mice following middle cerebral artery occlusion. METHODS: SH-SY5Y and human brain microvascular endothelial cells were exposed to oxygen-glucose deprivation-reoxygenation (3 hours 0.5%-1% oxygen and glucose-free media followed by 20-hour ambient air/regular media) without or with nanoliposomes (300 µg/mL). Viability was measured (calcein-acetoxymethyl fluorescence) and protein expression of antioxidant proteins HO-1 (heme oxygenase-1), NQO1 (NAD[P]H quinone dehydrogenase 1), and SOD1 (superoxide dismutase 1) were measured by Western blot. C57BL/6J mice were treated with saline (n=8) or nanoliposomes (10 mg/mL lipid, 200 µL, n=7) while undergoing 60-minute middle cerebral artery occlusion followed by reperfusion. Day 2 postinjury neurological impairment score and infarction size were compared. RESULTS: SH-SY5Y and human brain microvascular endothelial cells showed reduced viability post-oxygen-glucose deprivation-reoxygenation that was reversed by nanoliposomes. Nanoliposomes increased protein expressions of HO-1, NQO1 in both cell types and SOD1 in human brain microvascular endothelial cells. Nanoliposomes-treated mice showed reduced neurological impairment and brain infarct size (18.8±2% versus 27.3±2.3%, P=0.017) versus controls. CONCLUSIONS: Nanoliposomes reduced stroke injury in mice subjected to middle cerebral artery occlusion likely through induction of an antioxidant protective response. Nanoliposome is a candidate novel agent for stroke.


Assuntos
Infarto da Artéria Cerebral Média/tratamento farmacológico , Lipossomos/uso terapêutico , Nanopartículas/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Antioxidantes/metabolismo , Linhagem Celular , Endotélio Vascular/patologia , Glucose/deficiência , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Humanos , Hipóxia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/patologia , NAD(P)H Desidrogenase (Quinona)/biossíntese , NAD(P)H Desidrogenase (Quinona)/genética , Traumatismo por Reperfusão/patologia , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia , Superóxido Dismutase-1/biossíntese , Superóxido Dismutase-1/genética
15.
Kaohsiung J Med Sci ; 38(1): 38-48, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34369657

RESUMO

Proteins in Jumonji family function as histone demethylases and participate in cardiac development. Jumonji domain containing 5 (JMJD5) is responsible for the embryonic development through removing methyl moieties from H3K36me2 histone, and has pro-proliferative effect on heart and eye development. However, the protective role of JMJD5 against oxygen-glucose deprivation and reperfusion (OGD/R)-induced injury in cardiomyocytes has not been fully understood. Firstly, myocardial ischemia/reperfusion (I/R) rat model was established by ligation of left coronary artery. OGD/R was performed in non-transfected H9C2 or H9C2 transfected with pcDNA-JMJD5 plasmid to induce cell cytotoxicity. Data from qRT-PCR and western blot showed that JMJD5 was reduced in the heart tissues of myocardial I/R rat model and OGD/R-induced H9C2. Secondly, JMJD5 over-expression attenuated OGD/R-induced decrease in cell viability and increase in lactate dehydrogenase secretion and cell apoptosis in H9C2. Mitophagy was promoted by pcDNA-mediated over-expression of JMJD5 with enhanced protein expression of LC3-I, LC3-II, Atg5, and Beclin 1. Thirdly, knockdown of JMJD5 aggravated OGD/R-induced decrease in hypoxia-inducible factor-1α (HIF-1α), whereas JMJD5 over-expression enhanced BNIP3 (Bcl-2/adenovirus E1B 19-kDa interacting protein) through upregulation of HIF-1α. Lastly, BNIP3 silencing promoted cell apoptosis, suppressed mitophagy, and attenuated the protective effects of JMJD5 over-expression against OGD/R-induced injury in H9C2. In conclusion, JMJD5 exerted protective effects against OGD/R-induced injury in cardiomyocytes through upregulation of HIF-1α-BNIP3.


Assuntos
Glucose , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Oxigênio/metabolismo , Animais , Apoptose , Sobrevivência Celular , Modelos Animais de Doenças , Glucose/deficiência , Glucose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Histona Desmetilases com o Domínio Jumonji , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Mitofagia , Traumatismo por Reperfusão Miocárdica/induzido quimicamente , Traumatismo por Reperfusão Miocárdica/enzimologia , Miócitos Cardíacos/citologia , Substâncias Protetoras , Ratos
16.
Bioengineered ; 12(2): 12274-12293, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34898370

RESUMO

Stroke is a leading cause of morbidity and mortality worldwide. As the most common type of stroke cases, treatment effectiveness is still limited despite intensive research. Recently, traditional Chinese medicine has attracted attention because of potential benefits for stroke treatment. Among these, luteolin, a natural plant flavonoid compound, offers neuroprotection following against ischemic stroke, although the specific mechanisms are unknown. Here we used network pharmacology, molecular docking, and experimental verification to explore the mechanisms whereby luteolin can benefit stroke recovery. The pharmacological and molecular properties of luteolin were obtained from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform. The potential targets of luteolin and ischemic stroke were collected from interrogating public databases. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed by Funrich and Database for Annotation, Visualization and Integrated Discovery respectively, a luteolin-target-pathway network constructed using Cytoscape, Autodock vina was used for molecular docking simulation with Discovery Studio was used to visualize and analyze the docked conformations. Lastly, we employed an in vitro model of stroke injury to evaluate the effects of luteolin on cell survival and expression of the putative targets. From 95 candidate luteolin target genes, our analysis identified six core targets . KEGG analysis of the candidate targets identified that luteolin provides therapeutic effects on stroke through TNF signaling and other pathways. Our experimental analyses confirmed the conclusions analyzed above. In summary, the molecular and pharmacological mechanisms of luteolin against stroke are indicated in our study from a systematic perspective.


Assuntos
AVC Isquêmico/tratamento farmacológico , Luteolina/uso terapêutico , Simulação de Acoplamento Molecular , Farmacologia em Rede , Animais , Células CACO-2 , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Ontologia Genética , Glucose/deficiência , Humanos , AVC Isquêmico/genética , AVC Isquêmico/patologia , Luteolina/farmacologia , Oxigênio , Células PC12 , Mapas de Interação de Proteínas , Ratos , Reprodutibilidade dos Testes
17.
Bioengineered ; 12(2): 12917-12930, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34852714

RESUMO

Ischemia injury can cause cell death or impairment of neuron and astrocytes, and thus induce loss of nerve function. central nervous systems injury induces an aberrant activation of astrocytes called astrogliosis. Pyroptosis, which is a kind of programmed cell death, was proved play an important role in ischemia injury. Zinc Finger E-Box Binding Homeobox 2 (ZEB2) promoted neuron astrogliosis, which play a protected role in neuron regeneration. However, its precise mechanism remains unclear. This study investigated the mechanism of ZEB2 on astrogliosis and neuron regeneration after cerebral ischemia reperfusion condition. To confirm our hypothesis, Neurons and astrocytes were isolated from fetal Sprague Dawley rats, in vivo Middle Cerebral Artery Occlusion/reperfusion (MCAO/R) rat model and in vitro oxygen-glucose deprivation/reperfusion (OGD/R)-treated astrocytes and neurocytes model were constructed. Our results showed that ZEB2 was expressed in nucleus of astrocyte and upregulated after OGD/R induction, ZEB2 promoted astrogliosis. Further upregulation of ZEB2 promoted the astrogliosis, which promoted neuron proliferation and regeneration by decreased pyroptosis. Moreover, this finding was further confirmed in vivo MCAO/R rat model. Overexpression of ZEB2 promoted astrogliosis, which decreased infarct volume and accumulated recovery of neurological function by alleviated pyroptosis. This finding revealed that ZEB2 was a regulator of the astrogliosis after ischemia/reperfusion injury, and then astrogliosis promoted neuron regeneration via decreased neuron pyroptosis. Therefore, ZEB2 may be a potential therapeutic target for ischemia/reperfusion injury.


Assuntos
Isquemia Encefálica/patologia , Gliose/metabolismo , Gliose/patologia , Neuroproteção , Piroptose , Traumatismo por Reperfusão/patologia , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Gliose/complicações , Gliose/fisiopatologia , Glucose/deficiência , Regeneração Nervosa , Oxigênio , Ratos Sprague-Dawley , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia
18.
Cells ; 10(12)2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34944075

RESUMO

Protein aggregates of cofilin and actin have been found in neurons under oxygen-glucose deprivation. However, the regulatory mechanism behind the expression of Cfl1 during oxygen-glucose deprivation remains unclear. Here, we found that heterogeneous nuclear ribonucleoproteins (hnRNP) Q and hnRNP A1 regulate the translation of Cfl1 mRNA, and formation of cofilin-actin aggregates. The interaction between hnRNP A1 and Cfl1 mRNA was interrupted by hnRNP Q under normal conditions, while the changes in the expression and localization of hnRNP Q and hnRNP A1 increased such interaction, as did the translation of Cfl1 mRNA under oxygen-glucose deprived conditions. These findings reveal a new translational regulatory mechanism of Cfl1 mRNA in hippocampal neurons under oxygen-glucose deprivation.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Glucose/deficiência , Ribonucleoproteína Nuclear Heterogênea A1/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Hipocampo/patologia , Neurônios/metabolismo , Oxigênio/metabolismo , Biossíntese de Proteínas , Fatores de Despolimerização de Actina/genética , Animais , Isquemia Encefálica/patologia , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
19.
Front Immunol ; 12: 730672, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34737742

RESUMO

At sites of inflammation, monocytes carry out specific immune functions while facing challenging metabolic restrictions. Here, we investigated the potential of human monocytes to adapt to conditions of gradually inhibited oxidative phosphorylation (OXPHOS) under glucose free conditions. We used myxothiazol, an inhibitor of mitochondrial respiration, to adjust two different levels of decreased mitochondrial ATP production. At these levels, and compared to uninhibited OXPHOS, we assessed phagocytosis, production of reactive oxygen species (ROS) through NADPH oxidase (NOX), expression of surface activation markers CD16, CD80, CD11b, HLA-DR, and production of the inflammatory cytokines IL-1ß, IL-6 and TNF-α in human monocytes. We found phagocytosis and the production of IL-6 to be least sensitive to metabolic restrictions while surface expression of CD11b, HLA-DR, production of TNF-α, IL-1ß and production of ROS through NOX were most compromised by inhibition of OXPHOS in the absence of glucose. Our data demonstrate a short-term hierarchy of immune functions in human monocytes, which represents novel knowledge potentially leading to the development of new therapeutics in monocyte-mediated inflammatory diseases.


Assuntos
Metabolismo Energético , Glucose/deficiência , Interleucina-6/metabolismo , Mitocôndrias/metabolismo , Monócitos/metabolismo , Fagocitose , Trifosfato de Adenosina/metabolismo , Células Cultivadas , Metabolismo Energético/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/genética , Metacrilatos/farmacologia , Mitocôndrias/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Fagocitose/efeitos dos fármacos , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Tiazóis/farmacologia
20.
Oxid Med Cell Longev ; 2021: 6955628, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34824671

RESUMO

Oxidative stress and apoptosis contribute to the progression of cerebral ischemia/reperfusion (I/R) injury. Ubiquitin-specific protease 29 (USP29) is abundantly expressed in the brain and plays critical roles in regulating oxidative stress and cell apoptosis. The purpose of the present study is to investigate the role and underlying mechanisms of USP29 in cerebral I/R injury. Neuron-specific USP29 knockout mice were generated and subjected to cerebral I/R surgery. For USP29 overexpression, mice were stereotactically injected with the adenoassociated virus serotype 9 vectors carrying USP29 for 4 weeks before cerebral I/R. And primary cortical neurons were isolated and exposed to oxygen glucose deprivation/reperfusion (OGD/R) stimulation to imitate cerebral I/R injury in vitro. USP29 expression was elevated in the brain and primary cortical neurons upon I/R injury. Neuron-specific USP29 knockout significantly diminished, whereas USP29 overexpression aggravated cerebral I/R-induced oxidative stress, apoptosis, and neurological dysfunction in mice. In addition, OGD/R-induced oxidative stress and neuronal apoptosis were also attenuated by USP29 silence but exacerbated by USP29 overexpression in vitro. Mechanistically, neuronal USP29 enhanced p53/miR-34a-mediated silent information regulator 1 downregulation and then promoted the acetylation and suppression of brain and muscle ARNT-like protein, thereby aggravating oxidative stress and apoptosis upon cerebral I/R injury. Our findings for the first time identify that USP29 upregulation during cerebral I/R may contribute to oxidative stress, neuronal apoptosis, and the progression of cerebral I/R injury and that inhibition of USP29 may help to develop novel therapeutic strategies to treat cerebral I/R injury.


Assuntos
Apoptose , Neurônios/patologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/patologia , Proteases Específicas de Ubiquitina/fisiologia , Animais , Glucose/deficiência , Hipóxia , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...