Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.926
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nat Immunol ; 14(5): 461-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23525087

RESUMO

Inflammation is essential for host defense but can cause tissue damage and organ failure if unchecked. How the inflammation is resolved remains elusive. Here we report that the transcription factor Miz1 was required for terminating lipopolysaccharide (LPS)-induced inflammation. Genetic disruption of the Miz1 POZ domain, which is essential for the transactivation or repression activity of Miz1, resulted in hyperinflammation, lung injury and greater mortality in LPS-treated mice but a lower bacterial load and mortality in mice with Pseudomonas aeruginosa pneumonia. Loss of the Miz1 POZ domain prolonged the expression of proinflammatory cytokines. After stimulation, Miz1 was phosphorylated at Ser178, which was required for recruitment of the histone deacetylase HDAC1 to repress transcription of the gene encoding C/EBP-δ, an amplifier of inflammation. Our data provide a long-sought mechanism underlying the resolution of LPS-induced inflammation.


Assuntos
Lesão Pulmonar Aguda/imunologia , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Lesão Pulmonar Aguda/genética , Animais , Citocinas/metabolismo , Repressão Enzimática/genética , Histona Desacetilase 1/metabolismo , Tolerância Imunológica , Inflamação/genética , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Fosforilação , Proteínas Inibidoras de STAT Ativados/genética , Infecções por Pseudomonas/genética , Proteínas Repressoras/genética , Ativação Transcricional/genética , Ubiquitina-Proteína Ligases
2.
Physiol Rev ; 96(1): 307-64, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26681794

RESUMO

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated from aerobic metabolism, as a result of accidental electron leakage as well as regulated enzymatic processes. Because ROS/RNS can induce oxidative injury and act in redox signaling, enzymes metabolizing them will inherently promote either health or disease, depending on the physiological context. It is thus misleading to consider conventionally called antioxidant enzymes to be largely, if not exclusively, health protective. Because such a notion is nonetheless common, we herein attempt to rationalize why this simplistic view should be avoided. First we give an updated summary of physiological phenotypes triggered in mouse models of overexpression or knockout of major antioxidant enzymes. Subsequently, we focus on a series of striking cases that demonstrate "paradoxical" outcomes, i.e., increased fitness upon deletion of antioxidant enzymes or disease triggered by their overexpression. We elaborate mechanisms by which these phenotypes are mediated via chemical, biological, and metabolic interactions of the antioxidant enzymes with their substrates, downstream events, and cellular context. Furthermore, we propose that novel treatments of antioxidant enzyme-related human diseases may be enabled by deliberate targeting of dual roles of the pertaining enzymes. We also discuss the potential of "antioxidant" nutrients and phytochemicals, via regulating the expression or function of antioxidant enzymes, in preventing, treating, or aggravating chronic diseases. We conclude that "paradoxical" roles of antioxidant enzymes in physiology, health, and disease derive from sophisticated molecular mechanisms of redox biology and metabolic homeostasis. Simply viewing antioxidant enzymes as always being beneficial is not only conceptually misleading but also clinically hazardous if such notions underpin medical treatment protocols based on modulation of redox pathways.


Assuntos
Antioxidantes/metabolismo , Enzimas/metabolismo , Nível de Saúde , Estresse Oxidativo , Animais , Modelos Animais de Doenças , Indução Enzimática , Repressão Enzimática , Enzimas/biossíntese , Enzimas/genética , Técnicas de Silenciamento de Genes , Predisposição Genética para Doença , Humanos , Camundongos Transgênicos , Estado Nutricional , Oxirredução , Fenótipo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Risco
3.
J Cell Physiol ; 234(12): 22635-22647, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31102300

RESUMO

Calcium-activated nucleotidase 1 (CANT1, belongs to the apyrase family, is widely expressed in various organs. However, the biological function of CANT1 remains poorly explored. In this study, we aimed to investigate the expression profile and functions of CANT1 in clear cell renal cell carcinoma (ccRCC). Our data show that the protein level of CANT1 was significantly higher in tumor tissues than in adjacent normal tissues. CANT1 silencing suppressed cell proliferation, migration, and invasion obviously in 769-P and 786-O cells, arrested cell cycle in S phase and promoted apoptosis in 769-P cells. In conclusion, the present study shows the different expression mode of CANT1 in human ccRCC tumor tissue and adjacent normal tissue, denotes the function of CANT1 in ccRCC cells and provides potential molecular mechanisms and pathways of CANT1 antitumor function in ccRCC.


Assuntos
Carcinoma de Células Renais/enzimologia , Proliferação de Células , Neoplasias Renais/enzimologia , Nucleotidases/metabolismo , Interferência de RNA , Apoptose , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular , Repressão Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Invasividade Neoplásica , Nucleotidases/genética , Pontos de Checagem da Fase S do Ciclo Celular , Transdução de Sinais
4.
Mol Microbiol ; 108(3): 226-239, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29424946

RESUMO

Nucleoid-associated protein H-NS represses transcription by forming extended DNA-H-NS complexes. Repression by H-NS operates mostly at the level of transcription initiation. Less is known about how DNA-H-NS complexes interfere with transcription elongation. In vitro H-NS has been shown to enhance RNA polymerase pausing and to promote Rho-dependent termination, while in vivo inhibition of Rho resulted in a decrease of the genome occupancy by H-NS. Here we show that transcription directed across H-NS binding regions relieves H-NS (and H-NS/StpA) mediated repression of promoters in these regions. Further, we observed a correlation of transcription across the H-NS-bound region and de-repression. The data suggest that the transcribing RNA polymerase is able to remodel the H-NS complex and/or dislodge H-NS from the DNA and thus relieve repression. Such an interference of transcription and H-NS mediated repression may imply that poorly transcribed AT-rich loci are prone to be repressed by H-NS, while efficiently transcribed loci escape repression.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Bactérias/biossíntese , Sítios de Ligação/genética , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/biossíntese , RNA Polimerases Dirigidas por DNA/metabolismo , Repressão Enzimática , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/genética , Regiões Promotoras Genéticas/genética , Transcrição Gênica/genética , Transcrição Gênica/fisiologia
5.
J Pathol ; 244(4): 394-407, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29230817

RESUMO

Nasopharyngeal carcinoma (NPC) is a highly invasive epithelial malignancy that is prevalent in southern China and Southeast Asia. It is consistently associated with latent Epstein-Barr virus (EBV) infection. In NPC, miR-BARTs, the EBV-encoded miRNAs derived from BamH1-A rightward transcripts, are abundantly expressed and contribute to cancer development by targeting various cellular and viral genes. In this study, we establish a comprehensive transcriptional profile of EBV-encoded miRNAs in a panel of NPC patient-derived xenografts and an EBV-positive NPC cell line by small RNA sequencing. Among the 40 miR-BARTs, predominant expression of 22 miRNAs was consistently detected in these tumors. Among the abundantly expressed EBV-miRNAs, BART5-5p, BART7-3p, BART9-3p, and BART14-3p could negatively regulate the expression of a key DNA double-strand break (DSB) repair gene, ataxia telangiectasia mutated (ATM), by binding to multiple sites on its 3'-UTR. Notably, the expression of these four miR-BARTs represented more than 10% of all EBV-encoded miRNAs in tumor cells, while downregulation of ATM expression was commonly detected in all of our tested sequenced samples. In addition, downregulation of ATM was also observed in primary NPC tissues in both qRT-PCR (16 NP and 45 NPC cases) and immunohistochemical staining (35 NP and 46 NPC cases) analysis. Modulation of ATM expression by BART5-5p, BART7-3p, BART9-3p, and BART14-3p was demonstrated in the transient transfection assays. These findings suggest that EBV uses miRNA machinery as a key mechanism to control the ATM signaling pathway in NPC cells. By suppressing these endogenous miR-BARTs in EBV-positive NPC cells, we further demonstrated the novel function of miR-BARTs in inhibiting Zta-induced lytic reactivation. These findings imply that the four viral miRNAs work co-operatively to modulate ATM activity in response to DNA damage and to maintain viral latency, contributing to the tumorigenesis of NPC. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/genética , MicroRNAs/genética , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , RNA Viral/genética , Regiões 3' não Traduzidas , Animais , Proteínas Mutadas de Ataxia Telangiectasia/biossíntese , Sítios de Ligação , Linhagem Celular Tumoral , Dano ao DNA , Repressão Enzimática , Infecções por Vírus Epstein-Barr/diagnóstico , Feminino , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Interações Hospedeiro-Patógeno , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/enzimologia , Carcinoma Nasofaríngeo/patologia , Carcinoma Nasofaríngeo/virologia , Neoplasias Nasofaríngeas/enzimologia , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/virologia , Transcriptoma , Latência Viral
6.
Pharmacol Rev ; 68(1): 168-241, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26721703

RESUMO

During the last 10-15 years, cytochrome P450 (CYP) 2C8 has emerged as an important drug-metabolizing enzyme. CYP2C8 is highly expressed in human liver and is known to metabolize more than 100 drugs. CYP2C8 substrate drugs include amodiaquine, cerivastatin, dasabuvir, enzalutamide, imatinib, loperamide, montelukast, paclitaxel, pioglitazone, repaglinide, and rosiglitazone, and the number is increasing. Similarly, many drugs have been identified as CYP2C8 inhibitors or inducers. In vivo, already a small dose of gemfibrozil, i.e., 10% of its therapeutic dose, is a strong, irreversible inhibitor of CYP2C8. Interestingly, recent findings indicate that the acyl-ß-glucuronides of gemfibrozil and clopidogrel cause metabolism-dependent inactivation of CYP2C8, leading to a strong potential for drug interactions. Also several other glucuronide metabolites interact with CYP2C8 as substrates or inhibitors, suggesting that an interplay between CYP2C8 and glucuronides is common. Lack of fully selective and safe probe substrates, inhibitors, and inducers challenges execution and interpretation of drug-drug interaction studies in humans. Apart from drug-drug interactions, some CYP2C8 genetic variants are associated with altered CYP2C8 activity and exhibit significant interethnic frequency differences. Herein, we review the current knowledge on substrates, inhibitors, inducers, and pharmacogenetics of CYP2C8, as well as its role in clinically relevant drug interactions. In addition, implications for selection of CYP2C8 marker and perpetrator drugs to investigate CYP2C8-mediated drug metabolism and interactions in preclinical and clinical studies are discussed.


Assuntos
Indutores do Citocromo P-450 CYP2C8/farmacologia , Citocromo P-450 CYP2C8/metabolismo , Interações Medicamentosas/fisiologia , Repressão Enzimática/efeitos dos fármacos , Antimaláricos/farmacologia , Antineoplásicos/farmacologia , Interações Medicamentosas/genética , Humanos , Hipoglicemiantes/farmacologia , Hipolipemiantes/farmacologia , Farmacogenética
7.
J Bacteriol ; 200(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29339417

RESUMO

Septicemia-causing Vibrio vulnificus produces at least three exoproteases, VvpE, VvpS, and VvpM, all of which participate in interactions with human cells. Expression of VvpE and VvpS is induced in the stationary phase by multiple transcription factors, including sigma factor S, SmcR, and the cAMP-cAMP receptor protein (cAMP-CRP) complex. Distinct roles of VvpM, such as induction of apoptosis, lead us to hypothesize VvpM expression is different from that of the other exoproteases. Its transcription, which was found to be independent of sigma S, is induced at the early exponential phase and then becomes negligible upon entry into the stationary phase. SmcR and CRP were studied regarding the control of vvpM expression. Transcription of vvpM was repressed by SmcR and cAMP-CRP complex individually, which specifically bound to the regions -2 to +20 and +6 to +27, respectively, relative to the vvpM transcription initiation site. Derepression of vvpM gene expression was 10- to 40-fold greater in an smcR crp double mutant than in single-gene mutants. Therefore, these results show that the expression of V. vulnificus exoproteases is differentially regulated, and in this way, distinct proteases can engage in specific interactions with a host.IMPORTANCE An opportunistic human pathogen, Vibrio vulnificus produces multiple extracellular proteases that are involved in diverse interactions with a host. The total exoproteolytic activity is detected mainly in the supernatants of the high-cell-density cultures. However, some proteolytic activity derived from a metalloprotease, VvpM, was present in the supernatants of the low-cell-density cultures sampled at the early growth period. In this study, we present the regulatory mechanism for VvpM expression via repression by at least two transcription factors. This type of transcriptional regulation is the exact opposite of those for expression of the other V. vulnificus exoproteases. Differential regulation of each exoprotease's production then facilitates the pathogen's participation in the distinct interactions with a host.


Assuntos
Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Metaloendopeptidases/biossíntese , Metaloendopeptidases/genética , Percepção de Quorum , Vibrio vulnificus/genética , Apoptose , Proteína Receptora de AMP Cíclico/metabolismo , Repressão Enzimática/genética , Humanos , Proteólise , Fatores de Transcrição/genética , Vibrio vulnificus/enzimologia
8.
Mol Microbiol ; 106(2): 266-284, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28787542

RESUMO

Transmission of the malaria parasite occurs in an unpredictable moment, when a mosquito takes a blood meal. Plasmodium has therefore evolved strategies to prepare for transmission, including translationally repressing and protecting mRNAs needed to establish the infection. However, mechanisms underlying these critical controls are not well understood, including whether Plasmodium changes its translationally repressive complexes and mRNA targets in different stages. Efforts to understand this have been stymied by severe technical limitations due to substantial mosquito contamination of samples. Here using P. yoelii, for the first time we provide a proteomic comparison of a protein complex across asexual blood, sexual and sporozoite stages, along with a transcriptomic comparison of the mRNAs that are affected in these stages. We find that the Apicomplexan-specific ALBA4 RNA-binding protein acts to regulate development of the parasite's transmission stages, and that ALBA4 associates with both stage-specific and stage-independent partners to produce opposing mRNA fates. These efforts expand our understanding and ability to interrogate both sexual and sporozoite transmission stages and the molecular preparations they evolved to perpetuate their infectious cycle.


Assuntos
Plasmodium yoelii/fisiologia , RNA Mensageiro/biossíntese , Animais , Anopheles/parasitologia , Repressão Enzimática , Malária/parasitologia , Parasitos , Doenças Parasitárias/genética , Plasmodium yoelii/genética , Plasmodium yoelii/crescimento & desenvolvimento , Proteômica , Proteínas de Protozoários/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Esporozoítos/metabolismo , Transcriptoma
9.
RNA ; 22(4): 623-35, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26857222

RESUMO

The RNA exosome is essential for 3' processing of functional RNA species and degradation of aberrant RNAs in eukaryotic cells. Recent reports have defined the substrates of the exosome catalytic domains and solved the multimeric structure of the exosome complex. However, regulation of exosome activity remains poorly characterized, especially in response to physiological stress. Following the observation that cooling of mammalian cells results in a reduction in 40S:60S ribosomal subunit ratio, we uncover regulation of the nuclear exosome as a result of reduced temperature. Using human cells and an in vivo model system allowing whole-body cooling, we observe reduced EXOSC10 (hRrp6, Pm/Scl-100) expression in the cold. In parallel, both models of cooling increase global SUMOylation, leading to the identification of specific conjugation of SUMO1 to EXOSC10, a process that is increased by cooling. Furthermore, we define the major SUMOylation sites in EXOSC10 by mutagenesis and show that overexpression of SUMO1 alone is sufficient to suppress EXOSC10 abundance. Reducing EXOSC10 expression by RNAi in human cells correlates with the 3' preribosomal RNA processing defects seen in the cold as well as reducing the 40S:60S ratio, a previously uncharacterized consequence of EXOSC10 suppression. Together, this work illustrates that EXOSC10 can be modified by SUMOylation and identifies a physiological stress where this regulation is prevalent both in vitro and in vivo.


Assuntos
Exorribonucleases/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Sequência de Aminoácidos , Animais , Resposta ao Choque Frio , Repressão Enzimática , Exorribonucleases/genética , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Biossíntese de Proteínas , RNA Ribossômico/metabolismo , Proteína SUMO-1/metabolismo , Sumoilação
10.
Toxicol Appl Pharmacol ; 352: 77-86, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29802914

RESUMO

It is known that inhibiting 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) expression in the placenta can cause fetal over-exposure to maternal glucocorticoids and induce intrauterine growth restriction (IUGR); these effects ultimately increase the risk of adult chronic diseases. This study aimed to investigate the molecular mechanism of the prenatal ethanol exposure (PEE)-induced inhibition of placental 11ß-HSD2 expression. Pregnant Wistar rats were intragastrically administered ethanol (4 g/kg/d) from gestational days 9 to 20. The levels of maternal and fetal serum corticosterone and placental 11ß-HSD2-related gene expression were analyzed. Furthermore, we investigated the mechanism of reduced placental 11ß-HSD2 expression induced by ethanol treatment (15-60 mM) in HTR-8/SVneo cells. In vivo, PEE decreased fetal body weights and increased maternal and fetal serum corticosterone and early growth response factor 1 (EGR1) expression levels. Moreover, histone modification changes (decreased acetylation and increased di-methylation of H3K9) to the HSD11B2 promoter and lower 11ß-HSD2 expression levels were observed. In vitro, ethanol decreased cAMP/PKA signaling and 11ß-HSD2 expression and increased EGR1 expression in a concentration-dependent manner. A cAMP agonist and EGR1 siRNA reversed the ethanol-induced inhibition of 11ß-HSD2 expression. Together, PEE reduced placental 11ß-HSD2 expression, and the underlying mechanism is associated with ethanol-induced histone modification changes to the HSD11B2 promoter through the cAMP/PKA/EGR1 pathway.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Consumo de Bebidas Alcoólicas/efeitos adversos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Etanol/toxicidade , Placenta/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Acetilação , Animais , Linhagem Celular , Proteína 1 de Resposta de Crescimento Precoce/genética , Repressão Enzimática , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Histonas/metabolismo , Humanos , Metilação , Placenta/enzimologia , Gravidez , Regiões Promotoras Genéticas , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
11.
PLoS Biol ; 13(10): e1002283, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26506406

RESUMO

Work on rats and humans reveals a role for reduction in the levels of the stress-regulated protein kinase SGK1 in the development of post-traumatic stress disorder. Read the Research Article.


Assuntos
Transtorno Depressivo Maior/etiologia , Repressão Enzimática , Proteínas Imediatamente Precoces/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Animais , Feminino , Humanos , Masculino
12.
PLoS Biol ; 13(10): e1002282, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26506154

RESUMO

Exposure to extreme stress can trigger the development of major depressive disorder (MDD) as well as post-traumatic stress disorder (PTSD). The molecular mechanisms underlying the structural and functional alterations within corticolimbic brain regions, including the prefrontal cortex (PFC) and amygdala of individuals subjected to traumatic stress, remain unknown. In this study, we show that serum and glucocorticoid regulated kinase 1 (SGK1) expression is down-regulated in the postmortem PFC of PTSD subjects. Furthermore, we demonstrate that inhibition of SGK1 in the rat medial PFC results in helplessness- and anhedonic-like behaviors in rodent models. These behavioral changes are accompanied by abnormal dendritic spine morphology and synaptic dysfunction. Together, the results are consistent with the possibility that altered SGK1 signaling contributes to the behavioral and morphological phenotypes associated with traumatic stress pathophysiology.


Assuntos
Transtorno Depressivo Maior/etiologia , Repressão Enzimática , Proteínas Imediatamente Precoces/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Adulto , Animais , Comportamento Animal , Estudos de Coortes , Espinhas Dendríticas/enzimologia , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Feminino , Técnicas de Transferência de Genes , Hipocampo/enzimologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/genética , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Neurônios/enzimologia , Neurônios/patologia , Córtex Pré-Frontal/enzimologia , Córtex Pré-Frontal/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Ratos Sprague-Dawley , Transdução de Sinais , Transtornos de Estresse Pós-Traumáticos/patologia , Transtornos de Estresse Pós-Traumáticos/psicologia , Transmissão Sináptica , Bancos de Tecidos
13.
J Biol Chem ; 291(12): 6245-61, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26828066

RESUMO

The alcohol oxidase 1 (AOX1) promoter (P AOX1) of Pichia pastoris is the most powerful and commonly used promoter for driving protein expression. However, mechanisms regulating its transcriptional activity are unclear. Here, we identified a Zn(II)2Cys6-type methanol-induced transcription factor 1 (Mit1) and elucidated its roles in regulating PAOX1 activity in response to glycerol and methanol. Mit1 regulated the expression of many genes involved in methanol utilization pathway, including AOX1, but did not participate in peroxisome proliferation and transportation of peroxisomal proteins during methanol metabolism. Structural analysis of Mit1 by performing domain deletions confirmed its specific and critical role in the strict repression of P AOX1 in glycerol medium. Importantly, Mit1, Mxr1, and Prm1, which positively regulated P AOX1 in response to methanol, were bound to P AOX1 at different sites and did not interact with each other. However, these factors cooperatively activated P AOX1 through a cascade. Mxr1 mainly functioned during carbon derepression, whereas Mit1 and Prm1 functioned during methanol induction, with Prm1 transmitting methanol signal to Mit1 by binding to the MIT1 promoter (P MIT1), thus increasingly expressing Mit1 and subsequently activating P AOX1.


Assuntos
Oxirredutases do Álcool/genética , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Metanol/metabolismo , Pichia/enzimologia , Fatores de Transcrição/fisiologia , Oxirredutases do Álcool/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Repressão Enzimática , Proteínas Fúngicas/metabolismo , Dados de Sequência Molecular , Peroxissomos/metabolismo , Pichia/genética , Regiões Promotoras Genéticas , Transporte Proteico , Homologia de Sequência de Aminoácidos , Transdução de Sinais
14.
J Biol Chem ; 291(3): 1529-37, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26589799

RESUMO

MicroRNA regulation of protein expression plays an important role in mediating many cellular processes, from cell proliferation to cell death. The human microRNA miR-424 is up-regulated in response to anti-proliferative cytokines, such as transforming growth factor ß (TGFß), and directly represses cell cycle progression. Our laboratory recently established that microRNA can be used as a proxy to identify biological roles of glycosylation enzymes (glycogenes). Herein we identify MGAT4A, OGT, and GALNT13 as targets of miR-424. We demonstrate that MGAT4A, an N-acetylglucosaminyltransferase that installs the ß-1,4 branch of N-glycans, is directly regulated by miR-424 in multiple mammary epithelial cell lines and observe the loss of MGAT4A in response to TGFß, an inducer of miR-424. Knockdown of MGAT4A induces cell cycle arrest through decreasing CCND1 levels. MGAT4A does not affect levels of ß-1,6 branched N-glycans, arguing that this effect is specific to ß-1,4 branching and not due to gross changes in overall N-linked glycosylation. This work provides insight into the regulation of cell cycle progression by specific N-glycan branching patterns.


Assuntos
Glândulas Mamárias Humanas/metabolismo , MicroRNAs/metabolismo , N-Acetilgalactosaminiltransferases/antagonistas & inibidores , N-Acetilglucosaminiltransferases/antagonistas & inibidores , Interferência de RNA , Ciclo Celular , Linhagem Celular , Proliferação de Células , Ciclina D1/antagonistas & inibidores , Ciclina D1/genética , Ciclina D1/metabolismo , Repressão Enzimática , Genes Reporter , Glicosilação , Células HEK293 , Humanos , Luciferases/genética , Luciferases/metabolismo , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/enzimologia , MicroRNAs/agonistas , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , RNA Interferente Pequeno , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator de Crescimento Transformador beta/metabolismo
15.
PLoS Pathog ; 11(9): e1005171, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26402907

RESUMO

Kaposi's sarcoma (KS) is a highly disseminated angiogenic tumor of endothelial cells linked to infection by Kaposi's sarcoma-associated herpesvirus (KSHV). KSHV encodes more than two dozens of miRNAs but their roles in KSHV-induced tumor dissemination and metastasis remain unknown. Here, we found that ectopic expression of miR-K12-3 (miR-K3) promoted endothelial cell migration and invasion. Bioinformatics and luciferase reporter analyses showed that miR-K3 directly targeted G protein-coupled receptor (GPCR) kinase 2 (GRK2, official gene symbol ADRBK1). Importantly, overexpression of GRK2 reversed miR-K3 induction of cell migration and invasion. Furthermore, the chemokine receptor CXCR2, which was negatively regulated by GRK2, was upregulated in miR-K3-transduced endothelial cells. Knock down of CXCR2 abolished miR-K3-induced cell migration and invasion. Moreover, miR-K3 downregulation of GRK2 relieved its direct inhibitory effect on AKT. Both CXCR2 induction and the release of AKT from GRK2 were required for miR-K3 maximum activation of AKT and induction of cell migration and invasion. Finally, deletion of miR-K3 from the KSHV genome abrogated its effect on the GRK2/CXCR2/AKT pathway and KSHV-induced migration and invasion. Our data provide the first-line evidence that, by repressing GRK2, miR-K3 facilitates cell migration and invasion via activation of CXCR2/AKT signaling, which likely contribute to the dissemination of KSHV-induced tumors.


Assuntos
Endotélio Vascular/virologia , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno , MicroRNAs/metabolismo , RNA Viral/metabolismo , Internalização do Vírus , Movimento Celular , Células Cultivadas , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Repressão Enzimática , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Deleção de Genes , Herpesvirus Humano 8/imunologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Células Endoteliais da Veia Umbilical Humana/virologia , Humanos , Mutação , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-akt/agonistas , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA/metabolismo , Interferência de RNA , Receptores de Interleucina-8B/agonistas , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sarcoma de Kaposi/imunologia , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Transdução de Sinais
16.
Cancer Invest ; 35(3): 152-162, 2017 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-28267394

RESUMO

microRNAs are the post-transcriptional regulators implicated in the initiation and progression of various cancer types, including oral squamous cell carcinoma (OSCC). Here, we investigated the role of miR-377 in OSCC tumorigenesis. miR-377 expression was reduced in OSCC samples and cell line (UPCI-SCC-116), and was associated with patient survival. In vitro restoration of miR-377 repressed cell growth, induced apoptosis, and reduced cell migration. We identified HDAC9 as a target of miR-377 and found miR-377 to regulate HDAC9 and its pro-apoptotic target, NR4A1/Nur77. Our findings show that miR-377 targets HDAC9 pathway in OSCC, suggesting that miR-377-HDAC9 axis may provide a novel therapeutic target for OSCC therapy.


Assuntos
Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Bucais/genética , Interferência de RNA , Regiões 3' não Traduzidas , Apoptose , Sequência de Bases , Sítios de Ligação , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Repressão Enzimática , Histona Desacetilases , Humanos , Estimativa de Kaplan-Meier , MicroRNAs/metabolismo , Neoplasias Bucais/enzimologia , Neoplasias Bucais/mortalidade , Neoplasias Bucais/patologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Proteínas Repressoras
17.
Exp Cell Res ; 342(1): 62-71, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26902400

RESUMO

PHOX2B and its paralogue gene PHOX2A are two homeodomain proteins in the network regulating the development of autonomic ganglia that have been associated with the pathogenesis of neuroblastoma (NB), because of their over-expression in different NB cell lines and tumour samples. We used the SK-N-BE(2)C cell line to show that all-trans retinoic acid (ATRA), a drug that is widely used to inhibit growth and induce differentiation in NBs, regulates both PHOX2A and PHOX2B expression, albeit by means of different mechanisms: it up-regulates PHOX2A and down-regulates PHOX2B. Both mechanisms act at transcriptional level, but prolonged ATRA treatment selectively degrades the PHOX2A protein, whereas the corresponding mRNA remains up-regulated. Further, we show that PHOX2A is capable of modulating PHOX2B expression, but this mechanism is not involved in the PHOX2B down-regulation induced by retinoic acid. Our findings demonstrate that PHOX2A expression is finely controlled during retinoic acid differentiation and this, together with PHOX2B down-regulation, reinforces the idea that they may be useful biomarkers for NB staging, prognosis and treatment decision making.


Assuntos
Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Tretinoína/farmacologia , Sequência de Bases , Diferenciação Celular , Linhagem Celular Tumoral , Indução Enzimática/efeitos dos fármacos , Repressão Enzimática/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Humanos , Neuroblastoma , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Elementos de Resposta , Fatores de Transcrição/genética
18.
Artigo em Inglês | MEDLINE | ID: mdl-28089857

RESUMO

Survival of prolonged anoxia requires a balance between cellular ATP demand and anaerobic ATP supply from glycolysis, especially in critical tissues such as the brain. To add insight into the ATP demand of the brain of the anoxia-tolerant red-eared slider turtle (Trachemys scripta) during prolonged periods of anoxic submergence, we quantified and compared the number of Na+-K+-ATPase units and their molecular activity in brain tissue from turtles acclimated to either 21°C or 5°C and exposed to either normoxia or anoxia (6h 21°C; 14days at 5°C). Na+-K+-ATPase activity and density per g tissue were similar at 21°C and 5°C in normoxic turtles. Likewise, anoxia exposure at 21°C did not induce any change in Na+-K+-ATPase activity or density. In contrast, prolonged anoxia at 5°C significantly reduced Na+-K+-ATPase activity by 55%, which was largely driven by a 50% reduction of the number of Na+-K+-ATPase units without a change in the activity of existing Na+-K+-ATPase pumps or α-subunit composition. These findings are consistent with the "channel arrest" hypothesis to reduce turtle brain Na+-K+-ATPase activity during prolonged, but not short-term anoxia, a change that likely helps them overwinter under low temperature, anoxic conditions.


Assuntos
Encéfalo/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Proteínas de Répteis/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Tartarugas/fisiologia , Aclimatação , Animais , Ligação Competitiva , Hipóxia Celular , Temperatura Baixa/efeitos adversos , Inibidores Enzimáticos/farmacologia , Repressão Enzimática , Feminino , Hibernação , Cinética , Masculino , Proteínas do Tecido Nervoso/antagonistas & inibidores , Ouabaína/farmacologia , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/metabolismo , Proteínas de Répteis/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Trítio
19.
Environ Toxicol ; 32(4): 1390-1398, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27566995

RESUMO

The Warburg effect plays a critical role in tumorigenesis, suggesting that specific agents targeting Warburg effect key proteins may be a promising strategy for cancer therapy. Previous studies have shown that diallyl trisulfide (DATS) inhibits proliferation of breast cancer cells by inducing apoptosis in vitro and in vivo. However, whether the Warburg effect is involved with the apoptosis-promoting action of DATS is unclear. Here, we show that the action of DATS is associated with downregulation of lactate dehydrogenase A (LDHA), an essential protein of the Warburg effect whose upregulation is closely related to tumorigenesis. Interestingly, inhibition of the Warburg effect by DATS in breast cancer cells did not greatly affect normal cells. Furthermore, DATS inhibited growth of breast cancer cells, particularly in MDA-MB-231, a triple-negative breast cancer (TNBC) cell, and reduced proliferation and migration; invasion was reversed by over-expression of LDHA. These data suggest that DATS inhibits breast cancer growth and aggressiveness through a novel pathway targeting the key enzyme of the Warburg effect. Our study shows that LDHA downregulation is involved in the apoptotic effect of DATS on TNBC. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1390-1398, 2017.


Assuntos
Compostos Alílicos/farmacologia , Antineoplásicos/farmacologia , L-Lactato Desidrogenase/genética , Sulfetos/farmacologia , Neoplasias de Mama Triplo Negativas/enzimologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Metabolismo dos Carboidratos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Regulação para Baixo , Ensaios de Seleção de Medicamentos Antitumorais , Repressão Enzimática/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Desidrogenase/metabolismo , Lactato Desidrogenase 5 , Metástase Neoplásica , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia
20.
J Sci Food Agric ; 97(2): 551-555, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27097525

RESUMO

BACKGROUND: The present study aimed to investigate the in vivo antihypertensive effect on spontaneously hypertensive rats (SHRs) induced by egg protein-derived peptide QIGLF, which has been previously characterized in vitro as a potent angiotensin-converting enzyme inhibitor. RESULTS: In vivo antihypertensive effect of QIGLF orally administered was evaluated by the tail-cuff method. The systolic blood pressure and the diastolic blood pressure of rats were measured 0, 5, 10, 15 and 20 h after administration every day. Subsequently, the effect of QIGLF on angiotensin-converting enzyme mRNA expression in the kidney of SHRs was evaluated by a polymerase chain reaction. Systolic blood pressure was found to be reduced markedly in the SHRs after a single oral administration. CONCLUSION: The results show that the effect of QIGLF (50 mg kg-1 body weight) was similar to that of captopril (10 mg kg-1 body weight) with respect to lowering systolic blood pressure in SHRs. Therefore, egg white protein-derived peptide QIGLF may be useful in the prevention or treatment of hypertension. © 2016 Society of Chemical Industry.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Anti-Hipertensivos/uso terapêutico , Suplementos Nutricionais , Proteínas do Ovo/uso terapêutico , Hipertensão/dietoterapia , Rim/fisiopatologia , Oligopeptídeos/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Inibidores da Enzima Conversora de Angiotensina/efeitos adversos , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/efeitos adversos , Pressão Sanguínea/efeitos dos fármacos , Captopril/efeitos adversos , Captopril/uso terapêutico , Suplementos Nutricionais/efeitos adversos , Proteínas do Ovo/administração & dosagem , Proteínas do Ovo/efeitos adversos , Repressão Enzimática , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Rim/metabolismo , Masculino , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/efeitos adversos , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , RNA Mensageiro/metabolismo , Ratos Endogâmicos SHR , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA