Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167112, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38432455

RESUMO

The dysregulation of N6-methyladenosine (m6A) on mRNAs is involved in the pathogenesis of rheumatoid arthritis (RA). Methyltransferase-like 3 (METTL3), serving as a central m6A methyltransferase, is highly expressed in macrophages, synovial tissues and RA fibroblast-like synoviocytes (RA-FLS) of RA patients. However, METTL3-mediated m6A modification on target mRNAs and the molecular mechanisms involved in RA-FLS remain poorly defined. Our research demonstrated that METTL3 knockdown decreased the proliferation, migratory and invasive abilities of RA-FLS. Notably, we identified the adhesion molecule with Ig like domain 2 (AMIGO2) as a probable downstream target of both METTL3 and YTH Domain Containing 2 (YTHDC2) in RA-FLS. We revealed that AMIGO2 augmented the activation of RA-FLS and can potentially reverse the phenotypic effects induced by the knockdown of either METTL3 or YTHDC2. Mechanistically, METTL3 knockdown decreased m6A modification in the 5'-untranslated region (5'UTR) of AMIGO2 mRNA, which diminished its interaction with YTHDC2 in RA-FLS. Our findings unveiled that silencing of METTL3 inhibited the proliferation and aggressive behaviors of RA-FLS by downregulating AMIGO2 expression in an m6A-YTHDC2 dependent mechanism, thereby underscoring the pivotal role of the METTL3-m6A-YTHDC2-AMIGO2 axis in modulating RA-FLS phenotypes.


Assuntos
Artrite Reumatoide , Sinoviócitos , Humanos , Proliferação de Células , Artrite Reumatoide/patologia , Membrana Sinovial/metabolismo , Sinoviócitos/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , RNA Helicases/metabolismo , RNA Helicases/farmacologia
2.
Plant Physiol Biochem ; 205: 108202, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37995575

RESUMO

RNA helicases (RHs) are required for most aspects of RNA metabolism and play an important role in plant stress tolerance. Heat stress (HS) causes the deleterious effects on plant cells, such as membrane disruption and protein misfolding, which results in the inhibition of plant growth and development. In this study, CaRH57 was identified from pepper (Capsicum annuum) and encodes a DEAD-box RH. CaRH57 was induced by HS, and overexpression of CaRH57 in Atrh57-1 rescued the glucose-sensitive phenotype of Atrh57-1, suggesting the functional replacement of CaRH57 to AtRH57. The nucleolus-localized CaRH57 possessed a RH activity in vitro. CaRH57 knockdown impaired pepper heat tolerance, showing severe necrosis and enhanced ROS accumulation in the region of the shoot tip. Additionally, accumulation of aberrant-spliced CaHSFA1d and CaHSFA9d was enhanced, and the corresponding mature mRNA levels were reduced in the TRV2 (Tobacco rattle virus)-CaRH57-infected plants compared with the control plants under HS. Overall, these results suggested that CaRH57 acted as a RH to confer pepper heat tolerance and was required for the proper pre-mRNA splicing of some HS-related genes.


Assuntos
Capsicum , Termotolerância , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Helicases/farmacologia , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Resposta ao Choque Térmico , Termotolerância/genética , Glucose/farmacologia , Capsicum/fisiologia , Regulação da Expressão Gênica de Plantas , Estresse Fisiológico/genética , Plantas Geneticamente Modificadas/metabolismo
3.
Biochem Pharmacol ; 217: 115834, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37778447

RESUMO

The emergence of chemoresistance in cervical cancer is extremely challenging in chemotherapy. Oxidative stress has emerged as the regulatory factor in drug resistance, but the detailed mechanism is still unknown. Stress granules, are membrane-less ribonucleoprotein-based condensates, could enhance chemoresistance by sequestering proapoptotic proteins inhibition of cell death upon exposure to drug-induced oxidative stress. Galectin-7, a member of galectin family, exerts varied roles in tumor repression or progression in different cancers. However, its role in cervical cancer has not been sufficiently studied. Here, we found that galectin-7 promotes cisplatin (CDDP) induced apoptosis and associates with stress granule-nucleating protein G3BP1 degradation. With the treatment of cisplatin, galectin-7 could enhance apoptosis by upregulating cleaved-PARP1 and the generation of reactive oxygen species (ROS), promoting mitochondrial fission, and reducing mitochondrial membrane potential (MMP). Furthermore, galectin-7 also reduces resistance by facilitating cisplatin-induced stress granules clearance through galectin-7/RACK1/G3BP1 axis. All these data suggested that galectin-7 promotes cisplatin sensitivity, and it would be potential target for potentiating efficacy in cervical cancer chemotherapy.


Assuntos
Antineoplásicos , Neoplasias do Colo do Útero , Feminino , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , DNA Helicases , Neoplasias do Colo do Útero/tratamento farmacológico , Proteínas de Ligação a Poli-ADP-Ribose , RNA Helicases/farmacologia , RNA Helicases/uso terapêutico , Proteínas com Motivo de Reconhecimento de RNA , Galectinas/farmacologia , Galectinas/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos
4.
Avian Pathol ; 51(6): 574-589, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35917182

RESUMO

Baicalin, a flavonoid compound extracted from the dry root of Scutellaria baicalensis Georgi, has been shown to have anti-inflammation, anti-viral, anti-bacterial, and immunomodulatory activity. However, the effect of baicalin against avian infectious bronchitis virus (IBV) remains unknown. The purpose of this study was to investigate the anti-IBV activity and underlying mechanism of baicalin in vitro. The results showed that baicalin has a direct virucidal effect but no prophylactic effect on IBV infection. The mRNA and protein of IBV N were decreased significantly when IBV-infected cells were treated with baicalin during the multiple stages of the virus replication cycle, including viral adsorption, invasion, internalization, and release. Stress granule (SG) formation resulted from the increase of G3BP1 and the phosphorylation of the PKR/eIF2α due to the treatment of IBV-infected cells with baicalin. The inhibitory activity of baicalin on IBV replication was increased when G3BP1 expression was inhibited, and the down-regulation of G3BP1 expression occurred when the expression of PKR and eIF2α was inhibited. These findings revealed that baicalin activates phosphorylation of the PKR/eIF2α pathway and induces SG formation by targeting G3BP1, initiating the antiviral response to suppress IBV replication in Vero cells. The results suggest that baicalin is a promising candidate drug to treat or prevent IBV infection.RESEARCH HIGHLIGHTS Baicalin inhibits IBV replication by reducing IBV N protein and mRNA.Baicalin disturbs multiple stages of the IBV life cycle.Baicalin activates PKR/eIF2α pathway and induces stress granule formation to exert anti-IBV response.


Assuntos
Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas , Chlorocebus aethiops , Animais , Antivirais/farmacologia , Células Vero , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , DNA Helicases/metabolismo , DNA Helicases/farmacologia , Proteínas de Ligação a Poli-ADP-Ribose , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Helicases/farmacologia , Doenças das Aves Domésticas/tratamento farmacológico , Flavonoides/farmacologia , RNA Mensageiro , Replicação Viral
5.
Int Immunopharmacol ; 108: 108764, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35421804

RESUMO

The prevalence of avian infectious bronchitis virus (IBV) is still one of causes inducing severe losses of production in the poultry industry worldwide. Vaccination does not completely prevent IBV infection and spread due to immune failure and viral mutations. ForsythiaeFructus and its compounds have been widely used in a lot of prescriptions of the traditional Chinese medicine for a long history, and it is well-known as safety and efficiency in heat-clearing and detoxifying. This study aims to investigate the anti-IBV activity and mechanism of phillygenin. The results showed that phillygenin inhibited IBV replication by disturbing multiple stages of the virus life cycle, including viral adsorption, invasion, internalization, and release in Vero cells. After being treated with 100, 125 and 150 µg/mL phillygenin, the expression of G3BP1 was significantly increased and the phosphorylation of PKR/eIF2α was activated, which increased stress granule, thereby triggering the antiviral response in Vero cells. The anti-virus activity of PHI was decreased when G3BP1 was interfered by si-RNA, and G3BP1 was down-regulated when PKR/eIF2α was interfered by si-RNA. In conclusion, our findings indicate that phillygenin activates PKR/eIF2α pathway and induces stress granule formation to exert anti-IBV, which holds promise to develop into a novel anti-IBV drug. Further study in vivo is needed to explore phillygenin as a potential and effective drug to prevent IB in poultry.


Assuntos
Infecções por Coronavirus , Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas , Animais , Chlorocebus aethiops , DNA Helicases/metabolismo , DNA Helicases/farmacologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 2 em Eucariotos/farmacologia , Vírus da Bronquite Infecciosa/fisiologia , Lignanas , Proteínas de Ligação a Poli-ADP-Ribose , RNA , RNA Helicases/metabolismo , RNA Helicases/farmacologia , Proteínas com Motivo de Reconhecimento de RNA , Grânulos de Estresse , Células Vero
6.
Gut ; 71(5): 991-1005, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34021034

RESUMO

OBJECTIVE: RNA helicase DDX5 is downregulated during HBV replication and poor prognosis HBV-related hepatocellular carcinoma (HCC). The objective of this study is to investigate the role of DDX5 in interferon (IFN) signalling. We provide evidence of a novel mechanism involving DDX5 that enables translation of transcription factor STAT1 mediating the IFN response. DESIGN AND RESULTS: Molecular, pharmacological and biophysical assays were used together with cellular models of HBV replication, HCC cell lines and liver tumours. We demonstrate that DDX5 regulates STAT1 mRNA translation by resolving a G-quadruplex (rG4) RNA structure, proximal to the 5' end of STAT1 5'UTR. We employed luciferase reporter assays comparing wild type (WT) versus mutant rG4 sequence, rG4-stabilising compounds, CRISPR/Cas9 editing of the STAT1-rG4 sequence and circular dichroism determination of the rG4 structure. STAT1-rG4 edited cell lines were resistant to the effect of rG4-stabilising compounds in response to IFN-α, while HCC cell lines expressing low DDX5 exhibited reduced IFN response. Ribonucleoprotein and electrophoretic mobility assays demonstrated direct and selective binding of RNA helicase-active DDX5 to the WT STAT1-rG4 sequence. Immunohistochemistry of normal liver and liver tumours demonstrated that absence of DDX5 corresponded to absence of STAT1. Significantly, knockdown of DDX5 in HBV infected HepaRG cells reduced the anti-viral effect of IFN-α. CONCLUSION: RNA helicase DDX5 resolves a G-quadruplex structure in 5'UTR of STAT1 mRNA, enabling STAT1 translation. We propose that DDX5 is a key regulator of the dynamic range of IFN response during innate immunity and adjuvant IFN-α therapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Regiões 5' não Traduzidas/genética , Antivirais/farmacologia , Carcinoma Hepatocelular/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/farmacologia , Vírus da Hepatite B , Hepatócitos/metabolismo , Humanos , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Neoplasias Hepáticas/metabolismo , Biossíntese de Proteínas , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Helicases/farmacologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Replicação Viral
7.
mBio ; 12(5): e0131621, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34517762

RESUMO

Coronaviruses (CoVs) are emergent pathogens that may cause life-threatening respiratory diseases in humans. Understanding of CoV-host interactions may help to identify novel therapeutic targets. MOV10 is an RNA helicase involved in different steps of cellular RNA metabolism. Both MOV10 antiviral and proviral activities have been described in a limited number of viruses, but this protein has not been previously associated with CoVs. We found that during Middle East respiratory syndrome coronavirus (MERS-CoV) infection, MOV10 aggregated in cytoplasmic structures colocalizing with viral nucleocapsid (N) protein. MOV10-N interaction was confirmed by endogenous MOV10 coimmunoprecipitation, and the presence of other cellular proteins was also detected in MOV10 complexes. MOV10 silencing significantly increased both N protein accumulation and virus titer, with no changes in the accumulation of viral RNAs. Moreover, MOV10 overexpression caused a 10-fold decrease in viral titers. These data indicated that MOV10 has antiviral activity during MERS-CoV infection. We postulated that this activity could be mediated by viral RNA sequestration, and in fact, RNA immunoprecipitation data showed the presence of viral RNAs in the MOV10 cytoplasmic complexes. Expression of wild-type MOV10 or of a MOV10 mutant without helicase activity in MOV10 knockout cell lines, developed by CRISPR-Cas technology, indicated that the helicase activity of MOV10 was required for its antiviral effect. Interestingly MOV10-N interaction was conserved in other mildly or highly pathogenic human CoVs, including the recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), although MOV10 antiviral activity was found only in highly pathogenic CoVs, suggesting a potential role of MOV10 in the modulation of human CoVs pathogenesis. IMPORTANCE Coronaviruses (CoVs) are emerging pathogens causing life-threatening diseases in humans. Knowledge of virus-host interactions and viral subversion mechanisms of host pathways is required for the development of effective countermeasures against CoVs. The interaction between cellular RNA helicase MOV10 and nucleocapsid (N) protein from several human CoVs is shown. Using MERS-CoV as a model, we demonstrate that MOV10 has antiviral function, requiring its helicase activity, most likely mediated by viral RNA sequestration in cytoplasmic ribonucleoprotein structures. Furthermore, we found that MOV10 antiviral activity may act only in highly pathogenic human CoVs, suggesting a role for MOV10 in modulating CoVs pathogenesis. The present study uncovers a complex network of viral and cellular RNAs and proteins interaction modulating the antiviral response against CoVs.


Assuntos
Antivirais/farmacologia , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , RNA Helicases/metabolismo , RNA Helicases/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Chlorocebus aethiops , Imunofluorescência , Humanos , Imunoprecipitação , RNA Viral/metabolismo , Células Vero , Replicação Viral/efeitos dos fármacos
8.
Mil Med Res ; 8(1): 37, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-34148549

RESUMO

BACKGROUND: Acute kidney injury (AKI) is the main life-threatening complication of crush syndrome (CS), and myoglobin is accepted as the main pathogenic factor. The pattern recognition receptor retinoicacid-inducible gene I (RIG-I) has been reported to exert anti-viral effects function in the innate immune response. However, it is not clear whether RIG-I plays a role in CS-AKI. The present research was carried out to explore the role of RIG-I in CS-AKI. METHODS: Sprague-Dawley rats were randomly divided into two groups: the sham and CS groups (n = 12). After administration of anesthesia, the double hind limbs of rats in the CS group were put under a pressure of 3 kg for 16 h to mimic crush conditions. The rats in both groups were denied access to food and water. Rats were sacrificed at 12 h or 36 h after pressure was relieved. The successful establishment of the CS-AKI model was confirmed by serum biochemical analysis and renal histological examination. In addition, RNA sequencing was performed on rat kidney tissue to identify molecular pathways involved in CS-AKI. Furthermore, NRK-52E cells were treated with 200 µmol/L ferrous myoglobin to mimic CS-AKI at the cellular level. The cells and cell supernatant samples were collected at 6 h or 24 h. Small interfering RNAs (siRNA) was used to knock down RIG-I expression. The relative expression levels of molecules involved in the RIG-I pathway in rat kidney or cells samples were measured by quantitative Real-time PCR (qPCR), Western blotting analysis, and immunohistochemistry (IHC) staining. Tumor necrosis factor-α (TNF-α) was detected by ELISA. Co-Immunoprecipitation (Co-IP) assays were used to detect the interaction between RIG-I and myoglobin. RESULTS: RNA sequencing of CS-AKI rat kidney tissue revealed that the different expression of RIG-I signaling pathway. qPCR, Western blotting, and IHC assays showed that RIG-I, nuclear factor kappa-B (NF-κB) P65, p-P65, and the apoptotic marker caspase-3 and cleaved caspase-3 were up-regulated in the CS group (P < 0.05). However, the levels of interferon regulatory factor 3 (IRF3), p-IRF3 and the antiviral factor interferon-beta (IFN-ß) showed no significant changes between the sham and CS groups. Co-IP assays showed the interaction between RIG-I and myoglobin in the kidneys of the CS group. Depletion of RIG-I could alleviate the myoglobin induced expression of apoptosis-associated molecules via the NF-κB/caspase-3 axis. CONCLUSION: RIG-I is a novel damage-associated molecular patterns (DAMPs) sensor for myoglobin and participates in the NF-κB/caspase-3 signaling pathway in CS-AKI. In the development of CS-AKI, specific intervention in the RIG-I pathway might be a potential therapeutic strategy for CS-AKI.


Assuntos
Caspase 3/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , RNA Helicases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/fisiopatologia , Alarminas , Animais , China , Síndrome de Esmagamento/sangue , Síndrome de Esmagamento/complicações , Modelos Animais de Doenças , Masculino , Mioglobina/farmacologia , Mioglobina/uso terapêutico , RNA Helicases/uso terapêutico , Ratos , Ratos Sprague-Dawley
9.
Pharmacol Ther ; 221: 107783, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33307143

RESUMO

Hypoxia is a hallmark of cancer. Hypoxia-inducible factor (HIF), a master player for sensing and adapting to hypoxia, profoundly influences genome instability, tumor progression and metastasis, metabolic reprogramming, and resistance to chemotherapies and radiotherapies. High levels and activity of HIF result in poor clinical outcomes in cancer patients. Thus, HIFs provide ideal therapeutic targets for cancers. However, HIF biology is sophisticated, and currently available HIF inhibitors have limited clinical utility owing to their low efficacy or side effects. RNA helicases, which are master players in cellular RNA metabolism, are usually highly expressed in tumors to meet the increased oncoprotein biosynthesis demand. Intriguingly, recent findings provide convincing evidence that RNA helicases are crucial for the adaptive cellular response to hypoxia via a mutual regulation with HIFs. More importantly, some RNA helicase inhibitors may suppress HIF signaling by blocking the translation of HIF-responsive genes. Therefore, RNA helicase inhibitors may work synergistically with HIF inhibitors in cancer to improve treatment efficacy. In this review, we discuss current knowledge of how cells sense and adapt to hypoxia through HIFs. However, our primary focus is on the multiple functions of RNA helicases in the adaptive response to hypoxia. We also highlight how these hypoxia-related RNA helicases can be exploited for anti-cancer therapeutics.


Assuntos
Antineoplásicos , Hipóxia Celular , Neoplasias , RNA Helicases , Antineoplásicos/farmacologia , Hipóxia Celular/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias/tratamento farmacológico , RNA Helicases/farmacologia , Transdução de Sinais
10.
PLoS One ; 7(1): e30653, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22292009

RESUMO

INTRODUCTION: Persistent infection with GBV-C (GB Virus C), a non-pathogenic virus related to hepatitis C virus (HCV), prolongs survival in HIV infection. Two GBV-C proteins, NS5A and E2, have been shown previously to inhibit HIV replication in vitro. We investigated whether the GBV-C NS3 serine protease affects HIV replication. RESULTS: GBV-C NS3 protease expressed in a human CD4+ T lymphocyte cell line significantly inhibited HIV replication. Addition of NS4A or NS4A/4B coding sequence to GBV-C NS3 increased the effect on HIV replication. Inhibition of HIV replication was dose-dependent and was not mediated by increased cell toxicity. Mutation of the NS3 catalytic serine to alanine resulted in loss of both HIV inhibition and protease activity. GBV-C NS3 expression did not measurably decrease CD4 or CXCR4 expression. CONCLUSION: GBV-C NS3 serine protease significantly inhibited HIV replication without decreasing HIV receptor expression. The requirement for an intact catalytic serine at the active site indicates that inhibition was mediated by proteolytic cleavage of an unidentified target(s).


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Vírus GB C/enzimologia , HIV-1/efeitos dos fármacos , Receptores de HIV/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/farmacologia , Replicação Viral/efeitos dos fármacos , Sequência de Aminoácidos , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Vírus GB C/genética , Expressão Gênica/efeitos dos fármacos , Genoma Viral/genética , HIV-1/fisiologia , Humanos , Células Jurkat , Dados de Sequência Molecular , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Helicases/farmacologia , RNA Helicases/fisiologia , Receptores de HIV/metabolismo , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Serina Endopeptidases/farmacologia , Serina Endopeptidases/fisiologia , Transfecção , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/fisiologia , Replicação Viral/genética
11.
Glia ; 60(3): 414-21, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22161971

RESUMO

Spinal cord injury (SCI) induces a glial response in which astrocytes become activated and produce inflammatory mediators. The molecular basis for regulation of glial-innate immune responses remains poorly understood. Here, we examined the activation of retinoic acid-inducible gene (RIG)-like receptors (RLRs) and their involvement in regulating inflammation after SCI. We show that astrocytes express two intracellular RLRs: RIG-I and melanoma differentiation-associated gene 5. SCI and stretch injury of cultured astrocytes stimulated RLR signaling as determined by phosphorylation of interferon regulatory factor 3 (IRF3) leading to production of type I interferons (IFNs). RLR signaling stimulation with synthetic ribonucleic acid resulted in RLR activation, phosphorylation of IRF3, and increased expression of glial fibrillary acidic protein (GFAP) and vimentin, two hallmarks of reactive astrocytes. Moreover, mitochondrial E3 ubiquitin protein ligase 1, an RLR inhibitor, decreased production of GFAP and vimentin after RIG-I signaling stimulation. Our findings identify a role for RLR signaling and type I IFN in regulating astrocyte innate immune responses after SCI.


Assuntos
Astrócitos/fisiologia , Imunidade Inata/fisiologia , RNA Helicases/metabolismo , Transdução de Sinais/fisiologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/metabolismo , Análise de Variância , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Células Cultivadas , RNA Helicases DEAD-box/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Imunidade Inata/efeitos dos fármacos , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon , Poli I-C/farmacologia , RNA Helicases/farmacologia , RNA de Cadeia Dupla/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Estresse Mecânico , Fatores de Tempo , Vimentina/metabolismo
12.
Cell ; 122(6): 875-86, 2005 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-16179257

RESUMO

Translation and mRNA degradation are affected by a key transition where eukaryotic mRNAs exit translation and assemble an mRNP state that accumulates into processing bodies (P bodies), cytoplasmic sites of mRNA degradation containing non-translating mRNAs, and mRNA degradation machinery. We identify the decapping activators Dhh1p and Pat1p as functioning as translational repressors and facilitators of P body formation. Strains lacking both Dhh1p and Pat1p show strong defects in mRNA decapping and P body formation and are blocked in translational repression. Contrastingly, overexpression of Dhh1p or Pat1p causes translational repression, P body formation, and arrests cell growth. Dhh1p, and its human homolog, RCK/p54, repress translation in vitro, and Dhh1p function is bypassed in vivo by inhibition of translational initiation. These results identify a broadly acting mechanism of translational repression that targets mRNAs for decapping and functions in translational control. We propose this mechanism is competitively balanced with translation, and shifting this balance is an important basis of translational control.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Proteínas de Ligação a DNA/fisiologia , Inativação Gênica/fisiologia , RNA Helicases/fisiologia , Estabilidade de RNA/fisiologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , RNA Helicases DEAD-box , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/farmacologia , Humanos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , RNA Helicases/genética , RNA Helicases/farmacologia , RNA Nucleotidiltransferases/genética , RNA Nucleotidiltransferases/fisiologia , RNA Mensageiro/efeitos dos fármacos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/farmacologia , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/farmacologia , Fatores de Tempo
13.
Int J Oncol ; 26(3): 685-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15703824

RESUMO

DJ-1 was identified by us as a novel oncogene in cooperation with activated ras. Although over-expression of DJ-1 has been reported in several cancer cells, including cells in breast cancer, lung cancer and prostate cancer, the precise mechanism underlying transformation has not been clarified. In this study, we screened proteins by a yeast two-hybrid method and identified Abstrakt as a DJ-1-binding protein. Abstrakt is an RNA helicase, but it has not yet been characterized. Northern blot analysis showed that human Abstrakt was expressed ubiquitously in all tissues. Abstrakt was then found to bind to and to be colocalized in the nucleus with DJ-1 in human cells. Furthermore, Abstrakt was found to stimulate transforming activity of DJ-1 in rat 3Y1 cells transfected with DJ-1 with activated ras. These findings suggest that Abstrakt is a positive regulator for DJ-1.


Assuntos
Regulação da Expressão Gênica , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/metabolismo , RNA Helicases/farmacologia , Animais , Northern Blotting , RNA Helicases DEAD-box , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Oncogênicas/genética , Proteína Desglicase DJ-1 , Ratos , Transfecção , Transformação Genética , Leveduras
14.
Nucleic Acids Res ; 32(6): 2031-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15064363

RESUMO

Viruses are intracellular parasites that must use the host machinery to multiply. Identification of the host factors that perform essential functions in viral replication is thus of crucial importance to the understanding of virus-host interactions. Here we describe Ded1p, a highly conserved DExD/H-box translation factor, as a possible host factor recruited by the yeast L-A double-stranded RNA (dsRNA) virus. We found that Ded1p interacts specifically and strongly with Gag, the L-A virus coat protein. Further analysis revealed that Ded1p interacts with the L-A virus in an RNA-independent manner and, as a result, L-A particles can be affinity purified via this interaction. The affinity-purified L-A particles are functional, as they are capable of synthesizing RNA in vitro. Critically, using purified L-A particles, we demonstrated that Ded1p specifically promotes L-A dsRNA replication by accelerating the rate of negative-strand RNA synthesis in vitro. In light of these data, we suggest that Ded1p may be a part of the long sought after activity shown to promote yeast viral dsRNA replication. This and the fact that Ded1p is also required for translating brome mosaic virus RNA2 in yeast thus raise the intriguing possibility that Ded1p is one of the key host factors favored by several evolutionarily related RNA viruses, including the human hepatitis C virus.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Fúngicas/fisiologia , RNA Helicases/fisiologia , RNA Viral/biossíntese , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/virologia , Totivirus/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/farmacologia , Sequência Conservada , RNA Helicases DEAD-box , Proteínas Fúngicas/química , Proteínas Fúngicas/farmacologia , Produtos do Gene gag/metabolismo , RNA Helicases/química , RNA Helicases/farmacologia , Totivirus/metabolismo , Totivirus/ultraestrutura , Transcrição Gênica , Vírion/genética , Vírion/metabolismo , Vírion/ultraestrutura , Replicação Viral
15.
J Biol Chem ; 279(2): 1269-80, 2004 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-14585830

RESUMO

The nonstructural 3 (NS3) protein encoded by the hepatitis C virus possesses both an N-terminal serine protease activity and a C-terminal 3'-5' helicase activity. This study examines the effects of the protease on the helicase by comparing the enzymatic properties of the full-length NS3 protein with truncated versions in which the protease is either deleted or replaced by a polyhistidine (His tag) or a glutathione S-transferase fusion protein (GST tag). When the NS3 protein lacks the protease domain it unwinds RNA more slowly and does not unwind RNA in the presence of excess nucleic acid that acts as an enzyme trap. Some but not all of the RNA helicase activity can be restored by adding a His tag or GST tag to the N terminus of the truncated helicase, suggesting that the effects of the protease are both specific and nonspecific. Similar but smaller effects are also seen in DNA helicase and translocation assays. While translocating on RNA (or DNA) the full-length protein hydrolyzes ATP more slowly than the truncated protein, suggesting that the protease allows for more efficient ATP usage. Binding assays reveal that the full-length protein assembles on single-stranded DNA as a higher order oligomer than the truncated fragment, and the binding appears to be more cooperative. The data suggest that hepatitis C virus RNA helicase, and therefore viral replication, could be influenced by the rotations of the protease domain which likely occur during polyprotein processing.


Assuntos
RNA de Cadeia Dupla/química , Proteínas não Estruturais Virais/química , Adenosina Trifosfatases/química , Trifosfato de Adenosina/química , Sequência de Aminoácidos , DNA de Cadeia Simples/química , Endopeptidases/química , Deleção de Genes , Glutationa Transferase/metabolismo , Hidrólise , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Desnaturação de Ácido Nucleico , Oligonucleotídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , RNA/química , RNA Helicases/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Ribonucleases/química , Espectrometria de Fluorescência , Temperatura , Fatores de Tempo , Proteínas não Estruturais Virais/metabolismo
16.
J Biochem ; 131(5): 687-92, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11983075

RESUMO

The activity of a hammerhead ribozyme (Rz) in vivo depends on several factors, such as abundance, stability, and accessibility of Rz to its target mRNA. Among these factors, accessibility is believed to be the rate-limiting factor for Rz-mediated cleavage in vivo. As Rz and its substrate RNA are negatively charged, we examined whether cellular RNA-interacting proteins or artificial polycations might improve the accessibility of Rz to its substrate RNA. Specifically, we examined the effects of two kinds of cationic comb-type copolymer, alphaPLL-g-Dex, and a cellular RNA helicase on the accessibility of Rz to a model structured RNA in vitro. The cleavage activity of Rz was slightly enhanced by alphaPLL-g-Dex, probably due to an acceleration of the association/dissociation rate. And also, the RNA helicase-bound hybrid-Rz could cleave the target substrate at a significantly higher rate due to its unwinding activity for the duplex RNA substrate. These approaches should be useful in the development of efficient gene-inactivating reagents in the post-genomic era.


Assuntos
RNA Helicases/farmacologia , RNA Catalítico/metabolismo , Sequência de Bases , Cetrimônio , Compostos de Cetrimônio/farmacologia , Relação Dose-Resposta a Droga , Cinética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Poliaminas/metabolismo , Polieletrólitos , RNA de Cadeia Dupla/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
17.
Mol Endocrinol ; 15(1): 69-79, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11145740

RESUMO

The nuclear receptor steroidogenic factor-1 (SF-1) is essential for development of the gonads, adrenal gland, and the ventromedial hypothalamic nucleus. It also regulates the expression of pivotal steroidogenic enzymes and other important proteins in the reproductive system. We sought to elucidate the mechanisms that govern the transcriptional activity of SF-1. We demonstrate here that a previously uncharacterized domain, located C-terminal to the DNA binding domain of SF-1, exhibits transcriptional repression function. Point mutations in this domain markedly potentiate the transcriptional activity of native SF-1. Using an SF-1 region that spans this proximal repression domain as bait in a yeast two-hybrid system, we cloned an SF-1 interacting protein that is homologous to human DP103, a member of the DEAD box family of putative RNA helicases. DP103 directly interacts with the proximal repression domain of SF-1, and mutations in this domain abrogate its interaction with DP103. DP103 is expressed predominantly in the testis and is also expressed at a lower level in other steroidogenic and nonsteroidogenic tissues. Functionally, DP103 exhibits a native transcriptional repression function that localizes to the C-terminal region of the protein and represses the activity of wild-type, but not mutant, SF-1. Together, the physical and functional interaction of DP103 with a previously unrecognized repression domain within SF-1 represents a novel mechanism for regulation of SF-1 activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , RNA Helicases/fisiologia , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Clonagem Molecular , Proteína DEAD-box 20 , RNA Helicases DEAD-box , DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Feminino , Fatores de Transcrição Fushi Tarazu , Expressão Gênica , Proteínas de Homeodomínio , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Mutação Puntual , RNA Helicases/genética , RNA Helicases/farmacologia , Ratos , Receptores Citoplasmáticos e Nucleares , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/farmacologia , Proteínas Repressoras/fisiologia , Homologia de Sequência , Fator Esteroidogênico 1 , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção
18.
Antivir Ther ; 3(Suppl 3): 83-91, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-10726058

RESUMO

Considerable progress has been made in characterizing the proteins involved in hepatitis C virus (HCV) replication, despite the lack of a cell culture system. A number of systems have been developed to examine the processes involved in viral replication, including the initiation and processing of the viral proteins required for RNA replication, the unwinding activities of the RNA helicase and the synthesis of RNA by the viral polymerase. These processes have been examined using individually cloned proteins expressed in various in vitro systems, which may be suitable targets for antiviral agents. The viral helicase and protease domains have now been crystallized, which may enable the rational design of specific inhibitors. The recent developments in HCV research in understanding the function of the viral non-structural proteins and the establishment of in vitro screening assays may aid in the development of new antiviral agents.


Assuntos
Hepacivirus/enzimologia , Replicação Viral/fisiologia , Antivirais/farmacologia , Células Cultivadas , RNA Polimerases Dirigidas por DNA/farmacologia , Flavivirus/genética , Hepacivirus/genética , Humanos , RNA Helicases/farmacologia , RNA Viral/análise , RNA Viral/biossíntese , Transcrição Gênica/efeitos dos fármacos , Proteínas não Estruturais Virais/fisiologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA