Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Mol Med ; 27(1): 110, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34530730

RESUMO

Posttranslational modification (PTM) and regulation of protein stability are crucial to various biological processes. Histone deacetylase 6 (HDAC6), a unique histone deacetylase with two functional catalytic domains (DD1 and DD2) and a ZnF-UBP domain (ubiquitin binding domain, BUZ), regulates a number of biological processes, including gene expression, cell motility, immune response, and the degradation of misfolded proteins. In addition to the deacetylation of histones, other nonhistone proteins have been identified as substrates for HDAC6. Hsp90, a molecular chaperone that is a critical modulator of cell signaling, is one of the lysine deacetylase substrates of HDAC6. Intriguingly, as one of the best-characterized regulators of Hsp90 acetylation, HDAC6 is the client protein of Hsp90. In addition to regulating Hsp90 at the post-translational modification level, HDAC6 also regulates Hsp90 at the gene transcription level. HDAC6 mainly regulates the Hsp90-HSF1 complex through the ZnF-UBP domain, thereby promoting the HSF1 entry into the nucleus and activating gene transcription. The mutual interaction between HDAC6 and Hsp90 plays an important role in the regulation of protein stability, cell migration, apoptosis and other functions. Plenty of of studies have indicated that blocking HDAC6/Hsp90 has a vital regulatory role in multifarious diseases, mainly in cancers. Therefore, developing inhibitors or drugs against HDAC6/Hsp90 becomes a promising development direction. Herein, we review the current knowledge on molecular regulatory mechanisms based on the interaction of HDAC6 and Hsp90 and inhibition of HDAC6 and/or Hsp90 in oncogenesis and progression, antiviral and immune-related diseases and other vital biological processes.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Desacetilase 6 de Histona/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Animais , Desenvolvimento de Medicamentos , Descoberta de Drogas , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Desacetilase 6 de Histona/antagonistas & inibidores , Desacetilase 6 de Histona/genética , Histonas/metabolismo , Humanos , Isoenzimas , Ligação Proteica , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
2.
Crit Rev Microbiol ; 46(4): 381-396, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32715819

RESUMO

Alzheimer's disease (AD) is a multifactorial disease triggered by environmental factors in combination with genetic predisposition. Infectious agents, in particular herpes simplex virus type 1 (HSV-1), are gradually being recognised as important factors affecting the development of AD. However, the mechanism linking HSV-1 and AD remains unknown. Of note, HSV-1 manipulates the activity of cofilin-1 to ensure their efficient infection in neuron cells. Cofilin-1, the main regulator of actin cytoskeleton reorganization, is implicating for the plastic of dendritic spines and axon regeneration of neuronal cells. Moreover, dysfunction of cofilin-1 is observed in most AD patients, as well as in mice with AD and ageing. Further, inhibition of cofilin-1 activity ameliorates the host cognitive impairment in an animal model of AD. Together, dysregulation of cofilin-1 led by HSV-1 infection is a potential link between HSV-1 and AD. Herein, we critically summarize the role of cofilin-1-mediated actin dynamics in both HSV-1 infection and AD, respectively. We also propose several hypotheses regarding the connecting roles of cofilin-1 dysregulation in HSV-1 infection and AD. Our review provides a foundation for future studies targeting individuals carrying HSV-1 in combination with cofilin-1 to promote a more individualised approach for treatment and prevention of AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/virologia , Cofilina 1/metabolismo , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Doença de Alzheimer/genética , Animais , Axônios/metabolismo , Axônios/virologia , Cofilina 1/genética , Herpes Simples/genética , Herpesvirus Humano 1/genética , Humanos , Neurônios/metabolismo , Neurônios/virologia
3.
Crit Rev Microbiol ; 45(5-6): 581-594, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31512533

RESUMO

Microglia, as brain-resident macrophages, are the first line of defense against brain invading pathogens. Further, their dysfunction has been recognized to be closely associated with mounting CNS diseases. Of note, chronic HSV-1 infection leads to the persistent activation of microglia, which elicit a comprehensive response by generating certain factors with neurotoxic and neuroprotective effects. CNS infection with HSV-1 results in herpes simplex encephalitis and herpes simplex keratitis. Microglial immune response plays a crucial role in the development of these diseases. Moreover, HSV-1 infection is strongly associated with several CNS diseases, especially Alzheimer's disease and schizophrenia. These CNS diseases can be effectively ameliorated by eliciting an appropriate immune response, such as inhibition of microglial proliferation and activation. Therefore, it is crucial to reassess the positive and negative roles of microglia in HSV-1 CNS infection for a more comprehensive and detailed understanding of the relationship between microglia and CNS diseases. Hence, the present review focuses on the dual roles of microglia in mediating HSV-1 CNS infection, as well as on the strategy of targeting microglia to ameliorate CNS diseases. Further research in this field can help comprehensively elucidate the dual role of the microglial immune response in HSV-1 CNS infection, providing a theoretical basis for identifying therapeutic targets against overactive microglia in CNS diseases and HSV-1 infection.


Assuntos
Encefalite por Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Microglia/imunologia , Animais , Encefalite por Herpes Simples/imunologia , Herpesvirus Humano 1/genética , Humanos , Microglia/virologia
4.
Mol Med ; 24(1): 65, 2018 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-30577726

RESUMO

BACKGROUND: Numerous host cellular factors are exploited by viruses to facilitate infection. Our previous studies and those of others have shown heat-shock protein 90 (Hsp90), a cellular molecular chaperone, is involved in herpes simplex virus (HSV)-1 infection. However, the function of the dominant Hsp90 isoform and the relationship between Hsp90 and HSV-1 α genes remain unclear. METHODS AND RESULTS: Hsp90α knockdown or inhibition significantly inhibited the promoter activity of HSV-1 α genes and downregulated virion protein 16(VP16) expression from virus and plasmids. The Hsp90α knockdown-induced suppression of α genes promoter activity and downregulation of α genes was reversed by VP16 overexpression, indicating that Hsp90α is involved in VP16-mediated transcription of HSV-1 α genes. Co-immunoprecipitation experiments indicated that VP16 interacted with Hsp90α through the conserved core domain within VP16. Based on using autophagy inhibitors and the presence of Hsp90 inhibitors in ATG7-/- (autophagy-deficient) cells, Hsp90 inhibition-induced degradation of VP16 is dependent on macroautophagy-mediated degradation but not chaperone-mediated autophagy (CMA) pathway. In vivo studies demonstrated that treatment with gels containing Hsp90 inhibitor effectively reduced the level of VP16 and α genes, which may contribute to the amelioration of the skin lesions in an HSV-1 infection mediated zosteriform model. CONCLUSION: Our study provides new insights into the mechanisms by which Hsp90α facilitates the transactivation of HSV-1 α genes and viral infection, and highlights the importance of developing selective inhibitors targeting the interaction between Hsp90α and VP16 to reduce toxicity, a major challenge in the clinical use of Hsp90 inhibitors.


Assuntos
Proteínas de Choque Térmico HSP90/genética , Proteína Vmw65 do Vírus do Herpes Simples/genética , Herpesvirus Humano 1/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Feminino , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Herpes Simples/tratamento farmacológico , Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Ativação Transcricional
5.
Virus Genes ; 54(3): 343-350, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29541932

RESUMO

Herpes simplex virus 1 (HSV-1) encodes various microRNAs (miRNAs), whose targets are largely unknown. miR-H1 is the first discovered HSV-1 miRNA and is expressed predominantly in productive infection. Here we show that ubiquitin protein ligase E3 component n-recognin 1 (Ubr1) is a cellular target of miR-H1. Ubr1 is a RING-type E3 ubiquitin ligase of the Arg/N-end rule pathway, which causes the degradation of proteins bearing "destabilizing" N-terminal residues, such as neurodegeneration-associated protein fragment ß-amyloid. Using model substrates, we found that miR-H1 significantly repressed the expression and activity of Ubr1. Consequently, miR-H1-mediated Ubr1 silencing resulted in the accumulation of ß-amyloid, which might contribute to the neurodegenerative pathogenesis enhanced by HSV-1. Our results provide novel insights into the mechanism by which HSV-1-encoded miR-H1 functions in neurodegenerative pathogenesis through targeting Ubr1-mediated Arg/N-end rule degradation pathway.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Herpesvirus Humano 1/fisiologia , MicroRNAs/fisiologia , RNA Viral/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Células HEK293 , Humanos , MicroRNAs/biossíntese , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , RNA Viral/biossíntese , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores
6.
J Gen Virol ; 98(3): 322-337, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27959772

RESUMO

Histone deacetylase 6 (HDAC6) is a unique cytoplasmic deacetylase that regulates various important biological processes by preventing protein aggregation and deacetylating different non-histone substrates including tubulin, heat shock protein 90, cortactin, retinoic acid inducible gene I and ß-catenin. Growing evidence has indicated a dual role for HDAC6 in viral infection and pathogenesis: HDAC6 may represent a host defence mechanism against viral infection by modulating microtubule acetylation, triggering antiviral immune response and stimulating protective autophagy, or it may be hijacked by the virus to enhance proinflammatory response. In this review, we will highlight current data illustrating the complexity and importance of HDAC6 in viral pathogenesis. We will summarize the structure and functional specificity of HDAC6, and its deacetylase- and ubiquitin-dependent activity in key cellular events in response to virus infection. We will also discuss how HDAC6 exerts its direct or indirect histone modification ability in viral lytic-latency switch.


Assuntos
Histona Desacetilases/fisiologia , Viroses/enzimologia , Viroses/virologia , Fenômenos Fisiológicos Virais , Animais , Movimento Celular , Núcleo Celular/virologia , Desacetilase 6 de Histona , Histona Desacetilases/química , Histonas/metabolismo , Humanos , Imunidade Celular , Microtúbulos/metabolismo , Processamento de Proteína Pós-Traducional , Ubiquitina/metabolismo
7.
Arch Virol ; 162(11): 3269-3282, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28780632

RESUMO

The emergence of antiviral drug-resistant mutants is the most important issue in current antiviral therapy. As obligate parasites, viruses require host factors for efficient replication. An ideal therapeutic target to prevent drug-resistance development is represented by host factors that are crucial for the viral life cycle. Recent studies have indicated that heat shock protein 90 (HSP90) is a crucial host factor that is required by many viruses for multiple phases of their life cycle including viral entry, nuclear import, transcription, and replication. In this review, we summarize the most recent advances regarding HSP90 function, mechanisms of action, and molecular pathways that are associated with viral infection, and provide a comprehensive understanding of the role of HSP90 in the immune response and exosome-mediated viral transmission. In addition, several HSP90 inhibitors have entered clinical trials for specific cancers that are associated with viral infection, which further implies a crucial role for HSP90 in the malignant transformation of virus-infected cells; as such, HSP90 inhibitors exhibit excellent therapeutic potential. Finally, we describe the challenge of developing HSP90 inhibitors as anti-viral drugs.


Assuntos
Antivirais/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Viroses/tratamento farmacológico , Viroses/metabolismo , Animais , Humanos , Viroses/virologia , Replicação Viral/efeitos dos fármacos
8.
Crit Rev Microbiol ; 42(5): 677-95, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25853495

RESUMO

Actin-depolymerizing factor (ADF)/cofilin proteins are key players in controlling the temporal and spatial extent of actin dynamics, which is crucial for mediating host-pathogen interactions. Pathogenic microbes have evolved molecular mechanisms to manipulate cofilin activity to subvert the actin cytoskeletal system in host cells, promoting their internalization into the target cells, modifying the replication niche and facilitating their intracellular and intercellular dissemination. The study of how these pathogens exploit cofilin pathways is crucial for understanding infectious disease and providing potential targets for drug therapies.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Fenômenos Fisiológicos Bacterianos , Interações Hospedeiro-Patógeno , Citoesqueleto de Actina/genética , Fatores de Despolimerização de Actina/genética , Animais , Bactérias/genética , Infecções Bacterianas/genética , Destrina/genética , Destrina/metabolismo , Humanos
9.
Rev Med Virol ; 24(4): 274-86, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24888553

RESUMO

Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that regulates cellular homeostatic processes. Following ligand binding, EGFR activates different downstream signalling cascades that promote cell survival, proliferation, motility, and angiogenesis and induces F-actin-dependent EGFR endocytosis, which relocalises the activated receptors for degradation or recycling. The responses that are induced by ligand binding to EGFR, including cell signalling activation, protein kinase phosphorylation and cytoskeletal network rearrangement, resemble those induced by virus infection. Increasing evidence demonstrates that many viruses usurp EGFR endocytosis or EGFR-mediated signalling for entry, replication, inflammation, and viral antagonism to the host antiviral system. In addition, viruses have acquired sophisticated mechanisms to regulate EGFR functions by interrupting the EGFR-recycling process and modulating EGFR expression. In this review, we provide an overview of the mechanisms by which viruses alter EGFR signalling in favour of their continued survival.


Assuntos
Receptores ErbB/metabolismo , Interações Hospedeiro-Patógeno , Transdução de Sinais , Fenômenos Fisiológicos Virais , Humanos
10.
Biochem Biophys Res Commun ; 453(4): 821-5, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25450354

RESUMO

Influenza A virus (IAV) assembly and budding on host cell surface plasma membrane requires actin cytoskeleton reorganization. The underlying molecular mechanism involving actin reorganization remains unclarified. In this study, we found that the natural antiviral compound petagalloyl glucose (PGG) inhibits F-actin reorganization in the host cell membrane during the late stage of IAV infection, which are associated with the suppression of total cofilin-1 level and its phosphorylation. Knock-down of cofilin-1 reduces viral yields. These findings provide the first evidence that cofilin-1 plays an important role in regulating actin reorganization during IAV assembly and budding.


Assuntos
Actinas/metabolismo , Cofilina 1/metabolismo , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/ultraestrutura , Proteínas Virais/metabolismo , Montagem de Vírus/fisiologia , Liberação de Vírus/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Vírus da Influenza A/isolamento & purificação
11.
Biochem Biophys Res Commun ; 437(3): 482-8, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23850690

RESUMO

Autophagy plays a crucial role in a wide array of physiological processes. To uncover the complex regulatory networks and mechanisms underlying basal autophagy, we performed a quantitative proteomics analysis of autophagy-deficient mouse embryonic fibroblast cells (MEFs) using iTRAQ labeling coupled with on-line 2D LC/MS/MS. We quantified a total of 1234 proteins and identified 114 proteins that were significantly altered (90% confidence interval), including 48 up-regulated proteins and 66 down-regulated proteins. We determined that F-actin was disassembled in autophagy-deficient Atg7(-/-) MEFs. Treatment of the WT MEFs with cytochalasin D (CD), which induces F-actin depolymerization, significantly induced autophagosome formation. However, treatment with cytochalasin D also increased the protein level of p62 under starvation conditions, suggesting that depolymerization of F-actin impaired autophagosome maturation and that the intact F-actin network is required for basal and starvation-induced autophagy. Our results demonstrate a close relationship between F-actin and autophagy and provide the basis for further investigation of their interactions.


Assuntos
Actinas/fisiologia , Autofagia/genética , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas Associadas aos Microtúbulos/deficiência , Proteômica/métodos , Actinas/genética , Animais , Proteína 7 Relacionada à Autofagia , Linhagem Celular Transformada , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Camundongos , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/genética , Fagossomos/metabolismo , Fagossomos/patologia , Mapas de Interação de Proteínas/genética
12.
J Virol ; 86(16): 8440-51, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623803

RESUMO

Herpes simplex virus 1 (HSV-1) invades the nervous system and causes pathological changes. In this study, we defined the remodeling of F-actin and its possible mechanisms during HSV-1 infection of neuronal cells. HSV-1 infection enhanced the formation of F-actin-based structures in the early stage of infection, which was followed by a continuous decrease in F-actin during the later stages of infection. The disruption of F-actin dynamics by chemical inhibitors significantly reduced the efficiency of viral infection and intracellular HSV-1 replication. The active form of the actin-depolymerizing factor cofilin 1 was found to increase at an early stage of infection and then to continuously decrease in a manner that corresponded to the remodeling pattern of F-actin, suggesting that cofilin 1 may be involved in the biphasic F-actin dynamics induced by HSV-1 infection. Knockdown of cofilin 1 impaired HSV-1-induced F-actin assembly during early infection and inhibited viral entry; however, overexpression of cofilin 1 did not affect F-actin assembly or viral entry during early infection but decreased intracellular viral reproduction efficiently. Our results, for the first time, demonstrated the biphasic F-actin dynamics in HSV-1 neuronal infection and confirmed the association of F-actin with the changes in the expression and activity of cofilin 1. These results may provide insight into the mechanism by which HSV-1 productively infects neuronal cells and causes pathogenesis.


Assuntos
Actinas/metabolismo , Cofilina 1/metabolismo , Herpesvirus Humano 1/fisiologia , Neurônios/metabolismo , Neurônios/virologia , Internalização do Vírus , Replicação Viral , Linhagem Celular , Humanos
13.
J Proteome Res ; 10(2): 845-55, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21080727

RESUMO

Tumor reversion is defined as the process by which cancer cells lose their malignant phenotype. However, relatively little is known about the cellular proteome changes that occur during the reversion process. A biological model of multiple myeloma (MM) reversion was established by using the H-1 parvovirus as a tool to select for revertant cells from MM cells. Isolated revertant cells displayed a strongly suppressed malignant phenotype both in vitro and in vivo. To explore possible mechanisms of MM reversion, the protein profiles of the revertant and parental MM cells were compared using a quantitative proteomic strategy termed SILAC-MS. Our results revealed that 379 proteins were either activated or inhibited during the reversion process, with a much greater proportion of the proteins, including STAT3, TCTP, CDC2, BAG2, and PCNA, being inhibited. Of these, STAT3, which is significantly down regulated, was selected for further functional studies. Inhibition of STAT3 expression by RNA interference resulted in suppression of the malignant phenotype and concomitant down regulation of TCTP expression, suggesting that myeloma reversion operates, at least in part, through inhibition of STAT3. Our results provide novel insights into the mechanisms of tumor reversion and suggest new alternative approaches for MM treatment.


Assuntos
Biomarcadores Tumorais/análise , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/análise , Proteoma/análise , Proteômica/métodos , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Parvovirus H-1 , Humanos , Marcação por Isótopo , Proteínas de Neoplasias/metabolismo , Processos Neoplásicos , Fenótipo , Proteoma/metabolismo , Interferência de RNA , Fator de Transcrição STAT3 , Proteína Tumoral 1 Controlada por Tradução
14.
Biochem Biophys Res Commun ; 405(2): 186-91, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21216235

RESUMO

Pentagalloylglucose (PGG) is a natural polyphenolic compound with broad-spectrum anti-viral activity, however, the mechanisms underlying anti-viral activity remain undefined. In this study, we investigated the effects of PGG on anti-viral activity against Herpes simplex virus type 1 (HSV-1) associated with autophagy. We found that the PGG anti-HSV-1 activity was impaired significantly in MEF-atg7-/- cells (autophagy-defective cells) derived from an atg7-/- knockout mouse. Transmission electron microscopy revealed that PGG-induced autophagosomes engulfed HSV-1 virions. The mTOR signaling pathway, an essential pathway for the regulation of autophagy, was found to be suppressed following PGG treatment. Data presented in this report demonstrated for the first time that autophagy induced following PGG treatment contributed to its anti-HSV activity in vitro.


Assuntos
Antivirais/farmacologia , Autofagia , Herpesvirus Humano 1/efeitos dos fármacos , Taninos Hidrolisáveis/farmacologia , Fagossomos/efeitos dos fármacos , Animais , Proteína 7 Relacionada à Autofagia , Linhagem Celular , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Fagossomos/virologia , Fatores de Transcrição/genética
15.
Arch Virol ; 156(8): 1359-69, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21479599

RESUMO

Influenza A virus (IAV) infection is a major public health threat leading to significant morbidity and mortality. The emergence of drug-resistant virus strains highlights the urgent need to develop novel antiviral drugs with alternative modes of action. Pentagalloylglucose (PGG), a naturally occurring polyphenolic compound, possesses a broad spectrum of biological activities. In this study, we found that PGG has anti-influenza-virus activity, and investigated its possible mechanism(s) of action in vitro. Both pre-incubation of virus prior to infection and post-exposure of infected cells with PGG significantly inhibited virus yields. Influenza-virus-induced hemagglutination of chicken red blood cells was inhibited by PGG treatment, suggesting that PGG can inhibit IAV infection by interacting with the viral hemagglutinin. PGG did not affect viral protein synthesis or nuclear transport of viral nucleoprotein (NP) but greatly reduced plasma membrane accumulation of NP protein at the late stage of the replication cycle. Furthermore, PGG significantly reduced virus budding and progeny virus release from infected cells. This study revealed for the first time that PGG can inhibit IAV replication with a dual mode of action and offers new insights into its underlying mechanisms of antiviral action.


Assuntos
Antivirais/farmacologia , Taninos Hidrolisáveis/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Animais , Linhagem Celular , Galinhas , Cães , Eritrócitos , Hemaglutinação/efeitos dos fármacos , Humanos , Taninos Hidrolisáveis/química , Phyllanthus emblica/química , Estereoisomerismo , Internalização do Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
16.
Phytother Res ; 25(7): 975-82, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21213355

RESUMO

In this study, 1,2,4,6-tetra-O-galloyl-ß-D-glucose (1246TGG), a polyphenolic compound isolated from traditional Chinese medicine Phyllanthus emblica L. (Euphorbiaceae), was found to inhibit herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) infection at different magnitudes of activity in vitro. Further studies revealed that 1246TGG directly inactivated HSV-1 particles, leading to the failure of early infection, including viral attachment and penetration. 1246TGG also suppressed the intracellular growth of HSV-1 within a long period post-infection (from 0 h p.i. to 12 h p.i.), while it might exert an antiviral effect mainly before 3 h p.i. It inhibited HSV-1 E and L gene expressions as well as viral DNA replication but did not affect the RNA synthesis of IE gene in our study. Also, in the presence of 1246TGG, the synthesis of viral protein was reduced. Taken together, it was suggested that 1246TGG might exert anti-HSV activity both by inactivating extracellular viral particles and by inhibiting viral biosynthesis in host cells. These results warrant further studies on the antiviral mechanisms of 1246TGG and suggest that it might be a candidate for HSV therapy.


Assuntos
Antivirais/farmacologia , Ácido Gálico/análogos & derivados , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Monossacarídeos/farmacologia , Animais , Antivirais/isolamento & purificação , Chlorocebus aethiops , DNA Viral/biossíntese , Ácido Gálico/isolamento & purificação , Ácido Gálico/farmacologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 2/fisiologia , Monossacarídeos/isolamento & purificação , Phyllanthus emblica/química , Células Vero , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos
17.
J Asian Nat Prod Res ; 13(6): 498-504, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21623512

RESUMO

Further study on steam-treated notoginseng, the roots of Panax notoginseng (Burk.) F.H. Chen (Araliaceae), which is a famous traditional Chinese medicine that is used both in raw and treated forms for a long time, led to the isolation of a new dammarane-type saponin, namely notoginsenoside ST-4. Its structure was elucidated to be 3ß,12ß,20(S)-tri-hydroxydammar-24-ene-3-O-ß-d-xylopyranosyl-(1 â†’ 2)-ß-d-glucopyranosyl-(1 â†’ 2)-ß-d-glu-copyranoside, based on the detailed analyses of the 1D and 2D NMR spectral data and acidic hydrolysis. Notoginsenoside ST-4 was investigated for its antiviral activity on herpes simplex type 1 (HSV-1) and type 2 (HSV-2) in vitro. The 50% effective concentration (EC(50)) values, determined by plaque reduction assay, were 16.47 ± 0.67 and 19.44 ± 1.16 µM for HSV-1 and HSV-2, respectively, whereas the 50% cytotoxic concentration (CC(50)) determined by the XTT test on Vero cells was 510.64 ± 4.56 µM. As analyzed by attachment assay and penetration assay based on plaque reduction assay, the antiviral activity of notoginsenoside ST-4 was principally due to the penetration inhibition effects, which was confirmed by fluorescence microscopy observation that notoginsenoside ST-4 blocked the penetration of virus. Therefore, notoginsenoside ST-4 might be a promising agent for herpes simplex virus infection.


Assuntos
Antivirais/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Panax/química , Saponinas/farmacologia , Simplexvirus/efeitos dos fármacos , Triterpenos/farmacologia , Animais , Antivirais/química , Antivirais/isolamento & purificação , Chlorocebus aethiops , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/isolamento & purificação , Estrutura Molecular , Raízes de Plantas/química , Saponinas/química , Saponinas/isolamento & purificação , Triterpenos/química , Triterpenos/isolamento & purificação , Células Vero
18.
Theranostics ; 11(19): 9623-9651, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646390

RESUMO

Microglia are the primary cellular source of type I interferons (I-IFNs) in the brain upon neurotropic virus infection. Although the I-IFN-based antiviral innate immune response is crucial for eliminating viruses, overproduction led to immune disorders. Therefore, the relatively long-lasting I-IFNs must be precisely controlled, but the regulatory mechanism for the innate antiviral response in microglia remains largely unknown. Long non-coding RNAs (lncRNAs) are being recognized as crucial factors in numerous diseases, but their regulatory roles in the innate antiviral response in microglia are undefined. Methods: The high-throughput RNA sequencing was performed to obtain differentially expressed lncRNAs (DELs) in primary microglia infected with or without the neurotropic herpes simplex virus type 1 (HSV-1). We selected four DELs ranked in the top 15 in basic level and their fold change induced by HSV-1, i.e., FPKMHSV-1/FPKMCells.We subsequently found a key lncRNA affecting the innate antiviral response of microglia significantly. We next used dual-luciferase reporter assays, bioinformatical tools, and truncation mutants of both lncRNA and targeted proteins to elucidate the downstream and upstream mechanism of action of lncRNA. Further, we established microglia-specific knock-in (KI) mice to investigate the role of lncRNA in vivo. Results: We identified a long intergenic non-coding RNA, linc-AhRA, involved in regulating the innate antiviral response in murine microglia. linc-AhRA is activated by aryl hydrocarbon receptor (AhR) and restricts I-IFN production in microglia upon neurotropic herpesvirus infection and innate immune stimulation. Mechanistically, linc-AhRA binds to both tripartite motif-containing 27 (TRIM27) and TANK-binding kinase 1 (TBK1) through its conserved 117nt fragment as a molecular scaffold to enhance TRIM27-TBK1 interaction. This interaction facilitates the TRIM27-mediated ubiquitination of TBK1 and results in ubiquitin-proteasome-dependent degradation of TBK1. Consequently, linc-AhRA suppresses I-IFN production through facilitating TBK1 degradation and limits the microglial innate immune response against neurotropic herpesvirus infection. Microglia-specific KI of linc-AhRA mice shows a weakened antiviral immune response upon neurotropic herpesvirus challenge due to a reduction of TBK1 in microglia. Conclusion: Our findings indicate that linc-AhRA is a negative regulator of I-IFN production in microglia to avoid excessive autoimmune responses. These findings uncover a previously unappreciated role for lncRNA conserved fragments in the innate antiviral response, providing a strong foundation for developing nucleotide drugs based on conserved functional fragments within lncRNAs.


Assuntos
Infecções por Herpesviridae/genética , Microglia/imunologia , RNA Longo não Codificante/genética , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Herpesviridae/patogenicidade , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 1/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/genética , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação , Transdução de Sinais , Transcriptoma/genética
19.
Proteomics ; 10(12): 2337-47, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20391533

RESUMO

MIP-T3 (microtubule-interacting protein associated with TRAF3) is a microtubule-interacting protein that evolutionarily conserved from worms to humans, but whose cellular functions remains unknown. To get insight into the functions of MIP-T3, we set out to identify MIP-T3 interacting proteins by immunoprecipitation in human embryonic kidney 293 cells and MS analysis. As the results, a total of 34 proteins were identified and most of them were novel MIP-T3 putative partners. The MIP-T3-associated proteins could be grouped into nine clusters based on their molecule functions, including cytoskeleton, chaperone, nucleic acid binding, kinase and so on. Three MIP-T3-interacted proteins - actin, HSPA8 and tubulin - were further confirmed by reciprocal coimmunoprecipitations and colocalization analysis. The interaction of MIP-T3 with both actin filaments and microtubule suggested that MIP-T3 may play an important role in regulation of cytoskeleton dynamics in cells. Our results therefore not only uncover a large number of MIP-T3-associated proteins that possess a variety of cellular functions, but also provide new research directions for the study of the functions of MIP-T3.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteômica , Actinas/metabolismo , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Imunoprecipitação , Microscopia Confocal , Microtúbulos/metabolismo , Ligação Proteica
20.
Anal Biochem ; 397(2): 197-201, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19852926

RESUMO

A highly selective and facile assay of human immunodeficiency virus protease (HIV-PR) has been required for the screening of medicinal inhibitors and also for classifying the subtypes of HIV in the therapeutic treatment of acquired immune deficiency syndrome (AIDS). This article describes a novel assay method of HIV-PR based on the selective fluorogenic reaction of peptides. A peptide fragment generated from a substrate by the enzymatic digestion with HIV-PR could be selectively quantified by the spectrofluorometric detection after the fluorogenic reaction with catechol in the presence of sodium periodate and sodium borate (pH 7.0). This assay system uses an N-terminal acetyl peptide as the substrate and crude extracts from Escherichia coli expressing recombinant HIV-PR. The activity obtained by the proposed assay correlated with that obtained by a conventional HIV-PR assay based on fluorescence resonance energy transfer detection.


Assuntos
Protease de HIV/análise , Protease de HIV/metabolismo , Inibidores da Protease de HIV/farmacologia , HIV-1/enzimologia , Reprodutibilidade dos Testes , Espectrometria de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA