Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Antibiot (Tokyo) ; 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898184

RESUMO

The development of new therapeutic uses for existing drugs is important for the treatment of some diseases. Cephalosporin antibiotics stand as the most extensively utilized antibiotics in clinical practice, effectively combating bacterial infections. Here, we found that the antimicrobial drug ceftazidime strongly upregulates p27 protein levels by inhibiting p27 ubiquitination. The p27 protein is a classic negative regulator of the cell cycle. Next, we demonstrated that ceftazidime can impede the cell cycle from G1 to S phase, thus inhibiting cell proliferation. Furthermore, we found that ceftazidime promotes p27 expression and inhibits cell proliferation by reducing Skp2, which is a substrate recognition component of the Skp2-Cullin-F-box (SCF) ubiquitin ligase. Moreover, ceftazidime downregulates transcriptional expression of Skp2. Importantly, we demonstrated that ceftazidime inhibited the proliferation of tumor cells in vivo. These findings reveal ceftazidime-mediated inhibition of cell proliferation through the Skp2-p27 axis, and could provide a potential strategy for anti-tumor therapy.

2.
J Virol ; 97(10): e0078623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37796126

RESUMO

IMPORTANCE: EV71 poses a significant health threat to children aged 5 and below. The process of EV71 infection and replication is predominantly influenced by ubiquitination modifications. Our previous findings indicate that EV71 prompts the activation of host deubiquitinating enzymes, thereby impeding the host interferon signaling pathway as a means of evading the immune response. Nevertheless, the precise mechanisms by which the host employs ubiquitination modifications to hinder EV71 infection remain unclear. The present study demonstrated that the nonstructural protein 2Apro, which is encoded by EV71, exhibits ubiquitination and degradation mediated by the host E3 ubiquitin ligase SPOP. In addition, it is the first report, to our knowledge, that SPOP is involved in the host antiviral response.


Assuntos
Cisteína Endopeptidases , Enterovirus Humano A , Infecções por Enterovirus , Interações entre Hospedeiro e Microrganismos , Ubiquitina-Proteína Ligases , Ubiquitina , Ubiquitinação , Proteínas Virais , Criança , Humanos , Enterovirus Humano A/enzimologia , Enterovirus Humano A/fisiologia , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/virologia , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Cisteína Endopeptidases/metabolismo
3.
Immunology ; 170(4): 527-539, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37641430

RESUMO

Skp2 plays multiple roles in malignant tumours. Here, we revealed that Skp2 negatively regulates type-I interferon (IFN-I)-mediated antiviral activity. We first noticed that Skp2 can promote virus infection in cells. Further studies demonstrated that Skp2 interacts with IFN-I receptor 2 (IFNAR2) and promotes K48-linked polyubiquitination of IFNAR2, which accelerates the degradation of IFNAR2 proteins. Skp2-mediated downregulation of IFNAR2 levels inhibits IFN-I signalling and IFN-I-induced antiviral activity. In addition, we uncovered for the first time that the antibiotic ceftazidime can act as a repressor of Skp2. Ceftazidime reduces cellular Skp2 levels, thus enhancing IFNAR2 stability and IFN-I antiviral activity. This study reveals a new role of Skp2 in regulating IFN-I signalling and IFN-I antiviral activity and reports the antibiotic ceftazidime as a potential repressor of Skp2.


Assuntos
Interferon Tipo I , Interferon Tipo I/metabolismo , Ceftazidima , Linhagem Celular , Antivirais/farmacologia , Antibacterianos , Receptor de Interferon alfa e beta
4.
Int Immunopharmacol ; 114: 109595, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36700774

RESUMO

Methotrexate (MTX) is used to treat rheumatoid arthritis, acute leukemia, and psoriasis. MTX can cause certain side effects, such as myelosuppression, while the exact mechanism of myelosuppression caused by MTX is unknown. Notch signaling pathway has been considered to be essential to regulate hematopoietic stem cell (HSC) regeneration and homeostasis, thus contributing to bone marrow hematopoiesis. However, whether MTX affects Notch signaling remains unexplored. Here, our study provides evidence that MTX strongly suppresses the Notch signaling pathway. We found that MTX inhibited the interaction between Nedd4 with Numb, thus restricting K48-linked polyubiquitination of Numb and stabilizing Numb proteins. This in turn inhibited the Notch signaling pathway by reducing Notch1 protein levels. Interestingly, we found that a monomeric drug, Triptolide, is capable of alleviating the inhibitory effect of MTX on Notch signaling pathway. This study promotes our understanding of MTX-mediated regulation of Notch signaling and could provide ideas to alleviate MTX-induced myelosuppression.


Assuntos
Metotrexato , Receptores Notch , Proteínas de Membrana/metabolismo , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Receptor Notch1 , Receptores Notch/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
5.
STAR Protoc ; 2(3): 100708, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34386779

RESUMO

Innate immunity is the first line of host defense against viral infection. As one of the innate immune cell types, antigen-presenting cells play an important role in the process of antiviral immunity. This protocol describes the analysis of innate immunity induced by vesicular stomatitis virus infection of peritoneal macrophages in vitro and in vivo detection of IFN-ß production and lung injury. For complete details on the use and execution of this protocol, please refer to Shen et al. (2021).


Assuntos
Separação Celular/métodos , Imunidade Inata/fisiologia , Viroses/diagnóstico por imagem , Animais , Células Apresentadoras de Antígenos/imunologia , Interferon Tipo I/imunologia , Macrófagos/imunologia , Macrófagos Peritoneais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Viroses/imunologia , Replicação Viral/imunologia
6.
Cell Metab ; 33(5): 971-987.e6, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33798471

RESUMO

Serine metabolism promotes tumor oncogenesis and regulates immune cell functions, but whether it also contributes to antiviral innate immunity is unknown. Here, we demonstrate that virus-infected macrophages display decreased expression of serine synthesis pathway (SSP) enzymes. Suppressing the SSP key enzyme phosphoglycerate dehydrogenase (PHGDH) by genetic approaches or by treatment with the pharmaceutical inhibitor CBR-5884 and by exogenous serine restriction enhanced IFN-ß-mediated antiviral innate immunity in vitro and in vivo. Mechanistic experiments showed that virus infection or serine metabolism deficiency increased the expression of the V-ATPase subunit ATP6V0d2 by inhibiting S-adenosyl methionine-dependent H3K27me3 occupancy at the promoter. ATP6V0d2 promoted YAP lysosomal degradation to relieve YAP-mediated blockade of the TBK1-IRF3 axis and, thus, enhance IFN-ß production. These findings implicate critical functions of PHGDH and the key immunometabolite serine in blunting antiviral innate immunity and also suggest manipulation of serine metabolism as a therapeutic strategy against virus infection.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Imunidade Inata , Lisossomos/metabolismo , Serina/metabolismo , Fatores de Transcrição/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Histonas/metabolismo , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoglicerato Desidrogenase/antagonistas & inibidores , Fosfoglicerato Desidrogenase/genética , Fosfoglicerato Desidrogenase/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , S-Adenosilmetionina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , ATPases Vacuolares Próton-Translocadoras/genética , Vírus da Estomatite Vesicular Indiana/fisiologia
7.
Mol Immunol ; 135: 28-35, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33857816

RESUMO

Type I interferon (IFN-I) plays pivotal roles in defense against viral infection. HSV-1 has evolved multiple strategies to evade IFN-I antiviral response. In this study, we revealed a new mechanism that HSV-1-encoded ICP0 regulates the host deubiquitinase BRCC36 to inhibit IFN-I antiviral response. We found that HSV-1 infection rapidly downregulates BRCC36 proteins at the early stage of infection. Further studies demonstrated that HSV-1-encoded ICP0 induces K48-linked polyubiquitination and degradation of BRCC36. Importantly, HSV-1-induced BRCC36 degradation promotes downmodulation of IFN-I receptor IFNAR1, thus restricting host IFN-I antiviral response to facilitate HSV-1 early infection. These findings uncover a novel immune evasion mechanism exploited by HSV-1 and could provide potential strategies for anti-HSV-1 therapy.


Assuntos
Enzimas Desubiquitinantes/metabolismo , Herpesvirus Humano 1/imunologia , Proteínas Imediatamente Precoces/metabolismo , Evasão da Resposta Imune/imunologia , Interferon Tipo I/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação para Baixo , Células HEK293 , Células HeLa , Células Hep G2 , Herpes Simples/imunologia , Herpes Simples/terapia , Humanos , Interferon Tipo I/imunologia , Camundongos , Células RAW 264.7 , Receptor de Interferon alfa e beta/metabolismo , Ubiquitinação/fisiologia , Células Vero
8.
Eur J Immunol ; 51(2): 296-310, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32673428

RESUMO

Viral infection is a serious threat to both normal population and clinical patients. STAT1 plays central roles in host defense against viral infection. How STAT1 protein maintains stable in different conditions remains largely unknown. Here, we identified BRCC36 as a potent regulator of STAT1 protein stability. Mechanistically, BRCC36 maintains STAT1 levels by utilizing USP13 to form a balanced complex for antagonizing Smurf1-mediated degradation. Importantly, cellular BRCC36 deficiency results in rapid downregulation of STAT1 during viral infection, whereas a supplement of BRCC36 maintains STAT1 protein levels and host antiviral immunity in vivo. Moreover, we revealed that BRCC36 expression was downregulated in allogeneic HSC transplantation (allo-HSCT) mice that showed increased susceptibility to viral infection. Supplementing BRCC36 enhanced antiviral response of allo-HSCT mice by maintaining STAT1 stability. This study uncovers a critical role of BRCC36 in STAT1 protein stability and could provide potential strategies for enhancing clinical antiviral therapy.


Assuntos
Antivirais/metabolismo , Enzimas Desubiquitinantes/metabolismo , Fator de Transcrição STAT1/metabolismo , Viroses/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Feminino , Células HCT116 , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
9.
Mol Cell ; 77(4): 734-747.e7, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31812350

RESUMO

Mutation and prevalence of pathogenic viruses prompt the development of broad-spectrum antiviral strategies. Viperin is a potent antiviral protein that inhibits a broad range of viruses. Unexpectedly, we found that Viperin protein production in epithelium is defective in response to both viruses and interferons (IFNs). We further revealed that viruses and IFNs stimulate expression of the acetyltransferase HAT1, which induces Lys197-acetylation on Viperin. Viperin acetylation in turn recruits UBE4A that stimulates K6-linked polyubiquitination at Lys206 of Viperin, leading to Viperin protein degradation. Importantly, UBE4A deficiency restores Viperin protein production in epithelium. We then designed interfering peptides (IPs) to inhibit UBE4A binding with Viperin. We found that VIP-IP3 rescues Viperin protein production in epithelium and therefore enhances cellular antiviral activity. VIP-IP3 renders mice more resistant to viral infection. These findings could provide strategies for both enhancing host broad-spectrum antiviral response and improving the efficacy of IFN-based antiviral therapy.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/virologia , Proteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Acetilação , Animais , Linhagem Celular , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Humanos , Interferons/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Peptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Ubiquitinação
10.
Nat Microbiol ; 4(11): 1872-1884, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30988430

RESUMO

Outbreaks of viral infections are a global health burden. Although type I interferon (IFN-I) exerts broad-spectrum antiviral effects, its antiviral efficacy in host cells is largely restricted by viruses. How the antiviral efficacy of IFN-I can be improved remains to be explored. Here, we identified the ADP-ribosyltransferase poly(ADP-ribose) polymerase family member 11 (PARP11) as a potent regulator of IFN-I antiviral efficacy. PARP11 does not restrict IFN-I production induced by vesicular stomatitis virus or Sendai virus but inhibits the strength of IFN-I-activated signalling. Mechanistically, PARP11 mono-ADP-ribosylates the ubiquitin E3 ligase ß-transducin repeat-containing protein (ß-TrCP). Mono-ADP-ribosylation of ß-TrCP promotes IFNα/ß receptor subunit 1 (IFNAR1) ubiquitination and degradation. Moreover, PARP11 expression is upregulated by virus infections, including vesicular stomatitis virus, herpes simplex virus-1 and influenza A virus, thus promoting ADP-ribosylation-mediated viral evasion. We further highlight the potential for repurposing clinical ADP-ribosylation inhibitors. We found that rucaparib can target PARP11 to stabilize IFNAR1 and therefore exhibits efficient enhancement of IFN-I signalling and the host antiviral response. Consequently, rucaparib renders mice more resistant to viral infection. Our study updates the understanding of how ß-TrCP regulates its substrates and may provide a druggable target for improving IFN antiviral efficacy.


Assuntos
Interferon Tipo I/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/metabolismo , Viroses/imunologia , Proteínas Contendo Repetições de beta-Transducina/metabolismo , ADP-Ribosilação , Animais , Células Cultivadas , Chlorocebus aethiops , Modelos Animais de Doenças , Células HEK293 , Células Hep G2 , Humanos , Indóis/administração & dosagem , Indóis/farmacologia , Camundongos , Proteólise , Vírus Sendai/imunologia , Transdução de Sinais , Ubiquitinação , Células Vero , Vesiculovirus/imunologia , Viroses/tratamento farmacológico , Viroses/metabolismo
11.
Antiviral Res ; 144: 120-129, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28583475

RESUMO

Type-I interferons (IFN-I) are important antiviral drugs which are widely used in clinical therapy of diverse viral infections. However, understanding the detailed mechanisms for IFN-I antiviral signaling remains a major challenge, and may provide novel targets for IFN-based antiviral therapy. So far, the roles of deubiquitinases (DUBs) in regulating IFN-I antiviral activity are still largely unexplored. Here, we find that Ubiquitin C-terminal hydrolase-L3 (UCHL3) plays an important role in regulating type I-interferon (IFN-I) mediated antiviral response. Interestingly, we find that UCHL3 regulates COPS5-dependent deneddylation of Cullin1, which is an essential component of SCFß-TrCP complex and associated with SCFß-TrCP activities. Furthermore, we reveal that UCHL3 physically interacts with COPS5, and determines the level and protein stability of cellular COPS5 by deubiquitinating COPS5. We further demonstrate that UCHL3 upregulates the levels of SCFß-TrCP substrates including IFN-I receptor IFNAR1, which enhances IFN-I mediated signaling pathway and antiviral activity. These findings identify COPS5 as a novel in vivo substrate of UCHL3, and uncover the deubiquitination-deneddylation mediated regulation for IFN-I signaling and antiviral function, which may provide a novel strategy for improving IFN-based antiviral therapy.


Assuntos
Antivirais/metabolismo , Complexo do Signalossomo COP9/metabolismo , Cisteína Endopeptidases/metabolismo , Interferon Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeo Hidrolases/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Células HeLa , Humanos , Imunidade Inata , Ubiquitina Tiolesterase
12.
Exp Cell Res ; 356(1): 1-7, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28506875

RESUMO

Ubiquitin-mediated proteolysis regulates cellular levels of various proteins, and therefore plays important roles in controlling cell signaling and disease progression. The Skp1-Cul1-F-box ubiquitin ligase ß-TrCP is recognized as an important negative regulator for numerous key signaling proteins. Recently, the deubiquitinases (DUBs) have turned out to be essential to regulate signaling pathways related to human diseases. However, whether ß-TrCP is able to regulate the deubiquitinase family members remains largely unexplored. Here, we found that ß-TrCP downregulated cellular levels of endogenous USP33. We also revealed that ß-TrCP interacted with USP33 independently of the classic binding motif for ß-TrCP, and mediated USP33 degradation via the ubiquitin proteasome pathway. Furthermore, we found that the WD40 motif of ß-TrCP and 201-400 amino acid motif of USP33 are required for the interaction between ß-TrCP and USP33. Consequently, ß-TrCP attenuated USP33-mediated inhibition of cell proliferation and cell invasion. Taken together, our study clarified that the E3 ligase ß-TrCP regulates cellular USP33 levels by the ubiquitin-proteasomal proteolysis.


Assuntos
Regulação da Expressão Gênica/genética , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação/fisiologia , Repetições WD40/genética , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Proteólise , Transdução de Sinais/genética , Ubiquitina/metabolismo , Ubiquitina Tiolesterase/genética , Proteínas Contendo Repetições de beta-Transducina/genética
13.
J Biol Chem ; 291(48): 24974-24985, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27729454

RESUMO

Adenosine deaminase acting on RNA 1 (ADAR1) catalyzes RNA editing of cellular and viral RNAs. Besides RNA editing, ADAR1 has recently been shown to play important roles in maintaining the body balance, including tissue homoeostasis, organ development, and autoimmune regulations, by inhibiting both IFN production and subsequent IFN-activated pathways. Accordingly, the question was raised how IFN signaling induced by viral infections overcomes the inhibitory effect of constitutively expressed ADAR1 (ADAR1-P110) to execute efficient antiviral activity. Here we unexpectedly found that IFN signaling promoted Lys48-linked ubiquitination and degradation of ADAR1-P110. Furthermore, we identified the E3 ligase ß transducin repeat-containing protein responsible for IFN-mediated ADAR1-P110 down-regulation. IFN signaling promoted the interaction between ß transducin repeat-containing protein and ADAR1-P110 as well as protein turnover of ADAR1-P110. Moreover, we found that both lysine 574 and 576 are essential for ADAR1-P110 ubiquitination. Critically, we demonstrated that down-regulation of ADAR1-P110 is required for IFN signaling to execute efficient antiviral activity during viral infections. These findings renew the understanding of the mechanisms by which IFN signaling acts to achieve antiviral functions and may provide potential targets for IFN-based antiviral therapy.


Assuntos
Adenosina Desaminase/metabolismo , Interferon Tipo I/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Adenosina Desaminase/genética , Linhagem Celular , Humanos , Interferon Tipo I/genética , Proteínas de Ligação a RNA/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligases/genética
14.
Biochem Biophys Res Commun ; 474(3): 491-496, 2016 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-27133717

RESUMO

Ubiquitin-specific peptidase 5 (USP5) has been demonstrated to be critical for the production of Tumor Necrosis Factor-alpha (TNF-α), a pivotal mediator for inflammatory responses. Besides, USP5 regulates p53 activation and DNA repair. However, the mechanism underlying the regulation of USP5, especially its responsible E3 ligase is still unclear. Here we found that Smad ubiquitination regulatory factor 1 (Smurf1) down regulated protein expression of USP5, and the E3 enzyme activity of Smurf1 was required for this function. We also revealed that Smurf1 interacted with USP5 and mediated its degradation via the ubiquitin proteasome pathway. Consequently, Smurf1 inhibited the production of TNF-α through down-regulation of USP5. Taken together, our study for the first time clarified that the E3 ligase Smurf1 regulates USP5 protein stability and USP5-mediated TNF-α production through the ubiquitin proteasome pathway.


Assuntos
Endopeptidases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Regulação para Baixo/fisiologia , Estabilidade Enzimática , Células HEK293 , Células HeLa , Humanos
15.
BMC Mol Biol ; 13: 5, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22333459

RESUMO

BACKGROUND: The human papillomavirus (HPV) E2 protein is a multifunctional DNA-binding protein. The transcriptional activity of HPV E2 is mediated by binding to its specific binding sites in the upstream regulatory region of the HPV genomes. Previously we reported a HPV-2 variant from a verrucae vulgaris patient with huge extensive clustered cutaneous, which have five point mutations in its E2 ORF, L118S, S235P, Y287H, S293R and A338V. Under the control of HPV-2 LCR, co-expression of the mutated HPV E2 induced an increased activity on the viral early promoter. In the present study, a series of mammalian expression plasmids encoding E2 proteins with one to five amino acid (aa) substitutions for these mutations were constructed and transfected into HeLa, C33A and SiHa cells. RESULTS: CAT expression assays indicated that the enhanced promoter activity was due to the co-expressions of the E2 constructs containing A338V mutation within the DNA-binding domain. Western blots analysis demonstrated that the transiently transfected E2 expressing plasmids, regardless of prototype or the A338V mutant, were continuously expressed in the cells. To study the effect of E2 mutations on its DNA-binding activity, a serial of recombinant E2 proteins with various lengths were expressed and purified. Electrophoresis mobility shift assays (EMSA) showed that the binding affinity of E2 protein with A338V mutation to both an artificial probe with two E2 binding sites or HPV-2 and HPV-16 promoter-proximal LCR sequences were significantly stronger than that of the HPV-2 prototype E2. Furthermore, co-expression of the construct containing A338V mutant exhibited increased activities on heterologous HPV-16 early promoter P97 than that of prototype E2. CONCLUSIONS: These results suggest that the mutation from Ala to Val at aa 338 is critical for E2 DNA-binding and its transcriptional regulation.


Assuntos
Alphapapillomavirus/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Mutação Puntual , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Linhagem Celular , DNA/química , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Papillomavirus Humano 16/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Oncogênicas Virais/genética , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Transcrição Gênica
16.
Bing Du Xue Bao ; 26(3): 223-7, 2010 May.
Artigo em Chinês | MEDLINE | ID: mdl-20572344

RESUMO

HPV-2 is a very common type of HPV which causes common warts. The E2 protein of virus can repress the activity of the viral early promoter through binding to the specific binding sites in viral LCR. Previously we reported that the repression of a mutated E2 protein of HPV-2 isolated from a patient with huge common wart on the viral early promoter was obviously decreased, and A338V mutation located at the C terminal DNA binding region of E2 protein. In this study, we expressed and purified the recombinant mutated and prototype E2 fusion proteins, both in the contexts of the C terminal and the full length, by prokaryotic expression system. The electrophoretic mobility shift assay showed E2 protein could bind to double-stranded DNA oligos labeled with biotin that covered two E2 binding sites. The DNA binding abilities of both C terminal and full-length mutated E2 proteins were stronger than the prototype analogs. This result indicates that the enhancement of the mutated E2 DNA binding ability may be the molecular mechanism for its impact on the activity of viral promoter, which correlates with the phenotype of extensive common wart.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Mutação , Papillomaviridae , Proteínas Virais/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/isolamento & purificação , Eletroforese , Vetores Genéticos/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética , Proteínas Virais/isolamento & purificação
17.
Parasitol Res ; 106(6): 1315-20, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20306208

RESUMO

Human Blastocystis hominis were isolated from diarrhea patients' feces and cultured in vitro. Then the cultures were inoculated intraperitoneally to laboratory mice. The B. hominis in living mice were collected and inoculated again to healthy mice. The B. hominis showed dose-dependent pathogenicity in the primary inoculation. No pathogenicity was observed in the secondary inoculation. The protozoan existed in the living mice abdomen cavity for more than 6 months and the cyst was the only form. These results showed that encystation enable the parasite to avoid the immune attack in competent host and simultaneously decrease the pathogenicity to host. Intraperitoneal inoculation to laboratory mice is a good method to maintain and propagate B. hominis. This is also a good model to study the interaction of B. hominis and immune system.


Assuntos
Cavidade Abdominal/parasitologia , Blastocystis hominis/patogenicidade , Animais , Infecções por Blastocystis/parasitologia , Blastocystis hominis/imunologia , Blastocystis hominis/isolamento & purificação , Modelos Animais de Doenças , Fezes/parasitologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Análise de Sobrevida
18.
Biomed Environ Sci ; 22(1): 55-61, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19462689

RESUMO

OBJECTIVE: To study the circulation, distribution, and genomic diversity of HPVs in common warts in Beijing area of China. METHODS: Forty eight patients with pathologically diagnosed common warts were screened for the presence of HPV with HPV type-specific PCR and direct sequencing analysis. The genomic diversity of HPVs prevalent in Chinese patients was analyzed based on LCR. RESULTS: Forty one (85.5%) samples were positive for HPV DNA, 13 (31.7%)--HPV-57, 12 (29.3%)--HPV-1a, 7 (17%)--HPV-27 and 5(12.2%)--HPV-2a. Four cases were infected with two different HPV types, two (4.9%) with HPV-1a and HPV-27, one (2.4%) with HPV-1 and HPV-57 and one (2.4%) with HPV-27 and HPV-57. In contrast to the prevalence of single strain of novel HPV-57 variant and HPV-1 prototype, two HPV-2 and three HPV-27 novel variants were found to circulate in Beijing. CONCLUSION: HPV-1, -2, -27 and -57 are predominantly prevalent in patients with common warts in Beijing.


Assuntos
Papillomaviridae/isolamento & purificação , Verrugas/epidemiologia , Adolescente , Adulto , Idoso , China/epidemiologia , DNA Viral , Feminino , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Papillomaviridae/classificação , Papillomaviridae/genética , Filogenia , Prevalência , Verrugas/virologia
19.
Brain Res Bull ; 77(5): 233-6, 2008 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-18824078

RESUMO

The pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) is associated with the generation of inflammatory and neuropathic pain. The current study aims to investigate the expression of TNF-α in the brain of rats with spared nerve injury (SNI), a neuropathic pain model with the lesion of common peroneal and tibial nerves. Two weeks following SNI, the immunohistochemical results identified that the expression level of TNF-α in the Red nucleus (RN) of SNI rats was apparently higher than that of sham-operated rats. To further study the roles of TNF-α in the development of neuropathic pain, different doses of anti-TNF-α antibody (20, 2.0 and 0.2 µg/ml) were microinjected into the RN contralateral to the nerve injury side of SNI rats. The results showed that the 50% paw withdrawal threshold (von Frey test) of SNI rats were increased by 20 and 2.0 µg/ml anti-TNF-α antibody as compared with that of the basic value and control groups (P<0.05), the analgesic effect lasted for 50 and 30 min, respectively. However, no significant analgesic effect was observed after 0.2 µg/ml antibody was microinjected into the RN. These results suggest that the TNF-α of RN is involved in the development of neuropathic allodynia in SNI rats.


Assuntos
Hiperalgesia/fisiopatologia , Neuralgia/fisiopatologia , Nervos Periféricos/fisiopatologia , Núcleo Rubro/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Animais , Anticorpos/imunologia , Modelos Animais de Doenças , Masculino , Neuralgia/patologia , Nervos Periféricos/patologia , Ratos , Ratos Sprague-Dawley , Núcleo Rubro/patologia
20.
Bing Du Xue Bao ; 24(4): 268-71, 2008 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-18780628

RESUMO

Common warts are close associated with HPVs infection. In this study, we amplified and sequenced the LCR fragment and E2 gene of HPV-2 that infected the patient of extensive common wart with cutaneous horns, and we constructed the recombinant CAT-reporter plasmids pBLCAT-LCR containing HPV-2 prototype or variant LCR and mammalian expression plasmids pcDNA3. 1-E2 containing prototype or variant E2 ORF individually. The promoter activities of HPV-2 variant and the transcriptional repression activities of the mutated E2 protein were evaluated by transient transfection into HeLa cells. The results showed that there were several mutations in LCR and E2 gene of HPV-2 variant. Compared with the prototype, the viral early promoter activity of variant was significantly increased uder the control of LCR. Compared with the wild type E2 protein, the transcriptional repression activities of the mutated E2 protein was abolished partially. We speculate herein that increased promoter activities and decreased repression effect of the mutated E2 protein are linked, at least partially, with the clinical phenotypes of the uncommon huge common wart.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas Oncogênicas Virais/fisiologia , Papillomaviridae/genética , Proteínas Repressoras/fisiologia , Verrugas/virologia , Proteínas de Ligação a DNA/genética , Humanos , Mutação , Proteínas Oncogênicas Virais/genética , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA