Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Infect Immun ; 87(10)2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31383744

RESUMO

Reproductive tract pathology caused by Chlamydia trachomatis infection is an important global cause of human infertility. To better understand the mechanisms associated with Chlamydia-induced genital tract pathogenesis in humans, we used CRISPR genome editing to disrupt Toll-like receptor 3 (TLR3) function in the human oviduct epithelial (hOE) cell line OE-E6/E7 in order to investigate the possible role(s) of TLR3 signaling in the immune response to Chlamydia Disruption of TLR3 function in these cells significantly diminished the Chlamydia-induced synthesis of several inflammation biomarkers, including interferon beta (IFN-ß), interleukin-6 (IL-6), interleukin-6 receptor alpha (IL-6Rα), soluble interleukin-6 receptor beta (sIL-6Rß, or gp130), IL-8, IL-20, IL-26, IL-34, soluble tumor necrosis factor receptor 1 (sTNF-R1), tumor necrosis factor ligand superfamily member 13B (TNFSF13B), matrix metalloproteinase 1 (MMP-1), MMP-2, and MMP-3. In contrast, the Chlamydia-induced synthesis of CCL5, IL-29 (IFN-λ1), and IL-28A (IFN-λ2) was significantly increased in TLR3-deficient hOE cells compared to their wild-type counterparts. Our results indicate a role for TLR3 signaling in limiting the genital tract fibrosis, scarring, and chronic inflammation often associated with human chlamydial disease. Interestingly, we saw that Chlamydia infection induced the production of biomarkers associated with persistence, tumor metastasis, and autoimmunity, such as soluble CD163 (sCD163), chitinase-3-like protein 1, osteopontin, and pentraxin-3, in hOE cells; however, their expression levels were significantly dysregulated in TLR3-deficient hOE cells. Finally, we demonstrate using hOE cells that TLR3 deficiency resulted in an increased amount of chlamydial lipopolysaccharide (LPS) within Chlamydia inclusions, which is suggestive that TLR3 deficiency leads to enhanced chlamydial replication and possibly increased genital tract pathogenesis during human infection.


Assuntos
Chlamydia trachomatis/imunologia , Células Epiteliais/microbiologia , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/imunologia , Receptor 3 Toll-Like/imunologia , Fator Ativador de Células B/genética , Fator Ativador de Células B/imunologia , Linhagem Celular Transformada , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/patogenicidade , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/imunologia , Células Epiteliais/imunologia , Tubas Uterinas/imunologia , Tubas Uterinas/microbiologia , Feminino , Deleção de Genes , Células HeLa , Interações Hospedeiro-Patógeno/genética , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucinas/genética , Interleucinas/imunologia , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/imunologia , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética
2.
PLoS One ; 14(1): e0207422, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30625140

RESUMO

PROBLEM: Chlamydia trachomatis infections are often associated with acute syndromes including cervicitis, urethritis, and endometritis, which can lead to chronic sequelae such as pelvic inflammatory disease (PID), chronic pelvic pain, ectopic pregnancy, and tubal infertility. As epithelial cells are the primary cell type productively infected during genital tract Chlamydia infections, we investigated whether Chlamydia has any impact on the integrity of the host epithelial barrier as a possible mechanism to facilitate the dissemination of infection, and examined whether TLR3 function modulates its impact. METHOD OF STUDY: We used wild-type and TLR3-deficient murine oviduct epithelial (OE) cells to ascertain whether C. muridarum infection had any effect on the epithelial barrier integrity of these cells as measured by transepithelial resistance (TER) and cell permeability assays. We next assessed whether infection impacted the transcription and protein function of the cellular tight-junction (TJ) genes for claudins1-4, ZO-1, JAM1 and occludin via quantitative real-time PCR (qPCR) and western blot. RESULTS: qPCR, immunoblotting, transwell permeability assays, and TER studies show that Chlamydia compromises cellular TJ function throughout infection in murine OE cells and that TLR3 deficiency significantly exacerbates this effect. CONCLUSION: Our data show that TLR3 plays a role in modulating epithelial barrier function during Chlamydia infection of epithelial cells lining the genital tract. These findings propose a role for TLR3 signaling in maintaining the integrity of epithelial barrier function during genital tract Chlamydia infection, a function that we hypothesize is important in helping limit the chlamydial spread and subsequent genital tract pathology.


Assuntos
Infecções por Chlamydia/microbiologia , Chlamydia muridarum/fisiologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Oviductos/microbiologia , Oviductos/patologia , Infecções do Sistema Genital/microbiologia , Receptor 3 Toll-Like/deficiência , Animais , Permeabilidade da Membrana Celular , Infecções por Chlamydia/genética , Infecções por Chlamydia/patologia , Impedância Elétrica , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções do Sistema Genital/genética , Infecções do Sistema Genital/patologia , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/genética , Receptor 3 Toll-Like/metabolismo , Transcrição Gênica
3.
Methods Mol Biol ; 1855: 81-85, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30426408

RESUMO

The electrophoresis on cellulose acetate membrane is most widely used because of its simplicity, and is without the use of any sophisticated instrument other than electrophoresis apparatus and the cellulose acetate strip. Here we describe a modified version of cellulose acetate membrane electrophoresis for hemoglobin separation from blood sample. Sharp, clear bands without tailing effects can be obtained with this method. The method and apparatus described here would be appropriate to separate protein fractions under 1 h at voltages up to 60 V/cm measured between the electrodes.


Assuntos
Eletroforese em Acetato de Celulose/métodos , Hemoglobinas/isolamento & purificação , Eletroforese em Acetato de Celulose/instrumentação , Humanos , Peso Molecular
4.
Int J Microbiol Curr Res ; 1(1): 1-13, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31891165

RESUMO

Chlamydia trachomatis replicates primarily in the epithelial cells lining the genital tract and induces the innate immune response by triggering cellular pathogen recognition receptors (PRRs). Our previous studies showed that Toll-like receptor 3 (TLR3) is expressed in murine oviduct epithelial (OE) cells, is the primary PRR triggered by C. muridarum (Cm) early during infection to induce IFN-ß synthesis, and that TLR3 signaling regulates the chlamydial induced synthesis of a plethora of other innate inflammatory modulators including IL-6, CXCL10, CXCL16 and CCL5. We also showed that the expression of these cytokines induced by Chlamydia was severely diminished during TLR3 deficiency; however, the replication of Chlamydiain TLR3 deficient OE cells was more robust than in WT cells. These data suggested that TLR3 had a biological impact on the inflammatory response to Chlamydia infection; however, the global effects of TLR3 signaling in the cellular response to Chlamydia infection in murine OE cells has not yet been investigated. To determine the impact of TLR3 signaling on Chlamydia infection in OE cell at the transcriptome level, we infected wild-type (OE-WT) and TLR3-deficient (OE-TLR3KO) cells with Cm, and performed transcriptome analyses using microarray. Genome-wide expression and ingenuity pathway analysis (IPA) identified enhanced expression of host genes encoding for components found in multiple cellular processes encompassing: (1) pro-inflammatory, (2) cell adhesion, (3) chemoattraction, (4) cellular matrix and small molecule transport, (5) apoptosis, and (6) antigen-processing and presentation. These results support a role for TLR3 in modulating the host cellular responses to Cm infection that extend beyond inflammation and fibrosis, and shows that TLR3 could serve a potential therapeutic target for drug and/or vaccine development.

5.
PLoS One ; 13(4): e0195165, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29624589

RESUMO

Chlamydia trachomatis urogenital serovars primarily replicate in epithelial cells lining the reproductive tract. Epithelial cells recognize Chlamydia through cell surface and cytosolic receptors, and/or endosomal innate receptors such as Toll-like receptors (TLRs). Activation of these receptors triggers both innate and adaptive immune mechanisms that are required for chlamydial clearance, but are also responsible for the immunopathology in the reproductive tract. We previously demonstrated that Chlamydia muridarum (Cm) induces IFN-ß in oviduct epithelial cells (OE) in a TLR3-dependent manner, and that the synthesis of several cytokines and chemokines are diminished in Cm-challenged OE derived from TLR3-/- 129S1 mice. Furthermore, our in vitro studies showed that Cm replication in TLR3-/- OE is more efficient than in wild-type OE. Because TLR3 modulates the release inflammatory mediators involved in host defense during Cm infection, we hypothesized that TLR3 plays a protective role against Cm-induced genital tract pathology in congenic C57BL/6N mice. Using the Cm mouse model for human Chlamydia genital tract infections, we demonstrated that TLR3-/- mice had increased Cm shedding during early and mid-stage genital infection. In early stage infection, TLR3-/- mice showed a diminished synthesis of IFN-ß, IL-1ß, and IL-6, but enhanced production of IL-10, TNF-α, and IFN-γ. In mid-stage infection, TLR3-/- mice exhibited significantly enhanced lymphocytic endometritis and salpingitis than wild-type mice. These lymphocytes were predominantly scattered along the endometrial stroma and the associated smooth muscle, and the lamina propria supporting the oviducts. Surprisingly, our data show that CD4+ T-cells are significantly enhanced in the genital tract TLR3-/- mice during mid-stage Chlamydial infection. In late-stage infections, both mouse strains developed hydrosalpinx; however, the extent of hydrosalpinx was more severe in TLR3-/- mice. Together, these data suggest that TLR3 promotes the clearance of Cm during early and mid-stages of genital tract infection, and that loss of TLR3 is detrimental in the development hydrosalpinx.


Assuntos
Infecções por Chlamydia/genética , Infecções por Chlamydia/microbiologia , Chlamydia muridarum , Suscetibilidade a Doenças , Infecções do Sistema Genital/genética , Infecções do Sistema Genital/microbiologia , Receptor 3 Toll-Like/genética , Animais , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/patologia , Citocinas/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções do Sistema Genital/metabolismo , Infecções do Sistema Genital/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor 3 Toll-Like/metabolismo
6.
PLoS One ; 10(3): e0119235, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25798928

RESUMO

We previously reported that the IFN-ß secreted by Chlamydia muridarum-infected murine oviduct epithelial cells (OE cells) was mostly dependent on the TLR3 signaling pathway. To further characterize the mechanisms of IFN-ß synthesis during Chlamydia infection of OE cells in vitro, we utilized specific inhibitory drugs to clarify the roles of IRF3 and NF-κB on both early- and late-phase C. muridarum infections. Our results showed that the pathways involved in the early-phase of IFN-ß production were distinct from that in the late-phase of IFN-ß production. Disruption of IRF3 activation using an inhibitor of TBK-1 at early-phase Chlamydia infection had a significant impact on the overall synthesis of IFN-ß; however, disruption of IRF3 activation at late times during infection had no effect. Interestingly, inhibition of NF-κB early during Chlamydia infection also had a negative effect on IFN-ß production; however, its impact was not significant. Our data show that the transcription factor IRF7 was induced late during Chlamydia infection, which is indicative of a positive feedback mechanism of IFN-ß synthesis late during infection. In contrast, IRF7 appears to play little or no role in the early synthesis of IFN-ß during Chlamydia infection. Finally, we demonstrate that antibiotics that target chlamydial DNA replication are much more effective at reducing IFN-ß synthesis during infection versus antibiotics that target chlamydial transcription. These results provide evidence that early- and late-phase IFN-ß production have distinct signaling pathways in Chlamydia-infected OE cells, and suggest that Chlamydia DNA replication might provide a link to the currently unknown chlamydial PAMP for TLR3.


Assuntos
Chlamydia muridarum/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Interferon beta/biossíntese , Oviductos/citologia , Transdução de Sinais , Animais , Linhagem Celular , Chlamydia muridarum/efeitos dos fármacos , Chlamydia muridarum/genética , Replicação do DNA/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/metabolismo , NF-kappa B/metabolismo , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tiofenos/farmacologia , Fatores de Tempo , Receptor 3 Toll-Like/metabolismo
7.
Infect Immun ; 80(1): 254-65, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22006569

RESUMO

Because epithelial cells are the major cell type productively infected with Chlamydia during genital tract infections, the overall goal of our research was to understand the contribution of infected epithelial cells to the host defense. We previously showed that Toll-like receptor 3 (TLR3) is the critical pattern recognition receptor in oviduct epithelial (OE) cells that is stimulated during Chlamydia infection, resulting in the synthesis of beta interferon (IFN-ß). Here, we present data that implicates TLR3 in the expression of a multitude of other innate-inflammatory immune modulators including interleukin-6 (IL-6), CXCL10, CXCL16, and CCL5. We demonstrate that Chlamydia-induced expression of these cytokines is severely disrupted in TLR3-deficient OE cells, whereas Chlamydia replication in the TLR3-deficient cells is more efficient than in wild-type OE cells. Pretreatment of the TLR3-deficient OE cells with 50 U of IFN-ß/ml prior to infection diminished Chlamydia replication and restored the ability of Chlamydia infection to induce IL-6, CXCL10, and CCL5 expression in TLR3-deficient OE cells; however, CXCL16 induction was not restored by IFN-ß preincubation. Our findings were corroborated in pathway-focused PCR arrays, which demonstrated a multitude of different inflammatory genes that were defectively regulated during Chlamydia infection of the TLR3-deficient OE cells, and we found that some of these genes were induced only when IFN-ß was added prior to infection. Our OE cell data implicate TLR3 as an essential inducer of IFN-ß and other inflammatory mediators by epithelial cells during Chlamydia infection and highlight the contribution of TLR3 to the inflammatory cytokine response.


Assuntos
Chlamydia muridarum/imunologia , Citocinas/metabolismo , Células Epiteliais/imunologia , Imunidade Inata , Oviductos/imunologia , Receptor 3 Toll-Like/imunologia , Animais , Células Cultivadas , Infecções por Chlamydia/imunologia , Feminino , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 3 Toll-Like/deficiência
8.
J Immunol ; 185(11): 6689-97, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20974982

RESUMO

Epithelial cells lining the murine genital tract act as sentinels for microbial infection, play a major role in the initiation of the early inflammatory response, and can secrete factors that modulate the adaptive immune response when infected with Chlamydia. C. muridarum-infected murine oviduct epithelial cells secrete the inflammatory cytokines IL-6 and GM-CSF in a TLR2-dependent manner. Further, C. muridarum infection induces IFN-ß synthesis in the oviduct epithelial cells in a TRIF-dependent manner. Because murine oviduct epithelial cells express TLR3 but not TLRs 4, 7, 8, or 9, we hypothesized that TLR3 or an unknown TRIF-dependent pattern recognition receptor was the critical receptor for IFN-ß production. To investigate the role of TLR3 in the Chlamydia-induced IFN-ß response in oviduct epithelial cells, we used small interfering RNA, dominant-negative TLR3 mutants, and TLR3-deficient oviduct epithelial cells to show that the IFN-ß secreted during C. muridarum infection requires a functional TLR3. Interestingly, we demonstrate that the TLR3 signaling pathway is not required for IFN-ß synthesis in C. muridarum-infected macrophages, suggesting that there are alternate and redundant pathways to Chlamydia-induced IFN-ß synthesis that seem to be dependent upon the cell type infected. Finally, because there is no obvious dsRNA molecule associated with Chlamydia infection, the requirement for TLR3 in Chlamydia-induced IFN-ß synthesis in infected oviduct epithelial cells implicates a novel ligand that binds to and signals through TLR3.


Assuntos
Chlamydia muridarum/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Interferon Tipo I/biossíntese , Oviductos/imunologia , Oviductos/microbiologia , Receptor 3 Toll-Like/fisiologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/microbiologia , Linhagem Celular , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/patologia , Células Clonais , Células Epiteliais/metabolismo , Feminino , Interferon Tipo I/metabolismo , Ligantes , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oviductos/citologia , Oviductos/metabolismo , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/metabolismo
9.
Infect Immun ; 75(3): 1280-90, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17178782

RESUMO

Chlamydia trachomatis is the most common sexually transmitted bacterial infection in the United States. Utilizing cloned murine oviduct epithelial cell lines, we previously identified Toll-like receptor 2 (TLR2) as the principal epithelial pattern recognition receptor (PRR) for infection-triggered release of the acute inflammatory cytokines interleukin-6 and granulocyte-macrophage colony-stimulating factor. The infected oviduct epithelial cell lines also secreted the immunomodulatory cytokine beta interferon (IFN-beta) in a largely MyD88-independent manner. Although TLR3 was the only IFN-beta production-capable TLR expressed by the oviduct cell lines, we were not able to determine whether TLR3 was responsible for IFN-beta production because the epithelial cells were unresponsive to the TLR3 ligand poly(I-C), and small interfering RNA (siRNA) techniques were ineffective at knocking down TLR3 expression. To further investigate the potential role of TLR3 in the infected epithelial cell secretion of IFN-beta, we examined the roles of its downstream signaling molecules TRIF and IFN regulatory factor 3 (IRF-3) using a dominant-negative TRIF molecule and siRNA specific for TRIF and IRF-3. Antagonism of either IRF-3 or TRIF signaling significantly decreased IFN-beta production. These data implicate TLR3, or an unknown PRR utilizing TRIF, as the source of IFN-beta production by Chlamydia-infected oviduct epithelial cells.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Infecções por Chlamydia/metabolismo , Chlamydia muridarum/fisiologia , Células Epiteliais/metabolismo , Fator Regulador 3 de Interferon/fisiologia , Interferon beta/biossíntese , Animais , Linhagem Celular , Células Epiteliais/microbiologia , Tubas Uterinas/citologia , Tubas Uterinas/metabolismo , Tubas Uterinas/microbiologia , Feminino , Interferon beta/fisiologia , Camundongos
10.
J Immunol ; 175(9): 6065-75, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16237102

RESUMO

Chlamydia trachomatis is the most common bacterial sexually transmitted disease in the United States and a major cause of female infertility due to infection-induced Fallopian tube scarring. Epithelial cells are likely central to host defense and pathophysiology as they are the principal cell type productively infected by C. trachomatis. We generated cloned murine oviduct epithelial cell lines without viral or chemical transformation to investigate the role of the TLRs and cytosolic nucleotide binding site/leucine-rich repeat proteins Nod1 and Nod2 in epithelial responses to Chlamydia muridarum infection. RT-PCR assays detected mRNA for TLR2 (TLRs 1 and 6), TLR3, and TLR5. No mRNA was detected for TLRs 4, 7, 8, and 9. Messenger RNAs for Nod1 and Nod2 were present in the epithelial cell lines. Oviduct epithelial cell lines infected with C. muridarum or exposed to the TLR2 agonist peptidoglycan secreted representative acute phase cytokines IL-6 and GM-CSF in a MyD88-dependent fashion. Infected epithelial cell lines secreted the immunomodulatory cytokine IFN-beta, even though C. muridarum does not have a clear pathogen-associated molecular pattern (PAMP) for triggering IFN-beta transcription. The oviduct epithelial lines did not secrete IFN-beta in response to the TLR2 agonist peptidoglycan or to the TLR3 agonist poly(I:C). Our data identify TLR2 as the principal TLR responsible for secretion of acute phase cytokines by C. muridarum-infected oviduct epithelial cell lines. The pattern recognition molecule responsible for infection-induced IFN-beta secretion by oviduct epithelial cells remains to be determined.


Assuntos
Chlamydia muridarum/patogenicidade , Tubas Uterinas/imunologia , Receptores Toll-Like/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Antígenos de Diferenciação/fisiologia , Linhagem Celular , Citocinas/biossíntese , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Tubas Uterinas/microbiologia , Feminino , Interferon beta/fisiologia , Camundongos , Fator 88 de Diferenciação Mieloide , Poli I-C/farmacologia , RNA Mensageiro/análise , Receptores Imunológicos/fisiologia , Receptor 2 Toll-Like/fisiologia , Receptor 3 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Receptores Toll-Like/genética
11.
J Virol ; 77(4): 2675-85, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12552007

RESUMO

The equine herpesvirus 1 (EHV-1) immediate-early (IE) and EICP0 proteins are potent trans-activators of EHV-1 promoters; however, in transient-transfection assays, the IE protein inhibits the trans-activation function of the EICP0 protein. Assays with IE mutant proteins revealed that its DNA-binding domain, TFIIB-binding domain, and nuclear localization signal may be important for the antagonism between the IE and EICP0 proteins. In vitro interaction assays with the purified IE and EICP0 proteins indicated that these proteins interact directly. At late times postinfection, the IE and EICP0 proteins colocalized in the nuclei of infected equine cells. Transient-transfection assays showed that the EICP0 protein trans-activated EHV-1 promoters harboring only a minimal promoter region (TATA box and cap site), suggesting that the EICP0 protein trans-activates EHV-1 promoters by interactions with general transcription factor(s). In vitro interaction assays revealed that the EICP0 protein interacted directly with the basal transcription factors TFIIB and TBP and that the EICP0 protein (amino acids [aa] 143 to 278) mediated the interaction with aa 125 to 174 of TFIIB. Our unpublished data showed that the IE protein interacts with the same domain (aa 125 to 174) of TFIIB and with TBP. Taken together, these results suggested that interaction of the EICP0 protein with the IE protein, TFIIB, and TBP may mediate the antagonism between the IE and EICP0 proteins.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Equídeo 1/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Transativadores/metabolismo , Proteínas Virais/metabolismo , Animais , Fibroblastos , Herpesvirus Equídeo 1/genética , Cavalos , Humanos , Proteínas Imediatamente Precoces/genética , Camundongos , Regiões Promotoras Genéticas , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo , Transativadores/genética , Fator de Transcrição TFIIB/genética , Fator de Transcrição TFIIB/metabolismo , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Proteínas Virais/genética
12.
Virus Res ; 84(1-2): 1-15, 2002 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-11900834

RESUMO

The early 293 amino acid EICP22 protein (EICP22P) of equine herpesvirus 1 localizes within the nucleus and functions as an accessory regulatory protein (J. Virol. 68 (1994) 4329). Transient transfection assays indicated that although the EICP22P by itself only minimally trans-activates EHV-1 promoters, the EICP22P functions synergistically with the immediate-early protein (IEP) to enhance expression of EHV-1 early genes (J. Virol. 71 (1997) 1004). We previously showed that the EICP22 protein enhances the DNA-binding activity of the EHV-1 IEP and that it also physically interacts with the IEP (J. Virol. 74 (2000) 1425). In this communication, we employed transient trans-activation assays utilizing EICP22P deletion mutants to address whether the sequences required for EICP22P-IEP physical interactions are essential for EICP22P's ability to interact synergistically with the IEP. Assays employing various classes of the EHV-1 promoters fused to the chloramphenicol acetyl-transferase (CAT) reporter gene indicated that: (1) neither full length nor any of the EICP22P mutants tested was able to overcome repression of the IE promoter elicited by the IEP, (2) the full-length EICP22P interacted synergistically with the IEP to trans-activate the early and late promoters tested, and (3) all of the EICP22P mutants, including those that were able to physically interact with IEP and itself, failed to function synergistically with the IEP to trans-activate representative EHV-1 early and late promoters. The results suggest that EICP22P sequences required for its interaction with the IE protein are not sufficient to mediate its synergistic effect on the trans-activation function of the IEP. The possible explanations as to why sequences in addition to those that mediate EICP22P-IEP interaction and EICP22P self-interactions are essential for the synergistic function of EICP22P are discussed.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Equídeo 1/genética , Proteínas Imediatamente Precoces/genética , Ativação Transcricional , Proteínas Virais , Animais , Células Cultivadas , Cavalos , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Mutagênese , Regiões Promotoras Genéticas , Proteínas Virais Reguladoras e Acessórias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA