Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arch Virol ; 157(10): 1971-4, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22711259

RESUMO

The 90 % human cytomegalovirus inhibitory concentration of 17-allylamino-17-(demethoxy)geldanamycin (17-AAG) was 0.1 nM and 50 % cytotoxicity required at least a 10 µM concentration. Three molecular targets may explain the antiviral activities of this compound. These are (1) heat shock protein maturation complexes, (2) host cell cycle progression and (3) phosphatidylinositol 3-kinase signaling. However, the data suggested a mechanism of action where 17-AAG blocked immediate-early protein transactivation.


Assuntos
Benzoquinonas/farmacologia , Citomegalovirus/efeitos dos fármacos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Lactamas Macrocíclicas/farmacologia , Replicação Viral/efeitos dos fármacos , Células Cultivadas , Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Fibroblastos/virologia , Humanos , Proteínas Imediatamente Precoces/metabolismo , Ativação Transcricional/efeitos dos fármacos
2.
J Virol ; 83(23): 12388-98, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19776115

RESUMO

Infection of host cells with human cytomegalovirus (HCMV) induces cell cycle dysregulation. Two HCMV immediate-early (IE) proteins, IE1-72 and IE2-86, are promiscuous transactivators that have been implicated in the dysregulatory events. Cellular p53 protein is accumulated to high levels in HCMV-infected cells, but the indicative marker of p53 transcriptional activity, p21, is markedly decreased. Both IE1-72 and IE2-86 were able to transactivate the p53 promoter and interact with p53 protein in DNA-transfected or HCMV-infected cells. HCMV UL84, a multiregulatory protein expressed in early periods of HCMV infection, also interacted with p53. HCMV IE1-72 prevented or disrupted p53 binding to p53-specific DNA sequences, while IE2-86 and/or UL84 enhanced p53 binding and induced supershift of this DNA-protein complex. Both HCMV IE1-72 and IE2-86 were able to inhibit p53-dependent transcriptional activation in plasmid-transfected cells. IE1-72, rather than IE2-86, was found to be responsible for p21 downregulation in HCMV-infected HEL cells. DNA transfection analysis using IE1-72 mutants revealed that exon 2/3 and the zinc finger region of IE1-72 are essential for IE1-72's effect on the repression of p53-dependent transcriptional activation. These data suggest that HCMV IE1-72 and/or IE2-86 transactivates the p53 promoter and induces p53 accumulation, but HCMV IE1-72 represses the p53 transactivation activity by a unique binding hindrance mechanism different from that of IE2-86. Thus, various modes of viral IE proteins and p53 interactions might result in multiple outcomes, such as stimulation of cellular DNA synthesis, cell cycle progression and cell cycle arrest, and prevention of program cell death.


Assuntos
Citomegalovirus/patogenicidade , Interações Hospedeiro-Patógeno , Proteínas Imediatamente Precoces/metabolismo , Transativadores/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/antagonistas & inibidores , Linhagem Celular , Imunoprecipitação da Cromatina , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Virais/metabolismo
3.
J Virol ; 80(8): 3833-43, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16571800

RESUMO

Levels of the p53 tumor suppressor protein are increased in human cytomegalovirus (HCMV)-infected cells and may be important for HCMV pathogenesis. In normal cells p53 levels are kept low due to an autoregulatory feedback loop where p53 activates the transcription of mdm2 and mdm2 binds and ubiquitinates p53, targeting p53 for proteasomal degradation. Here we report that, in contrast to uninfected cells, mdm2 was undetectable upon treatment of infected fibroblasts with the proteasome inhibitor MG132. Cellular depletion of mdm2 was reproducible in p53-null cells transfected with the HCMV IE2-86 protein, but not with IE172, independently of the endogenous mdm2 promoter. IE2-86 also prevented the emergence of presumably ubiquitinated species of p53. The regions of IE2-86 important for mdm2 depletion were those containing the sequences corresponding to the putative zinc finger and C-terminal acidic motifs. mdm2 and IE2-86 coimmunoprecipitated in transfected and infected cell lysates and in a cell-free system. IE2-86 blocked mdm2's p53-independent transactivation of the cyclin A promoter in transient-transfection experiments. Pulse-chase experiments revealed that IE2-86 but not IE1-72 or several loss-of-function IE2-86 mutants increased the half-life of p53 and reduced the half-life of mdm2. Short interfering RNA-mediated depletion of IE2-86 restored the ability of HCMV-infected cells to accumulate mdm2 in response to proteasome inhibition. Taken together, the data suggest that specific interactions between IE2-86 and mdm2 cause proteasome-independent degradation of mdm2 and that this may be important for the accumulation of p53 in HCMV-infected cells.


Assuntos
Proteínas Imediatamente Precoces/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transativadores/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Cricetinae , Ciclina A/genética , Meia-Vida , Humanos , Leupeptinas/farmacologia , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina/metabolismo
4.
Antiviral Res ; 68(3): 124-34, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16188329

RESUMO

We report antiviral activity against human cytomegalovirus for certain dietary flavonoids and their likely biochemical mechanisms of action. Nine out of ten evaluated flavonoids blocked HCMV replication at concentrations that were significantly lower than those producing cytotoxicity against growing or stationary phase host cells. Baicalein was the most potent inhibitor in this series (IC(50)=0.4-1.2 microM), including positive control ganciclovir. Baicalein and genistein were chosen as model compounds to study the antiviral mechanism(s) of action for this series. Both flavonoids significantly reduced the levels of HCMV early and late proteins, as well as viral DNA synthesis. Baicalein reduced the levels of HCMV immediate-early proteins to nearly background levels while genistein did not. The antiviral effects of genistein, but not baicalein, were fully reversible in cell culture. Pre-incubation of concentrated virus stocks with either flavonoid did not inhibit HCMV replication, suggesting that baicalein did not directly inactivate virus particles. Baicalein functionally blocked epidermal growth factor receptor tyrosine kinase activity and HCMV nuclear translocation, while genistein did not. At 24h post infection HCMV-infected cells treated with genistein continued to express immediate-early proteins and efficiently phosphorylate IE1-72. However, HCMV induction of NF-kappaB and increases in the levels of cell cycle regulatory proteins--events that are associated with immediate-early protein functioning--were absent. The data suggested that the primary mechanism of action for baicalein may be to block HCMV infection at entry while the primary mechanism of action for genistein may be to block HCMV immediate-early protein functioning.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Flavonoides/química , Flavonoides/farmacologia , Replicação Viral/efeitos dos fármacos , Técnicas de Cultura de Células , Citomegalovirus/genética , Citomegalovirus/fisiologia , Humanos , Proteínas Imediatamente Precoces/genética
5.
Nat Med ; 11(5): 515-21, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15834425

RESUMO

Human cytomegalovirus (HCMV) is a widespread opportunistic pathogen that causes birth defects in newborns and severe disease in immunocompromised individuals. The broad tropism of HCMV infection suggests that it uses multiple receptors. We recently showed that the epidermal growth factor receptor (EGFR) serves as a receptor for HCMV. Here we show that HCMV also uses integrin alphavbeta3 as a coreceptor. Upon infection, HCMV glycoproteins gB and gH independently bind to EGFR and alphavbeta3, respectively, to initiate viral entry and signaling. Alphavbeta3 then translocates to lipid rafts where it interacts with EGFR to induce coordinated signaling. The coordination between EGFR and alphavbeta3 is essential for the early events of HCMV infection, including viral entry, RhoA downregulation, stress-fiber disassembly and viral nuclear trafficking. Our findings support a model in which EGFR and alphavbeta3 work together as coreceptors for HCMV entry and signaling. This discovery is fundamental to understanding HCMV pathogenesis and developing treatment strategies targeted to viral receptors.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Integrina alfaVbeta3/metabolismo , Transdução de Sinais/fisiologia , Proteínas do Envelope Viral/metabolismo , Linhagem Celular , Primers do DNA , Receptores ErbB/metabolismo , Humanos , Microdomínios da Membrana/metabolismo , Microscopia de Fluorescência , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína rhoA de Ligação ao GTP/metabolismo
6.
J Med Chem ; 48(2): 534-46, 2005 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-15658867

RESUMO

A series of natural product analogues based on helioxanthin (2), with particular attention to modification of the lactone ring and methylenedioxy group, were synthesized and evaluated for their antiviral activities. Among them, lactam derivative 18 and helioxanthin cyclic hydrazide 28 exhibited significant in vitro antiviral activity against hepatitis B virus (EC(50) = 0.08 and 0.03 microM, respectively). Compound 18 showed the most potent antiviral activity against hepatitis C virus (55% inhibition at 1.0 microM). Compound 12, an acid-hydrolyzed product of helioxanthin cyclic imide derivative 9, was found to exhibit broad-spectrum antiviral activity against hepatitis B virus (EC(50) = 0.8 microM), herpes simplex virus type 1 (EC(50) = 0.15 microM) and type 2 (EC(50) < 0.1 microM), Epstein-Barr virus (EC(50) = 9.0 microM), and cytomegalovirus (EC(50) = 0.45 microM). Helioxanthin lactam derivative 18 also showed marked inhibition of herpes simplex virus type 1 (EC(50) = 0.29 microM) and type 2 (EC(50) = 0.16 microM). The cyclic hydrazide derivative of helioxanthin 28 and its brominated product 42 exhibited moderately potent activities against human immunodeficiency virus (EC(50) = 2.7 and 2.5 microM, respectively). Collectively, these molecules represent a novel set of antiviral compounds with unique structural features.


Assuntos
Antivirais/síntese química , Benzofuranos/síntese química , Dioxóis/síntese química , Lactonas/síntese química , Lignanas/química , Naftalenos/síntese química , Antivirais/química , Antivirais/farmacologia , Benzofuranos/química , Benzofuranos/farmacologia , Citomegalovirus/efeitos dos fármacos , Dioxóis/química , Dioxóis/farmacologia , HIV/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Vírus da Hepatite B/efeitos dos fármacos , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Herpesvirus Humano 4/efeitos dos fármacos , Lactonas/química , Lactonas/farmacologia , Naftalenos/química , Naftalenos/farmacologia , Relação Estrutura-Atividade
7.
Antiviral Res ; 65(1): 1-12, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15652966

RESUMO

FK778 (Fujisawa Healthcare Inc.) is an immunosuppressant structurally similar to A771726, the active metabolite of leflunomide (Aventis Pharmaceuticals), but with a clinically relevant shorter serum half-life. Leflunomide, a tolerated and efficacious immunosuppressive agent in patients receiving allograft transplantations, was reported to be active against HCMV and HSV-1. Here we report that FK778 is a potent and effective inhibitor of HCMV, and that its mode of antiviral action appears to mirror the biochemical mechanisms elsewhere described to be responsible for its immunosuppressive properties: inhibition of protein tyrosine phosphorylation and inhibition of cellular de novo pyrimidine biosynthesis. Initial HCMV-mediated activation of the EGF receptor/phosphatidylinositol 3-kinase (PI3-K) pathways and Sp1 and NF-kappaB were partially inhibited by FK778. The second tier (phase) of PI3-K, Sp1, and NF-kappaB induction by HCMV was more sensitive to FK778. Treatment of HCMV-infected cells with FK778 prevented the appearance of HCMV proteins some 12-24h post infection, and inhibited viral DNA synthesis. In our assays, leflunomide also reduced HCMV DNA levels. The antiviral activity of FK778 was reversed in cell culture by treatment with uridine, consistent with specific inhibition of dihydroorotate dehydrogenase (DHODH), a required enzyme in the de novo biosynthesis of pyrimidines. This report substantiates the clinical possibility of a single drug treatment to achieve immunosuppression and inhibit opportunistic herpesvirus infections. Our results differ from descriptions of leflunomide acting as an inhibitor of HCMV cytoplasmic capsid formation. Additionally, this study indicates that DHODH may be an effective cellular antiviral target.


Assuntos
Antivirais/farmacologia , Imunossupressores/farmacologia , Isoxazóis/farmacologia , Alcinos , Antivirais/toxicidade , Linhagem Celular , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Humanos , Imunossupressores/química , Imunossupressores/toxicidade , Isoxazóis/química , Isoxazóis/toxicidade , Leflunomida , Nitrilas , Transdução de Sinais/efeitos dos fármacos , Ensaio de Placa Viral , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
8.
Microbes Infect ; 6(12): 1084-93, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15380778

RESUMO

Human cytomegalovirus (HCMV) receptor-ligand interactions and viral entry excite cellular responses such as receptor tyrosine kinase and mitogen-activated protein kinase signaling, cytoskeletal rearrangement, and the induction of transcription factors, prostaglandins, and cytokines. Bi-phasic stimulation of these pathways, excepting interferon, facilitates productive viral infection and likely contributes to viral pathogenesis.


Assuntos
Citomegalovirus/patogenicidade , Resposta ao Choque Térmico , Transdução de Sinais , Linhagem Celular , Citocinas/metabolismo , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/microbiologia , Regulação da Expressão Gênica , Humanos , Proteínas/genética , Proteínas/metabolismo
9.
Antiviral Res ; 63(2): 85-95, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15302137

RESUMO

Resveratrol is a polyphenolic natural product that is present in red wine and peanuts and has inhibitory activity against inflammation, heart disease, and cancer. Here we describe its inhibition of human cytomegalovirus replication (IC50 = 1-2 microM). At least 50-fold higher concentrations of compound were required to produce cytotoxicity against growing or stationary human embryonic lung fibroblasts. Mechanism of action studies determined that resveratrol blocked virus-induced activation of the epidermal growth factor receptor (EGFR) and phosphatidylinositol-3-kinase signal transduction as well as NF-kappaB and Sp1 transcription factor activation shortly following infection. Resveratrol prevented the appearance of immediate-early, early, and late viral proteins. Human cytomegalovirus DNA replication was reduced to undetectable levels by treatment with resveratrol, as were the second (late) phases of virus-induced phosphatidylinositol-3-kinase signaling and transcription factor activation. Resveratrol lost substantial antiviral activity when its addition was delayed until 4 h postinfection. Compound reversibility and preincubation studies were inconsistent with a virucidal mechanism of action. These data indicated that this compound likely operated during attachment and entry. We hypothesize that the primary molecular target for resveratrol may be blockage of epidermal growth factor receptor activation and its downstream effectors.


Assuntos
Citomegalovirus/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Estilbenos/farmacologia , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Sobrevivência Celular , Células Cultivadas , Citomegalovirus/crescimento & desenvolvimento , Replicação do DNA/efeitos dos fármacos , DNA Viral/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Receptores ErbB/metabolismo , Fibroblastos/virologia , Humanos , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Resveratrol , Fator de Transcrição Sp1/metabolismo , Fatores de Tempo , Ensaio de Placa Viral , Proteínas Virais/biossíntese
10.
J Virol ; 77(23): 12660-70, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14610188

RESUMO

Previous work has demonstrated that the human cytomegalovirus IE1-72 protein is able to bind to the N terminus of p107, and IE1-72 alone is sufficient for alleviation of p107-mediated cell growth suppression. However, the mechanism of this alleviation is unclear. Here, we show that IE1-72 can alleviate p107 inhibition of cyclin E/cdk2 kinase activity. We cotransfected various IE1-72 and p107 constructs into C33A cells and demonstrated that IE1-72 could activate the kinase activity of cyclin E/cdk2. Conversely, IE2-86 did not activate this activity, suggesting that the interaction between p107 and IE1-72 and the subsequent kinase activation are specific. By the use of a series of deletion and point mutants of IE1-72 and p107, we observed that a mutation of the loop region of helix-loop-helix-turn-helix in exon 3 of IE1-72 as well as a mutation of the leucine zipper-2 region in exon 4 of IE1-72 abolished binding to p107. In addition, these two IE1-72 mutants did not alleviate p107 inhibition of cyclin E/cdk2 kinase activity and also failed to alleviate p107 inhibition of the E2F-responsive promoter. Meanwhile, deletion of the N-terminal aa 1 to 175 of p107 abolished both p107 binding with IE1-72 and p107 inhibition of cyclin E/cdk2 kinase activity. This result confirms that the N-terminus aa 1 to 175 region of p107 is a common region where both IE1-72 protein and cyclin E/cdk2 bind. We propose a mechanism in which binding of IE1-72 to p107 displaces cyclin E/cdk2 from p107. Once released from p107, cyclin E/cdk2 is able to function as an active kinase.


Assuntos
Quinases relacionadas a CDC2 e CDC28/metabolismo , Ciclina E/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Regulação para Cima , Proteínas Virais , Sequência de Bases , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina , Primers do DNA , Humanos , Ligação Proteica
11.
Nature ; 424(6947): 456-61, 2003 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-12879076

RESUMO

Human cytomegalovirus (HCMV) is a widespread opportunistic herpesvirus that causes severe and fatal diseases in immune-compromised individuals, including organ transplant recipients and individuals with AIDS. It is also a leading cause of virus-associated birth defects and is associated with atherosclerosis and coronary restenosis. HCMV initiates infection and intracellular signalling by binding to its cognate cellular receptors and by activating several signalling pathways including those mediated by mitogen-activated protein kinase, phosphatidylinositol-3-OH kinase, interferons, and G proteins. But a cellular receptor responsible for viral entry and HCMV-induced signalling has yet to be identified. Here we show that HCMV infects cells by interacting with epidermal growth factor receptor (EGFR) and inducing signalling. Transfecting EGFR-negative cells with an EGFR complementary DNA renders non-susceptible cells susceptible to HCMV. Ligand displacement and crosslinking analyses show that HCMV interacts with EGFR through gB, its principal envelope glycoprotein. gB preferentially binds EGFR and EGFR-ErbB3 oligomeric molecules in Chinese hamster ovary cells transfected with erbB family cDNAs. Taken together, these data indicate that EGFR is a necessary component for HCMV-triggered signalling and viral entry.


Assuntos
Citomegalovirus/metabolismo , Receptores Virais/metabolismo , Animais , Células CHO , Cálcio/metabolismo , Cricetinae , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fibroblastos , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama , Fosforilação , Receptores Virais/genética , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas , Fosfolipases Tipo C/metabolismo
12.
J Mol Med (Berl) ; 80(4): 233-42, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11976732

RESUMO

Antiviral therapy of primary and recurrent infections with human cytomegalovirus is reserved for severe manifestations and faces several limitations. Presently candidates for novel drugs with lower adverse side effects and a minimized frequency of resistance formation are under investigation. Here we demonstrate that artesunate, an antimalaria drug with highly valuable pharmacological properties, possesses antiviral activity. A concentration-dependent inhibition of the replication of human cytomegaloviruses with wild-type phenotype was demonstrated in several cell lines. Inhibition was quantified using recombinant green fluorescent protein expressing virus variants. The IC50 values were in the same range for ganciclovir-sensitive and ganciclovir-resistant human cytomegalovirus, as calculated with 5.8+/-0.4 microM and 6.9+/-0.2 microM, respectively. This indicated a strong antiviral potential and a lack of cross-resistance. The optimal antiviral concentrations of artesunate were separable from those inducing cytotoxicity. In addition, the replication of viruses from three genera was seen to be artesunate-sensitive to varying degrees. This suggests a mechanism linked to cellular activation pathways. Both the protein levels and the DNA binding activity of the two virus-induced cellular transcription factors Sp1 and NF-kappaB were found to be markedly reduced in the presence of artesunate. We also analyzed the cellular signaling kinase phosphoinositide 3-kinase, required for the activation of factors such as Sp1 and NF-kappaB in infected fibroblasts. The phosphorylation of two downstream effectors of phosphoinositide 3-kinase, Akt and p70S6K, was markedly inhibited in the presence of artesunate. Thus, artesunate possesses attractive antiviral characteristics which are suggestively based on the interference with essential steps in the host cell kinase cascades.


Assuntos
Antivirais/farmacologia , Artemisininas , Citomegalovirus/efeitos dos fármacos , Ganciclovir/farmacologia , Inibidores de Proteínas Quinases , Proteínas Serina-Treonina Quinases , Sesquiterpenos/farmacologia , Artesunato , Células Cultivadas , Citomegalovirus/genética , DNA Viral/efeitos dos fármacos , DNA Viral/metabolismo , Farmacorresistência Viral , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes , Testes de Sensibilidade Microbiana , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases S6 Ribossômicas/metabolismo , Simplexvirus/efeitos dos fármacos , Fator de Transcrição Sp1/metabolismo , Ensaio de Placa Viral , Replicação Viral/efeitos dos fármacos
13.
J Med Virol ; 67(1): 33-40, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11920815

RESUMO

Inhibition of virus-induced intracellular signaling pathways and viral infectivity are our ultimate goals in the development of effective antiviral agents to control human cytomegalovirus (HCMV) infections. The HCMV hyperimmune globulin may meet such criteria. In a human embryonic lung (HEL) fibroblast culture model, pretreatment of Towne strain HCMV with HCMV hyperimmune globulin was shown to inhibit viral infectivity successfully, as measured by a standard plaque assay. The extracellular viral titers and extracellular viral DNA, as measured by plaque assay and PCR, respectively, were also decreased. In addition, the HCMV hyperimmune globulin prevented HCMV from inducing the intracellular activation of NF-kappaB, Sp-1, and PI3-K signaling pathways. The PI3-K pathway was examined by following phosphorylation (activation) of two of its downstream kinases, Akt and p70S6K. HCMV hyperimmune globulin also prevented the production of immediate early, early, and late viral proteins. These studies show that HCMV hyperimmune globulin neutralization of HCMV prevents the earliest known events observed after viral envelope glycoproteins bind their cell membrane receptors, i.e., NF-kappaB, Sp-1 and PI3-K activation. This suggests that HCMV hyperimmune globulin not only can inhibit viral infectivity, but can also prevent the abnormal cellular signaling that may induce unwanted cellular proliferation or cytokine synthesis.


Assuntos
Proteínas do Capsídeo , Infecções por Citomegalovirus/imunologia , Imunoglobulinas/imunologia , Glicoproteínas de Membrana , NF-kappa B/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Serina-Treonina Quinases , Transdução de Sinais/imunologia , Fator de Transcrição Sp1/imunologia , Transativadores , Proteínas do Envelope Viral , Capsídeo/biossíntese , Ativação Enzimática , Humanos , Proteínas Imediatamente Precoces/biossíntese , Imunoglobulinas Intravenosas , Líquido Intracelular , Testes de Neutralização , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Proteínas Virais/biossíntese
14.
J Gen Virol ; 82(Pt 3): 493-497, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11172089

RESUMO

Human cytomegalovirus infection of quiescent fibroblasts was found to induce a bi-phasic activation of mitogen-activated protein kinase (MAPK) kinase 1 and 2 (MKK1/2) and two of their downstream targets, extracellular signal regulated kinase 1 and 2 (ERK1/2), as determined by Western blot analysis using phospho-specific antibodies. Treatment of infected fibroblasts with U0126, a potent and specific inhibitor of MKK1/2 kinase activity, completely blocked ERK1/2 activation following HCMV infection without affecting cell viability. Anti-viral studies demonstrate that in the presence of U0126, viral titres are reduced and viral DNA replication is inhibited. In addition, protein levels of two viral early genes that are required for viral DNA replication, UL44 and UL84, are significantly decreased in the presence of U0126. These results suggest that HCMV-mediated activation of MKK1/2 kinase activity enhances virus infectivity by ensuring timely initiation of viral DNA replication, possibly by regulating early gene expression.


Assuntos
Citomegalovirus/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/fisiologia , Butadienos/farmacologia , Células Cultivadas , Citomegalovirus/genética , Citomegalovirus/metabolismo , Replicação do DNA/efeitos dos fármacos , DNA Viral/biossíntese , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/virologia , Humanos , MAP Quinase Quinase 1 , MAP Quinase Quinase 2 , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Nitrilas/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...