Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-34502053

RESUMO

In recent decades, recombinant antibodies against specific antigens have shown great promise for the therapy of infectious diseases and cancer. Human papillomaviruses (HPVs) are involved in the development of around 5% of all human cancers and HPV16 is the high-risk genotype with the highest prevalence worldwide, playing a dominant role in all HPV-associated cancers. Here, we describe the main biological activities of the HPV16 E6, E7, and E5 oncoproteins, which are involved in the subversion of important regulatory pathways directly associated with all known hallmarks of cancer. We then review the state of art of the recombinant antibodies targeted to HPV oncoproteins developed so far in different formats, and outline their mechanisms of action. We describe the advantages of a possible antibody-based therapy against the HPV-associated lesions and discuss the critical issue of delivery to tumour cells, which must be addressed in order to achieve the desired translation of the antibodies from the laboratory to the clinic.


Assuntos
Anticorpos Antivirais/uso terapêutico , Neoplasias/tratamento farmacológico , Anticorpos de Domínio Único/uso terapêutico , Animais , Anticorpos Antivirais/imunologia , Humanos , Neoplasias/virologia , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Proteínas Repressoras/imunologia , Anticorpos de Domínio Único/imunologia
2.
Cell Microbiol ; 17(9): 1391-404, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25856684

RESUMO

Indomethacin, a cyclooxygenase-1 and -2 inhibitor widely used in the clinic for its potent anti-inflammatory/analgesic properties, possesses antiviral activity against several viral pathogens; however, the mechanism of antiviral action remains elusive. We have recently shown that indomethacin activates the double-stranded RNA (dsRNA)-dependent protein kinase R (PKR) in human colon cancer cells. Because of the important role of PKR in the cellular defence response against viral infection, herein we investigated the effect of indomethacin on PKR activity during infection with the prototype rhabdovirus vesicular stomatitis virus. Indomethacin was found to activate PKR in an interferon- and dsRNA-independent manner, causing rapid (< 5 min) phosphorylation of eukaryotic initiation factor-2 α-subunit (eIF2α). These events resulted in shutting off viral protein translation and blocking viral replication (IC50 = 2 µM) while protecting host cells from virus-induced damage. Indomethacin did not affect eIF2α kinases PKR-like endoplasmic reticulum-resident protein kinase (PERK) and general control non-derepressible-2 (GCN2) kinase, and was unable to trigger eIF2α phosphorylation in the presence of PKR inhibitor 2-aminopurine. In addition, small-interfering RNA-mediated PKR gene silencing dampened the antiviral effect in indomethacin-treated cells. The results identify PKR as a critical target for the antiviral activity of indomethacin and indicate that eIF2α phosphorylation could be a key element in the broad spectrum antiviral activity of the drug.


Assuntos
Antivirais/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Indometacina/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Vesiculovirus/efeitos dos fármacos , Proteínas Virais/biossíntese , eIF-2 Quinase/metabolismo , Linhagem Celular , Ativadores de Enzimas/metabolismo , Humanos , Concentração Inibidora 50 , Fosforilação , Processamento de Proteína Pós-Traducional
3.
Int J Cancer ; 134(11): 2742-7, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24226851

RESUMO

Human papillomavirus (HPV)-associated tumors still represent an urgent problem of public health in spite of the efficacy of the prophylactic HPV vaccines. Specific antibodies in single-chain format expressed as intracellular antibodies (intrabodies) are valid tools to counteract the activity of target proteins. We previously showed that the M2SD intrabody, specific for the E7 oncoprotein of HPV16 and expressed in the endoplasmic reticulum of the HPV16-positive SiHa cells, was able to inhibit cell proliferation. Here, we showed by confocal microscopy that M2SD and E7 colocalize in the endoplasmic reticulum of SiHa cells, suggesting that the E7 delocalization mediated by M2SD could account for the anti-proliferative activity of the intrabody. We then tested the M2SD antitumor activity in two mouse models for HPV tumors based respectively on TC-1 and C3 cells. The M2SD intrabody was delivered by retroviral vector to tumor cells before cell injection into C57BL/6 mice. In both models, a marked delay of tumor onset with respect to the controls was observed in all the mice injected with the M2SD-expressing tumor cells and, importantly, a significant percentage of mice remained tumor-free permanently. This is the first in vivo demonstration of the antitumor activity of an intrabody directed towards an HPV oncoprotein. We consider that these results could contribute to the development of new therapeutic molecules based on antibodies in single-chain format, to be employed against the HPV-associated lesions even in combination with other drugs.


Assuntos
Anticorpos Antivirais/administração & dosagem , Papillomavirus Humano 16/imunologia , Proteínas E7 de Papillomavirus/antagonistas & inibidores , Infecções por Papillomavirus/terapia , Anticorpos de Cadeia Única/administração & dosagem , Neoplasias do Colo do Útero/terapia , Animais , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Proliferação de Células , Feminino , Terapia Genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/imunologia , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/virologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Taxa de Sobrevida , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/virologia
4.
Biochem J ; 443(2): 379-86, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22268531

RESUMO

The NSAID (non-steroidal anti-inflammatory drug) indomethacin, a cyclo-oxygenase-1 and -2 inhibitor with anti-inflammatory and analgesic properties, is known to possess anticancer activity against CRC (colorectal cancer) and other malignancies in humans; however, the mechanism underlying the anticancer action remains elusive. In the present study we show that indomethacin selectively activates the dsRNA (double-stranded RNA)-dependent protein kinase PKR in a cyclo-oxygenase-independent manner, causing rapid phosphorylation of eIF2α (the α-subunit of eukaryotic translation initiation factor 2) and inhibiting protein synthesis in colorectal carcinoma and other types of cancer cells. The PKR-mediated translational block was followed by inhibition of CRC cell proliferation and apoptosis induction. Indomethacin did not affect the activity of the eIF2α kinases PERK (PKR-like endoplasmic reticulum-resident kinase), GCN2 (general control non-derepressible-2) and HRI (haem-regulated inhibitor kinase), and induced eIF2α phosphorylation in PERK-knockout and GCN2-knockout cells, but not in PKR-knockout cells or in human PKR-silenced CRC cells, identifying PKR as a selective target for indomethacin-induced translational inhibition. The fact that indomethacin induced PKR activity in vitro, an effect reversed by the PKR inhibitor 2-aminopurine, suggests a direct effect of the drug in kinase activation. The results of the present study identify PKR as a novel target of indomethacin, suggesting new scenarios on the molecular mechanisms underlying the pleiotropic activity of this traditional NSAID.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Neoplasias do Colo/enzimologia , Indometacina/farmacologia , eIF-2 Quinase/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Neoplasias do Colo/patologia , Ativação Enzimática , Humanos , Biossíntese de Proteínas/efeitos dos fármacos
5.
J Exp Clin Cancer Res ; 40(1): 37, 2021 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-33485370

RESUMO

BACKGROUND: The oncogenic activity of the high risk human papillomavirus type 16 (HPV16) is fully dependent on the E6 and E7 viral oncoproteins produced during viral infection. The oncoproteins interfere with cellular homeostasis by promoting proliferation, inhibiting apoptosis and blocking epithelial differentiation, driving the infected cells towards neoplastic progression. The causal relationship between expression of E6/E7 and cellular transformation allows inhibiting the oncogenic process by hindering the activity of the two oncoproteins. We previously developed and characterized some antibodies in single-chain format (scFvs) against the HPV16 E6 and E7 proteins, and demonstrated both in vitro and in vivo their antitumor activity consisting of protective efficacy against tumor progression of HPV16-positive cells. METHODS: Envisioning clinical application of the best characterized anti-HPV16 E6 and -HPV16 E7 scFvs, we verified their activity in the therapeutic setting, on already implanted tumors. Recombinant plasmids expressing the anti-HPV16 E6 scFvI7 with nuclear targeting sequence, or the anti-HPV16 E7 scFv43M2 with endoplasmic reticulum targeting sequence were delivered by injection followed by electroporation to three different preclinical models using C57/BL6 mice, and their effect on tumor growth was investigated. In the first model, the HPV16+ TC-1 Luc cells were used to implant tumors in mice, and tumor growth was measured by luciferase activity; in the second model, a fourfold number of TC-1 cells was used to obtain more aggressively growing tumors; in the third model, the HPV16+ C3 cells where used to rise tumors in mice. To highlight the scFv possible mechanism of action, H&E and caspase-3 staining of tumor section were performed. RESULTS: We showed that both the anti-HPV16 E6 and HPV16 E7 scFvs tested were efficacious in delaying tumor progression in the three experimental models and that their antitumor activity seems to rely on driving tumor cells towards the apoptotic pathway. CONCLUSION: Based on our study, two scFvs have been identified that could represent a safe and effective treatment for the therapy of HPV16-associated lesions. The mechanism underlying the scFv effectiveness appears to be leading cells towards death by apoptosis. Furthermore, the validity of electroporation, a methodology allowed for human treatment, to deliver scFvs to tumors was confirmed.


Assuntos
Papillomavirus Humano 16/imunologia , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos
6.
Cancers (Basel) ; 12(7)2020 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-32640530

RESUMO

Human Papillomavirus 16-associated cancer, affecting primarily the uterine cervix but, increasingly, other body districts, including the head-neck area, will long be a public health problem, despite there being a vaccine. Since the virus oncogenic activity is fully ascribed to the viral E6 and E7 oncoproteins, one of the therapeutic approaches for HPV16 cancer is based on specific antibodies in single-chain format targeting the E6/E7 activity. We analyzed the Complementarity Determining Regions, repositories of antigen-binding activity, of four anti-HPV16 E6 and -HPV16 E7 scFvs, to highlight possible conformity to biophysical properties, recognized to be advantageous for therapeutic use. By epitope mapping, using E7 mutants with amino acid deletions or variations, we investigated differences among the anti-16E7 scFvs in terms of antigen-binding capacity. We also performed computational analyses to determine whether length, total net charge, surface hydrophobicity, polarity and charge distribution conformed well to those of the antibodies that had already reached clinical use, through the application of developability guidelines derived from recent literature on clinical-stage antibodies, and the Therapeutic Antibodies Profiler software. Overall, our findings show that the scFvs investigated may represent valid candidates to be developed as therapeutic molecules for clinical use, and highlight characteristics that could be improved by molecular engineering.

7.
Antivir Ther ; 11(8): 995-1004, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17302369

RESUMO

BACKGROUND: Herpes simplex virus type 1 (HSV-1) is a potent inducer of nuclear factor-KB (NF-kappaB), a cellular transcription factor with a crucial role in promoting inflammation and controlling cell proliferation and survival. OBJECTIVES: On the basis of the recent demonstration that HSV-1-induced NF-kappaB is actively recruited to KB-binding sites in the HSV-1 infected-cell protein 0 (ICPO) promoter enhancing viral transcription and replication, we investigated the effect of proteasome inhibitors MG132, MG115 and epoxomicin, which block NF-kappaB function by preventing the degradation of the inhibitory proteins IkappaBalpha, on HSV-1-induced NF-kappaB activation and viral replication. METHODS: Antiviral activity of proteasome inhibitors was analysed in HSV-1-infected HEp2 cells by determining infective virus titres by CPE50%, viral RNA synthesis by RT-PCR, and viral protein synthesis by immunoblot analysis or immunofluorescence. ICPO transcription was studied in transient transfection experiments using the ICPO promoter-luciferase IE1-Luc construct. IkappaBalpha degradation and NF-kappaB activity were determined by immunoblot analysis and EMSA, respectively. RESULTS: Proteasome inhibitors were found to prevent HSV-1-induced NF-kappaB activation in the early phase of infection. Block of virus-induced NF-kappaB activation resulted in inhibiting HSV-1 ICPO gene expression, in decreasing the level of immediate-early and late viral proteins, and ultimately in greatly suppressing viral replication. The antiviral effect was lost if treatment was started after NF-kappaB activation, and appeared to be independent of the HSV-1-induced activation of the JNK pathway. CONCLUSIONS: Proteasome inhibitors possess NF-kappaB-dependent antiherpetic activity. The results described further identify the IKK/NF-kappaB pathway as a suitable target for novel antiherpetic drugs.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , NF-kappa B/metabolismo , Inibidores de Proteassoma , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação para Baixo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Leupeptinas/farmacologia , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Fator de Transcrição AP-1/metabolismo , Células Vero
8.
Antivir Ther ; 11(8): 1021-30, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17302372

RESUMO

UNLABELLED: Severe acute respiratory syndrome (SARS) is a newly emerging, highly transmissible and fatal disease caused by a previously unknown coronavirus (SARS-CoV). Existing in non-identified animal reservoirs, SARS-CoV continues to represent a threat to humans because there is no effective specific antiviral therapy for coronavirus infections. OBJECTIVES: Starting from the observation that cyclopentenone cyclooxygenase (COX) metabolites are active against several RNA viruses, we investigated the effect of the COX inhibitor indomethacin on coronavirus replication. METHODS: Work involving infectious SARS-CoV was performed in biosafety level 3 facilities. SARS-CoV was grown in monkey VERO cells and human lung epithelial A549 cells, while canine coronavirus (CCoV) was grown in A72 canine cells. Antiviral activity was analysed by determining infective virus titres by TCID50, viral RNA synthesis by Northern blot analysis and real-time RT-PCR, and viral protein synthesis by SDS-PAGE analysis after 35S-methionine-labelling. Antiviral efficacy in vivo was determined by evaluating virus titres in CCoV-infected dogs treated orally with 1 mg/kg body weight indomethacin (INDO). RESULTS: Unexpectedly, we found that INDO has a potent direct antiviral activity against the coronaviruses SARS-CoV and CCoV. INDO does not affect coronavirus binding or entry into host cells, but acts by blocking viral RNA synthesis at cytoprotective doses. This effect is independent of cyclooxygenase inhibition. INDO's potent antiviral activity (>1,000-fold reduction in virus yield) was confirmed in vivo in CCoV-infected dogs. CONCLUSIONS: The results identify INDO as a potent inhibitor of coronavirus replication and suggest that, having both anti-inflammatory and antiviral activity, INDO could be beneficial in SARS therapy.


Assuntos
Antivirais/farmacologia , Indometacina/farmacologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Aspirina/farmacologia , Linhagem Celular , Chlorocebus aethiops , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/veterinária , Coronavirus Canino/efeitos dos fármacos , Doenças do Cão/tratamento farmacológico , Cães , Relação Dose-Resposta a Droga , Fezes/virologia , Humanos , Indometacina/uso terapêutico , Interferon-alfa/farmacologia , RNA Viral/isolamento & purificação , Ribavirina/farmacologia , Replicação Viral/efeitos dos fármacos
9.
Oncotarget ; 7(13): 15539-53, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26788990

RESUMO

Single-chain variable fragments (scFvs) expressed as "intracellular antibodies" (intrabodies) can target intracellular antigens to hamper their function efficaciously and specifically. Here we use an intrabody targeting the E6 oncoprotein of Human papillomavirus 16 (HPV16) to address the issue of a non-invasive therapy for HPV cancer patients.A scFv against the HPV16 E6 was selected by Intracellular Antibody Capture Technology and expressed as I7nuc in the nucleus of HPV16-positive SiHa, HPV-negative C33A and 293T cells. Colocalization of I7nuc and recombinant E6 was observed in different cell compartments, obtaining evidence of E6 delocalization ascribable to I7nuc. In SiHa cells, I7nuc expressed by pLNCX retroviral vector was able to partially inhibit degradation of the main E6 target p53, and induced p53 accumulation in nucleus. When analyzing in vitro activity on cell proliferation and survival, I7nuc was able to decrease growth inducing late apoptosis and necrosis of SiHa cells.Finally, I7nuc antitumor activity was demonstrated in two pre-clinical models of HPV tumors. C57BL/6 mice were injected subcutaneously with HPV16-positive TC-1 or C3 tumor cells, infected with pLNCX retroviral vector expressing or non-expressing I7nuc. All the mice injected with I7nuc-expressing cells showed a clear delay in tumor onset; 60% and 40% of mice receiving TC-1 and C3 cells, respectively, remained tumor-free for 17 weeks of follow-up, whereas 100% of the controls were tumor-bearing 20 days post-inoculum. Our data support the therapeutic potential of E6-targeted I7nuc against HPV tumors.


Assuntos
Imunoterapia/métodos , Neoplasias Experimentais/virologia , Proteínas Oncogênicas Virais/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Anticorpos de Cadeia Única/farmacologia , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Papillomavirus/complicações
10.
FASEB J ; 18(13): 1609-11, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15289449

RESUMO

Glutathione depletion by inhibition of its synthesis with buthionine sulfoximine (BSO) is a focus of the current research in antitumor therapy, BSO being used as chemosensitizer. We had previously shown that two human tumor cell lines (U937 and HepG2) survive to treatment with BSO: BSO can elicit an apoptotic response, but the apoptotic process is aborted after cytochrome c release and before caspase activation, suggesting the development of an adaptive response (FASEB J., 1999, 13, 2031-2036). Here, we investigate the mechanisms of such an adaptation. We found that following BSO, U937 up-regulate Bcl-2 mRNA and protein levels, by a mechanism possibly involving NF-kappaB transcription factor; the increase in protein level is limited by a rapid decay of Bcl-2 in BSO-treated cells, suggesting that redox imbalance speeds up Bcl-2 turnover. BSO-dependent Bcl-2 up-regulation is associated with the ability to survive to BSO. Indeed, 1) its abrogation by CAPE or protein synthesis inhibition sensitizes U937 to BSO; 2) in a panel of four tumor lines, BSO-resistant (U937, HepG2, and HGB1) but not BSO-sensitive (BL41) cells can up-regulate Bcl-2 following GSH depletion; remarkably, only the latter are chemosensitized by BSO.


Assuntos
Butionina Sulfoximina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Glutationa/deficiência , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Regulação para Cima/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células U937
11.
AIDS ; 18(9): 1271-80, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15362659

RESUMO

OBJECTIVES: Herpes simplex virus (HSV) infections have been associated with reactivation of HIV-1 replication and increases of HIV-1-load in plasma of co-infected individuals. The present authors have previously reported that in epithelial cells HSV-1 induces the IkappaB-kinase (IKK) causing persistent activation of NF-kappaB, a critical regulator of HIV-1 replication. The present study was performed to investigate whether HSV-1-infection could induce IKK-mediated NF-kappaB activation and enhance HIV-1 expression in human T cells, and to analyze the effect of the IKK-inhibitor prostaglandin A1 (PGA1) and other prostanoids on the NF-kappaB-mediated HSV-HIV interaction. DESIGN AND METHODS: Induction of IKK and NF-kappaB activity was determined in lymphoblastoid Jurkat cells and HIV-1 chronically-infected H9 and ACH-2 cells by kinase assay and electrophoretic mobility shift assay, respectively. The effect of HSV-1 and different prostanoids on HIV-1 expression and replication was determined in Jurkat cells transfected with HIV-1-LTR-driven reporter genes, and in H9 and ACH-2 cells by p24-antigen level evaluation. The role of NF-kappaB in HSV-1-induced HIV-1 expression was investigated by using the IkappaBalpha dominant-negative IkappaBalpha-AA in co-transfection experiments. RESULTS: In human T lymphoblastoid cells HSV-1 potently induces IKK activity, causing a persistent induction of NF-kappaB. HSV-1-induced IKK and NF-kappaB function results in transactivation of HIV-1-LTR-regulated genes and induction of HIV-1 replication in chronically-infected T cells. The cyclopentenone PGA1 inhibits HSV-1-induced IKK and NF-kappaB activities, blocking HIV-1-LTR-driven expression and preventing HSV-1-induced HIV-1 replication in co-infected cells. CONCLUSIONS: The results indicate that IKK is a key factor in triggering HSV-1-induced HIV-1 transcription in chronically-infected cells and identify cyclopentenone prostanoids as potent inhibitors of HSV-1-induced HIV-1 reactivation.


Assuntos
Infecções por HIV/virologia , HIV-1/fisiologia , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Prostaglandinas A/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Ensaio de Desvio de Mobilidade Eletroforética , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/metabolismo , Herpes Simples/metabolismo , Humanos , Quinase I-kappa B , Células Jurkat , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T/enzimologia , Transcrição Gênica , Células Vero , Carga Viral , Ativação Viral/efeitos dos fármacos
12.
Pathog Glob Health ; 106(1): 12-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22595270

RESUMO

Bovine lactoferrin (bLf) is a multifunctional glycoprotein that plays an important role in innate immunity against infections, including influenza. Here we have dissected bLf into its C- and N-lobes and show that inhibition of influenza virus hemagglutination and cell infection is entirely attributable to the C-lobe and that all major virus subtypes, including H1N1 and H3N2, are inhibited. By far-western blotting and sequencing studies, we demonstrate that bLf C-lobe strongly binds to the HA(2) region of viral hemagglutinin, precisely the highly conserved region containing the fusion peptide. By molecular docking studies, three C-lobe fragments were identified which inhibited virus hemagglutination and infection at fentomolar concentration range. Besides contributing to explain the broad anti-influenza activity of bLf, our findings lay the foundations for exploiting bLf fragments as source of potential anti-influenza therapeutics.


Assuntos
Antivirais/farmacologia , Lactoferrina/farmacologia , Orthomyxoviridae/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antivirais/metabolismo , Células Cultivadas , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Hemaglutininas/metabolismo , Lactoferrina/genética , Lactoferrina/metabolismo , Dados de Sequência Molecular , Orthomyxoviridae/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Ligação Proteica , Alinhamento de Sequência
13.
Blood ; 109(7): 2718-26, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17138827

RESUMO

Human herpesvirus 8 (HHV-8) is considered the causative agent of Kaposi sarcoma, a highly vascularized neoplasm characterized by spindle-shaped cells of endothelial origin and inflammatory cell infiltration. The cell transforming ability of HHV-8 has been associated with the activation of NF-kappaB, a nuclear factor playing a pivotal role in promoting inflammation and cell proliferation; however, little is known about NF-kappaB activation during acute HHV-8 infection. In the present study, we used a recently established in vitro model of HHV-8 acute productive infection in endothelial cells to investigate the effect of HHV-8 on NF-kappaB activity and function. HHV-8 rapidly and potently induced NF-kappaB activity in endothelial cells via stimulation of the IkappaB kinase (IKK). Following IKK activation, HHV-8 selectively triggered the production of high levels of monocyte chemoattractant protein 1 (MCP-1), whereas it did not affect the expression of other NF-kappaB-dependent proinflammatory proteins, including TNF-alpha, IL-8, and RANTES. Deletion of NF-kappaB-binding sites in the MCP-1 enhancer resulted in significant inhibition of HHV-8-induced transcription. Furthermore, MCP-1 production was accompanied by virus-induced capillary-like structure formation at early stages of infection. The results suggest that HHV-8-induced MCP-1 may play an important role in promoting inflammation and pathogenic angiogenesis typical of HHV-8-associated lesions.


Assuntos
Quimiocina CCL2/fisiologia , Células Endoteliais/fisiologia , Herpesvirus Humano 8/patogenicidade , Quinase I-kappa B/fisiologia , NF-kappa B/fisiologia , Capilares/patologia , Células Cultivadas , Quimiocina CCL2/genética , Células Endoteliais/patologia , Expressão Gênica , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Humanos , Neovascularização Patológica , Sarcoma de Kaposi/irrigação sanguínea , Sarcoma de Kaposi/etiologia , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia
14.
J Biol Chem ; 281(11): 7110-7, 2006 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-16407234

RESUMO

Herpes simplex viruses (HSVs) are able to hijack the host-cell IkappaB kinase (IKK)/NF-kappaB pathway, which regulates critical cell functions from apoptosis to inflammatory responses; however, the molecular mechanisms involved and the outcome of the signaling dysregulation on the host-virus interaction are mostly unknown. Here we show that in human keratinocytes HSV-1 attains a sophisticated control of the IKK/NF-kappaB pathway, inducing two distinct temporally controlled waves of IKK activity and disrupting the NF-kappaB autoregulatory mechanism. Using chromatin immunoprecipitation we demonstrate that dysregulation of the NF-kappaB-response is mediated by a virus-induced block of NF-kappaB recruitment to the promoter of the IkappaBalpha gene, encoding the main NF-kappaB-inhibitor. We also show that HSV-1 redirects NF-kappaB recruitment to the promoter of ICP0, an immediate-early viral gene with a key role in promoting virus replication. The results reveal a new level of control of cellular functions by invading viruses and suggest that persistent NF-kappaB activation in HSV-1-infected cells, rather than being a host response to the virus, may play a positive role in promoting efficient viral replication.


Assuntos
Regulação da Expressão Gênica , Genes Virais , Proteínas I-kappa B/genética , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Simplexvirus/metabolismo , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Primers do DNA/química , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Inflamação , Queratinócitos/metabolismo , Modelos Genéticos , Inibidor de NF-kappaB alfa , Plasmídeos/metabolismo , Prostaglandinas A/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção , Ubiquitina-Proteína Ligases/metabolismo , Raios Ultravioleta
15.
J Biol Chem ; 280(25): 24127-34, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15837793

RESUMO

Influenza A viruses continue to represent a severe threat worldwide, causing large epidemics and pandemics responsible for thousands of deaths every year. Excessive inflammation due to overabundant production of proinflammatory cytokines by airway epithelial cells is considered an important factor in disease pathogenesis. Here we report that influenza A virus induced IkappaB kinase (IKK) activity in human airway epithelial A549 cells, resulting in persistent activation of nuclear factor-kappaB (NF-kappaB), a critical regulator of the inflammatory response. Although lung epithelial cells are highly sensitive to stimulation of the IKK/NF-kappaB pathway by influenza virus infection, NF-kappaB was not activated in several non-pulmonary cells permissive to the virus, indicating a cell-specific response. Moreover, NF-kappaB was not essential for virus replication but triggered the expression of proinflammatory cytokines in infected lung cells and was directly responsible for production of high levels of interleukin-8, a chemokine associated with influenza-induced inflammation and airway pathology. We also report that 9-deoxy-delta9,delta12-13,14-dihydro-prostaglandin D2, a cyclopentenone prostanoid with therapeutic efficacy against influenza in preclinical studies, was a powerful inhibitor of influenza virus-induced IKK activity and interleukin-8 production by human pulmonary cells. The results identify IKK as an important factor in triggering influenza virus-induced inflammatory reactions in pulmonary epithelium, suggesting novel therapeutic approaches in the treatment of influenza.


Assuntos
Mediadores da Inflamação/metabolismo , Vírus da Influenza A/metabolismo , Interleucina-8/biossíntese , Pulmão/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Cães , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Quinase I-kappa B , Pulmão/citologia , Pulmão/virologia , NF-kappa B/metabolismo
16.
Mol Pharmacol ; 67(2): 572-81, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15548765

RESUMO

Methylation of N3-adenine represents a novel pharmacological strategy for the treatment of resistant tumors. However, little is known about the biochemical pathways involved in cell death induced by N3-methyladenine. In the present study, we show that MeOSO(2) (CH(2))(2)-lexitropsin (Me-Lex), a compound generating almost exclusively N3-methyladenine (>99%), provoked a burst of poly(ADP-ribosylation) and loss of mitochondrial membrane potential in leukemia cells. These events were followed by a marked decrease in nuclear poly(ADP-ribose) polymerase-1 (PARP-1) expression and nuclear factor-kappaB (NF-kappaB) activity. Moreover, DNA damage generated by N3-methyladenine induced a marked decrease in telomerase in the cytosol that was accompanied by a transient up-regulation of activity in the nucleus, as a consequence of nuclear translocation of telomerase in response to genotoxic damage. PARP-1 inhibition blocked ADP-ribose polymer formation, preserved mitochondrial membrane integrity, and counteracted the reduction of NF-kappaB activity, thus preventing the appearance of necrosis. On the other hand, because PARP-1 is a component of the base excision repair (BER), the combination of Me-Lex + PARP-1 inhibitor triggered apoptosis as a result of disruption of BER process. In conclusion, the present study provides new insight into the cellular response to N3-adenine-selective methylating agents that can be exploited for the treatment of tumors unresponsive to classical wide-spectrum methylating agents. Moreover, the results underline the central and paradoxical role of PARP-1 in cell death induced by N3-methyladenine: effector of necrosis and coordinator of methylpurine repair.


Assuntos
Adenina/análogos & derivados , Adenina/farmacologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , DNA/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Telomerase/biossíntese , Linhagem Celular Tumoral , Núcleo Celular/genética , DNA/antagonistas & inibidores , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Humanos , Poli Adenosina Difosfato Ribose/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Telomerase/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
17.
EMBO J ; 22(11): 2552-60, 2003 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-12773372

RESUMO

Among the different definitions of viruses, 'pirates of the cell' is one of the most picturesque, but also one of the most appropriate. Viruses have been known for a long time to utilize a variety of strategies to penetrate cells and, once inside, to take over the host nucleic acid and protein synthesis machinery to build up their own components and produce large amounts of viral progeny. As their genomes carry a minimal amount of information, encoding only a few structural and regulatory proteins, viruses are largely dependent on their hosts for survival; however, despite their apparent simplicity, viruses have evolved different replicative strategies that are regulated in a sophisticated manner. During the last years, the study of the elaborate relationship between viruses and their hosts has led to the understanding of how viral pathogens not only are able to alter the host metabolism via their signaling proteins, but are also able to hijack cellular signaling pathways and transcription factors, and control them to their own advantage. In particular, the nuclear factor-kappaB (NF-kappaB) pathway appears to be an attractive target for common human viral pathogens. This review summarizes what is known about the control of NF-kappaB by viruses, and discusses the possible outcome of NF-kappaB activation during viral infection, which may benefit either the host or the pathogen.


Assuntos
NF-kappa B/fisiologia , Viroses/fisiopatologia , Animais , Humanos , Modelos Biológicos , Transdução de Sinais , Viroses/virologia , Fenômenos Fisiológicos Virais , Vírus/patogenicidade
18.
J Biol Chem ; 278(38): 36059-67, 2003 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-12844494

RESUMO

Signals involved in protection against apoptosis by herpes simplex virus 1 (HSV-1) were investigated. Using U937 monocytoid cells as an experimental model, we have demonstrated that HSV-1 rendered these cells resistant to Fas-induced apoptosis promptly after infection. UV-inactivated virus as well as the envelope glycoprotein D (gD) of HSV-1, by itself, exerted a protective effect on Fas-induced apoptosis. NF-kappaB was activated by gD, and protection against Fas-mediated apoptosis by gD was abolished in cells stably transfected with a dominant negative mutant I-kappaBalpha, indicating that NF-kappaB activation plays a role in the antiapoptotic activity of gD in our experimental model. Moreover, NF-kappaB-dependent protection against Fas-mediated apoptosis was associated with decreased levels of caspase-8 activity and with the up-regulation of intracellular antiapoptotic proteins.


Assuntos
Apoptose , NF-kappa B/fisiologia , Proteínas do Envelope Viral/fisiologia , Receptor fas/metabolismo , Animais , Western Blotting , Caspase 8 , Caspase 9 , Caspases/metabolismo , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Genes Dominantes , Humanos , Proteínas I-kappa B/metabolismo , Marcação In Situ das Extremidades Cortadas , Cinética , Camundongos , Mutação , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fatores de Tempo , Transfecção , Células U937 , Raios Ultravioleta , Regulação para Cima , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA