Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 529(3): 805-811, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32736711

RESUMO

Zika virus (ZIKV) is a mosquito-borne flavivirus associated with Congenital Zika Syndrome (CZS), reflecting a wide range of congenital abnormalities in fetuses and infants infected with ZIKV before birth. ZIKV infections have also been associated with the neurological autoimmune disorder known as Guillian-Barré syndrome (GBS). To date, no vaccines or antiviral strategies are licensed for ZIKV. We used rational design to develop a novel ZIKV vaccine candidate using a Woodchuck Hepatitis core Antigen (WHcAg) Virus-Like Particle (VLP) scaffold for displaying selected antigens from the ZIKV Envelope (E) protein. A Zika-VLP vaccine candidate containing the CD Loop sub-structural domain from ZIKV E protein Domain III (WHcAg CD Loop) elicited a strong immune response in a murine model. Analysis of serum immunoglobulins demonstrated induction of both Th1- and Th2- mediated immune response. No cross-reacting antibodies were detected between Zika, dengue and yellow fever virus, demonstrating a high level of specificity for the ZIKV CD Loop antigen. Immunization with the WHcAg CD Loop vaccine candidate demonstrated immunoprotection in a murine model of ZIKV infection, stimulating protective antibodies associated with antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activities. The WHcAg CD Loop candidate may represent a safer vaccine for preventing antibody dependent enhancement (ADE).


Assuntos
Vacinas de Partículas Semelhantes a Vírus/uso terapêutico , Proteínas do Envelope Viral/uso terapêutico , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Animais , Modelos Animais de Doenças , Feminino , Imunidade , Camundongos , Camundongos Endogâmicos C57BL , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/imunologia , Infecção por Zika virus/imunologia
2.
Clin Immunol ; 183: 99-108, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28780375

RESUMO

The development of virus-like particle (VLP) technology has had an enormous impact on modern vaccinology. In order to optimize the efficacy and safety of VLP-based vaccines, adjuvants are included in most vaccine formulations. To date, most licensed VLP-based vaccines utilize the classic aluminum adjuvant compositions. Certain challenging pathogens and weak immune responder subjects may require further optimization of the adjuvant formulation to maximize the magnitude and duration of the protective immunity. Indeed, novel classes of adjuvants such as liposomes, agonists of pathogen recognition receptors, polymeric particles, emulsions, cytokines and bacterial toxins, can be used to further improve the immunostimulatory activity of a VLP-based vaccine. This review describes the current advances in adjuvant technology for VLP-based vaccines directed at viral diseases, and discusses the basic principles for designing adjuvant formulations for enhancing the vaccine immunogenicity.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Imunogenicidade da Vacina/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/uso terapêutico , Citocinas/imunologia , Citocinas/uso terapêutico , Composição de Medicamentos , Descoberta de Drogas , Humanos , Lipossomos/imunologia , Lipossomos/uso terapêutico , Polímeros/uso terapêutico , Vacinas de Partículas Semelhantes a Vírus/uso terapêutico
3.
J Virol ; 88(4): 2246-59, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24390324

RESUMO

Hantaviruses successfully replicate in primary human endothelial cells by restricting the early induction of beta interferon (IFN-ß) and interferon-stimulated genes (ISGs). Gn proteins from NY-1V, ANDV, and TULV, but not PHV, harbor elements in their 142-residue cytoplasmic tails (GnTs) that inhibit RIG-I/MAVS/TBK1-TRAF3-directed IFN-ß induction. Here, we define GnT interactions and residues required to inhibit TRAF3-TBK1-directed IFN-ß induction and IRF3 phosphorylation. We observed that GnTs bind TRAF3 via residues within the TRAF-N domain (residues 392 to 415) and that binding is independent of the MAVS-interactive TRAF-C domain (residues 415 to 568). We determined that GnT binding to TRAF3 is mediated by C-terminal degrons within NY-1V or ANDV GnTs and that mutations that add degrons to TULV or PHV GnTs confer TRAF3 binding. Further analysis of GnT domains revealed that TRAF3 binding is a discrete GnT function, independent of IFN regulation, and that residues 15 to 42 from the NY-1V GnT C terminus are required for inhibiting TBK1-directed IFN-ß transcription. Mutagenesis of the NY-1V GnT revealed that altering tyrosine 627 (Y627A/S/F) abolished GnT regulation of RIG-I/TBK1-directed IRF3 phosphorylation and transcriptional responses of ISRE, κB, and IFN-ß promoters. Moreover, GnTs from NY-1V, ANDV, and TULV, but not PHV, inhibited RIG-I-directed IRF3 phosphorylation. Collectively, these findings suggest a novel role for GnTs in regulating RIG-I/TBK1 pathway-directed IRF3 phosphorylation and IFN-ß induction and define virulence determinants within GnTs that may permit the attenuation of pathogenic hantaviruses. IMPORTANCE These findings provide a mechanism for selected hantavirus GnT interactions to regulate RIG-I/TBK1 signaling responses required for IFN-ß induction by inhibiting TBK1 phosphorylation of IRF3. These studies culminate in showing that a single GnT residue, Y627, is required for the NY-1V GnT to inhibit RIG-I/TBK1-directed IRF3 phosphorylation and IFN-ß induction. These findings define a potential virulence determinant within the NY-1V GnT that may permit hantavirus attenuation.


Assuntos
Regulação Viral da Expressão Gênica/imunologia , Interferon beta/antagonistas & inibidores , Orthohantavírus/genética , Transdução de Sinais/imunologia , Fator 3 Associado a Receptor de TNF/metabolismo , Proteínas do Envelope Viral/metabolismo , Replicação Viral/fisiologia , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Células HEK293 , Orthohantavírus/fisiologia , Humanos , Imunoprecipitação , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Mutagênese , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais/genética
4.
Proc Natl Acad Sci U S A ; 108(1): 319-24, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21173237

RESUMO

Rhabdoid tumors (RTs) are rare, highly aggressive pediatric malignancies with poor prognosis and with no standard or effective treatment strategies. RTs are characterized by biallelic inactivation of the INI1 tumor suppressor gene. INI1 directly represses CCND1 and activates cyclin-dependent kinase (cdk) inhibitors p16(Ink4a) and p21(CIP). RTs are exquisitely dependent on cyclin D1 for genesis and survival. To facilitate translation of unique therapeutic strategies, we have used genetically engineered, Ini1(+/-) mice for therapeutic testing. We found that PET can be used to noninvasively and accurately detect primary tumors in Ini1(+/-) mice. In a PET-guided longitudinal study, we found that treating Ini1(+/-) mice bearing primary tumors with the pan-cdk inhibitor flavopiridol resulted in complete and stable regression of some tumors. Other tumors showed resistance to flavopiridol, and one of the resistant tumors overexpressed cyclin D1, more than flavopiridol-sensitive cells. The concentration of flavopiridol used was not sufficient to down-modulate the high level of cyclin D1 and failed to induce cell death in the resistant cells. Furthermore, FISH and PCR analyses indicated that there is aneuploidy and increased CCND1 copy number in resistant cells. These studies indicate that resistance to flavopiridol may be correlated to elevated cyclin D1 levels. Our studies also indicate that Ini1(+/-) mice are valuable tools for testing unique therapeutic strategies and for understanding mechanisms of drug resistance in tumors that arise owing to loss of Ini1, which is essential for developing effective treatment strategies against these aggressive tumors.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Ciclina D1/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/genética , Tumor Rabdoide/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Primers do DNA/genética , Flavonoides/uso terapêutico , Inativação Gênica , Técnicas Histológicas , Immunoblotting , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Estudos Longitudinais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Piperidinas/uso terapêutico , Reação em Cadeia da Polimerase , Tomografia por Emissão de Pósitrons , Tumor Rabdoide/genética , Tumor Rabdoide/ultraestrutura , Proteína SMARCB1
5.
J Biol Chem ; 287(25): 20904-12, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22547065

RESUMO

Breast tumor kinase (Brk) was originally isolated from a human metastatic breast tumor, but also is found expressed in other epithelial tumors and in a subset of normal epithelia. Brk is a tyrosine kinase and its expression in breast carcinoma has been linked to tumor progression. The signal transducer and activator of transcription 3 (STAT3) is one of the substrate targets of Brk, and elevated tyrosine phosphorylation of STAT3 is known to contribute to oncogenesis. Conventional activation of STAT3 occurs in response to cytokine stimulation of Janus tyrosine kinases (JAK). One of the negative regulators discovered in cytokine signaling of the JAK-STAT pathway is the suppressor of cytokine signaling 3 (SOCS3). In this report we describe the finding that SOCS3 can also inhibit the unconventional target, Brk. Investigation of the mechanism by which SOCS3 inhibits Brk reveals the SOCS3 protein binds to Brk primarily via its SH2 domain, and its main inhibitory effect is mediated by the SOCS3 kinase inhibitory region (KIR). SOCS3 has only a modest effect on promoting Brk degradation, and this requires the C-terminal SOCS box domain. SOCS3 is the only known inhibitor of Brk, and knowledge of the mechanisms by which SOCS3 inhibits Brk may lead to methods that block Brk in cancer progression.


Assuntos
Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteólise , Fator de Transcrição STAT3/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/genética , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Neoplasias/genética , Ligação Proteica , Proteínas Tirosina Quinases/genética , Fator de Transcrição STAT3/genética , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Domínios de Homologia de src
6.
Expert Rev Vaccines ; 20(11): 1483-1498, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34148481

RESUMO

Introduction: Zika virus disease received little attention until its recent explosive emergence around the globe. The devastating consequences of this pandemic include congenital Zika syndrome (CZS) and the neurological autoimmune disorder Guillain-Barré syndrome. These potential outcomes prompted massive efforts to understand the course of Zika infection and to develop therapeutic and prophylactic strategies for treatment and prevention of disease.Area covered: Preclinical and clinical data demonstrate that a safe and efficacious vaccine for protection against Zika virus infection is possible in the near future. Nevertheless, significant knowledge gaps regarding the outcome of a mass vaccination strategy exist and must be addressed. Zika virus circulates in flavivirus-endemic regions, an ideal Zika vaccine should avoid the potential of antibody-dependent enhancement from exposure to dengue virus. Prevention of CZS is the primary goal for immunization, and the vaccine must provide protection against intrauterine transmission for use during pregnancy and in women of childbearing age. Ideally, a vaccine should also prevent sexual transmission of the virus through mucosal protection.Expert opinion: This review describes current vaccine approaches against Zika virus with particular attention to the application of virus-like particle (VLP) technology as a strategy for solving the challenges of Zika virus immunization.


Assuntos
Vacinas de Partículas Semelhantes a Vírus , Vacinas Virais , Infecção por Zika virus , Zika virus , Feminino , Humanos , Gravidez , Tecnologia , Infecção por Zika virus/prevenção & controle
7.
PLoS One ; 16(3): e0247394, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33651821

RESUMO

The inflammatory cytokine IL-6 is known to play a causal role in the promotion of cancer, although the underlying mechanisms remain to be completely understood. Interplay between endogenous and environmental cues determines the fate of cancer development. The Eµ-myc transgenic mouse expresses elevated levels of c-Myc in the B cell lineage and develops B cell lymphomas with associated mutations in p53 or other genes linked to apoptosis. We generated Eµ-myc mice that either lacked the IL-6 gene, or lacked the STAT3 gene specifically in B cells to determine the role of the IL-6/JAK/STAT3 pathway in tumor development. Using the Eµ-myc lymphoma mouse model, we demonstrate that IL-6 is a critical tumor promoter during early stages of B cell lymphomagenesis. IL-6 is shown to inhibit the expression of tumor suppressors, notably BIM and PTEN, and this may contribute to advancing MYC-driven B cell tumorigenesis. Several miRNAs known to target BIM and PTEN are upregulated by IL-6 and likely lead to the stable suppression of pro-apoptotic pathways early during the tumorigenic process. STAT3, a classical downstream effector of IL-6, appears dispensable for Eµ-myc driven lymphomagenesis. We conclude that the growth-promoting and anti-apoptotic mechanisms activated by IL-6 are critically involved in Eµ-myc driven tumor initiation and progression, but the B cell intrinsic expression of STAT3 is not required.


Assuntos
Interleucina-6/metabolismo , Linfoma de Células B/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose/genética , Linfócitos B/metabolismo , Morte Celular/genética , Genes myc , Interleucina-6/imunologia , Janus Quinases/metabolismo , Linfoma/patologia , Linfoma de Células B/genética , Linfoma de Células B/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Transcrição STAT3/fisiologia , Proteína Supressora de Tumor p53/metabolismo
8.
Pathogens ; 10(6)2021 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-34072726

RESUMO

Powassan virus (POWV) is a tick-borne flavivirus circulating in North America and the Russian Far East that can cause severe neuroinvasive diseases, including encephalitis, meningitis, and meningoencephalitis. The reported neuroinvasive case fatality is about 10%, and approximately 50% of the survivors from the neuroinfection exhibit long-lasting or permanent neurological sequelae. Currently, treatment of POWV infection is supportive, and no FDA-approved vaccines or specific therapeutics are available. A novel Powassan vaccine candidate was created using virus-like particle technology (POW-VLP) and assembled with the viral structural proteins pre-Membrane (prM) and Envelope (E). Western blot immunoassay demonstrated high antigenicity of POW-VLP structural proteins. Transmission electron microscopy indicated that the POW-VLP exhibited icosahedral morphology typical of flaviviruses. A dose-escalation study in a murine model was performed to test immunogenicity and safety. Serum antibody was tested by ELISA, demonstrating that POW-VLP afforded 100% seroconversion to the E protein. Reporter viral-particle neutralization assay demonstrated high levels of neutralizing antibodies in the serum of immunized mice. Hybridomas expressing monoclonal antibodies were produced following POW-VLP immunization. The POW-VLP vaccine candidate created in this study provides a strategy for inducing protective antibodies against Powassan neuroinvasive infection.

9.
BMC Cancer ; 10: 634, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-21092078

RESUMO

BACKGROUND: Rhabdoid Tumors (RTs) are highly aggressive pediatric malignancies with poor prognosis. There are currently no standard or effective treatments for RTs in part because treatments are not designed to specifically target these tumors. Our previous studies indicated that targeting the cyclin/cdk pathway is a novel therapeutic strategy for RTs and that a pan-cdk inhibitor, flavopiridol, inhibits RT growth. Since the toxicities and narrow window of activity associated with flavopiridol may limit its clinical use, we tested the effect of combining flavopiridol with 4-hydroxy-Tamoxifen (4OH-Tam) in order to reduce the concentration of flavopiridol needed for inhibition of RTs. METHODS: The effects of flavopiridol, 4OH-Tam, and their combination on RT cell cycle regulation and apoptosis were assessed by: i) cell survival assays, ii) FACS analysis, iii) caspase activity assays, and iv) immunoblot analysis. Furthermore, the role of p53 in flavopiridol- and 4OH-Tam-mediated induction of cell cycle arrest and apoptosis was characterized using RNA interference (siRNA) analysis. The effect of p53 on flavopiridol-mediated induction of caspases 2, 3, 8 and 9 was also determined. RESULTS: We found that the combination of flavopiridol and 4OH-Tam potently inhibited the growth of RT cells. Low nanomolar concentrations of flavopiridol induced G2 arrest, which was correlated to down-modulation of cyclin B1 and up-regulation of p53. Addition of 4OH-Tam did not affect flavopiridol-mediated G2 arrest, but enhanced caspase 3,7-mediated apoptosis induced by the drug. Abrogation of p53 by siRNA abolished flavopiridol-induced G2 arrest, but enhanced flavopiridol- (but not 4OH-Tam-) mediated apoptosis, by enhancing caspase 2 and 3 activities. CONCLUSIONS: Combining flavopiridol with 4OH-Tam potently inhibited the growth of RT cells by increasing the ability of either drug alone to induce caspases 2 and 3 thereby causing apoptosis. The potency of flavopiridol was enhanced by abrogation of p53. Our results warrant further studies investigating the combinatorial effects of flavopiridol and 4OH-Tam as a novel therapeutic strategy for RTs and other tumors that have been shown to respond to flavopiridol.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Flavonoides/administração & dosagem , Piperidinas/administração & dosagem , Tumor Rabdoide/tratamento farmacológico , Tamoxifeno/análogos & derivados , Apoptose , Caspases/biossíntese , Ciclo Celular , Sobrevivência Celular , Ensaios de Seleção de Medicamentos Antitumorais , Citometria de Fluxo , Fase G2 , Humanos , Interferência de RNA , Tamoxifeno/administração & dosagem , Proteína Supressora de Tumor p53/metabolismo
10.
Clin Cancer Res ; 14(2): 523-32, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18223228

RESUMO

PURPOSE: Rhabdoid tumors are aggressive and incurable pediatric malignancies. INI1/hSNF5, a tumor suppressor biallelically deleted/inactivated in rhabdoid tumors, directly represses cyclin D1. Rhabdoid tumors and cells are exquisitely dependent on cyclin D1 for genesis and survival, suggesting that targeting the cyclin/cyclin-dependent kinase (cdk) axis may be an effective therapeutic strategy for these tumors. Because cdk inhibitors have not been used for preclinical or clinical testing on rhabdoid tumors, we investigated the effect of flavopiridol, a pan-cdk inhibitor with promising clinical activity, on rhabdoid tumors. EXPERIMENTAL DESIGN: The effect of flavopiridol on rhabdoid cells was tested in vitro using survival, cell cycle, and apoptosis assays. Its effect was assessed in vivo using xenografted rhabdoid tumor models. Immunoblot and immunohistochemical analysis was used to assess the effect of flavopiridol on cyclin D1 and p21 expression in vitro and in vivo, respectively. RESULTS: Nanomolar concentrations of flavopiridol inhibited rhabdoid cell growth (IC(50) approximately 200 nmol/L), induced G(1) and G(2) arrest, and apoptosis in vitro in a concentration-dependent manner. These effects were correlated with the down-modulation of cyclin D1, up-regulation of p21, and induction of caspase 3/7 activities. Flavopiridol (at 7.5 mg/kg) significantly inhibited the growth of xenografted rhabdoid tumors, and its effect was correlated with the induction of p21 and down-modulation of cyclin D1. CONCLUSIONS: Flavopiridol is effective in inducing cell cycle arrest and cytotoxicity in rhabdoid tumors. Its effects are correlated with the down-regulation of cyclin D1 and the up-regulation of p21. Flavopiridol is potentially a novel chemotherapeutic agent for rhabdoid tumors.


Assuntos
Antineoplásicos/farmacologia , Flavonoides/farmacologia , Piperidinas/farmacologia , Tumor Rabdoide/tratamento farmacológico , Tumor Rabdoide/patologia , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Camundongos SCID , Transplante de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , Tumor Rabdoide/enzimologia , Tumor Rabdoide/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Cancer Res ; 13(16): 4721-30, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699849

RESUMO

PURPOSE: Rhabdoid tumors are rare but aggressive pediatric malignancies characterized by biallelic loss of INI1/hSNF5. Reintroduction of INI1 causes cell arrest and senescence in rhabdoid cells. Our purpose was to identify INI1-downstream genes and to determine their functional and therapeutic significance for rhabdoid tumors. EXPERIMENTAL DESIGN: INI1 downstream targets in rhabdoid cells were identified using a cDNA microarray analysis and the expression of selected INI1 targets was confirmed by quantitative reverse transcription-PCR, Western analysis, and/or immunohistochemical analysis of rhabdoid cells and primary rhabdoid tumors. To determine the functional significance of downstream targets, activated targets of INI1 were induced and repressed targets of INI1 were knocked down (by using RNA interference) in rhabdoid cells, in the absence of INI1. Consequence of altered expression of INI1 downstream targets for rhabdoid cell survival, cell cycle, and apoptosis was assessed. RESULTS: Microarray studies indicated that INI1 activated IFN-stimulated genes at early time points and senescence markers at late time points and repressed mitotic genes such as Polo like kinase 1 (PLK1), selectively in rhabdoid cells. Treatment of rhabdoid cells with recombinant IFNs resulted in induction of IFN-stimulated genes, G1 arrest, and flat cell formation. PLK1 was overexpressed in primary human and mouse rhabdoid tumors. RNA interference-mediated knock down of PLK1 in rhabdoid cells resulted in mitotic arrest, aberrant nuclear division, decreased survival, and induction of apoptosis. CONCLUSIONS: Targeting downstream effectors of INI1 such as IFN pathway and mitotic genes leads to antiproliferative effects in rhabdoid cells. IFN treatment and down-modulation of PLK1 constitute potential novel therapeutic strategies for rhabdoid tumors.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Proteínas de Ligação a DNA/fisiologia , Interferons/farmacologia , Tumor Rabdoide/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/fisiologia , Apoptose , Ciclo Celular , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Mitose , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Receptores de Interferon/fisiologia , Proteínas Recombinantes/farmacologia , Tumor Rabdoide/patologia , Proteína SMARCB1 , Fuso Acromático/metabolismo , Quinase 1 Polo-Like
12.
Vaccine ; 36(50): 7728-7736, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30377067

RESUMO

A dengue vaccine capable of rapidly eliciting a robust and balanced immunity against the four virus serotypes after only a few immunizations is greatly needed. We describe a new strategy to develop dengue vaccines based on the assembly of virus-like particles (VLPs) utilizing the structural proteins CprME together with a modified complex of the NS2B/NS3 protease, which enhances particle formation and yield. These VLPs are produced in mammalian cells and resemble native dengue virus as demonstrated by negative staining and immunogold labelling electron microscopy (EM). We found that VLPs produced at lower temperature (31 °C) were recognized by conformational monoclonal antibodies (MAbs) 4G2, 3H5 and C10 whereas VLPs produced at higher temperature (37 °C) were not recognized by these MAbs. To investigate the significance of these conformational discrepancies in vaccine performance, we tested the immunogenicity of VLP vaccines produced at 31 °C or 37 °C. Mice immunized with the VLP vaccine produced at 31 °C (VLP-31 °C) elicited the highest titer of neutralizing antibodies when compared to those elicited by equivalent doses of the vaccine produced at 37 °C (VLP-37 °C), inactivated dengue virus vaccine or to the titer of a human anti-dengue-2 convalescence serum reference. Our results demonstrate that the conformation of the E protein displayed on the VLP vaccine plays a critical role in the induction of highly neutralizing antibodies. These findings will guide development of a tetravalent vaccine capable of eliciting a robust and balanced neutralizing response against the four-dengue serotypes regardless of background immunity.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Dengue/imunologia , Tecnologia Farmacêutica/métodos , Temperatura , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/administração & dosagem , Antígenos Virais/imunologia , Dengue/prevenção & controle , Vacinas contra Dengue/administração & dosagem , Camundongos Endogâmicos BALB C , Conformação Proteica , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia
13.
Clin Vaccine Immunol ; 23(6): 451-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27030590

RESUMO

Respiratory syncytial virus (RSV) is the leading cause of severe respiratory disease in infants and children and represents an important global health burden for the elderly and the immunocompromised. Despite decades of research efforts, no licensed vaccine for RSV is available. We have developed virus-like particle (VLP)-based RSV vaccines assembled with the human metapneumovirus (hMPV) matrix protein (M) as the structural scaffold and the RSV fusion glycoprotein (F) in either the postfusion or prefusion conformation as its prime surface immunogen. Vaccines were composed of postfusion F, prefusion F, or a combination of the two conformations and formulated with a squalene-based oil emulsion as adjuvant. Immunization with these VLP vaccines afforded full protection against RSV infection and prevented detectable viral replication in the mouse lung after challenge. Analyses of lung cytokines and chemokines showed that VLP vaccination mostly induced the production of gamma interferon (IFN-γ), a marker of the Th1-mediated immune response, which is predominantly required for viral protection. Conversely, immunization with a formalin-inactivated RSV (FI-RSV) vaccine induced high levels of inflammatory chemokines and cytokines of the Th2- and Th17-mediated types of immune responses, as well as severe lung inflammation and histopathology. The VLP vaccines showed restricted production of these immune mediators and did not induce severe bronchiolitis or perivascular infiltration as seen with the FI-RSV vaccine. Remarkably, analysis of the serum from immunized mice showed that the VLP vaccine formulated using a combination of postfusion and prefusion F elicited the highest level of neutralizing antibody and enhanced the Th1-mediated immune response.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/química , Vírus Sinciciais Respiratórios/imunologia , Vacinas de Partículas Semelhantes a Vírus/química , Proteínas Virais de Fusão/química , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Citocinas/imunologia , Humanos , Imunização , Interferon gama/imunologia , Pulmão/imunologia , Pulmão/virologia , Metapneumovirus/química , Camundongos , Camundongos Endogâmicos BALB C , Conformação Proteica , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/química , Vírus Sinciciais Respiratórios/genética , Células Th17/imunologia , Células Th2/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/química , Vacinas de Produtos Inativados/genética , Vacinas de Produtos Inativados/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas Virais de Fusão/efeitos adversos , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Carga Viral , Proteínas da Matriz Viral/imunologia
14.
mBio ; 6(3): e00553-15, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25968648

RESUMO

UNLABELLED: Dengue virus (DENV) replication is inhibited by the prior addition of type I interferon or by RIG-I agonists that elicit RIG-I/MAVS/TBK1/IRF3-dependent protective responses. DENV infection of primary human endothelial cells (ECs) results in a rapid increase in viral titer, which suggests that DENV inhibits replication-restrictive RIG-I/interferon beta (IFN-ß) induction pathways within ECs. Our findings demonstrate that DENV serotype 4 (DENV4) nonstructural (NS) proteins NS2A and NS4B inhibited RIG-I-, MDA5-, MAVS-, and TBK1/IKKε-directed IFN-ß transcription (>80%) but failed to inhibit IFN-ß induction directed by STING or constitutively active IRF3-5D. Expression of NS2A and NS4B dose dependently inhibited the phosphorylation of TBK1 and IRF3, which suggests that they function at the level of TBK1 complex activation. NS2A and NS4B from DENV1/2/4, as well as the West Nile virus NS4B protein, commonly inhibited TBK1 phosphorylation and IFN-ß induction. A comparative analysis of NS4A proteins across DENVs demonstrated that DENV1, but not DENV2 or DENV4, NS4A proteins uniquely inhibited TBK1. These findings indicate that DENVs contain conserved (NS2A/NS4B) and DENV1-specific (NS4A) mechanisms for inhibiting RIG-I/TBK1-directed IFN responses. Collectively, our results define DENV NS proteins that restrict IRF3 and IFN responses and thereby facilitate DENV replication and virulence. Unique DENV1-specific NS4A regulation of IFN induction has the potential to be a virulence determinant that contributes to the increased severity of DENV1 infections and the immunodominance of DENV1 responses during tetravalent DENV1-4 vaccination. IMPORTANCE: Our findings demonstrate that NS2A and NS4B proteins from dengue virus serotypes 1, 2, and 4 are inhibitors of RIG-I/MDA5-directed interferon beta (IFN-ß) induction and that they accomplish this by blocking TBK1 activation. We determined that IFN inhibition is functionally conserved across NS4B proteins from West Nile virus and DENV1, -2, and -4 viruses. In contrast, DENV1 uniquely encodes an extra IFN regulating protein, NS4A, that inhibits TBK1-directed IFN induction. DENV1 is associated with an increase in severe patient disease, and added IFN regulation by the DENV1 NS4A protein may contribute to increased DENV1 replication, immunodominance, and virulence. The regulation of IFN induction by nonstructural (NS) proteins suggests their potential roles in enhancing viral replication and spread and as potential protein targets for viral attenuation. DENV1-specific IFN regulation needs to be considered in vaccine strategies where enhanced DENV1 replication may interfere with DENV2-4 seroconversion within coadministered tetravalent DENV1-4 vaccines.


Assuntos
Vírus da Dengue/imunologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Fator Regulador 3 de Interferon/metabolismo , Interferons/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas não Estruturais Virais/metabolismo , Vírus da Dengue/fisiologia , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transdução de Sinais
15.
mBio ; 5(1)2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24549848

RESUMO

UNLABELLED: Andes virus (ANDV) is the only hantavirus known to spread from person to person and shown to cause highly lethal hantavirus pulmonary syndrome (HPS) in patients and Syrian hamsters. Hantaviruses replicate in human endothelial cells and accomplish this by restricting the early induction of beta interferon (IFN-ß)- and IFN-stimulated genes (ISGs). Our studies reveal that the ANDV nucleocapsid (N) protein uniquely inhibits IFN signaling responses directed by cytoplasmic double-stranded RNA (dsRNA) sensors RIG-I and MDA5. In contrast, N proteins from Sin Nombre, New York-1, and Prospect Hill hantaviruses had no effect on RIG-I/MDA5-directed transcriptional responses from IFN-ß-, IFN-stimulated response element (ISRE)-, or κB-containing promoters. Ablating a potential S-segment nonstructural open reading frame (ORF) (NSs) within the ANDV plasmid expressing N protein failed to alter IFN regulation by ANDV N protein. Further analysis demonstrated that expressing the ANDV N protein inhibited downstream IFN pathway activation directed by MAVS, TBK1, and IκB kinase ε (IKKε) but failed to inhibit transcriptional responses directed by constitutive expression of active interferon regulatory factor IRF3-5D or after stimulation by alpha interferon (IFN-α) or tumor necrosis factor alpha (TNF-α). Consistent with IFN pathway-specific regulation, the ANDV N protein inhibited TBK1-directed IRF3 phosphorylation (phosphorylation of serine 396 [pS396]) and TBK1 autophosphorylation (pS172). Collectively, these findings indicate that the ANDV N inhibits IFN signaling responses by interfering with TBK1 activation, upstream of IRF3 phosphorylation and NF-κB activation. Moreover, our findings reveal that ANDV uniquely carries a gene encoding a virulence determinant within its N protein that is capable of restricting ISG and IFN-ß induction and provide a rationale for the novel pathogenesis and spread of ANDV. IMPORTANCE: Andes virus (ANDV) is distinguished from other hantaviruses by its unique ability to spread from person to person and cause lethal hantavirus pulmonary syndrome (HPS)-like disease in Syrian hamsters. However, virulence determinants that distinguish ANDV from other pathogenic hantaviruses have yet to be defined. Here we reveal that ANDV uniquely contains a virulence determinant within its nucleocapsid (N) protein that potently inhibits innate cellular signaling pathways. This novel function of the N protein provides a new mechanism for hantaviruses to regulate interferon (IFN) and IFN-stimulated gene (ISG) induction that is likely to contribute to the enhanced ability of ANDV to replicate, spread, and cause disease. These findings differentiate ANDV from other HPS-causing hantaviruses and provide a potential target for viral attenuation that needs to be considered in vaccine development.


Assuntos
Interações Hospedeiro-Patógeno , Imunidade Inata , Interferon beta/antagonistas & inibidores , Proteínas do Nucleocapsídeo/imunologia , Orthohantavírus/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Fatores de Virulência/imunologia , Linhagem Celular , Humanos , Evasão da Resposta Imune , Transdução de Sinais
16.
Virus Res ; 187: 65-71, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24412542

RESUMO

Hantaviruses predominantly replicate in primary human endothelial cells and cause 2 diseases characterized by altered barrier functions of vascular endothelium. Most hantaviruses restrict the early induction of interferon-ß (IFNß) and interferon stimulated genes (ISGs) within human endothelial cells to permit their successful replication. PHV fails to regulate IFN induction within human endothelial cells which self-limits PHV replication and its potential as a human pathogen. These findings, and the altered regulation of endothelial cell barrier functions by pathogenic hantaviruses, suggest that virulence is determined by the ability of hantaviruses to alter key signaling pathways within human endothelial cells. Our findings indicate that the Gn protein from ANDV, but not PHV, inhibits TBK1 directed ISRE, kB and IFNß induction through virulence determinants in the Gn cytoplasmic tail (GnT) that inhibit TBK1 directed IRF3 phosphorylation. Further studies indicate that in response to hypoxia induced VEGF, ANDV infection enhances the permeability and adherens junction internalization of microvascular and lymphatic endothelial cells. These hypoxia/VEGF directed responses are rapamycin sensitive and directed by mTOR signaling pathways. These results demonstrate the presence of at least two hantavirus virulence determinants that act on endothelial cell signaling pathways: one that regulates antiviral IFN signaling responses, and a second that enhances normal hypoxia-VEGF-mTOR signaling pathways to facilitate endothelial cell permeability. These findings suggest signaling pathways as potential targets for therapeutic regulation of vascular deficits that contribute to hantavirus diseases and viral protein targets for attenuating pathogenic hantaviruses.


Assuntos
Infecções por Hantavirus/imunologia , Hipóxia/imunologia , Fatores Reguladores de Interferon/genética , Interferon beta/antagonistas & inibidores , Orthohantavírus/patogenicidade , Antígenos CD/genética , Antígenos CD/imunologia , Caderinas/genética , Caderinas/imunologia , Capilares/patologia , Capilares/virologia , Permeabilidade Capilar , Células Endoteliais/imunologia , Células Endoteliais/patologia , Células Endoteliais/virologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Endotélio Vascular/virologia , Regulação da Expressão Gênica , Orthohantavírus/genética , Infecções por Hantavirus/complicações , Infecções por Hantavirus/genética , Infecções por Hantavirus/virologia , Interações Hospedeiro-Patógeno , Humanos , Hipóxia/complicações , Hipóxia/genética , Hipóxia/virologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Fatores Reguladores de Interferon/imunologia , Interferon beta/biossíntese , Interferon beta/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Virulência , Replicação Viral
17.
Methods Mol Biol ; 967: 189-202, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296731

RESUMO

The ability to observe the dynamic localization of a protein in living cells can provide critical insight to its mode of action and functional molecular interactions. To this purpose, green fluorescent protein (GFP) has served as a powerful tool to tag STAT proteins for microscopic visualization. Live cell imaging with STAT-GFP proteins has contributed to our understanding of signal transduction and the complexities of nuclear transport of STAT proteins. In this report we summarize recent approaches that use GFP-based techniques with live cell imaging to study the mechanisms of STAT nuclear import and export: photoactivation, fluorescence recovery after photobleaching (FRAP), and fluorescence loss in photobleaching (FLIP).


Assuntos
Núcleo Celular/metabolismo , Imagem Molecular/métodos , Fatores de Transcrição STAT/metabolismo , Transporte Ativo do Núcleo Celular/efeitos da radiação , Núcleo Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células HeLa , Humanos , Luz , Fotodegradação , Espectrometria de Fluorescência
18.
PLoS One ; 6(5): e20188, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21625522

RESUMO

The signal transducer and activator of transcription-3 (STAT3) induces transcription of genes that control differentiation, inflammation, proliferation, and tumor cell invasion. Cytokines such as interleukin-6 and interferon stimulate the specific tyrosine phosphorylation of STAT3, which confers its ability to bind consensus DNA targets. In addition, unphosphorylated STAT3 has been demonstrated to induce specific gene expression. STAT3 must gain entrance to the nucleus to impact transcription, however access to the nucleus is a tightly regulated process. Because nuclear trafficking is critical to the function of STAT3, we investigated the molecular mechanisms by which STAT3 is imported to the nucleus. Live cell imaging techniques were used with STAT3 tagged with green fluorescence protein (GFP) or photoactivatable GFP to follow the cellular dynamics of both unphosphorylated and tyrosine phosphorylated forms. Cytokine activation did not alter the rate of STAT3 nuclear import or nuclear export. In addition, Förster resonance energy transfer experiments revealed homomeric interaction of unphosphorylated STAT3 dependent on its amino terminus, but this dimerization is not necessary for its nuclear import. Previous work demonstrated the adapter importin-α3 binds to STAT3 and is required for nuclear import. To determine whether STAT3 nuclear import is mediated by the importin-α/importin-ß1 heterodimer, the effects of siRNA to importin-ß1 were evaluated. Results indicate STAT3 nuclear import is dependent on the function of importin-ß1. Since the Ran GTPase is necessary to bind importin-ß1 in the nucleus for release of importin-α-cargo, the effect of a GTPase deficient mutant of Ran was tested. Expression of the Ran interfering mutant inhibited STAT3 nuclear import. This study defines importin-α/importin-ß1/Ran as the molecular mechanism by which STAT3 traffics to the nucleus.


Assuntos
Núcleo Celular/metabolismo , Fator de Transcrição STAT3/metabolismo , beta Carioferinas/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Fosforilação , Transporte Proteico , Tirosina/metabolismo
19.
Cancer Res ; 71(9): 3225-35, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21521802

RESUMO

Rhabdoid tumors (RT) are aggressive pediatric malignancies with poor prognosis. INI1/hSNF5 is a component of the chromatin remodeling SWI/SNF complex and a tumor suppressor deleted in RT. Previous microarray studies indicated that reintroduction of INI1/hSNF5 into RT cells leads to repression of a high degree of mitotic genes including Aurora Kinase A (Aurora A, STK6). Here, we found that INI1/SNF5 represses Aurora A transcription in a cell-type-specific manner. INI1-mediated repression was observed in RT and normal cells but not in non-RT cell lines. Chromatin immunoprecipitation (ChIP) assay indicated that INI1/hSNF5 associates with Aurora A promoter in RT and normal cells but not in non-RT cells. Real-time PCR and immunohistochemical analyses of primary human and mouse RTs harboring mutations in INI1/hSNF5 gene indicated that Aurora A was overexpressed/derepressed in these tumor cells, confirming that INI1/hSNF5 represses Aurora A in vivo. Knockdown of Aurora A impaired cell growth, induced mitotic arrest and aberrant nuclear division leading to decreased survival, and increased cell death and caspase 3/7-mediated apoptosis in RT cells (but not in normal cells). These results indicated that Aurora A is a direct downstream target of INI1/hSNF5-mediated repression in RT cells and that loss of INI1/hSNF5 leads to aberrant overexpression of Aurora A in these tumors, which is required for their survival. We propose that a high degree of Aurora A expression may play a role in aggressive behavior of RTs and that targeting expression or activity of this gene is a novel therapeutic strategy for these tumors.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tumor Rabdoide/enzimologia , Fatores de Transcrição/metabolismo , Animais , Apoptose/fisiologia , Aurora Quinase A , Aurora Quinases , Caspase 3/metabolismo , Caspase 7/metabolismo , Processos de Crescimento Celular , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/biossíntese , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Células Jurkat , Camundongos , Mitose/fisiologia , Terapia de Alvo Molecular , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Tumor Rabdoide/genética , Tumor Rabdoide/patologia , Proteína SMARCB1 , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transfecção , Transplante Heterólogo
20.
Biochem Biophys Res Commun ; 360(3): 545-52, 2007 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-17606220

RESUMO

We have applied serial analysis of gene expression for studying the molecular mechanism of the rat liver regeneration in the model of 70% partial hepatectomy. We generated three SAGE libraries from a normal control liver (NL library: 52,343 tags), from a sham control operated liver (Sham library: 51,028 tags), and from a regenerating liver (PH library: 53,061 tags). By SAGE bioinformatics analysis we identified 40 induced genes and 20 repressed genes during the liver regeneration. We verified temporal expression of such genes by real time PCR during the regeneration process and we characterized 13 induced genes and 3 repressed genes. We found connective tissue growth factor transcript and protein induced very early at 4h after PH operation before hepatocytes proliferation is triggered. Our study suggests CTGF as a growth factor signaling mediator that could be involved directly in the mechanism of liver regeneration induction.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Regeneração Hepática/genética , Fígado/fisiologia , Animais , Biologia Computacional , Fator de Crescimento do Tecido Conjuntivo , Perfilação da Expressão Gênica/métodos , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Fígado/crescimento & desenvolvimento , Masculino , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA