Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
Curr Opin Virol ; 7: 134-40, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25063986

RESUMO

The endothelial lining of the vasculature performs a vital role in maintaining fluid barrier functions despite balancing nutrient and fluid content of tissues, repairing localized damage, coordinating responses of a plethora of factors, immune cells and platelets through a multitude of endothelial cell surface receptors. Viruses that nonlytically cause lethal hemorrhagic or edematous diseases engage receptors on vascular and lymphatic endothelial cells, altering normal cellular responses that control capillary leakage and fluid clearance functions with lethal consequences. Recent studies indicate that receptors directing dengue virus and hantavirus infection of the endothelium contribute to the dysregulation of normal endothelial cell signaling responses that control capillary permeability and immune responses that contribute to pathogenesis. Here we present recent studies of virally altered endothelial functions that provide new insight into targeting barrier functions of the endothelium as a potential therapeutic approach.


Assuntos
Vírus da Dengue/metabolismo , Dengue/metabolismo , Células Endoteliais/virologia , Infecções por Hantavirus/metabolismo , Orthohantavírus/metabolismo , Receptores Virais/metabolismo , Animais , Dengue/virologia , Vírus da Dengue/genética , Vírus da Dengue/patogenicidade , Orthohantavírus/genética , Orthohantavírus/patogenicidade , Infecções por Hantavirus/virologia , Humanos
3.
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
4.
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
5.
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
6.
Lymphat Res Biol ; 11(3): 128-35, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24024573

RESUMO

BACKGROUND: Hantaviruses in the Americas cause a highly lethal acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). Hantaviruses nonlytically infect microvascular and lymphatic endothelial cells and cause dramatic changes in barrier functions without disrupting the endothelium. Hantaviruses cause changes in the function of infected endothelial cells that normally regulate fluid barrier functions. The endothelium of arteries, veins, and lymphatic vessels are unique and central to the function of vast pulmonary capillary beds that regulate pulmonary fluid accumulation. RESULTS: We have found that HPS-causing hantaviruses alter vascular barrier functions of microvascular and lymphatic endothelial cells by altering receptor and signaling pathway responses that serve to permit fluid tissue influx and clear tissue edema. Infection of the endothelium provides several mechanisms for hantaviruses to cause acute pulmonary edema, as well as potential therapeutic targets for reducing the severity of HPS disease. CONCLUSIONS: Here we discuss interactions of HPS-causing hantaviruses with the endothelium, roles for unique lymphatic endothelial responses in HPS, and therapeutic targeting of the endothelium as a means of reducing the severity of HPS disease.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais/fisiologia , Síndrome Pulmonar por Hantavirus/tratamento farmacológico , Animais , Anticorpos/imunologia , Anticorpos/uso terapêutico , Cricetinae , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Cloridrato de Fingolimode , Orthohantavírus/fisiologia , Síndrome Pulmonar por Hantavirus/metabolismo , Síndrome Pulmonar por Hantavirus/fisiopatologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imunossupressores/uso terapêutico , Mesocricetus , Propilenoglicóis/uso terapêutico , Edema Pulmonar/tratamento farmacológico , Edema Pulmonar/metabolismo , Edema Pulmonar/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/uso terapêutico , Esfingosina/análogos & derivados , Esfingosina/uso terapêutico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
Adv Virol ; 2012: 840654, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952474

RESUMO

Dengue viruses cause two severe diseases that alter vascular fluid barrier functions, dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). The endothelium is the primary fluid barrier of the vasculature and ultimately the effects of dengue virus infection that cause capillary leakage impact endothelial cell (EC) barrier functions. The ability of dengue virus to infect the endothelium provides a direct means for dengue to alter capillary permeability, permit virus replication, and induce responses that recruit immune cells to the endothelium. Recent studies focused on dengue virus infection of primary ECs have demonstrated that ECs are efficiently infected, rapidly produce viral progeny, and elicit immune enhancing cytokine responses that may contribute to pathogenesis. Furthermore, infected ECs have also been implicated in enhancing viremia and immunopathogenesis within murine dengue disease models. Thus dengue-infected ECs have the potential to directly contribute to immune enhancement, capillary permeability, viremia, and immune targeting of the endothelium. These effects implicate responses of the infected endothelium in dengue pathogenesis and rationalize therapeutic targeting of the endothelium and EC responses as a means of reducing the severity of dengue virus disease.

8.
J Virol ; 86(12): 6408-15, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22496214

RESUMO

Dengue viruses cause two severe diseases that alter vascular fluid barrier functions, dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Preexisting antibodies to dengue virus disposes patients to immune-enhanced edema (DSS) or hemorrhagic (DHF) disease following infection by a discrete dengue virus serotype. Although the endothelium is the primary vascular fluid barrier, direct effects of dengue virus on endothelial cells (ECs) have not been considered primary factors in pathogenesis. Here, we show that dengue virus infection of human ECs elicits immune-enhancing EC responses. Our results suggest that rapid early dengue virus proliferation within ECs is permitted by dengue virus regulation of early, but not late, beta interferon (IFN-ß) responses. The analysis of EC responses following synchronous dengue virus infection revealed the high-level induction and secretion of immune cells (T cells, B cells, and mast cells) as well as activating and recruiting cytokines BAFF (119-fold), IL-6/8 (4- to 7-fold), CXCL9/10/11 (45- to 338-fold), RANTES (724-fold), and interleukin-7 (IL-7; 128-fold). Moreover, we found that properdin factor B, an alternative pathway complement activator that directs chemotactic anaphylatoxin C3a and C5a production, was induced 34-fold. Thus, dengue virus-infected ECs evoke key inflammatory responses observed in dengue virus patients which are linked to DHF and DSS. Our findings suggest that dengue virus-infected ECs directly contribute to immune enhancement, capillary permeability, viremia, and immune targeting of the endothelium. These data implicate EC responses in dengue virus pathogenesis and further rationalize therapeutic targeting of the endothelium as a means of reducing the severity of dengue virus disease.


Assuntos
Vírus da Dengue/fisiologia , Dengue/imunologia , Dengue/virologia , Células Endoteliais/imunologia , Aedes , Animais , Linhagem Celular , Citocinas/genética , Citocinas/imunologia , Dengue/genética , Vírus da Dengue/imunologia , Células Endoteliais/virologia , Feminino , Humanos
9.
Biochemistry ; 43(31): 9939-49, 2004 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-15287721

RESUMO

Eg5 is a kinesin-like motor protein required for mitotic progression in higher eukaryotes. It is thought to cross-link antiparallel microtubules, and provides a force required for the formation of a bipolar spindle. Monastrol causes the catastrophic collapse of the mitotic spindle through the allosteric inhibition of Eg5. Utilizing a truncated Eg5 protein, we employ difference infrared spectroscopy to probe structural changes that occur in the motor protein with monastrol in the presence of either ADP or ATP. Difference FT-IR spectra of Eg5-monastrol-nucleotide complexes demonstrate that there are triggered conformational changes corresponding to an interconversion of secondary structural elements in the motor upon interaction with nucleotides. Notably, conformational changes elicited in the presence of ADP are different from those in the presence of ATP. In Eg5-monastrol complexes, exchange of ADP is associated with a decrease in random structure and an increase in alpha-helical content. In contrast, formation of the Eg5-monastrol-ATP complex is associated with a decrease in alpha-helical content and a concomitant increase in beta-sheet content. One or more carboxylic acid residues in Eg5 undergo unique changes when ATP, but not ADP, interacts with the motor domain in the presence of monastrol. This first direct dissection of inhibitor-protein interactions, using these methods, demonstrates a clear disparity in the structural consequences of monastrol in the presence of ADP versus ATP.


Assuntos
Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/química , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/química , Cinesinas/metabolismo , Nitrobenzenos/química , Pirimidinas/metabolismo , Tionas/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Amidas/química , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/química , Substâncias Macromoleculares , Dados de Sequência Molecular , Nitrobenzenos/metabolismo , Fosfatos/química , Fosfatos/metabolismo , Fotólise , Ligação Proteica , Pirimidinas/química , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Tionas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...