Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(7): e0046923, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37310242

RESUMO

Due to increased and broadened screening efforts, the last decade has seen a rapid expansion in the number of viral species classified into the Hepacivirus genus. Conserved genetic features of hepaciviruses suggest that they have undergone specific adaptation and have evolved to hijack similar host proteins for efficient propagation in the liver. Here, we developed pseudotyped viruses to elucidate the entry factors of GB virus B (GBV-B), the first hepacivirus described in an animal after hepatitis C virus (HCV). GBV-B-pseudotyped viral particles (GBVBpp) were shown to be uniquely sensitive to the sera of tamarins infected with GBV-B, validating their usefulness as a surrogate for GBV-B entry studies. We screened GBVBpp infection of human hepatoma cell lines that were CRISPR/Cas9 engineered to ablate the expression of individual HCV receptors/entry factors and found that claudin-1 is essential for GBV-B infection, indicating the GBV-B and HCV share an entry factor. Our data suggest that claudin-1 facilitates HCV and GBV-B entry through distinct mechanisms since the former requires the first extracellular loop and the latter is reliant on a C-terminal region containing the second extracellular loop. The observation that claudin-1 is an entry factor shared between these two hepaciviruses suggests that the tight junction protein is of fundamental mechanistic importance during cell entry. IMPORTANCE Hepatitis C virus (HCV) is a major public health burden; approximately 58 million individuals have chronic HCV infection and are at risk of developing cirrhosis and liver cancer. To achieve the World Health Organization's target of eliminating hepatitis by 2030, new therapeutics and vaccines are needed. Understanding how HCV enters cells can inform the design of new vaccines and treatments targeting the first stage of infection. However, the HCV cell entry mechanism is complex and has been sparsely described. Studying the entry of related hepaciviruses will increase the knowledge of the molecular mechanisms of the first stages of HCV infection, such as membrane fusion, and inform structure-guided HCV vaccine design; in this work, we have identified a protein, claudin-1, that facilitates the entry of an HCV-related hepacivirus but with a mechanism not described for HCV. Similar work on other hepaciviruses may unveil a commonality of entry factors and, possibly, new mechanisms.


Assuntos
Vírus GB B , Hepatite C , Animais , Humanos , Hepacivirus/genética , Claudina-1/genética
2.
mSphere ; 8(1): e0056822, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36719225

RESUMO

Having varied approaches to the design and manufacture of vaccines is critical in being able to respond to worldwide needs and newly emerging pathogens. Virus-like particles (VLPs) form the basis of two of the most successful licensed vaccines (against hepatitis B virus [HBV] and human papillomavirus). They are produced by recombinant expression of viral structural proteins, which assemble into immunogenic nanoparticles. VLPs can be modified to present unrelated antigens, and here we describe a universal "bolt-on" platform (termed VelcroVax) where the capturing VLP and the target antigen are produced separately. We utilize a modified HBV core (HBcAg) VLP with surface expression of a high-affinity binding sequence (Affimer) directed against a SUMO tag and use this to capture SUMO-tagged gp1 glycoprotein from the arenavirus Junín virus (JUNV). Using this model system, we have solved the first high-resolution structures of VelcroVax VLPs and shown that the VelcroVax-JUNV gp1 complex induces superior humoral immune responses compared to the noncomplexed viral protein. We propose that this system could be modified to present a range of antigens and therefore form the foundation of future rapid-response vaccination strategies. IMPORTANCE The hepatitis B core protein (HBc) forms noninfectious virus-like particles, which can be modified to present a capturing molecule, allowing suitably tagged antigens to be bound on their surface. This system can be adapted and provides the foundation for a universal "bolt-on" vaccine platform (termed VelcroVax) that can be easily and rapidly modified to generate nanoparticle vaccine candidates.


Assuntos
Vacinas , Humanos , Antígenos do Núcleo do Vírus da Hepatite B/genética , Vírus da Hepatite B , Glicoproteínas , Vacinação
3.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31413132

RESUMO

Vaccines aimed at inducing T cell responses to protect against human immunodeficiency virus (HIV) infection have been under development for more than 15 years. Replication-defective adenovirus (rAd) vaccine vectors are at the forefront of this work and have been tested extensively in the simian immunodeficiency virus (SIV) challenge macaque model. Vaccination with rAd vectors coding for SIV Gag or other nonenvelope proteins induces T cell responses that control virus load but disappointingly is unsuccessful so far in preventing infection, and attention has turned to inducing antibodies to the envelope. However, here we report that Mauritian cynomolgus macaques (MCM), Macaca fascicularis, vaccinated with unmodified SIV gag alone in a DNA prime followed by an rAd boost exhibit increased protection from infection by repeated intrarectal challenge with low-dose SIVmac251. There was no evidence of infection followed by eradication. A significant correlation was observed between cytokine expression by CD4 T cells and delayed infection. Vaccination with gag fused to the ubiquitin gene or fragmented, designed to increase CD8 magnitude and breadth, did not confer resistance to challenge or enhance immunity. On infection, a significant reduction in peak virus load was observed in all vaccinated animals, including those vaccinated with modified gag These findings suggest that a nonpersistent viral vector vaccine coding for internal virus proteins may be able to protect against HIV type 1 (HIV-1) infection. The mechanisms are probably distinct from those of antibody-mediated virus neutralization or cytotoxic CD8 cell killing of virus-infected cells and may be mediated in part by CD4 T cells.IMPORTANCE The simian immunodeficiency virus (SIV) macaque model represents the best animal model for testing new human immunodeficiency virus type 1 (HIV-1) vaccines. Previous studies employing replication-defective adenovirus (rAd) vectors that transiently express SIV internal proteins induced T cell responses that controlled virus load but did not protect against virus challenge. However, we show for the first time that SIV gag delivered in a DNA prime followed by a boost with an rAd vector confers resistance to SIV intrarectal challenge. Other partially successful SIV/HIV-1 protective vaccines induce antibody to the envelope and neutralize the virus or mediate antibody-dependent cytotoxicity. Induction of CD8 T cells which do not prevent initial infection but eradicate infected cells before infection becomes established has also shown some success. In contrast, the vaccine described here mediates resistance by a different mechanism from that described above, which may reflect CD4 T cell activity. This could indicate an alternative approach for HIV-1 vaccine development.


Assuntos
Produtos do Gene gag/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Produtos do Gene gag/genética , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Macaca fascicularis , Masculino , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vacinação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Carga Viral
4.
J Gen Virol ; 99(12): 1717-1728, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30311877

RESUMO

Intra-host evolution of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) has been shown by viral RNA analysis in subjects who naturally suppress plasma viremia to low levels, known as controllers. However, little is known about the variability of proviral DNA and the inter-relationships among contained systemic viremia, rate of reservoir reseeding and specific major histocompatibility complex (MHC) genotypes, in controllers. Here, we analysed the proviral DNA quasispecies of the env V1-V2 region, in PBMCs and in anatomical compartments of 13 long-term controller monkeys after 3.2 years of infection with simian/human immunodeficiency virus (SHIV)SF162P4cy. A considerable variation in the genetic diversity of proviral quasispecies was present among animals. Seven monkeys exhibited env V1-V2 proviral populations composed of both clusters of identical ancestral sequences and new variants, whereas the other six monkeys displayed relatively high env V1-V2 genetic diversity with a large proportion of diverse novel sequences. Our results demonstrate that in SHIVSF162P4cy-infected monkeys there exists a disparate pattern of intra-host viral diversity and that reseeding of the proviral reservoir occurs in some animals. Moreover, even though no particular association has been observed between MHC haplotypes and the long-term control of infection, a remarkably similar pattern of intra-host viral diversity and divergence was found within animals carrying the M3 haplotype. This suggests that in animals bearing the same MHC haplotype and infected with the same virus, viral diversity follows a similar pattern with similar outcomes and control of infection.


Assuntos
Produtos do Gene env/genética , Variação Genética , HIV/genética , Provírus/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Animais , Genótipo , Leucócitos Mononucleares/virologia , Macaca fascicularis , Complexo Principal de Histocompatibilidade/genética , Quase-Espécies
5.
J Virol ; 89(23): 12131-44, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26401036

RESUMO

UNLABELLED: Hepatitis C virus (HCV) only infects humans and chimpanzees, while GB virus B (GBV-B), another hepatotropic hepacivirus, infects small New World primates (tamarins and marmosets). In an effort to develop an immunocompetent small primate model for HCV infection to study HCV pathogenesis and vaccine approaches, we investigated the HCV life cycle step(s) that may be restricted in small primate hepatocytes. First, we found that replication-competent, genome-length chimeric HCV RNAs encoding GBV-B structural proteins in place of equivalent HCV sequences designed to allow entry into simian hepatocytes failed to induce viremia in tamarins following intrahepatic inoculation, nor did they lead to progeny virus in permissive, transfected human Huh7.5 hepatoma cells upon serial passage. This likely reflected the disruption of interactions between distantly related structural and nonstructural proteins that are essential for virion production, whereas such cross talk could be restored in similarly designed HCV intergenotypic recombinants via adaptive mutations in NS3 protease or helicase domains. Next, HCV entry into small primate hepatocytes was examined directly using HCV-pseudotyped retroviral particles (HCV-pp). HCV-pp efficiently infected tamarin hepatic cell lines and primary marmoset hepatocyte cultures through the use of the simian CD81 ortholog as a coreceptor, indicating that HCV entry is not restricted in small New World primate hepatocytes. Furthermore, we observed genomic replication and modest virus secretion following infection of primary marmoset hepatocyte cultures with a highly cell culture-adapted HCV strain. Thus, HCV can successfully complete its life cycle in primary simian hepatocytes, suggesting the possibility of adapting some HCV strains to small primate hosts. IMPORTANCE: Hepatitis C virus (HCV) is an important human pathogen that infects over 150 million individuals worldwide and leads to chronic liver disease. The lack of a small animal model for this infection impedes the development of a preventive vaccine and pathogenesis studies. In seeking to establish a small primate model for HCV, we first attempted to generate recombinants between HCV and GB virus B (GBV-B), a hepacivirus that infects small New World primates (tamarins and marmosets). This approach revealed that the genetic distance between these hepaciviruses likely prevented virus morphogenesis. We next showed that HCV pseudoparticles were able to infect tamarin or marmoset hepatocytes efficiently, demonstrating that there was no restriction in HCV entry into these simian cells. Furthermore, we found that a highly cell culture-adapted HCV strain was able to achieve a complete viral cycle in primary marmoset hepatocyte cultures, providing a promising basis for further HCV adaptation to small primate hosts.


Assuntos
Vírus GB B/fisiologia , Hepacivirus/fisiologia , Estágios do Ciclo de Vida/fisiologia , Modelos Animais , Primatas/virologia , Internalização do Vírus , Animais , Sequência de Bases , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Células HEK293 , Hepacivirus/genética , Hepatócitos/virologia , Especificidade de Hospedeiro , Humanos , Immunoblotting , Dados de Sequência Molecular , Plasmídeos/genética , Análise de Sequência de DNA , Viremia
6.
PLoS One ; 9(8): e104390, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25162725

RESUMO

Vaccination of Mauritian cynomolgus macaques with the attenuated nef-truncated C8 variant of SIVmac251/32H (SIVmacC8) induces early, potent protection against pathogenic, heterologous challenge before the maturation of cognate immunity. To identify processes that contribute to early protection in this model the pathogenesis, anatomical distribution and viral vaccine kinetics were determined in relation to localised innate responses triggered by vaccination. The early biodistribution of SIVmacC8 was defined by rapid, widespread dissemination amongst multiple lymphoid tissues, detectable after 3 days. Cell-associated viral RNA dynamics identified mesenteric lymph nodes (MLN) and spleen, as well as the gut mucosae, as early major contributors of systemic virus burden. Rapid, localised infection was populated by discrete foci of persisting virus-infected cells. Localised productive infection triggered a broad innate response, with type-1 interferon sensitive IRF-7, STAT-1, TRIM5α and ApoBEC3G genes all upregulated during the acute phase but induction did not prevent viral persistence. Profound changes in vaccine-induced cell-surface markers of immune activation were detected on macrophages, B-cells and dendritic cells (DC-SIGN, S-100, CD40, CD11c, CD123 and CD86). Notably, high DC-SIGN and S100 staining for follicular and interdigitating DCs respectively, in MLN and spleen were detected by 3 days, persisting 20 weeks post-vaccination. Although not formally evaluated, the early biodistribution of SIVmacC8 simultaneously targets multiple lymphoid tissues to induce strong innate immune responses coincident at the same sites critical for early protection from wild-type viruses. HIV vaccines which stimulate appropriate innate, as well as adaptive responses, akin to those generated by live attenuated SIV vaccines, may prove the most efficacious.


Assuntos
Imunidade Inata/efeitos dos fármacos , Vacinas contra a SAIDS/imunologia , Vacinas contra a SAIDS/farmacocinética , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Imunidade Adaptativa/efeitos dos fármacos , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/virologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/virologia , Macaca fascicularis , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/virologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Proteínas S100/genética , Proteínas S100/imunologia , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/biossíntese , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Baço/citologia , Baço/imunologia , Baço/virologia , Vacinas Atenuadas , Carga Viral/efeitos dos fármacos , Dedos de Zinco/genética , Dedos de Zinco/imunologia
7.
PLoS One ; 9(4): e93235, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24695530

RESUMO

Little is known about the effects of Major Histocompatibility Complex (MHC) haplotypes on immunity to primate lentiviruses involving both acquired and innate immune responses. We present statistical evidence of the influence of MHC polymorphism on antiviral immunity of Mauritian cynomolgus macaques (MCM) following simian/human immunodeficiency virus SHIVSF162P4cy infection, involving the production of pro- and anti-inflammatory cytokines and α-defensins, which may modulate acquired immune responses. During the acute phase of infection, IL-10 correlated positively with viral load and negatively with CD4+T cell counts. Furthermore, α-defensins production was directly correlated with plasma viral RNA, particularly at peak of viral load. When the effects of the MHC were analyzed, a significant association between lower anti-Env binding and neutralizing antibody levels with class IB M4 haplotype and with class IA, IB M4 haplotype, respectively, was observed in the post-acute phase. Lower antibody responses may have resulted into a poor control of infection thus explaining the previously reported lower CD4 T cell counts in these monkeys. Class II M3 haplotype displayed significantly lower acute and post-acute IL-10 levels. In addition, significantly lower levels of α-defensins were detected in class IA M3 haplotype monkeys than in non-M3 macaques, in the post-acute phase of infection. These data indicate that the MHC could contribute to the delicate balance of pro-inflammatory mechanisms, particularly with regard to the association between IL-10 and α-defensins in lentivirus infection. Our results show that host genetic background, virological and immunological parameters should be considered for the design and interpretation of HIV-1 vaccine efficacy studies.


Assuntos
Haplótipos/imunologia , Macaca fascicularis/imunologia , Macaca/imunologia , Complexo Principal de Histocompatibilidade/genética , Complexo Principal de Histocompatibilidade/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Anticorpos Neutralizantes/genética , Linfócitos T CD4-Positivos/imunologia , HIV/imunologia , Haplótipos/genética , Interleucina-10/genética , Interleucina-10/imunologia , Macaca/genética , Macaca/virologia , Macaca fascicularis/genética , Masculino , RNA Viral/genética , RNA Viral/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Carga Viral/genética , Carga Viral/imunologia , alfa-Defensinas/genética
8.
Virol J ; 10: 326, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24180225

RESUMO

BACKGROUND: Foamy viruses are non-pathogenic in vivo and naturally infect all species of non-human primates (NHP). Simian foamy viruses (SFV) are highly prevalent in both free ranging and captive NHP but few longitudinal studies have been performed to assess the prevalence and biodistribution of SFV within captive NHP. METHOD: LTR and pol gene along with Gag antibody detection were undertaken to identify infection in a cohort of over 80 captive macaques. RESULTS: The prevalence of SFV was between 64% and 94% in different groups. Access to 23 dam-infant pairs allowed us to reveal horizontal transfer as the dominant route of SFV transmission in our cohort. Further, analysis of SFV from a range of tissues and blood revealed that macaques as young as six months old can be infected and that proviral biodistribution increases with age. CONCLUSIONS: These are the first data of this type for a captive cohort of cynomolgus macaques.


Assuntos
Transmissão de Doença Infecciosa , Macaca fascicularis/virologia , Infecções por Retroviridae/veterinária , Spumavirus/classificação , Spumavirus/genética , Animais , Anticorpos Antivirais/sangue , Análise por Conglomerados , Feminino , Produtos do Gene gag/imunologia , Produtos do Gene pol/genética , Masculino , Epidemiologia Molecular , Dados de Sequência Molecular , Filogenia , Prevalência , RNA Viral/genética , Infecções por Retroviridae/epidemiologia , Infecções por Retroviridae/transmissão , Infecções por Retroviridae/virologia , Análise de Sequência de DNA , Spumavirus/isolamento & purificação , Sequências Repetidas Terminais
9.
PLoS One ; 7(11): e50397, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226275

RESUMO

Live attenuated measles virus is one of the most efficient and safest vaccines available, making it an attractive candidate vector for a HIV/AIDS vaccine aimed at eliciting cell-mediated immune responses (CMI). Here we have characterized the potency of CMI responses generated in mice and non-human primates after intramuscular immunisation with a candidate recombinant measles vaccine carrying an HIV-1 insert encoding Clade B Gag, RT and Nef (MV1-F4). Eight Mauritian derived, MHC-typed cynomolgus macaques were immunised with 10(5) TCID(50) of MV1-F4, four of which were boosted 28 days later with the same vaccine. F4 and measles virus (MV)-specific cytokine producing T cell responses were detected in 6 and 7 out of 8 vaccinees, respectively. Vaccinees with either M6 or recombinant MHC haplotypes demonstrated the strongest cytokine responses to F4 peptides. Polyfunctional analysis revealed a pattern of TNFα and IL-2 responses by CD4+ T cells and TNFα and IFNγ responses by CD8+ T cells to F4 peptides. HIV-specific CD4+ and CD8+ T cells expressing cytokines waned in peripheral blood lymphocytes by day 84, but CD8+ T cell responses to F4 peptides could still be detected in lymphoid tissues more than 3 months after vaccination. Anti-F4 and anti-MV antibody responses were detected in 6 and 8 out of 8 vaccinees, respectively. Titres of anti-F4 and MV antibodies were boosted in vaccinees that received a second immunisation. MV1-F4 carrying HIV-1 Clade B inserts induces robust boostable immunity in non-human primates. These results support further exploration of the MV1-F4 vector modality in vaccination strategies that may limit HIV-1 infectivity.


Assuntos
Vacinas contra a AIDS/imunologia , Síndrome da Imunodeficiência Adquirida/prevenção & controle , Anticorpos Antivirais/biossíntese , HIV-1/imunologia , Imunização Secundária , Vacina contra Sarampo/genética , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Síndrome da Imunodeficiência Adquirida/imunologia , Síndrome da Imunodeficiência Adquirida/virologia , Animais , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Produtos do Gene gag/genética , Produtos do Gene gag/imunologia , Humanos , Imunidade Celular , Memória Imunológica , Injeções Intramusculares , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-2/biossíntese , Interleucina-2/imunologia , Macaca fascicularis , Masculino , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/imunologia , Camundongos , DNA Polimerase Dirigida por RNA/genética , DNA Polimerase Dirigida por RNA/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia , Vacinas Sintéticas , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia
10.
Virology ; 413(2): 161-8, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21349567

RESUMO

Immunopathology during early simian retrovirus type 2 (SRV-2) infection is poorly characterized. Here, viral dynamics, immune response and disease progression in transiently- or persistently-infected cynomolgus macaques are assessed. Viral nucleic acids were detected in selected lymphoid tissues of both persistently- and transiently-infected macaques, even after viral clearance from the periphery. Immunohistochemical staining of lymphoid tissues revealed alterations in a number of immune cell populations in both transiently- and persistently-infected macaques. The precise pattern depended upon the infection status of the macaque and the marker studied. Gross immunopathological changes in lymphoid tissues were similar between SRV infection and those observed for other simian retroviruses SIV and STLV, suggesting a common immunopathological response to infection with these agents.


Assuntos
Macaca fascicularis , Vírus dos Macacos de Mason-Pfizer , Doenças dos Macacos/imunologia , Infecções por Retroviridae/veterinária , Infecções Tumorais por Vírus/veterinária , Animais , DNA Viral , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Vírus dos Macacos de Mason-Pfizer/genética , Vírus dos Macacos de Mason-Pfizer/imunologia , Vírus dos Macacos de Mason-Pfizer/isolamento & purificação , Mesentério/patologia , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Baço/imunologia , Baço/patologia , Baço/virologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia , Carga Viral
11.
J Biol Chem ; 284(1): 505-514, 2009 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18990693

RESUMO

Rhesus rhadinovirus (RRV) is currently the closest known, fully sequenced homolog of human Kaposi sarcoma-associated herpesvirus. Both these viruses encode complement inhibitors as follows: Kaposi sarcoma-associated herpesvirus-complement control protein (KCP) and RRV-complement control protein (RCP). Previously we characterized in detail the functional properties of KCP as a complement inhibitor. Here, we performed comparative analyses for two variants of RCP protein, encoded by RRV strains H26-95 and 17577. Both RCP variants and KCP inhibited human and rhesus complement when tested in hemolytic assays measuring all steps of activation via the classical and the alternative pathway. RCP variants from both RRV strains supported C3b and C4b degradation by factor I and decay acceleration of the classical C3 convertase, similar to KCP. Additionally, the 17577 RCP variant accelerated decay of the alternative C3 convertase, which was not seen for KCP. In contrast to KCP, RCP showed no affinity to heparin and is the first described complement inhibitor in which the binding site for C3b/C4b does not interact with heparin. Molecular modeling shows a structural disruption in the region of RCP that corresponds to the KCP-heparin-binding site. This makes RRV a superior model for future in vivo investigations of complement evasion, as RCP does not play a supportive role in viral attachment as KCP does.


Assuntos
Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Herpesvirus Saimiriíneo 2/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Proteínas Virais/metabolismo , Animais , Sítios de Ligação , Células CHO , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/genética , Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/genética , Cricetinae , Cricetulus , Heparina/química , Heparina/metabolismo , Herpesvirus Saimiriíneo 2/química , Herpesvirus Saimiriíneo 2/genética , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Macaca mulatta , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Especificidade da Espécie , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/genética , Ligação Viral
12.
Retrovirology ; 4: 24, 2007 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-17407584

RESUMO

BACKGROUND: Rapamycin is an immunosuppressive which represses translation of transcripts harbouring a polypyrimidine motif downstream of the mRNA cap site through the mammalian target of rapamycin complex. It inhibits the abnormal autologous proliferation of T-cell clones containing a transcriptionally active human T-lymphotropic virus, type I (HTLV-I) provirus, generated from infected subjects. We showed previously that this effect is independent of the polypyrimidine motifs in the viral long terminal repeat (LTR) R region suggesting that HTLV-I transcription, and not translation, is being affected. Here we studied whether rapamycin is having an effect on a specific transcription factor pathway. Further, we investigated whether mRNAs encoding transcription factors involved in HTLV-I transcriptional activation, specifically CREB, Ets and c-Myb, are implicated in the rapamycin-sensitivity of the HTLV-I LTR. RESULTS: An in vitro analysis of the role of SRE- and NF-kappaB-mediated transcription highlighted the latter as rapamycin sensitive. Over-expression of c-Myb reversed the rapamycin effect. CONCLUSION: The sensitivity of HTLV-I transcription to rapamycin may be effected through an NF-kappaB-pathway associated with the rapamycin-sensitive mTORC1 cellular signalling network.


Assuntos
Vírus Linfotrópico T Tipo 1 Humano/efeitos dos fármacos , Vírus Linfotrópico T Tipo 1 Humano/genética , Proteínas Proto-Oncogênicas c-myb/fisiologia , Sirolimo/farmacologia , Sequências Repetidas Terminais/efeitos dos fármacos , Fusão Gênica Artificial , Cloranfenicol O-Acetiltransferase/biossíntese , Deltaretrovirus , Genes Reporter , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , NF-kappa B/antagonistas & inibidores , Transcrição Gênica/efeitos dos fármacos
13.
J Gen Virol ; 82(Pt 2): 435-439, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11161283

RESUMO

The immunosuppressant rapamycin can regulate the translation of a subset of messenger RNAs, a phenotype which has been linked to the presence of a polypyrimidine motif [C(N)(4-14)] downstream of the mRNA cap structure. T-cell clones naturally infected with transcriptionally active human T-cell leukaemia virus, type I (HTLV-I) undergo autologous proliferation; this phenotype is inhibited by rapamycin but not FK506, which reverses the rapamycin effect. Within the R region of the HTLV-I 5' long terminal repeat (LTR) there are seven polypyrimidine motifs. We sought to determine if these were involved in the sensitivity of proliferation to the presence of rapamycin. Here we illustrate the generation of an in vitro model of this rapamycin-sensitivity and the analysis of LTR mutants which were created to determine the importance of the polypyrimidine motifs. Reporter gene assays suggest the effect is independent of the polypyrimidine motifs in the virus leader sequence.


Assuntos
Vírus Linfotrópico T Tipo 1 Humano/efeitos dos fármacos , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Imunossupressores/farmacologia , Sirolimo/farmacologia , Linfócitos T/efeitos dos fármacos , Sequências Repetidas Terminais/genética , Animais , Sequência de Bases , Células COS , Divisão Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Produtos do Gene rex/genética , Produtos do Gene rex/metabolismo , Produtos do Gene tax/genética , Produtos do Gene tax/metabolismo , Genes Reporter/genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Células Jurkat , Modelos Biológicos , Mutação/genética , Sirolimo/antagonistas & inibidores , Linfócitos T/citologia , Tacrolimo/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA