Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(4): e0240495, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33914754

RESUMO

RATIONALE/STUDY DESIGN: A major challenge in the development of HIV vaccines is finding immunogens that elicit protection against a broad range of viral strains. Immunity to a narrow range of viral strains may protect infants of HIV-infected women or partners discordant for HIV. We hypothesized that immunization to the relevant viral variants could be achieved by exposure to infectious virus during prophylaxis with antiretroviral drugs. To explore this approach in an animal model, macaques were exposed to live virus (SIVmne or HIV-2287) during prophylaxis with parenteral tenofovir and humoral and cellular immune responses were quantified. Subsequently, experimental animals were challenged with homologous virus to evaluate protection from infection, and if infection occurred, the course of disease was compared to control animals. Experimental animals uninfected with SIVmne were challenged with heterologous HIV-2287 to assess resistance to retroviral infection. METHODOLOGY/PRINCIPAL FINDINGS: Juvenile female Macaca nemestrina (N = 8) were given ten weekly intravaginal exposures with either moderately (SIVmne) or highly (HIV-2287) pathogenic virus during tenofovir prophylaxis. Tenofovir protected all 8 experimental animals from infection, while all untreated control animals became infected. Specific non-neutralizing antibodies were elicited in blood and vaginal secretions of experimental animals, but no ELISPOT responses were detected. Six weeks following the cessation of tenofovir, intravaginal challenge with homologous virus infected 2/4 (50%) of the SIVmne-immunized animals and 4/4 (100%) of the HIV-2287-immunized animals. The two SIVmne-infected and 3 (75%) HIV-2287-infected had attenuated disease, suggesting partial protection. CONCLUSIONS/SIGNIFICANCE: Repeated exposure to SIVmne or HIV-2287, during antiretroviral prophylaxis that blocked infection, induced binding antibodies in the blood and mucosa, but not neutralizing antibodies or specific cellular immune responses. Studies to determine whether antibodies are similarly induced in breastfeeding infants and sexual partners discordant for HIV infection and receiving pre-exposure antiretroviral prophylaxis are warranted, including whether these antibodies appear to confer partial or complete protection from infection.


Assuntos
Antirretrovirais/uso terapêutico , Infecções por HIV/prevenção & controle , HIV-2/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Tenofovir/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Infecções por HIV/imunologia , HIV-2/imunologia , Humanos , Imunização/métodos , Macaca nemestrina , Profilaxia Pré-Exposição/métodos , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia
2.
J Virol ; 87(24): 13676-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109218

RESUMO

The complete sequence of retroperitoneal fibromatosis-associated herpesvirus Macaca nemestrina (RFHVMn), the pig-tailed macaque homolog of Kaposi's sarcoma-associated herpesvirus (KSHV), was determined by next-generation sequence analysis of a Kaposi's sarcoma (KS)-like macaque tumor. Colinearity of genes was observed with the KSHV genome, and the core herpesvirus genes had strong sequence homology to the corresponding KSHV genes. RFHVMn lacked homologs of open reading frame 11 (ORF11) and KSHV ORFs K5 and K6, which appear to have been generated by duplication of ORFs K3 and K4 after the divergence of KSHV and RFHV. RFHVMn contained positional homologs of all other unique KSHV genes, although some showed limited sequence similarity. RFHVMn contained a number of candidate microRNA genes. Although there was little sequence similarity with KSHV microRNAs, one candidate contained the same seed sequence as the positional homolog, kshv-miR-K12-10a, suggesting functional overlap. RNA transcript splicing was highly conserved between RFHVMn and KSHV, and strong sequence conservation was noted in specific promoters and putative origins of replication, predicting important functional similarities. Sequence comparisons indicated that RFHVMn and KSHV developed in long-term synchrony with the evolution of their hosts, and both viruses phylogenetically group within the RV1 lineage of Old World primate rhadinoviruses. RFHVMn is the closest homolog of KSHV to be completely sequenced and the first sequenced RV1 rhadinovirus homolog of KSHV from a nonhuman Old World primate. The strong genetic and sequence similarity between RFHVMn and KSHV, coupled with similarities in biology and pathology, demonstrate that RFHVMn infection in macaques offers an important and relevant model for the study of KSHV in humans.


Assuntos
Genoma Viral , Herpesvirus Humano 8/genética , Macaca nemestrina/virologia , Doenças dos Primatas/virologia , Sarcoma de Kaposi/veterinária , Sequência de Aminoácidos , Animais , Sequência de Bases , Feminino , Herpesvirus Humano 8/química , Herpesvirus Humano 8/classificação , Herpesvirus Humano 8/isolamento & purificação , Sequenciamento de Nucleotídeos em Larga Escala , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Rhadinovirus/química , Rhadinovirus/classificação , Rhadinovirus/genética , Sarcoma de Kaposi/virologia , Alinhamento de Sequência , Proteínas Virais/química , Proteínas Virais/genética
3.
PLoS Pathog ; 8(10): e1002962, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23055934

RESUMO

Two gammaherpesviruses, Epstein-Barr virus (EBV) (Lymphocryptovirus genus) and Kaposi's sarcoma-associated herpesvirus (KSHV) (Rhadinovirus genus) have been implicated in the etiology of AIDS-associated lymphomas. Homologs of these viruses have been identified in macaques and other non-human primates. In order to assess the association of these viruses with non-human primate disease, archived lymphoma samples were screened for the presence of macaque lymphocryptovirus (LCV) homologs of EBV, and macaque rhadinoviruses belonging to the RV1 lineage of KSHV homologs or the more distant RV2 lineage of Old World primate rhadinoviruses. Viral loads were determined by QPCR and infected cells were identified by immunolabeling for different viral proteins. The lymphomas segregated into three groups. The first group (n = 6) was associated with SIV/SHIV infections, contained high levels of LCV (1-25 genomes/cell) and expressed the B-cell antigens CD20 or BLA.36. A strong EBNA-2 signal was detected in the nuclei of the neoplastic cells in one of the LCV-high lymphomas, indicative of a type III latency stage. None of the lymphomas in this group stained for the LCV viral capsid antigen (VCA) lytic marker. The second group (n = 5) was associated with D-type simian retrovirus-2 (SRV-2) infections, contained high levels of RV2 rhadinovirus (9-790 genomes/cell) and expressed the CD3 T-cell marker. The third group (n = 3) was associated with SIV/SHIV infections, contained high levels of RV2 rhadinovirus (2-260 genomes/cell) and was negative for both CD20 and CD3. In both the CD3-positive and CD3/CD20-negative lymphomas, the neoplastic cells stained strongly for markers of RV2 lytic replication. None of the lymphomas had detectable levels of retroperitoneal fibromatosis herpesvirus (RFHV), the macaque RV1 homolog of KSHV. Our data suggest etiological roles for both lymphocryptoviruses and RV2 rhadinoviruses in the development of simian AIDS-associated lymphomas and indicate that the virus-infected neoplastic lymphoid cells are derived from different lymphocyte lineages and differentiation stages.


Assuntos
Herpesvirus Humano 4 , Herpesvirus Humano 8 , Lymphocryptovirus/isolamento & purificação , Linfoma Relacionado a AIDS/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Animais , Antígenos CD20/biossíntese , Antígenos de Neoplasias/biossíntese , Complexo CD3/biossíntese , Antígenos Nucleares do Vírus Epstein-Barr/biossíntese , Herpesvirus Humano 4/classificação , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/classificação , Herpesvirus Humano 8/genética , Lymphocryptovirus/genética , Macaca , Vírus dos Macacos de Mason-Pfizer/genética , Vírus dos Macacos de Mason-Pfizer/isolamento & purificação , Rhadinovirus/isolamento & purificação , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/isolamento & purificação , Células Tumorais Cultivadas , Carga Viral , Proteínas Virais/biossíntese , Proteínas Virais/genética
4.
AIDS Res Ther ; 6: 23, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19891783

RESUMO

BACKGROUND: Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are an important category of drugs for both chemotherapy and prevention of human immunodeficiency virus type 1 (HIV-1) infection. However, current non-human primate (NHP) models utilizing simian immunodeficiency virus (SIV) or commonly used chimeric SHIV (SIV expressing HIV-1 envelope) are inadequate due to the insensitivity to NNRTIs. To develop a NHP model for evaluation of NNRTI compounds, we characterized a RT-SHIV virus that was assembled by replacing the SIV mac239 reverse transcriptase (RT) with that of HIV-1HXB2. Since RT-SHIV exhibited in vitro characteristics of high infectivity, CCR5-usage, and sensitivity to HIV-1 specific NNRTIs, this virus was thought to be suitable for mucosal transmission and then was used to carry out a vaginal transmission study in pigtail macaques (Macaca nemestrina). RESULTS: RT-SHIV exhibited in vitro characteristics of an infectious CCR5-tropic chimeric virus. This virus was not only highly sensitive to HIV-1 RT specific NNRTIs; its replication was also inhibited by a variety of NRTIs and protease inhibitors. For in vivo vaginal transmission studies, macaques were either pretreated with a single dose of DMPA (depot medroxyprogesterone acetate) or left untreated before intravaginal inoculation with 500 or 1,000 TCID50 of RT-SHIV. All macaques became systemically infected by 2 or 3 weeks post-inoculation exhibiting persistent high viremia, marked CD4+T cell depletion, and antiviral antibody response. DMPA-pretreated macaques showed a higher mean plasma viral load after the acute infection stage, highly variable antiviral antibody response, and a higher incidence of AIDS-like disease as compared with macaques without DMPA pretreatment. CONCLUSION: This chimeric RT-SHIV has exhibited productive replication in both macaque and human PBMCs, predominantly CCR5-coreceptor usage for viral entry, and sensitivity to NNRTIs as well as other anti-HIV compounds. This study demonstrates rapid systemic infection in macaques following intravaginal exposure to RT-SHIV. This RT-SHIV/macaque model could be useful for evaluation of NNRTI-based therapies, microbicides, or other preventive strategies.

5.
Drug Metab Dispos ; 35(9): 1459-62, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17591677

RESUMO

P-glycoprotein (P-gp) expression at the rodent blood-brain barrier (BBB) limits the central nervous system (CNS) distribution of anti-human immunodeficiency virus (HIV) protease inhibitors (PIs). However, it is not clear whether P-gp activity at the human BBB is as effective as that in rodents in preventing the distribution of PIs into the CNS. If it is, inhibition of P-gp at the human BBB could increase the distribution of the PIs into the CNS and, therefore, their efficacy against HIV-associated dementia. Because the distribution of the PIs into the human brain cannot be directly measured, we conducted studies in a more representative animal, the nonhuman primate. Specifically we investigated the distribution of nelfinavir (a PI and a P-gp substrate; 6 mg/kg i.v.) into the brain and cerebrospinal fluid (CSF) of nonhuman primates (cynomolgus monkeys, Macaca fascicularis) in the presence and absence of the potent and selective P-gp inhibitor, zosuquidar, and whether changes in brain nelfinavir concentration, after inhibition of P-gp, paralleled those in the CSF. Our data indicate that nelfinavir has poor penetration into the macaque's brain and CSF, and P-gp inhibition at the BBB by zosuquidar enhanced the distribution of nelfinavir into the brain by 146-fold. However, the concentration of nelfinavir in the CSF was unaffected by coadministration of zosuquidar (p > 0.05). In conclusion, P-gp inhibition at the nonhuman primate BBB significantly enhanced the distribution of nelfinavir into the brain, and this effect was not observed in the CSF. Therefore, as is common in human studies investigating P-gp inhibition at the BBB, CSF concentration of a drug should not be used as a surrogate marker for brain drug concentration.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Barreira Hematoencefálica/fisiologia , Encéfalo/metabolismo , Inibidores da Protease de HIV/farmacocinética , Nelfinavir/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Área Sob a Curva , Dibenzocicloeptenos/farmacologia , Inibidores da Protease de HIV/líquido cefalorraquidiano , Injeções Intravenosas , Macaca fascicularis , Nelfinavir/líquido cefalorraquidiano , Quinolinas/farmacologia
6.
Cell Microbiol ; 9(1): 120-30, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16879452

RESUMO

Novel therapeutic approaches are needed to combat the rapid increase in HIV sexual transmission in women. The probiotic organism Lactobacillus reuteri RC-14 which safely colonizes the human vagina and prevents microbial infections, has been genetically modified to produce anti-HIV proteins which were capable of blocking the three main steps of HIV entry into human peripheral blood mononuclear cells. The HIV entry or fusion inhibitors were fused to the native expression and secretion signals of BspA, Mlp or Sep in L. reuteri RC-14 and the expression cassettes were stably inserted into the chromosome. L. reuteri RC-14 expressed the HIV inhibitors in cell wall-associated and secreted forms. L. reuteri RC-14 expressing CD4D1D2-antibody-like fusion proteins were able to bind single or dual tropic coreceptor-using HIV-1 primary isolates. This is the first study to show that a well-documented and proven human vaginal probiotic strain can express potent functional viral inhibitors, which may potentially lower the sexual transmission of HIV.


Assuntos
Inibidores da Fusão de HIV/metabolismo , Infecções por HIV/virologia , HIV/metabolismo , Limosilactobacillus reuteri/genética , Probióticos/metabolismo , Vagina/microbiologia , Animais , Fusão Gênica Artificial , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Cobaias , HIV/efeitos dos fármacos , Humanos , Limosilactobacillus fermentum/isolamento & purificação , Limosilactobacillus reuteri/crescimento & desenvolvimento , Limosilactobacillus reuteri/metabolismo , Leucócitos Mononucleares/microbiologia , Leucócitos Mononucleares/virologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ligação Viral
7.
J Gen Virol ; 87(Pt 12): 3529-3538, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17098967

RESUMO

Two distinct lineages of rhadinoviruses related to Kaposi's sarcoma (KS)-associated herpesvirus (KSHV; Human herpesvirus 8), the causative agent of KS, have been identified. In macaques, the RV1 lineage is represented by retroperitoneal fibromatosis (RF) herpesvirus (RFHV), the homologue of KSHV, whilst the RV2 lineage is represented by rhesus rhadinovirus (RRV), a more distantly related virus. Real-time quantitative PCR was used to estimate the loads of RV1 and RV2 rhadinoviruses in simian acquired immunodeficiency syndrome-associated RF (SAIDS-RF), a neoplasm of macaques with similarities to AIDS-associated KS. Both RV1 and RV2 rhadinoviruses were detected in macaques with RF. The RV1 loads were 220- to 4300-fold higher in RF tumours than in spleen, showing a strong tumour association (mean loads of 1 800 000 vs 2900 copies per 10(6) cells in tumours and spleen, respectively). In contrast, RV2 loads in the RF tumours were 100-fold lower than RV1 loads and showed similar levels in tumours and spleen (mean loads of 16 000 vs 24 000 copies per 10(6) cells, respectively). Immunostaining with antibodies reactive against RFHV ORF73 latency-associated nuclear antigen (LANA) showed intense nuclear staining of the spindleoid RF tumour cells. Correlation of viral load and the number of LANA-positive cells indicated that RF tumour cells contained multiple copies of the RFHV genome per cell. This pattern of infectivity is similar to that seen in KS tumours latently infected with KSHV. Our study demonstrates similarities in the biology of KSHV and RFHV and supports a role for RFHV in the aetiology of SAIDS-RF.


Assuntos
Antígenos Virais/biossíntese , DNA Viral/análise , Infecções por Herpesviridae/virologia , Proteínas Nucleares/biossíntese , Neoplasias Retroperitoneais/virologia , Rhadinovirus/fisiologia , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Infecções Tumorais por Vírus/virologia , Animais , Antígenos Virais/imunologia , DNA Viral/genética , Modelos Animais de Doenças , Infecções por Herpesviridae/complicações , Imuno-Histoquímica , Macaca mulatta , Macaca nemestrina , Proteínas Nucleares/imunologia , Reação em Cadeia da Polimerase/métodos , Neoplasias Retroperitoneais/complicações , Neoplasias Retroperitoneais/patologia , Rhadinovirus/genética , Rhadinovirus/metabolismo , Baço/virologia , Estatística como Assunto , Infecções Tumorais por Vírus/complicações
8.
AIDS Res Ther ; 3: 29, 2006 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-17132170

RESUMO

BACKGROUND: A 4-week, uninterrupted treatment with 9-(2-phosphonyl-methoxypropyly)adenine (PMPA, commonly called tenofovir) completely prevents simian immunodeficiency virus (SIVmne) infection in cynomolgus macaques if treatment begins within 24 hours after SIVmne inoculation, but is less effective if treatment is delayed or duration of treatment is shortened. Critical factors for efficacy include timing and duration of treatment, potency of antiretroviral drug and a contribution from antiviral immune responses. Therefore, we evaluated the impact of one or more treatment interruptions plus SIVmne re-exposures on efficacy of PMPA treatment to prevent SIVmne infection in cynomolgus macaques. We also evaluated whether macaques with pre-existing SIV immune responses show increased efficacy of treatment. Eight PMPA-treated, virus-negative and seronegative macaques, and five PMPA-treated, virus-negative but weakly or strongly seropositive macaques were re-inoculated with SIVmne and treated with PMPA starting 24 hr post inoculation. Thereafter, they received either a 5-week treatment involving one interruption plus one SIVmne challenge or a 10-week treatment involving six interruptions plus six SIVmne challenges early during treatment. Parameters measured were plasma SIV RNA, SIV-antibody response, CD4+ T lymphocyte subsets and in vivo CD8+ cell-suppression of virus infection. RESULTS: All seronegative macaques developed persistent antibody response beginning 4 to 8 weeks after stopping PMPA-treatment in absence of viremia in a majority of macaques and coinciding with onset of intermittent viremia in other macaques. In contrast, all weakly or strongly seropositive macaques showed immediate increase in titers (> 1600) of SIV antibodies, even before the end of PMPA-treatment, and in absence of detectable viremia. However, in vivo CD8+-cell depletion revealed CD8 cell-suppression of viremia and persistence of virus in the macaques as long as 2 years after PMPA-treatment, even in aviremic macaques. Unlike untreated macaques, a treated macaque controlled viral replication and blocked CD4+ T cell depletion when challenged with a heterologous chimeric SIV/HIV-1 virus called SHIV89.6P. CONCLUSION: A single interruption plus one SIVmne challenge was as sufficient as six interruptions plus six SIVmne challenges in reducing efficacy of PMPA, but results in long-term persistence of virus infection suppressed by CD8+ cells. Efficacy of PMPA treatment was highest in macaques with pre-existing SIV immune responses.

9.
Virology ; 354(1): 103-15, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-16879850

RESUMO

Retroperitoneal fibromatosis herpesvirus (RFHV), the macaque homolog of the human rhadinovirus, Kaposi's sarcoma-associated herpesvirus (KSHV), was first identified in retroperitoneal fibromatosis (RF) tumor lesions of macaques with simian AIDS. We cloned and sequenced the ORF73 latency-associated nuclear antigen (LANA) of RFHVMn from the pig-tailed macaque. RFHVMn LANA is structurally analogous to KSHV ORF73 LANA and contains an N-terminal serine-proline-rich region, a large internal glutamic acidic-rich repeat region and a conserved C-terminal domain. RFHVMn LANA reacts with monoclonal antibodies specific for a glutamic acid-proline dipeptide motif and a glutamic acid-glutamine-rich motif in the KSHV LANA repeat region. Immunohistochemical and immunofluorescence analysis revealed that RFHVMn LANA is a nuclear antigen which is highly expressed in RF spindloid tumor cells. These data suggest that RFHV LANA is an ortholog of KSHV LANA and will function similarly to maintain viral latency and play a role in tumorigenicity in macaques.


Assuntos
Antígenos Virais/genética , Fibroma/virologia , Proteínas Nucleares/genética , Fases de Leitura Aberta , Neoplasias Retroperitoneais/virologia , Rhadinovirus/genética , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Antígenos Virais/química , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Núcleo Celular/química , Clonagem Molecular , DNA Viral/química , DNA Viral/genética , Fibroma/patologia , Imuno-Histoquímica , Macaca nemestrina , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Rhadinovirus/isolamento & purificação , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas
10.
J Acquir Immune Defic Syndr ; 42(2): 155-61, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16760797

RESUMO

In HIV-infected persons on highly active antiretroviral therapy, residual virus is found in lymphoid tissues. Indinavir concentrations in lymph node mononuclear cells of patients on highly active antiretroviral therapy were approximately 25% to 35% of those in blood mononuclear cells, suggesting that drug insufficiency contributes to residual virus in lymphoid tissues. Therefore, we developed novel lipid-indinavir nanoparticles targeted to lymphoid tissues. Given subcutaneously, these nanoparticles provided indinavir concentrations 250% to 2270% higher than plasma indinavir concentrations in both peripheral and visceral lymph nodes. Improved indinavir delivery was reflected in reduced viral RNA and CD4(+) T-cell rebound. This study optimized lipid nanoparticle formulation with respect to indinavir in lymphoid tissues of HIV-infected macaques. Regardless of lipid characteristic tested (charge, fluidity, and steric modification), indinavir binds completely to lipid at pH 7.4 but is reversed at pH 5.5 or lower. Compared with previous formulations, nanoparticles composed of disteroyl phosphatidylcholine and methyl polyethylene glycol-disteroyl phosphatidylethanolamine (DSPC:mPEG-DSPE) provided 6-fold higher indinavir levels in lymph nodes and enhanced drug exposure in blood. Enhanced anti-HIV activity paralleled improved intracellular drug accumulation. Collectively, these data suggest that indinavir nanoparticles composed of DSPC:mPEG-DSPE provided the most effective lymphoid delivery and could maximally suppress the virus in lymphoid tissues.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Infecções por HIV/tratamento farmacológico , HIV-2 , Indinavir/administração & dosagem , Indinavir/farmacocinética , Tecido Linfoide/química , Animais , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Contagem de Linfócito CD4 , Linhagem Celular , Modelos Animais de Doenças , Portadores de Fármacos , Infecções por HIV/virologia , Concentração de Íons de Hidrogênio , Indinavir/metabolismo , Indinavir/farmacologia , Injeções Subcutâneas , Lipídeos/administração & dosagem , Lipídeos/farmacocinética , Linfonodos/química , Macaca nemestrina , Nanoestruturas/química , Fosfatidilcolinas/farmacocinética , RNA Viral/sangue , Distribuição Tecidual
11.
Virol J ; 3: 11, 2006 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-16515713

RESUMO

BACKGROUND: D-type simian retrovirus-2 (SRV-2) causes an AIDS-like immune deficiency syndrome (SAIDS) in various macaque species. SAIDS is often accompanied by retroperitoneal fibromatosis (RF), an aggressive fibroproliferative disorder reminiscent of Kaposi's sarcoma in patients with HIV-induced AIDS. In order to determine the association of SRV-2 subtypes with SAIDS-RF, and study the evolution and transmission of SRV-2 in captive macaque populations, we have molecularly characterized the env gene of a number of SRV-2 isolates from different macaque species with and without RF. RESULTS: We sequenced the env gene from eighteen SRV-2 isolates and performed sequence comparisons and phylogenetic analyses. Our studies revealed the presence of six distinct subtypes of SRV-2, three of which were associated with SAIDS-RF cases. We found no association between SRV-2 subtypes and a particular macaque species. Little sequence variation was detected in SRV-2 isolates from the same individual, even after many years of infection, or from macaques housed together or related by descent from a common infected parent. Seventy-two amino acid changes were identified, most occurring in the larger gp70 surface protein subunit. In contrast to the lentiviruses, none of the amino acid variations involved potential N-linked glycosylation sites. Structural analysis of a domain within the gp22/gp20 transmembrane subunit that was 100% conserved between SRV-2 subtypes, revealed strong similarities to a disulfide-bonded loop that is crucial for virus-cell fusion and is found in retroviruses and filoviruses. CONCLUSION: Our study suggests that separate introductions of at least six parental SRV-2 subtypes into the captive macaque populations in the U.S. have occurred with subsequent horizontal transfer between macaque species and primate centers. No specific association of a single SRV-2 subtype with SAIDS-RF was seen. The minimal genetic variability of the env gene within a subtype over time suggests that a strong degree of adaptation to its primate host has occurred during evolution of the virus.


Assuntos
Variação Genética , Macaca/virologia , Vírus dos Macacos de Mason-Pfizer/genética , Fibrose Retroperitoneal/complicações , Fibrose Retroperitoneal/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Regulação Viral da Expressão Gênica , Vírus dos Macacos de Mason-Pfizer/fisiologia , Dados de Sequência Molecular , Filogenia , Especificidade da Espécie , Proteínas do Envelope Viral/química
12.
AIDS Res Hum Retroviruses ; 21(3): 207-13, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15795526

RESUMO

SPL7013 is a dendrimer with a polyanionic outer surface that allows multiple interactions with target sites. It potently binds and blocks HIV-1 and chimeric simian/HIV-1 viruses (SHIVs) replication in vitro. Gels containing different concentrations of SPL7013 were used as topical microbicides in female pigtailed macaques (Macaca nemestrina) to study their ability to prevent vaginal transmission of SHIV(89,6P). On virus challenge, all untreated macaques (8/8) and seven of eight macaques treated with placebo gel were infected within 2 weeks postinfection (PI) and showed high plasma viremia and dramatic CD4(+) cell decline within 4 weeks PI. In contrast, 6/6 macaques, 5/6 macaques, and 2/6 macaques treated with 5% w/w (50 mg/ml), 3% w/w (30 mg/ml), and 1% w/w (10 mg/ml) SPL7013 gels, respectively, resisted viral challenge. The results showed that animals treated with SPL7013 showed a dose-dependent resistance to virus challenge. Neither SPL7013 nor placebo gels produced any adverse effects following the single application in the study. These results showed that 3-5% w/w SPL7013 gels were effective in blocking vaginal transmission of SHIV in macaques after single gel application followed by single virus challenge. These results suggest that SPL7013 gel may be a promising anti-HIV microbicide formulation for further evaluation.


Assuntos
Anti-Infecciosos Locais/administração & dosagem , HIV-1/efeitos dos fármacos , Polilisina/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vagina/virologia , Animais , Dendrímeros , Feminino , Géis , Macaca nemestrina , Polilisina/efeitos adversos , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão
13.
J Med Primatol ; 33(2): 105-8, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15061723

RESUMO

Preclinical studies of topical microbicide products, using appropriate animal models for assessing the safety of repeated use are essential. The pig-tailed macaque (Macaca nemestrina) model has been used to assess the safety of vaginally and rectally applied topical microbicide products. The availability of sexually mature female pig-tailed macaques has become extremely restricted. Currently, M. fascicularis is more readily available, and was therefore evaluated as an alternative model for topical microbicide pre-clinical evaluation. Twenty sexually mature M. fascicularis were assessed for feasibility to mimic the established models. The rectal and cervicovaginal microenvironments of the M. fascicularis were determined to be similar to those of M. nemestrina and humans. The gross anatomy was significantly smaller than that of the pig-tailed macaque, such that colposcopic examinations and multiple biopsies would not be possible. Thus, the M. fascicularis may not be useful for vaginally applied topical microbicide safety studies yet adequate for assessing safety of rectally applied topical microbicide products.


Assuntos
Anti-Infecciosos Locais/normas , Macaca fascicularis/microbiologia , Macaca nemestrina/anatomia & histologia , Modelos Animais , Animais , Avaliação Pré-Clínica de Medicamentos/veterinária , Feminino , Concentração de Íons de Hidrogênio , Macaca fascicularis/anatomia & histologia , Macaca nemestrina/microbiologia , Reto/anatomia & histologia , Reto/microbiologia , Vagina/anatomia & histologia , Vagina/microbiologia
14.
AIDS Res Hum Retroviruses ; 20(1): 11-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15000694

RESUMO

The cyanobacterial protein cyanovirin-N (CV-N) potently inactivates diverse strains of HIV-1 and other lentiviruses due to irreversible binding of CV-N to the viral envelope glycoprotein gp120. In this study, we show that recombinant CV-N effectively blocks HIV-1(Ba-L) infection of human ectocervical explants. Furthermore, we demonstrate the in vivo efficacy of CV-N gel in a vaginal challenge model by exposing CV-N-treated female macaques (Macaca fascicularis) to a pathogenic chimeric SIV/HIV-1 virus, SHIV89.6P. All of the placebo-treated and untreated control macaques (8 of 8) became infected. In contrast, 15 of 18 CV-N-treated macaques showed no evidence of SHIV infection. Further, CV-N produced no cytotoxic or clinical adverse effects in either the in vitro or in vivo model systems. Together these studies suggest that CV-N is a good candidate for testing in humans as an anti-HIV topical microbicide.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Proteínas de Bactérias , Proteínas de Transporte/administração & dosagem , Infecções por HIV/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vagina/virologia , Administração Intravaginal , Animais , Fármacos Anti-HIV/uso terapêutico , Proteínas de Transporte/uso terapêutico , Colo do Útero/virologia , Técnicas de Cultura , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/genética , HIV-1/patogenicidade , Humanos , Macaca fascicularis , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/patogenicidade
15.
J Acquir Immune Defic Syndr ; 34(4): 387-97, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14615656

RESUMO

Analysis of indinavir levels in HIV-positive patients indicated that drug concentrations in lymph node mononuclear cells (LNMCs) were about 25-35% of mononuclear cells in blood. To enhance lymphatic delivery of anti-HIV drugs, a novel drug delivery strategy was designed consisting of lipid-associated indinavir (50-80 nm in diameter) complexes in suspension for subcutaneous (SC) injection. Due to the pH-dependent lipophilicity of indinavir, practically all the drug molecules are incorporated into lipid phase when formulated at pH 7.4 and 5:1 lipid-to-drug (m/m) ratio. At pH 5.5, about 20% of drugs were found in lipid-drug complexes. Effects of lipid association on the time course of plasma indinavir concentrations were determined in macaques (Macaca nemestrina) administered with either soluble or lipid-associated formulation of indinavir (10 mg/kg, SC). Results yielded about a 10-fold reduction in peak plasma concentration and a 6-fold enhancement in terminal half-life (t1/2beta = 12 vs. 2 hours). In addition, indinavir concentrations in both peripheral and visceral lymph nodes were 250-2270% higher than plasma (compared with <35% with soluble lipid-free drug administration in humans). Administration of lipid-associated indinavir (20 mg/kg daily) to HIV-2287-infected macaques (at 30-33 weeks after infection) resulted in significantly reduced viral RNA load and increased CD4 T cell number concentrations. Collectively, these data indicate that lipid association greatly enhances delivery of the anti-HIV drug indinavir to lymph nodes at levels that cannot be achieved with soluble drug, provides significant virus load reduction, and could potentially reverse CD4 T cell depletion due to HIV infection.


Assuntos
Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/administração & dosagem , Inibidores da Protease de HIV/farmacocinética , HIV-2/fisiologia , Indinavir/administração & dosagem , Indinavir/farmacocinética , Tecido Linfoide/metabolismo , Animais , Contagem de Linfócito CD4 , Colesterol/sangue , Progressão da Doença , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Humanos , Hibridização In Situ , Lipossomos , Tecido Linfoide/virologia , Macaca , RNA Viral/química , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Viral , Viremia/tratamento farmacológico , Viremia/metabolismo , Replicação Viral/efeitos dos fármacos
16.
AIDS Res Hum Retroviruses ; 19(7): 535-41, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12921090

RESUMO

Cyanovirin-N (CV-N), an 11-kDa cyanobacterial protein, potently inactivates diverse strains of HIV-1, HIV-2, and simian immunodeficiency virus (SIV) and also prevents virus-to-cell fusion, virus entry, and infection of cells in vitro. These properties make CV-N an attractive candidate for use as a topical microbicide to prevent the sexual transmission of HIV. We evaluated the efficacy of gel-formulated, recombinant CV-N gel asa topical microbicide in male macaques (Macaca fascicularis) that were rectally challenged with a chimeric SIV/HIV-1 virus known as SHIV89.6P. All of the untreated macaques were infected and experienced CD4+T cell depletion. In contrast, none of the macaques that received either 1% or 2% CV-N gel showed evidence of SHIV89.6P infection. Neither CV-N nor placebo gels produced any adverse effects in any macaque following the rectal application. These results indicate that CV-N gel as a topical microbicide can prevent rectal transmission of SHIV in macaques. These studies encourage clinical evaluation of CV-N as a topical microbicide to prevent sexual transmission of HIV in humans.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Proteínas de Bactérias , Proteínas de Transporte/uso terapêutico , HIV-1/efeitos dos fármacos , Vírus Reordenados/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Administração Retal , Animais , Fármacos Anti-HIV/administração & dosagem , Contagem de Linfócito CD4 , Proteínas de Transporte/administração & dosagem , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Géis , HIV-1/isolamento & purificação , Linfonodos/virologia , Macaca fascicularis , Masculino , Modelos Animais , Vírus Reordenados/isolamento & purificação , Segurança , Comportamento Sexual , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/isolamento & purificação
17.
J Virol ; 77(9): 5084-97, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12692211

RESUMO

We previously identified retroperitoneal fibromatosis-associated herpesvirus (RFHV) as a simian homolog of Kaposi's sarcoma-associated herpesvirus (KSHV) in a fibroproliferative malignancy of macaques that has similarities to Kaposi's sarcoma. In this report, we cloned 4.3 kb of divergent locus B (DL-B) flanking the DNA polymerase gene from two variants of RFHV from different species of macaque with a consensus degenerate hybrid oligonucleotide primer approach. Within the DL-B region of RFHV, viral homologs of the cellular interleukin-6, dihydrofolate reductase, and thymidylate synthase genes were identified, along with a homolog of the gammaherpesvirus open reading frame (ORF) 10. In addition, a homolog of the KSHV ORF K3, the modulator of immune recognition-1, was identified. Our data show a close similarity in sequence conservation, gene content, and genomic structure between RFHV and KSHV which strongly supports the grouping of these viral species within the same RV-1 rhadinovirus lineage and the hypothesis that RFHV is the macaque homolog of KSHV.


Assuntos
Evolução Molecular , Herpesvirus Humano 8/classificação , Herpesvirus Humano 8/genética , Rhadinovirus , Rhadinovirus/classificação , Rhadinovirus/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Polimerase Dirigida por DNA/genética , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/química , Humanos , Macaca mulatta , Macaca nemestrina , Dados de Sequência Molecular , Doenças dos Macacos/virologia , Filogenia , Fibrose Retroperitoneal/veterinária , Fibrose Retroperitoneal/virologia , Neoplasias Retroperitoneais/veterinária , Neoplasias Retroperitoneais/virologia , Rhadinovirus/química , Sarcoma de Kaposi/virologia , Análise de Sequência de DNA , Infecções Tumorais por Vírus/veterinária , Infecções Tumorais por Vírus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...