Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Phytomedicine ; 87: 153591, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34029937

RESUMO

BACKGROUND: The outbreak of coronavirus (SARS-CoV-2) disease caused more than 100,000,000 people get infected and over 2,200,000 people being killed worldwide. However, the current developed vaccines or drugs may be not effective in preventing the pandemic of COVID-19 due to the mutations of coronavirus and the severe side effects of the newly developed vaccines. Chinese herbal medicines and their active components play important antiviral activities. Corilagin exhibited antiviral effect on human immunodeficiency virus (HIV), hepatitis C virus (HCV) and Epstein-Barr virus (EBV). However, whether it blocks the interaction between SARS-CoV-2 RBD and hACE2 has not been elucidated. PURPOSE: To characterize an active compound, corilagin derived from Phyllanthus urinaria as potential SARS-CoV-2 entry inhibitors for its possible preventive application in daily anti-virus hygienic products. METHODS: Computational docking coupled with bio-layer interferometry, BLI were adopted to screen more than 1800 natural compounds for the identification of SARS-CoV-2 spike-RBD inhibitors. Corilagin was confirmed to have a strong binding affinity with SARS-CoV-2-RBD or human ACE2 (hACE2) protein by the BLI, ELISA and immunocytochemistry (ICC) assay. Furthermore, the inhibitory effect of viral infection of corilagin was assessed by in vitro pseudovirus system. Finally, the toxicity of corilagin was examined by using MTT assay and maximal tolerated dose (MTD) studies in C57BL/6 mice. RESULTS: Corilagin preferentially binds to a pocket that contains residues Cys 336 to Phe 374 of spike-RBD with a relatively low binding energy of -9.4 kcal/mol. BLI assay further confirmed that corilagin exhibits a relatively strong binding affinity to SARS-CoV-2-RBD and hACE2 protein. In addition, corilagin dose-dependently blocks SARS-CoV-2-RBD binding and abolishes the infectious property of RBD-pseudotyped lentivirus in hACE2 overexpressing HEK293 cells, which mimicked the entry of SARS-CoV-2 virus in human host cells. Finally, in vivo studies revealed that up to 300 mg/kg/day of corilagin was safe in C57BL/6 mice. Our findings suggest that corilagin could be a safe and potential antiviral agent against the COVID-19 acting through the blockade of the fusion of SARS-CoV-2 spike-RBD to hACE2 receptors. CONCLUSION: Corilagin could be considered as a safe and environmental friendly anti-SARS-CoV-2 agent for its potential preventive application in daily anti-virus hygienic products.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Antivirais/farmacologia , Glucosídeos/farmacologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Taninos Hidrolisáveis/farmacologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Enzima de Conversão de Angiotensina 2/química , Animais , Antivirais/química , Antivirais/toxicidade , COVID-19 , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Glucosídeos/química , Glucosídeos/toxicidade , Células HEK293 , Humanos , Taninos Hidrolisáveis/química , Taninos Hidrolisáveis/toxicidade , Infecções por Lentivirus/tratamento farmacológico , Masculino , Dose Máxima Tolerável , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Glicoproteína da Espícula de Coronavírus/química
3.
Arq. Inst. Biol ; 86: e0752018, 2019. ilus, tab
Artigo em Inglês | LILACS, VETINDEX | ID: biblio-1046025

RESUMO

Infections by small ruminant lentiviruses (SRLVs) affect goats and sheep causing chronic multisystemic diseases that generate great economic losses. The caprine lentivirus (CLV) and the ovine lentivirus (OLV) present tropism for cells of the monocyte/macrophage lineage, which are directly associated with the main route of transmission through the ingestion of milk and colostrum from infected animals. In this manner, controlling this route is of paramount importance. Currently, researches have investigated the use of chemical additives in milk that can preserve colostrum or milk and inactivate microbiological agents. Among the compounds, sodium dodecyl sulfate (SDS) has been shown to be satisfactory in the chemical inactivation of HIV and CLV in milk, and also as a biocide in goat colostrum.(AU)


As lentiviroses de pequenos ruminantes (LVPRs) são infecções que afetam caprinos e ovinos, causando doenças multissistêmicas crônicas, ocasionando grandes perdas econômicas. Os agentes causadores, lentivírus caprino (LVC) e o lentivírus ovino (LVO), apresentam tropismo por células da linhagem monocítico--fagocitária, as quais estão diretamente associadas à principal via de transmissão, por meio da ingestão de leite e colostro provindos de animais infectados. Desse modo, o controle por esta via é de suma importância. Atualmente, pesquisas vêm sendo desenvolvidas para o uso de aditivos químicos no leite, que possam conservar o colostro ou leite, e inativar agentes microbiológicos presentes. Dentre estes, o dodecil sulfato de sódio (SDS) vem apresentando resultados satisfatórios na inativação química do HIV e LVC em leite, e ainda como biocida em colostro caprino.(AU)


Assuntos
Animais , Dodecilsulfato de Sódio/farmacologia , Ruminantes/virologia , Infecções por Lentivirus/tratamento farmacológico , Lentivirus Ovinos-Caprinos/efeitos dos fármacos , Ovinos/virologia , Infecções por Lentivirus/transmissão , Colostro/virologia , Leite/virologia
4.
Methods Mol Biol ; 1836: 139-158, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30151572

RESUMO

Our understanding of infection biology is based on experiments in which pathogen or host proteins are perturbed by small compound inhibitors, mutation, or depletion. This approach has been remarkably successful, as, for example, demonstrated by the independent identification of the endosomal membrane protein Niemann-Pick C1 as an essential factor for Ebola virus infection in both small compound and insertional mutagenesis screens (Côté, Nature 477:344-348, 2011; Carette et al., Nature 477:340-343, 2011). However, many aspects of host-pathogen interactions are poorly understood because we cannot target all of the involved molecules with small molecules, or because we cannot deplete essential proteins. Single domain antibody fragments expressed in the cytosol or other organelles constitute a versatile alternative to perturb the function of any given protein by masking protein-protein interaction interfaces, by stabilizing distinct conformations, or by directly interfering with enzymatic activities. The variable domains of heavy chain-only antibodies (VHHs) from camelid species can be cloned from blood samples of animals immunized with the desired target molecules. We can thus exploit the ability of the camelid immune system to generate affinity-matured single domain antibody fragments to obtain highly specific tools. Interesting VHH candidates are typically identified based on their affinity toward immobilized antigens using techniques such as phage display.The phenotypical screening approach described here allows the direct identification of VHHs that prevent infection of cells with influenza A virus (IAV) or other pathogens. The VHH repertoire is cloned into a lentiviral vector, which is used to generate pseudo-typed lentivirus particles. Target cells are transduced with the lentivirus, so that every cell inducibly expresses a different VHH. This cell collection is then challenged with a lethal dose of virus. Only the cells which express a VHH that prevents infection by targeting virus proteins or host cell components essential for infection will survive. We can thus identify critical target molecules including vulnerable epitopes and conformations, render target molecules accessible to informative perturbation studies, and stabilize intermediates of virus entry for detailed analysis.


Assuntos
Antirretrovirais/farmacologia , Lentivirus/efeitos dos fármacos , Fenótipo , Anticorpos de Domínio Único/farmacologia , Sequência de Aminoácidos , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Biblioteca Gênica , Vetores Genéticos/genética , Humanos , Vírus da Influenza A/genética , Lentivirus/genética , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/virologia , Testes de Sensibilidade Microbiana , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/genética
5.
PLoS Pathog ; 14(1): e1006849, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29364950

RESUMO

Reverse transcriptase (RT) is the target for the majority of anti-HIV-1 drugs. As with all anti-AIDS treatments, continued success of RT inhibitors is persistently disrupted by the occurrence of resistance mutations. To explore latent resistance mechanisms potentially accessible to therapeutically challenged HIV-1 viruses, we examined RT from the related feline immunodeficiency virus (FIV). FIV closely parallels HIV-1 in its replication and pathogenicity, however, is resistant to all non-nucleoside inhibitors (NNRTI). The intrinsic resistance of FIV RT is particularly interesting since FIV harbors the Y181 and Y188 sensitivity residues absent in both HIV-2 and SIV. Unlike RT from HIV-2 or SIV, previous efforts have failed to make FIV RT susceptible to NNRTIs concluding that the structure or flexibility of the feline enzyme must be profoundly different. We report the first crystal structure of FIV RT and, being the first structure of an RT from a non-primate lentivirus, enrich the structural and species repertoires available for RT. The structure demonstrates that while the NNRTI binding pocket is conserved, minor subtleties at the entryway can render the FIV RT pocket more restricted and unfavorable for effective NNRTI binding. Measuring NNRTI binding affinity to FIV RT shows that the "closed" pocket configuration inhibits NNRTI binding. Mutating the loop residues rimming the entryway of FIV RT pocket allows for NNRTI binding, however, it does not confer sensitivity to these inhibitors. This reveals a further layer of resistance caused by inherent FIV RT variances that could have enhanced the dissociation of bound inhibitors, or, perhaps, modulated protein plasticity to overcome inhibitory effects of bound NNRTIs. The more "closed" conformation of FIV RT pocket can provide a template for the development of innovative drugs that could unlock the constrained pocket, and the resilient mutant version of the enzyme can offer a fresh model for the study of NNRTI-resistance mechanisms overlooked in HIV-1.


Assuntos
Farmacorresistência Viral , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina , Infecções por Lentivirus/tratamento farmacológico , DNA Polimerase Dirigida por RNA/química , DNA Polimerase Dirigida por RNA/fisiologia , Inibidores da Transcriptase Reversa/uso terapêutico , Sequência de Aminoácidos , Animais , Gatos , Cristalografia por Raios X , Farmacorresistência Viral/genética , Síndrome de Imunodeficiência Adquirida Felina/enzimologia , Vírus da Imunodeficiência Felina/enzimologia , Vírus da Imunodeficiência Felina/genética , Infecções por Lentivirus/enzimologia , Modelos Moleculares , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
6.
JCI Insight ; 2(4): e91214, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28239657

RESUMO

SIV DNA can be detected in lymphoid tissue-resident macrophages of chronically SIV-infected Asian macaques. These macrophages also contain evidence of recently phagocytosed SIV-infected CD4+ T cells. Here, we examine whether these macrophages contain replication-competent virus, whether viral DNA can be detected in tissue-resident macrophages from antiretroviral (ARV) therapy-treated animals and humans, and how the viral sequences amplified from macrophages and contemporaneous CD4+ T cells compare. In ARV-naive animals, we find that lymphoid tissue-resident macrophages contain replication-competent virus if they also contain viral DNA in ARV-naive Asian macaques. The genetic sequence of the virus within these macrophages is similar to those within CD4+ T cells from the same anatomic sites. In ARV-treated animals, we find that viral DNA can be amplified from lymphoid tissue-resident macrophages of SIV-infected Asian macaques that were treated with ARVs for at least 5 months, but we could not detect replication-competent virus from macrophages of animals treated with ARVs. Finally, we could not detect viral DNA in alveolar macrophages from HIV-infected individuals who received ARVs for 3 years and had undetectable viral loads. These data demonstrate that macrophages can contain replication-competent virus, but may not represent a significant reservoir for HIV in vivo.


Assuntos
DNA Viral/metabolismo , Infecções por HIV/virologia , HIV-1/genética , Macrófagos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Replicação Viral , Animais , Antirretrovirais/uso terapêutico , Terapia Antirretroviral de Alta Atividade , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , HIV-1/fisiologia , Humanos , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/virologia , Tecido Linfoide/citologia , Macaca , Macaca mulatta , Macaca nemestrina , Macrófagos/imunologia , Macrófagos/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Carga Viral
7.
JCI Insight ; 2(4): e91230, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28239658

RESUMO

The conditioning regimen used as part of the Berlin patient's hematopoietic cell transplant likely contributed to his eradication of HIV infection. We studied the impact of conditioning in simian-human immunodeficiency virus-infected (SHIV-infected) macaques suppressed by combination antiretroviral therapy (cART). The conditioning regimen resulted in a dramatic, but incomplete depletion of CD4+ and CD8+ T cells and CD20+ B cells, increased T cell activation and exhaustion, and a significant loss of SHIV-specific Abs. The disrupted T cell homeostasis and markers of microbial translocation positively correlated with an increased viral rebound after cART interruption. Quantitative viral outgrowth and Tat/rev-induced limiting dilution assays showed that the size of the latent SHIV reservoir did not correlate with viral rebound. These findings identify perturbations of the immune system as a mechanism for the failure of autologous transplantation to eradicate HIV. Thus, transplantation strategies may be improved by incorporating immune modulators to prevent disrupted homeostasis, and gene therapy to protect transplanted cells.


Assuntos
Linfócitos T CD4-Positivos/efeitos da radiação , Linfócitos T CD8-Positivos/efeitos da radiação , Infecções por HIV/imunologia , HIV-1/efeitos da radiação , Transplante de Células-Tronco Hematopoéticas/métodos , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/efeitos da radiação , Condicionamento Pré-Transplante/métodos , Irradiação Corporal Total , Animais , Antirretrovirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/tratamento farmacológico , Homeostase/efeitos da radiação , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/imunologia , Macaca nemestrina , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Transplante Autólogo , Carga Viral/efeitos da radiação
8.
J Virol ; 90(16): 7541-7551, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27279614

RESUMO

UNLABELLED: Simian immunodeficiency virus (SIV)-infected sooty mangabeys (SMs) do not develop AIDS despite high levels of viremia. Key factors involved in the benign course of SIV infection in SMs are the absence of chronic immune activation and low levels of infection of CD4(+) central memory (TCM) and stem cell memory (TSCM) T cells. To better understand the role of virus replication in determining the main features of SIV infection in SMs, we treated 12 SMs with a potent antiretroviral therapy (ART) regimen for 2 to 12 months. We observed that ART suppressed viremia to <60 copies/ml of plasma in 10 of 12 animals and induced a variable decrease in the level of cell-associated SIV DNA in peripheral blood (average changes of 0.9-, 1.1-, 1.5-, and 3.7-fold for CD4(+) transitional memory [TTM], TCM, effector memory [TEM], and TSCM cells, respectively). ART-treated SIV-infected SMs showed (i) increased percentages of circulating CD4(+) TCM cells, (ii) increased levels of CD4(+) T cells in the rectal mucosa, and (iii) significant declines in the frequencies of HLA-DR(+) CD8(+) T cells in the blood and rectal mucosa. In addition, we observed that ART interruption resulted in rapid viral rebound in all SIV-infected SMs, indicating that the virus reservoir persists for at least a year under ART despite lower infection levels of CD4(+) TCM and TSCM cells than those seen in pathogenic SIV infections of macaques. Overall, these data indicate that ART induces specific immunological changes in SIV-infected SMs, thus suggesting that virus replication affects immune function even in the context of this clinically benign infection. IMPORTANCE: Studies of natural, nonpathogenic simian immunodeficiency virus (SIV) infection of African monkeys have provided important insights into the mechanisms responsible for the progression to AIDS during pathogenic human immunodeficiency virus (HIV) infection of humans and SIV infection of Asian macaques. In this study, for the first time, we treated SIV-infected sooty mangabeys, a natural host for the infection, with a potent antiretroviral therapy (ART) regimen for periods ranging from 2 to 12 months and monitored in detail how suppression of virus replication affected the main virological and immunological features of this nonpathogenic infection. The observed findings provide novel information on both the pathogenesis of residual immunological disease under ART during pathogenic infection and the mechanisms involved in virus persistence during primate lentiviral infections.


Assuntos
Antirretrovirais/uso terapêutico , Cercocebus atys , Infecções por Lentivirus/veterinária , Doenças dos Primatas/tratamento farmacológico , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Animais , Sangue/imunologia , Sangue/virologia , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Doenças dos Primatas/patologia , Doenças dos Primatas/virologia , Vírus da Imunodeficiência Símia/isolamento & purificação , Carga Viral
9.
Vet J ; 200(1): 44-50, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24662027

RESUMO

Neutropenia can often be corrected by treatment with granulocyte-colony stimulating factor (G-CSF) and off-label use of commercial human G-CSF (HuG-CSF) is a commonly used treatment for neutropenic animals. However, long-term HuG-CSF treatment can be associated with adverse effects, including neutropenia. Here, feline (Fe) G-CSF was produced in Pichia pastoris, pegylated (Peg) FeG-CSF and tested in cats. A randomized controlled clinical trial was conducted to evaluate the efficacy of PegFeG-CSF compared to FeG-CSF or HuG-CSF in FIV-infected (n=14), FIV-uninfected healthy cats (n=19), and in HuG-CSF-induced neutropenic cats (n=4). Daily FeG-CSF doses induced higher neutrophil production than HuG-CSF after the second week of treatment (P ⩽ 0.002). Weekly doses of PegFeG-CSF induced higher neutrophil counts and showed greater sustained activity than weekly doses of FeG-CSF. PegFeG-CSF provided the most therapeutic and sustainable neutrophil production (P<0.001) in both FIV-uninfected and FIV-infected cats, without the development of neutralizing antibodies. Conversely, all HuG-CSF-treated cats developed neutralizing antibodies, suggesting cross-reactive antibodies to endogenous G-CSF in a majority of the cases with severe neutropenia. Strikingly, when PegFeG-CSF was used to rescue cats with HuG-CSF-induced neutropenia, clinically normal neutrophil numbers returned. Thus, PegFeG-CSF appears to be a superior treatment for neutropenia in feline patients.


Assuntos
Anticorpos Neutralizantes/imunologia , Fator Estimulador de Colônias de Granulócitos/imunologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Vírus da Imunodeficiência Felina/imunologia , Infecções por Lentivirus/tratamento farmacológico , Polietilenoglicóis/uso terapêutico , Animais , Gatos , Feminino , Humanos , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Infecções por Lentivirus/imunologia , Masculino , Neutropenia/induzido quimicamente , Neutropenia/metabolismo , Neutrófilos/virologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Organismos Livres de Patógenos Específicos
10.
J Small Anim Pract ; 55(1): 39-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24279640

RESUMO

OBJECTIVES: Recombinant feline interferon-ω therapy is an immunomodulator currently used in the treatment of different retroviral diseases including feline immune deficiency virus and feline leukaemia virus. Although its mechanism of action remains unclear, this drug appears to potentiate the innate response. Acute phase proteins are one of the key components of innate immunity and studies describing their use as a monitoring tool for the immune system in animals undergoing interferon-ω therapy are lacking. This study aimed to determine whether interferon-ω therapy influences acute phase protein concentrations namely serum amyloid-A, α-1-glycoprotein and C-reactive protein. METHODS: A single-arm study was performed using 16 cats, living in an animal shelter, naturally infected with retroviruses and subjected to the interferon-ω therapy licensed protocol. Samples were collected before (D0), during (D10 and D30) and after therapy (D65). Serum amyloid-A and C-reactive protein were measured by specific enzyme-linked immunosorbent assay kits and α-1-glycoprotein by single radial immunodiffusion. RESULTS: All the acute phase proteins significantly increased in cats undergoing interferon-ω therapy (D0/D65: P<0·05) CLINICAL SIGNIFICANCE: Acute phase proteins appear to be reasonable predictors of innate-immune stimulation and may be useful in the individual monitoring of naturally retroviral infected cats undergoing interferon-ω therapy.


Assuntos
Proteína C-Reativa/análise , Doenças do Gato/tratamento farmacológico , Interferon Tipo I/uso terapêutico , Orosomucoide/análise , Infecções por Retroviridae/veterinária , Proteína Amiloide A Sérica/análise , Animais , Doenças do Gato/sangue , Doenças do Gato/virologia , Gatos , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Vírus da Imunodeficiência Felina , Infecções por Lentivirus/sangue , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/veterinária , Vírus da Leucemia Felina , Leucemia Felina/sangue , Leucemia Felina/tratamento farmacológico , Masculino , Infecções por Retroviridae/sangue , Infecções por Retroviridae/tratamento farmacológico
11.
J Med Primatol ; 42(5): 237-46, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24025078

RESUMO

BACKGROUND: Nonhuman primates (NHPs) are an important model organism for studies of HIV pathogenesis and preclinical evaluation of anti-HIV therapies. The successful translation of NHP-derived data to clinically relevant anti-HIV studies will require better understanding of the viral strains and NHP species used and their responses to existing antiretroviral therapies (ART). METHODS: Five pigtailed macaques (Macaca nemestrina) were productively infected with the SIV/HIV chimeric virus SHIV-1157 ipd3N4 following intravenous challenge. After 8 or 27 weeks, ART (PMPA, FTC, raltegravir) was initiated. Viral load, T-cell counts, and production of SHIV-specific antibodies were monitored throughout the course of infection and ART. RESULTS: ART led to a rapid and sustained decrease in plasma viral load. Suppression of plasma viremia by ART was independent of the timing of initiation during chronic infection. CONCLUSIONS: We present a new NHP model of HIV infection on antiretroviral therapy, which should prove applicable to multiple clinically relevant anti-HIV approaches.


Assuntos
Antirretrovirais/administração & dosagem , Modelos Animais de Doenças , Infecções por Lentivirus/tratamento farmacológico , Lentivirus de Primatas/efeitos dos fármacos , Macaca nemestrina , Animais , Doença Crônica/tratamento farmacológico , Quimioterapia Combinada , Carga Viral , Viremia/tratamento farmacológico , Replicação Viral/efeitos dos fármacos
12.
J Vet Intern Med ; 26(6): 1267-73, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23039914

RESUMO

BACKGROUND: (R)-9-(2-phosphonylmethoxypropyl)-2,6-diaminopurine (PMPDAP) is active against feline immunodeficiency virus (FIV) in vitro, and is less toxic than other acyclic nucleoside phosphonates. Its efficacy in naturally infected cats has not been evaluated in large controlled studies. HYPOTHESIS/OBJECTIVES: PMPDAP is effective in naturally FIV-infected cats with minimal adverse effects. ANIMALS: Forty-five privately owned cats naturally infected with FIV. METHODS: Prospective, randomized, placebo-controlled, double-blinded clinical study. Cats were randomly assigned to be treated with PMPDAP (25 mg/kg) daily, PMPDAP 3 times a week, or placebo for a period of 6 weeks. RESULTS: Administration of PMPDAP to FIV-infected cats did not lead to detectable improvements in clinical, virological, or immunological variables. Proviral load (FIV copies/10(6) cells) did not change significantly during treatment (placebo group: from 9505 ± 10119 to 8564 ± 8615; PMPDAP 3 times a week: from 4818 ± 4426 to 5041 ± 6197; PMPDAP daily: from 3525 ± 5038 to 3167 ± 5824). There was a significant decrease of red blood cell counts (×10(12) /L) (from 8.91 ± 1.82 to 7.34 ± 1.79 in cats treated 3 times per week (P < .001), and from 8.96 ± 1.13 to 6.01 ± 1.36 in cats treated daily (P < .001)), as well as of packed cell volume, and hemoglobin in both groups receiving PMPDAP. CONCLUSIONS AND CLINICAL IMPORTANCE: Administration of PMPDAP was not associated with significant improvements in clinical, immunological, or virological parameters, but treatment was associated with adverse effects, mainly anemia. Thus, PMPDAP, as administered in this study, cannot be recommended for treatment of FIV-infected cats.


Assuntos
Adenina/análogos & derivados , Antivirais/uso terapêutico , Doenças do Gato/tratamento farmacológico , Vírus da Imunodeficiência Felina , Infecções por Lentivirus/veterinária , Compostos Organofosforados/uso terapêutico , Adenina/efeitos adversos , Adenina/uso terapêutico , Animais , Antivirais/efeitos adversos , Gatos , Método Duplo-Cego , Esquema de Medicação , Feminino , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/virologia , Masculino , Compostos Organofosforados/efeitos adversos , Carga Viral
13.
J Vet Intern Med ; 25(3): 413-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21457319

RESUMO

BACKGROUND: Feline immunodeficiency virus (FIV) is a lentivirus that infects domestic and wild felidae and the course of disease is similar to that of human immunodeficiency virus infection. The thymidine nucleoside analog fozivudine (FZD) tidoxil is a lipid-zidovudine (ZDV) conjugate and member of the family of nucleoside reverse transcriptase (RT) inhibitors (NRTIs). HYPOTHESIS: FZD administration to cats during acute FIV infection produces antiviral activity with fewer adverse effects than its parent compound ZDV (AZT). ANIMALS: Male, neutered cats approximately 7 months of age (n = 12). METHODS: FZD (45 mg/kg q12h, n = 6) or placebo (n = 6) was administered PO in a nonblinded trial for 6 weeks to cats infected with the NCSU(1) isolate of FIV. Peripheral blood was collected preinfection and at 2, 4, and 6 weeks postinfection for CBC, evaluation of CD4(+) and CD8(+) cell counts by flow cytometry, and quantification of plasma and cell-associated viremia by real time RT-PCR. RESULTS: Treatment of cats with FZD during the acute stage of FIV infection decreased plasma and cell-associated viremia during the first 2 weeks of infection, but was not protective against FIV, as all cats were infected by 6 weeks. CONCLUSIONS: At the dosage used in this study, treatment with FZD results in a short-term decrease in viral load with no adverse effects. Further investigation of FZD is warranted to assess pharmacokinetics, optimal dosage, and to directly compare the antiviral activity of FZD to ZDV in naturally infected cats.


Assuntos
Antivirais/uso terapêutico , Doenças do Gato/tratamento farmacológico , Vírus da Imunodeficiência Felina , Infecções por Lentivirus/tratamento farmacológico , Lipídeos/uso terapêutico , Viremia/veterinária , Zidovudina/análogos & derivados , Animais , Doenças do Gato/virologia , Gatos , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Masculino , Organismos Livres de Patógenos Específicos , Viremia/tratamento farmacológico , Zidovudina/uso terapêutico
14.
Arch Virol ; 155(11): 1877-81, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20972598

RESUMO

In this study, strains of feline immunodeficiency virus (FIV), designated TR-D, TR-Mo and TR-Mi, isolated from three cats in Turkey, were characterized. PCR products (859 bp) from the envelope (env) gene region were amplified and sequenced, and possible geographical differences in the env gene region of Turkish FIV strains are discussed. Phylogenetic analysis of two strains showed that FIV subtype B was present in Turkey. Phylogenetic analysis showed that one new Turkish FIV strain occupies a separate branch from known clusters (subtypes A to E) from the USA, Canada, Europe and Japan.


Assuntos
Vírus da Imunodeficiência Felina/genética , Infecções por Lentivirus/veterinária , Sequência de Aminoácidos , Animais , Antibacterianos/uso terapêutico , Gatos , Hidratação , Genes env/genética , Genes env/fisiologia , Vírus da Imunodeficiência Felina/classificação , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/epidemiologia , Infecções por Lentivirus/virologia , Masculino , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase/veterinária , Turquia/epidemiologia
15.
Cell ; 141(3): 392-8, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20434978

RESUMO

Mammalian cells are equipped with so-called "restriction factors" that suppress virus replication and help to prevent virus transmission from one species to another. This Essay discusses the host restriction factor tetherin, which blocks the release of enveloped viruses like HIV-1, and the factors evolved by primate lentiviruses, such as Vpu and Nef, that antagonize tetherin's action.


Assuntos
Antígenos CD/metabolismo , HIV-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Proteínas Ligadas por GPI , Produtos do Gene nef/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Lentivirus/genética , Infecções por Lentivirus/tratamento farmacológico , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Proteínas Virais Reguladoras e Acessórias/metabolismo
16.
FASEB J ; 23(9): 2928-41, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19380511

RESUMO

Previous studies have implicated CXCL12 in the neuropathogenesis of HIV infection. Proteolysis of CXCL12 generates a neurotoxic molecule, CXCL12(5-67), which engages and activates CXCR3, in addition to exhibiting increased expression in the brains of patients with HIV-associated dementia (HAD). Herein, we investigated CXCR3-mediated neuronal injury, particularly, its contribution to autophagy suppression and the concomitant effects of antiretroviral therapy using human brain samples and models of HIV neuropathogenesis. Neurons in the brains of HAD patients and feline immunodeficiency virus (FIV)-infected animals, as well as cultured human neurons, expressed CXCR3, which was modulated in a ligand-specific manner. Exposure of human neurons to CXCL12(5-67) caused a reduction in the autophagy-associated molecule LC3 (P<0.05) and neuronal survival (P<0.05), which recapitulated findings in FIV- and HIV-infected brains (P<0.05). Oral didanosine (ddI) treatment of FIV-infected animals reduced neurobehavioral abnormalities in conjunction with diminished plasma viral load (P<0.05). F4/80 transcript abundance and CXCL12(5-67) immunoreactivity were reduced with restored neuronal LC3 expression in the brains of FIV-infected animals after ddI treatment (P<0.05). ddI treatment also prevented microglial activation and depletion of synaptic proteins in the cortex of FIV-infected animals (P<0.05). These findings indicate that the beneficial effects of ddI might be a consequence of a reduced systemic viral burden and concurrent leukocyte activation, leading to diminished neuroinflammation with preservation of neuronal autophagy by regulating CXCR3 activation.


Assuntos
Autofagia , Didanosina/farmacologia , Infecções por Lentivirus/tratamento farmacológico , Neurônios/virologia , Receptores CXCR3/metabolismo , Animais , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico , Encéfalo/patologia , Encéfalo/virologia , Gatos , Didanosina/uso terapêutico , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida Felina/patologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/patologia , Humanos , Infecções por Lentivirus/patologia , Neurônios/patologia , Fármacos Neuroprotetores , Receptores CXCR3/fisiologia
17.
J Immunol ; 177(9): 6405-14, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17056572

RESUMO

Patients with HIV infection exhibit deficits in bacterial and fungal clearance, and possibly depressed innate immunity. In this study, we observed that neutrophils from HIV-infected patients have a profound defect in chemotaxis in response to endogenous (IL-8) and bacterial (fMLP) chemoattractants, which was directly correlated with peripheral CD4(+) lymphocyte levels but not plasma viral load. A similar chemotactic defect was observed in the feline immunodeficiency virus (FIV) model of HIV infection. Intravital microscopy of FIV-infected animals revealed marked impairment in the in vivo recruitment of leukocytes; specifically integrin-dependent neutrophil adhesion and emigration induced by bacterial products. Treatment of FIV-infected animals with GM-CSF re-established both neutrophil recruitment (rolling, adhesion, and emigration) and in vitro chemotaxis to the levels seen in uninfected animals. This restoration of neutrophil responses was not due to GM-CSF-mediated priming. Rather, HIV and FIV infections resulted in defective neutrophil development, with an ensuing reduction in neutrophil granularity and chemotactic receptor expression. GM-CSF therapy restored neutrophil granularity, implying restoration of normal neutrophil development. Together, our findings underscore the fundamental defects in innate immunity caused by lentivirus infections, while also indicating that GM-CSF may be a potential immunorestorative therapy for HIV-infected patients.


Assuntos
Quimiotaxia de Leucócito , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Infecções por HIV/imunologia , Neutrófilos/imunologia , Animais , Gatos , Quimiotaxia de Leucócito/efeitos dos fármacos , Apresentação Cruzada , Grânulos Citoplasmáticos/ultraestrutura , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Infecções por HIV/tratamento farmacológico , Humanos , Vírus da Imunodeficiência Felina , Interleucina-8/farmacologia , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/veterinária , Lipopolissacarídeos/imunologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/ultraestrutura
18.
Neurobiol Dis ; 20(1): 12-26, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16137563

RESUMO

Complement activation in the brain contributes to the pathology of neuroinflammatory and neurodegenerative diseases such as neuro-AIDS. Using semiquantitative in situ hybridization and immunohistochemistry, we observed an early and sustained increase in the expression of C1q, the initial recognition subcomponent of the classical complement cascade, in the CNS during simian immunodeficiency virus (SIV) infection of rhesus macaques. Cells of the microglial/macrophage lineage were the sources for C1q protein and transcripts. C1q expression was observed in proliferating and infiltrating cells in SIV-encephalitic brains. All SIV-positive cells were also C1q-positive. Treatment with the CNS-permeant antiretroviral agent 6-chloro-2',3'-dideoxyguanosine decreased C1q synthesis along with SIV burden and focal inflammatory reactions in the brains of AIDS-symptomatic monkeys. Thus, activation of the classical complement arm of innate immunity is an early event in neuro-AIDS and a possible target for intervention.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Complemento C1q/metabolismo , Didesoxinucleosídeos/uso terapêutico , Infecções por Lentivirus/tratamento farmacológico , Macrófagos/imunologia , Microglia/imunologia , Complexo AIDS Demência/imunologia , Complexo AIDS Demência/metabolismo , Animais , Antirretrovirais/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/virologia , Modelos Animais de Doenças , Encefalite/tratamento farmacológico , Encefalite/imunologia , Encefalite/metabolismo , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/metabolismo , Macaca mulatta , Macrófagos/metabolismo , Macrófagos/virologia , Microglia/metabolismo , Microglia/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia , Carga Viral
19.
Vet Immunol Immunopathol ; 108(3-4): 357-71, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16098604

RESUMO

The hematological and virological effects of recombinant human granulocyte colony-stimulating factor (rHuG-CSF) were evaluated in feline immunodeficiency virus (FIV)-infected cats. Six age-matched, FIV-infected cats used in this cross-over study were injected subcutaneously with 5 microg/kg of rHuG-CSF daily for 3 weeks, while six control cats received a placebo. Five of six rHuG-CSF-treated cats had significant increases in neutrophil counts that peaked on days 11-21 of treatment. All rHuG-CSF-treated cats exhibited an increase in myeloid:erythroid ratios of the bone marrow cells without significant changes in lymphocyte, CD4 counts, CD4/CD8 ratios, RBC counts, FIV antibody titers, and FIV loads in peripheral blood, and without clinical and hematological toxicities. Five of six rHuG-CSF-treated cats developed antibodies to rHuG-CSF by 14-21 days of treatment, which correlated with decreasing neutrophil counts and increasing neutralizing antibodies to rHuG-CSF. Three cats re-treated with rHuG-CSF rapidly developed neutralizing antibodies to rHuG-CSF, while one cat also developed neutralizing antibodies to recombinant feline G-CSF (rFeG-CSF). Overall, rHuG-CSF treatment increased neutrophil counts in FIV-infected cats without affecting the infection status of cats. However, long-term use of rHuG-CSF is not recommended in cats because of the neutralizing antibody production to rHuG-CSF that affects the drug activity. In addition, a preliminary finding suggests that repeated treatment cycle can also induce cross-neutralizing antibodies to rFeG-CSF, which may potentially affect the homeostasis of endogenous FeG-CSF.


Assuntos
Anticorpos/imunologia , Fator Estimulador de Colônias de Granulócitos/imunologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Vírus da Imunodeficiência Felina/imunologia , Infecções por Lentivirus/tratamento farmacológico , Animais , Medula Óssea/virologia , Gatos , Fator Estimulador de Colônias de Granulócitos/antagonistas & inibidores , Humanos , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Infecções por Lentivirus/imunologia , Neutrófilos/virologia , Proteínas Recombinantes , Organismos Livres de Patógenos Específicos , Fatores de Tempo , Replicação Viral/efeitos dos fármacos
20.
Eur J Neurosci ; 19(11): 2997-3005, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15182307

RESUMO

Increased kynurenine pathway metabolism has been implicated in the aetiology of lentiviral encephalopathy. Indoleamine-2,3-dioxygenase (IDO) initiates the increased production of kynurenine pathway metabolites like quinolinic acid (QUIN). QUIN itself is elevated in AIDS-diseased monkey and human brain parenchyma and cerebrospinal fluid at levels excitotoxic for neurons in vitro. This study investigates the cellular origin of IDO biosynthesis in the brain of rhesus monkeys infected with simian immunodeficiency virus (SIV) and explores the effects of CNS-permeant antiretroviral treatment. IDO transcript and protein were absent from the brain of non-infected and SIV-infected asymptomatic monkeys. IDO biosynthesis was induced in the brain of monkeys exhibiting AIDS. Nodule and multinucleated giant cell-forming macrophages were the main sources of IDO synthesis. Treatment with the lipophilic 6-chloro-2',3'-dideoxyguanosine suppressed IDO expression in the brain of AIDS-diseased monkeys. The effectiveness of this treatment was confirmed by the reduction of virus burden and SIV-induced perivascular infiltrates, mononuclear nodules and multinucleated giant cells. Our data demonstrate that brain IDO biosynthesis is induced in a subset of monocyte-derived cells, depends on viral burden and is susceptible to antiretroviral treatment. Thus, IDO induction is associated with reversible overt inflammatory events localized to areas of active viral replication in the SIV-infected brain.


Assuntos
Antivirais/uso terapêutico , Encéfalo/enzimologia , Proteínas de Ligação a DNA , Didesoxinucleosídeos/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Triptofano Oxigenase/biossíntese , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Proteínas de Ligação ao Cálcio/metabolismo , Cromogranina A , Cromograninas/metabolismo , Didanosina/uso terapêutico , Proteína Glial Fibrilar Ácida/metabolismo , Histocitoquímica/métodos , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Indolamina-Pirrol 2,3,-Dioxigenase , Infecções por Lentivirus/tratamento farmacológico , Macaca mulatta , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos , Microscopia Confocal/métodos , RNA Mensageiro/biossíntese , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/líquido cefalorraquidiano , Fatores de Tempo , Triptofano Oxigenase/sangue , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...