Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
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
2.
J Neurovirol ; 24(2): 220-228, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29247305

RESUMO

Feline immunodeficiency virus (FIV) is a lentivirus that causes immunosuppression through virus-mediated CD4+ T cell depletion in feline species. FIV infection is complicated by virus-induced disease in the nervous system. FIV enters the brain soon after primary infection and is detected as FIV-encoded RNA, DNA, and proteins in microglia, macrophages, and astrocytes. FIV infection activates neuroinflammatory pathways including cytokines, chemokines, proteases, and ROS with accompanying neuronal injury and loss. Neurobehavioral deficits during FIV infection are manifested as impaired motor and cognitive functions. Several treatment strategies have emerged from studies of FIV neuropathogenesis including the therapeutic benefits of antiretroviral therapies, other protease inhibitors, anti-inflammatory, and neurotrophic compounds. Recently, insulin's antiviral, anti-inflammatory, and neuroprotective effects were investigated in models of lentivirus brain infection. Insulin suppressed HIV-1 replication in human microglia as well as FIV replication of lymphocytes. Insulin treatment diminished cytokine and chemokine activation in HIV-infected microglia while also protecting neurons from HIV-1 Vpr protein-mediated neurotoxicity. Intranasal (IN) insulin delivery for 6 weeks suppressed FIV expression in the brains of treated cats. IN insulin also reduced neuroinflammation and protected neurons in the hippocampus, striatum, and neocortex of FIV-infected animals. These morphological and molecular effects of IN insulin were confirmed by neurobehavioral studies that showed IN insulin-treated FIV-infected animals displayed improved motor and cognitive performance compared to sham-treated FIV-infected animals. Thus, FIV infection of the nervous system provides a valuable comparative in vivo model for discovering and evaluating disease mechanisms as well as developing therapeutic strategies for NeuroAIDS in humans.


Assuntos
Antivirais/farmacologia , Disfunção Cognitiva/tratamento farmacológico , Modelos Animais de Doenças , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Insulina/farmacologia , Administração Intranasal , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/virologia , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Gatos , Cognição/efeitos dos fármacos , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/virologia , Síndrome de Imunodeficiência Adquirida Felina/imunologia , Síndrome de Imunodeficiência Adquirida Felina/fisiopatologia , Síndrome de Imunodeficiência Adquirida Felina/virologia , Humanos , Vírus da Imunodeficiência Felina/patogenicidade , Vírus da Imunodeficiência Felina/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Microglia/efeitos dos fármacos , Microglia/virologia , Desempenho Psicomotor/efeitos dos fármacos , Latência Viral/efeitos dos fármacos , Latência Viral/fisiologia , Replicação Viral/efeitos dos fármacos
3.
J Neurovirol ; 24(2): 137-140, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29582355
4.
Nat Prod Res ; 36(16): 4159-4164, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34586005

RESUMO

The feline immunodeficiency virus (FIV) is a widespread lentivirus of felids. Due to its worldwide diffusion and the lack of an effective preventive and therapeutic protocol, it has a high impact on the cats' health. Several therapeutical protocols have been proposed, among those, phytotherapeutic compounds have been tested with the purpose to find a possible natural treatment. The most studied active compounds are derived from Ganoderma lucidum, Cordyceps sinensis, and Trametes versicolor. The present study aims to investigate in vitro antiviral effects of a commercially available compound HELP-TH1 (Camon, S.p.A., Italy) against FIV. The antiviral effect of HELP-TH1 was evaluated by quantifying and comparing the viral load of control groups, infected and not-treated cells, vs both experimental groups, infected and treated cells. Our data indicate that HELP-TH1 reduce the viral load in the experimental conditions demonstrating its antiviral effect.


Assuntos
Síndrome de Imunodeficiência Adquirida Felina , Vírus da Imunodeficiência Felina , Animais , Antivirais/farmacologia , Gatos , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Itália , Trametes
5.
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
6.
J Virol ; 82(16): 7863-74, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550661

RESUMO

Feline immunodeficiency virus (FIV) is the Lentivirus responsible for an immunodeficiency-like disease in domestic cats (Felis catus). FIV is divided into five phylogenetic subtypes (A, B, C, D, and E), based on genetic diversity. Knowledge of the geographical distribution of subtypes is relevant for understanding different disease progressions and for vaccine development. In this study, viral sequences of 26 infected cats from Rio de Janeiro, 8 undergoing treatment with zidovudine (AZT) for at least 5 years, were successfully amplified from blood specimens. gag capsid (CA), pol reverse transcriptase (RT), and env gp120 (V3-V4) regions were analyzed to determine subtypes and to evaluate potential mutations related to antiretroviral drug resistance among treated cats. Subtyping based on phylogenetic analysis was performed by the neighbor-joining and maximum likelihood methods. All of the sequences clustered with subtype B in the three regions, exhibiting low genetic variability. Additionally, we found evidence that the same virus is circulating in animals in close contact. The analysis of FIV RT sequences identified two new putative mutations related to drug resistance located in the RT "finger" domain, which has 60% identity to human immunodeficiency virus (HIV) sequence. Amino acid change K-->R at codons 64 and 69 was found in 25% and 37.5% of the treated animals, respectively. These signatures were comparable to K65R and K70R thymidine-associated mutations found in the HIV-1 HXB2 counterpart. This finding strongly suggests a position correlation between the mutations found in FIV and the K65R and K70R substitutions from drug-resistant HIV-1 strains.


Assuntos
Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Produtos do Gene env/genética , Produtos do Gene gag/genética , Produtos do Gene pol/genética , Vírus da Imunodeficiência Felina/metabolismo , Inibidores da Transcriptase Reversa/farmacologia , Animais , Brasil , Gatos , Farmacorresistência Viral , Síndrome de Imunodeficiência Adquirida Felina/virologia , Feminino , Produtos do Gene env/metabolismo , Produtos do Gene gag/metabolismo , Produtos do Gene pol/metabolismo , Técnicas Genéticas , Variação Genética , Masculino , Filogenia
7.
Vet Microbiol ; 136(1-2): 155-9, 2009 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-19036536

RESUMO

For the entry of lentivirus into target cells, fusion between its viral membrane and cellular membrane is essential. The present study was conducted to examine the inhibitory effect of modified peptides corresponding to heptad repeats (HR) 1 and 2 of feline immunodeficiency virus (FIV) envelope gp40 on the fusion between the viral and cellular membranes. FIV-N36 and FIV-C35 were synthesized as authentic peptides of the N-terminal HR1 domain and C-terminal HR2 domain of FIV gp40, respectively. FIV-C35EK1, FIV-C35EK2, and FIV-C35EK3 were peptides synthesized by modifying FIV-C35 as the X-EE-XX-KK concept to increase their solubility in water and the stability of their alpha-helicity. FIV-C35 and FIV-C35EK1 inhibited the cell membrane fusion mediated by FIV-infected cells and the replication of FIV. FIV-N36, FIV-C35EK2, and FIV-C35EK3 did not show any apparent inhibitory effect. These results indicated that the newly developed membrane fusion inhibitors could facilitate the development of novel anti-lentiviral chemotherapies.


Assuntos
Antivirais/farmacologia , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Glicoproteínas/química , Vírus da Imunodeficiência Felina/fisiologia , Fragmentos de Peptídeos/farmacologia , Proteínas do Envelope Viral/química , Internalização do Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antivirais/síntese química , Gatos , Síndrome de Imunodeficiência Adquirida Felina/virologia , Células Gigantes/virologia , Glicoproteínas/metabolismo , Células HeLa , Histocitoquímica , Humanos , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/química , Proteínas do Envelope Viral/metabolismo
8.
Viruses ; 11(9)2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31514435

RESUMO

Specific treatments for the long-life infections by feline leukemia virus (FeLV) and feline immunodeficiency virus (FIV) are either toxic, expensive or not too effective. Interferon α (IFN-α) is an immunomodulatory molecule which has been shown in vitro to decrease the release of infective particles. The aim of this study was to follow the progress of the clinical score and viral parameters of FeLV- and FIV-naturally infected privately owned cats treated with recombinant human IFN-α (rHuIFN-α, Roferon-A). Twenty-seven FeLV-infected cats (FeLV+) and 31 FIV-infected cats (FIV+) were enrolled in the study. Owners were instructed to orally administer 1 mL/day of 60 IU rHuIFN-α/mL in alternating weeks for four months. Blood samples were taken at the beginning of the study (M0), mid-treatment (M2), end of treatment (M4), and 6-10 months later (M10). Clinical status at these time points improved notably with rHuIFN-α treatment, regardless of the initial severity of the disease, an effect which lasted throughout the study in most animals (15 of the 16 FeLV+ symptomatic cats; 20 of the 22 FIV+ symptomatic cats) improved markedly their clinical situation. In FeLV+ cats plasma antigenemia (p27CA), reverse transcriptase (RT) activity, and proviral load decreased at M2 and M4 but increased again at M10 ("rebound effect"). The level of antigenemia or RT activity was below the detection limits in FIV+ cats, and the effect on proviral load was less marked than in FeLV+ cats. Taken together, these results indicate that rHuIFN-α is a good candidate for treating FeLV+ cats, but the "rebound effect" seen when treatment was discontinued suggests that additional studies should be conducted to clarify its effect on progression of the infection in cats.


Assuntos
Antivirais/administração & dosagem , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Interferon alfa-2/administração & dosagem , Vírus da Leucemia Felina/efeitos dos fármacos , Leucemia Felina/tratamento farmacológico , Administração Oral , Animais , Antígenos Virais/sangue , Gatos/virologia , Síndrome de Imunodeficiência Adquirida Felina/imunologia , Feminino , Seguimentos , Humanos , Leucemia Felina/imunologia , Masculino , Animais de Estimação/virologia , DNA Polimerase Dirigida por RNA/metabolismo , Carga Viral
9.
Viruses ; 11(9)2019 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-31480322

RESUMO

Feline immunodeficiency virus (FIV) induces opportunistic disease in chronically infected cats, and both prednisolone and cyclosporine A (CsA) are clinically used to treat complications such as lymphoma and stomatitis. However, the impact of these compounds on FIV infection are still unknown and understanding immunomodulatory effects on FIV replication and persistence is critical to guide safe and effective therapies. To determine the immunologic and virologic effects of prednisolone and CsA during FIV infection, FIV-positive cats were administered immunosuppressive doses of prednisolone (2 mg/kg) or CsA (5 mg/kg). Both prednisolone and CsA induced acute and transient increases in FIV DNA and RNA loads as detected by quantitative PCR. Changes in the proportion of lymphocyte immunophenotypes were also observed between FIV-infected and naïve cats treated with CsA and prednisolone, and both treatments caused acute increases in CD4+ lymphocytes that correlated with increased FIV RNA. CsA and prednisolone also produced alterations in cytokine expression that favored a shift toward a Th2 response. Pre-treatment with CsA slightly enhanced the efficacy of antiretroviral therapy but did not enhance clearance of FIV. Results highlight the potential for drug-induced perturbation of FIV infection and underscore the need for more information regarding immunopathologic consequences of therapeutic agents on concurrent viral infections.


Assuntos
Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Imunossupressores/uso terapêutico , Replicação Viral/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Gatos , Ciclosporina/uso terapêutico , Citocinas/sangue , Síndrome de Imunodeficiência Adquirida Felina/imunologia , Síndrome de Imunodeficiência Adquirida Felina/virologia , Vírus da Imunodeficiência Felina/imunologia , Vírus da Imunodeficiência Felina/fisiologia , Contagem de Linfócitos , Prednisolona/uso terapêutico , Carga Viral/efeitos dos fármacos
10.
Brain ; 130(Pt 8): 2011-23, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17616550

RESUMO

Antiretroviral toxic neuropathy (ATN) has become a common peripheral neuropathy among HIV/AIDS patients, for which the underlying pathogenesis is uncertain. Indeed, no models exist for ATN that assess the interaction between retroviral infection and antiretroviral therapy. Herein, we developed ex vivo and in vivo models of ATN induced by didanosine (ddI) following infection by the lentivirus, feline immunodeficiency virus (FIV), permitting us to address the working hypothesis that ddI mediates ATN through mitochondrial injury in neurons. We investigated neuronal morphology, neurobehavioural testing, viral load, mitochondrial and neurotrophic factor gene expression after ddI treatment of FIV-infected and uninfected animals or dorsal root ganglia (DRG) cultures. ddI caused concentration-dependent neuronal injury in cultured feline DRGs (P < 0.05), together with reduced viral replication and diminished expression of mitochondrial cytochrome C oxidase subunit I gene (mtCOX I) and the neurotrophin, brain-derived neurotrophic factor (BDNF). Indeed, BDNF treatment reversed neuronal injury caused by FIV infection in the presence or absence of ddI exposure (P < 0.05). In vivo FIV infection revealed delays in withdrawal latency to a noxious stimulus, which were exacerbated by ddI treatment. Epidermal density of nerve endings was reduced after FIV infection (P < 0.05), especially with ddI treatment. Although viral replication in blood was suppressed in ddI-treated animals (P < 0.05), ddI had a limited effect on viral abundance in DRGs of the same animals. ddI decreased mtCOX I expression in DRG neurons of FIV-infected animals (P < 0.05). BDNF expression was downregulated by ddI in DRG Schwann cells following FIV infection. Thus, ddI treatment during FIV infection resulted in additive pathogenic effects contributing to the development of ATN, which was associated with mitochondrial injury on neurons and reduced BDNF production by Schwann cells in DRGs, highlighting the convergent pathogenic effects that antiretroviral drugs might have in patients with HIV infection.


Assuntos
Fármacos Anti-HIV/toxicidade , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Didanosina/toxicidade , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Animais , Western Blotting , Gatos , Células Cultivadas , Modelos Animais de Doenças , Síndrome de Imunodeficiência Adquirida Felina/metabolismo , Síndrome de Imunodeficiência Adquirida Felina/virologia , Gânglios Espinais/efeitos dos fármacos , Mitocôndrias/patologia , Doenças do Sistema Nervoso Periférico/metabolismo , Reação em Cadeia da Polimerase/métodos , Carga Viral
12.
Vet Immunol Immunopathol ; 109(3-4): 245-54, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16169599

RESUMO

Feline immunodeficiency virus sustains an AIDS-like syndrome in cats, which is considered a relevant model for human AIDS. Under precise enrolment requirements, 30 naturally infected cats showing overt disease were included in a trial of low-dose, oral human interferon-alpha treatment. Twenty-four of them received 10 IU/Kg of human interferon-alpha and 6 placebo only on a daily basis under veterinary supervision. The low-dose human interferon-alpha treatment significantly prolonged the survival of virus-infected cats (p<0.01) and brought to a rapid improvement of disease conditions in the infected hosts. Amelioration of clinical conditions was neither correlated with plasma viremia, nor with proviral load in leukocytes. A good survival of CD4+ T cells and a slow increase of CD8+ T cells were also observed in human interferon-alpha-treated cats. Interestingly, the improvement of the total leukocyte counts showed a much stronger correlation with the recovery from serious opportunistic infections. As shown in other models of low-dose interferon-alpha treatment, there was a rapid regression of overt immunopathological conditions in virus-infected cats. This hints at a major role of interferon-alpha in the control circuits of inflammatory cytokines, which was probably the very foundation of the improved clinical score and survival despite the unabated persistence of virus and virus-infected cells.


Assuntos
Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/crescimento & desenvolvimento , Interferon-alfa/administração & dosagem , Viremia/veterinária , Animais , Contagem de Linfócito CD4 , Relação CD4-CD8 , Linfócitos T CD8-Positivos , Gatos , Síndrome de Imunodeficiência Adquirida Felina/imunologia , Síndrome de Imunodeficiência Adquirida Felina/virologia , Feminino , Citometria de Fluxo/veterinária , Vírus da Imunodeficiência Felina/genética , Vírus da Imunodeficiência Felina/imunologia , Masculino , RNA Viral/química , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sobrevida , Viremia/tratamento farmacológico , Viremia/imunologia
13.
Tierarztl Prax Ausg K Kleintiere Heimtiere ; 44(6): 417-423, 2016 Dec 05.
Artigo em Inglês, Alemão | MEDLINE | ID: mdl-27808347

RESUMO

OBJECTIVE: The bee venom melittin shows an antiviral efficacy against the human immunodeficiency virus in cell culture. It was shown to be non-toxic for cats. Aim of this pilot study was to investigate the clinical efficacy and side-effects of melittin in cats naturally infected with feline immunodeficiency virus (FIV). MATERIAL AND METHODS: The study was performed as a prospective, placebo-controlled double-blinded trial. Twenty cats were included, of which 10 cats each were treated with either melittin (500 µg/kg body weight) or phosphate-buffered saline (placebo) subcutaneously twice per week. During the treatment period of 6 weeks, the cats' general health status, determined by the Karnofsky's score, and the severity of clinical signs (conjunctivitis and stomatitis) using a clinical scoring system were evaluated. Haematology, biochemistry profiles, lymphocyte subpopulations, CD4/CD8 ratio, and pterines (biopterine, 7-xanthopterine) as surrogate parameters were also compared. RESULTS: The general health status and the clinical scores for conjunctivitis and stomatitis improved in cats treated with melittin. A statistically significant improvement however could only be detected for conjunctivitis in cats treated with melittin compared to cats treated with placebo which was likely due to different scores between both groups at the beginning. No influence on the lymphocyte subpopulations, CD4/CD8 ratio, and pterine concentrations was observed. No side effects occurred in this study. CONCLUSION AND CLINICAL RELEVANCE: In the protocol used in the present study, no significant efficacy of melittin could be detected. However, efficacy of melittin, especially if applied in a higher dosage as in the present study or for a longer period, could be evaluated in further studies. Synergistic effects if used in combination with classic antiretroviral drugs could be an interesting future approach.


Assuntos
Antivirais/uso terapêutico , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/isolamento & purificação , Meliteno/uso terapêutico , Animais , Relação CD4-CD8 , Gatos , Método Duplo-Cego , Subpopulações de Linfócitos , Projetos Piloto , Estudos Prospectivos , Resultado do Tratamento
14.
J Feline Med Surg ; 18(4): 264-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25855689

RESUMO

OBJECTIVES: Feline immunodeficiency virus (FIV) is a lentivirus that induces AIDS-like disease in cats. Some of the antiretroviral drugs available to treat patients with HIV type 1 are used to treat FIV-infected cats; however, antiretroviral therapy (ART) is not used in cats as a long-term treatment. In this study, the effects of long-term ART were evaluated in domestic cats treated initially with the nucleoside transcriptase reverse inhibitor (NTRI) zidovudine (AZT) over a period ranging from 5-6 years, followed by a regimen of the NTRI lamivudine (3TC) plus AZT over 3 years. METHODS: Viral load, sequencing of pol (reverse transcriptase [RT]) region and CD4:CD8 lymphocyte ratio were evaluated during and after treatment. Untreated cats were evaluated as a control group. RESULTS: CD4:CD8 ratios were lower, and uncharacterized resistance mutations were found in the RT region in the group of treated cats. A slight increase in viral load was observed in some cats after discontinuing treatment. CONCLUSIONS AND RELEVANCE: The data strongly suggest that treated cats were resistant to therapy, and uncharacterized resistance mutations in the RT gene of FIV were selected for by AZT. Few studies have been conducted to evaluate the effect of long-term antiretroviral therapy in cats. To date, resistance mutations have not been described in vivo.


Assuntos
Linfócitos T CD8-Positivos , Doenças do Gato/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Lamivudina/uso terapêutico , Inibidores da Transcriptase Reversa/uso terapêutico , Animais , Gatos , Seguimentos , Dados de Sequência Molecular , Carga Viral , Zidovudina/uso terapêutico
15.
AIDS ; 19(16): 1781-91, 2005 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-16227785

RESUMO

OBJECTIVE: To study the effects of HIV-1 and feline immunodeficiency virus (FIV) on neural stem cell viability, together with the neurotrophic properties of growth hormone (GH) in models of pediatric neuroAIDS. DESIGN AND METHODS: Mouse neural stem cells were infected in vitro with a Sindbis virus vector (SIN-HIVenv) expressing the envelope protein from the brain-derived HIV-1 strain JR-FL using a vector expressing enhanced green fluorescent protein (SIN-EGFP) as control. Cell survival and alterations in expression of neural stem cell markers upon GH treatment was assessed. Neonatal cats were infected with a neurovirulent FIV strain and 6 weeks after infection treated with GH for 6 weeks. Twelve weeks post-infection, neural progenitor cell marker expression, neuronal loss and neuroinflammation in brain were examined using real time reverse transcription-PCR and immunohistochemical analyses. RESULTS: HIV-1 envelope expression in neural stem cells reduced nestin expression (P < 0.05) and induced cell death (P < 0.001), which was blocked by GH. In the frontal cortex of FIV-infected cats neuroinflammation, loss of differentiated neurons (P < 0.01) and aberrant neuronal progenitor cell gene expression (P < 0.05) were observed. FIV envelope expression was detected in neural progenitor and monocytoid cells. GH treatment of FIV-infected animals induced insulin-like growth factor-1 expression in neurons (P < 0.01), enhanced neuronal survival (P < 0.01) and increased nestin expression (P < 0.05). Moreover, improved neurobehavioral performance (P < 0.01) and immunological status (P < 0.001) were observed, among GH-treated animals infected with FIV. CONCLUSION: GH protects neural stem cells that are susceptible to lentivirus-mediated injury. Thus, GH may be a potential treatment for pediatric neuroAIDS because of its neurotrophic actions.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Hormônio do Crescimento/uso terapêutico , Proteínas de Filamentos Intermediários/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Células-Tronco/patologia , Complexo AIDS Demência/patologia , Animais , Gatos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida Felina/patologia , Genes env , Vírus da Imunodeficiência Felina , Camundongos , Modelos Biológicos , Nestina , Neurônios/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos
16.
Antivir Ther ; 10(5): 671-80, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16152761

RESUMO

The Trp-rich motif (TrpM) of the transmembrane glycoprotein (TM) of lentiviruses is an attractive domain on which to design new potential cell entry peptide inhibitors. We recently demonstrated that an octapeptide reproducing the TrpM of feline immunodeficiency virus (FIV), designated C8, broadly inhibited this virus in vitro and that the retroinverso analogue of this peptide (riC8) was almost as inhibitory and exhibited features suggestive of a much increased stability. Here, we demonstrated that riC8 is indeed highly stable, maintaining its concentration unchanged for at least 24 h in cat serum in vitro. Furthermore, once inoculated into cats, riC8 produced no major acute toxic effects and exhibited satisfactory pharmacokinetic properties. Finally, we report the results of a short-term monotherapy experiment in chronically FIV-infected cats showing that riC8 is well tolerated and also has substantial antiviral activity in vivo. In particular, the mean viral load of riC8-treated animals declined progressively with increasing time of treatment, whereas that of control animals given C8 or solvent alone did not. These results provide the first evidence that clinically useful inhibition of virus replication with a small peptide derived from a functional domain of the TM of a lentivirus can be achieved in vivo.


Assuntos
Antivirais/uso terapêutico , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Oligopeptídeos/uso terapêutico , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Antivirais/química , Antivirais/farmacologia , Gatos , Linhagem Celular Tumoral , Doença Crônica , Avaliação Pré-Clínica de Medicamentos , Síndrome de Imunodeficiência Adquirida Felina/virologia , Feminino , Vírus da Imunodeficiência Felina/isolamento & purificação , Dados de Sequência Molecular , Oligopeptídeos/química , Oligopeptídeos/genética , Oligopeptídeos/farmacologia , Triptofano , Carga Viral
17.
J Feline Med Surg ; 17(11): 925-39, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26486979

RESUMO

GLOBAL IMPORTANCE: The two feline retroviruses, feline immunodeficiency virus (FIV) and feline leukaemia virus (FeLV), are global and widespread, but differ in their potential to cause disease. VIRAL INFECTION - FIV: FIV, a lentivirus that shares many properties with human immunodeficiency virus (HIV), can cause an acquired immune deficiency syndrome, which predisposes cats to other infections, stomatitis, neurological disorders and tumours. Although secondary infections are common, specific opportunistic infections or acquired immunodeficiency virus-defining infections, such as those that occur with HIV, are not commonly reported in FIV-infected cats. In most naturally infected cats, FIV does not cause a severe clinical syndrome; with appropriate care, FIV-infected cats can live many years before succumbing to conditions unrelated to their FIV infection. Thus, overall survival time is not necessarily shorter than in uninfected cats, and quality of life is usually high over many years or lifelong. VIRAL INFECTION - FELV: FeLV, an oncornavirus, is more pathogenic than FIV. Historically, it was considered to account for more disease-related deaths and clinical syndromes in cats than any other infectious agent. Recently, the prevalence and importance of FeLV have been decreasing, mainly because of testing and eradication programmes and the use of FeLV vaccines. Progressive FeLV infection can cause tumours, bone marrow suppression and immunosuppression, as well as neurological and other disorders, and leads to a decrease in life expectancy. However, with appropriate care, many FeLV-infected cats can also live several years with a good quality of life. PRACTICAL RELEVANCE: A decision regarding treatment or euthanasia should never be based solely on the presence or absence of a retrovirus infection. Antiviral chemotherapy is of increasing interest in veterinary medicine, but is still not used commonly. EVIDENCE BASE: This article reviews the current literature on antiviral chemotherapy in retrovirus-infected cats, focusing on drugs that are currently available on the market and, thus, could potentially be used in cats.


Assuntos
Antivirais/uso terapêutico , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Proteínas Oncogênicas de Retroviridae/uso terapêutico , Vacinação/veterinária , Vacinas Virais/uso terapêutico , Animais , Gatos , Síndrome de Imunodeficiência Adquirida Felina/diagnóstico , Síndrome de Imunodeficiência Adquirida Felina/patologia , Vírus da Imunodeficiência Felina/isolamento & purificação , Guias de Prática Clínica como Assunto
18.
J Feline Med Surg ; 17(4): 375-80, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25425600

RESUMO

This report describes a 6-year-old neutered male feline immunodeficiency-positive cat with repeated abdominal and thoracic effusions. The cat was diagnosed with and treated for lymphosarcoma but remission was short-lived and, on re-evaluation, a fungal peritoneal exudate was noted. Cytology of the organisms is described and the culture elucidated Cladosporium carrionii, an important cause of chromoblastomycosis. Treatment with itraconazole was unsuccessful in this case.


Assuntos
Cromoblastomicose/microbiologia , Cromoblastomicose/veterinária , Síndrome de Imunodeficiência Adquirida Felina/complicações , Linfoma/complicações , Linfoma/veterinária , Animais , Antifúngicos/administração & dosagem , Gatos , Cromoblastomicose/tratamento farmacológico , Cladosporium/isolamento & purificação , Evolução Fatal , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/isolamento & purificação , Itraconazol/administração & dosagem , Masculino , África do Sul
19.
J Feline Med Surg ; 17(2): 79-86, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24782459

RESUMO

Feline immunodeficiency virus (FIV), the causative agent of an acquired immunodeficiency syndrome in cats (feline AIDS), is a ubiquitous health threat to the domestic and feral cat population, also triggering disease in wild animals. No registered antiviral compounds are currently available to treat FIV-infected cats. Several human antiviral drugs have been used experimentally in cats, but not without the development of serious adverse effects. Here we report on the treatment of six naturally FIV-infected cats, suffering from moderate to severe disease, with the antiretroviral compound (R)-9-(2-phosphonylmethoxypropyl)-2,6-diaminopurine ([R]-PMPDAP), a close analogue of tenofovir, a widely prescribed anti-HIV drug in human medicine. An improvement in the average Karnofsky score (pretreatment 33.2 ± 9.4%, post-treatment 65±12.3%), some laboratory parameters (ie, serum amyloid A and gammaglobulins) and a decrease of FIV viral load in plasma were noted in most cats. The role of concurrent medication in ameliorating the Karnofsky score, as well as the possible development of haematological side effects, are discussed. Side effects, when noted, appeared mild and reversible upon cessation of treatment. Although strong conclusions cannot be drawn owing to the small number of patients and lack of a placebo-treated control group, the activity of (R)-PMPDAP, as observed here, warrants further investigation.


Assuntos
Adenina/análogos & derivados , Antivirais/uso terapêutico , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina/efeitos dos fármacos , Compostos Organofosforados/uso terapêutico , Adenina/uso terapêutico , Animais , Gatos , Síndrome de Imunodeficiência Adquirida Felina/virologia , Humanos , Injeções Subcutâneas/veterinária , Avaliação de Estado de Karnofsky , Resultado do Tratamento , Replicação Viral/efeitos dos fármacos
20.
Res Vet Sci ; 99: 87-95, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25747956

RESUMO

This study assesses viremia, provirus and blood cytokine profile in naturally FIV-infected cats treated with two distinct protocols of interferon omega (rFeIFN-ω). Samples from FIV-cats previously submitted to two single-arm studies were used: 7/18 received the licensed/subcutaneous protocol (SC) while 11/18 were treated orally (PO). Viremia, provirus and blood mRNA expression of interleukin (IL)-1, IL-4, IL-6, IL-10, IL-12p40, Interferon-γ and Tumor Necrosis Factor-α were monitored by Real-Time qPCR. Concurrent plasma levels of IL-6, IL-12p40 and IL-4 were assessed by ELISA. IL-6 plasma levels decreased in the SC group (p = 0.031). IL-6 mRNA expression (p = 0.037) decreased in the PO group, albeit not sufficiently to change concurrent plasma levels. Neither viremia nor other measured cytokines changed with therapy. Proviral load increased in the SC group (p = 0.031), which can be justified by a clinically irrelevant increase of lymphocyte count. Independently of the protocol, rFeIFN-ω seems to act on innate immunity by reducing pro-inflammatory stimulus.


Assuntos
Doenças do Gato/virologia , Gatos/virologia , Citocinas/sangue , Síndrome de Imunodeficiência Adquirida Felina/tratamento farmacológico , Vírus da Imunodeficiência Felina , Interferon Tipo I/uso terapêutico , Carga Viral , Viremia/veterinária , Administração Oral , Animais , Doenças do Gato/sangue , Doenças do Gato/imunologia , Síndrome de Imunodeficiência Adquirida Felina/sangue , Síndrome de Imunodeficiência Adquirida Felina/imunologia , Vírus da Imunodeficiência Felina/imunologia , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Injeções Subcutâneas , Interferon Tipo I/administração & dosagem , Interferon Tipo I/farmacologia , Subunidade p40 da Interleucina-12/sangue , Interleucina-4/sangue , Interleucina-6/sangue , RNA Mensageiro/sangue , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Resultado do Tratamento , Carga Viral/efeitos dos fármacos , Carga Viral/imunologia , Viremia/sangue , Viremia/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA