Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
J Neuroimmune Pharmacol ; 16(2): 437-453, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32627098

RESUMO

HIV affects 37 million people worldwide, 25-69% of which develop HIV-associated neurocognitive disorders (HAND) regardless of antiviral treatment. HIV infection of the brain decreases cognitive function, disrupts/impairs learning and memory, and reduces quality of life for those affected. HIV-induced neuroinflammation has been associated with viral proteins such as gp120 and Tat, which remain elevated in the CNS even in patients with low peripheral viremia counts. In this study, we examined the effects of gp120 on neuroinflammation in immunodeficient vs. immunocompetent states by examining neuroinflammatory markers in gp120tg mice with or without systemic immunodeficiency caused by murine retroviral administration (LP-BM5 murine AIDS). Changes in inflammatory cytokine/chemokine mRNA expression was complex and dependent upon expression of gp120 protein, immunodeficiency status, brain region (hippocampus, frontal lobe, or striatum), and age. Gp120 expression reduced hippocampal synaptophysin expression but did not affect animals' learning/memory on the spontaneous T-maze test in our experimental conditions. Our results emphasize the critical role of the neuroinflammatory micro-environment and the peripheral immune system context in which gp120 acts. Multiple factors, particularly system-level differences in the immune response of different brain regions, need to be considered when developing treatment for HAND. Graphical Abstract.


Assuntos
Complexo AIDS Demência/imunologia , Complexo AIDS Demência/patologia , Proteína gp120 do Envelope de HIV , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Camundongos , Camundongos Transgênicos
2.
J Virol ; 90(16): 7118-7130, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226373

RESUMO

UNLABELLED: Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE: The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.


Assuntos
Glutationa/deficiência , Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/complicações , Síndrome de Imunodeficiência Adquirida Murina/etiologia , Infecções por Retroviridae/complicações , Células Th2/imunologia , Infecções Tumorais por Vírus/complicações , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Ativação Linfocitária , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Baço/imunologia , Baço/metabolismo , Baço/virologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th2/metabolismo , Células Th2/virologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia
3.
J Med Food ; 18(8): 882-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26076116

RESUMO

The immunomodulatory effects of a dietary supplement of purple sweet potato extract (PSPE) in LP-BM5 murine leukemia virus (MuLV)-induced immune-deficient mice were investigated. Mice were divided into six groups: normal control, infected control (LP-BM5 MuLV infection), positive control (LP-BM5 MuLV infection+dietary supplement of red ginseng 300 mg/kg), purple sweet potato water extract (PSPWE) (LP-BM5 MuLV infection+dietary supplement of PSPE 300 mg/kg), PSP10EE (LP-BM5 MuLV infection+dietary supplement of 10% ethanol PSPE 300 mg/kg), and PSP80EE (LP-BM5 MuLV infection+dietary supplement of 80% ethanol PSPE 300 mg/kg). Dietary supplementation began on the day of LP-BM5 MuLV infection and continued for 12 weeks. Dietary supplementation of PSPE inhibited LP-BM5 MuLV-induced splenomegaly and lymphadenopathy and attenuated the suppression of T- and B-cell proliferation and T helper 1/T helper 2 cytokine imbalance in LP-BM5 MuLV-infected mice. Dietary supplement of PSPE increased the activity of the antioxidant enzymes, superoxide dismutase and glutathione peroxidase. The data suggest that PSPE may ameliorate immune dysfunction due to LP-BM5 MuLV infection by modulating antioxidant defense systems.


Assuntos
Antioxidantes/farmacologia , Imunomodulação/efeitos dos fármacos , Ipomoea batatas/química , Ipomoea batatas/imunologia , Vírus da Leucemia Murina , Síndrome de Imunodeficiência Adquirida Murina/dietoterapia , Animais , Antioxidantes/química , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/análise , Modelos Animais de Doenças , Glutationa Peroxidase/metabolismo , Vírus da Leucemia Murina/patogenicidade , Doenças Linfáticas/dietoterapia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Extratos Vegetais/farmacologia , Preparações de Plantas/química , Esplenomegalia/dietoterapia , Superóxido Dismutase/metabolismo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos
4.
J Neuroinflammation ; 10: 98, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23902750

RESUMO

BACKGROUND: Highly active antiretroviral therapy (HAART) restores inflammatory immune responses in AIDS patients which may unmask previous subclinical infections or paradoxically exacerbate symptoms of opportunistic infections. In resource-poor settings, 25% of patients receiving HAART may develop CNS-related immune reconstitution inflammatory syndrome (IRIS). Here we describe a reliable mouse model to study underlying immunopathological mechanisms of CNS-IRIS. METHODS: Utilizing our HSV brain infection model and mice with MAIDS, we investigated the effect of immune reconstitution on MAIDS mice harboring opportunistic viral brain infection. Using multi-color flow cytometry, we quantitatively measured the cellular infiltrate and microglial activation. RESULTS: Infection with the LP-BM5 retroviral mixture was found to confer susceptibility to herpes simplex virus (HSV)-1 brain infection to normally-resistant C57BL/6 mice. Increased susceptibility to brain infection was due to severe immunodeficiency at 8 wks p.i. and a marked increase in programmed death-1 (PD-1) expression on CD4+ and CD8+ T-cells. Both T-cell loss and opportunistic brain infection were associated with high level PD-1 expression because PD-1-knockout mice infected with LP-BM5 did not exhibit lymphopenia and retained resistance to HSV-1. In addition, HSV-infection of MAIDS mice stimulated peripheral immune cell infiltration into the brain and its ensuing microglial activation. Interestingly, while opportunistic herpes virus brain infection of C57BL/6 MAIDS mice was not itself lethal, when T-cell immunity was reconstituted through adoptive transfer of virus-specific CD3+ T-cells, it resulted in significant mortality among recipients. This immune reconstitution-induced mortality was associated with exacerbated neuroinflammation, as determined by MHC class II expression on resident microglia and elevated levels of Th1 cytokines in the brain. CONCLUSIONS: Taken together, these results indicate development of an immune reconstitution disease within the central nervous system (CNS-IRD). Experimental immune reconstitution disease of the CNS using T-cell repopulation of lymphopenic murine hosts harboring opportunistic brain infections may help elucidate neuroimmunoregulatory networks that produce CNS-IRIS in patients initiating HAART.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Viroses do Sistema Nervoso Central/imunologia , Herpes Simples/imunologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Infecções Oportunistas Relacionadas com a AIDS/mortalidade , Infecções Oportunistas Relacionadas com a AIDS/patologia , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Viroses do Sistema Nervoso Central/mortalidade , Viroses do Sistema Nervoso Central/patologia , Herpes Simples/mortalidade , Herpes Simples/patologia , Inflamação/imunologia , Inflamação/mortalidade , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/mortalidade , Síndrome de Imunodeficiência Adquirida Murina/patologia
5.
J Neuroimmunol ; 261(1-2): 37-43, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23726765

RESUMO

B6 mice infected with LP-BM5 develop severe immunodeficiency (termed murine acquired immunodeficiency syndrome (MAIDS)) and peripheral neuropathy. To determine whether microglial CD40 is involved in LP-BM5-induced peripheral neuropathy, B6-CD40 knockout (KO) mice and B6-CD40 KO mice adoptively transferred either total leukocytes or B cells were examined for behavioral sensitivity, tissue viral loads, cytokine responses, and the development of MAIDS. All three CD40 KO groups developed MAIDS, the severity of which was correlated with peripheral cytokine responses. CD40 KO mice displayed significantly reduced mechanical hypersensitivity post-infection compared to wild-type mice regardless of cell transfer. These findings support microglial CD40 involvement in LP-BM5-induced peripheral neuropathy.


Assuntos
Antígenos CD40/imunologia , Vírus da Leucemia Murina/imunologia , Microglia/imunologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Doenças do Sistema Nervoso Periférico/metabolismo , Retroviridae/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos B/virologia , Antígenos CD40/deficiência , Leucócitos/imunologia , Leucócitos/patologia , Leucócitos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Microglia/virologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Doenças do Sistema Nervoso Periférico/imunologia , Doenças do Sistema Nervoso Periférico/virologia , Distribuição Aleatória
6.
Virol J ; 10: 154, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23680027

RESUMO

BACKGROUND: Indoleamine 2,3-dioxygenase (IDO) is an immunomodulatory intracellular enzyme involved in tryptophan degradation. IDO is induced during cancer and microbial infections by cytokines, ligation of co-stimulatory molecules and/or activation of pattern recognition receptors, ultimately leading to modulation of the immune response. LP-BM5 murine retroviral infection induces murine AIDS (MAIDS), which is characterized by profound and broad immunosuppression of T- and B-cell responses. Our lab has previously described multiple mechanisms regulating the development of immunodeficiency of LP-BM5-induced disease, including Programmed Death 1 (PD-1), IL-10, and T-regulatory (Treg) cells. Immunosuppressive roles of IDO have been demonstrated in other retroviral models, suggesting a possible role for IDO during LP-BM5-induced retroviral disease progression and/or development of viral load. METHODS: Mice deficient in IDO (B6.IDO-/-) and wildtype C57BL/6 (B6) mice were infected with LP-BM5 murine retrovirus. MAIDS and LP-BM5 viral load were assessed at termination. RESULTS: As expected, IDO was un-inducible in B6.IDO-/- during LP-BM5 infection. B6.IDO-/- mice infected with LP-BM5 retrovirus succumbed to MAIDS as indicated by splenomegaly, serum hyper IgG2a and IgM, decreased responsiveness to B- and T-cell mitogens, conversion of a proportion of CD4+ T cells from Thy1.2+ to Thy1.2-, and increased percentages of CD11b+Gr-1+ cells. LP-BM5 infected B6.IDO-/- mice also demonstrated the development of roughly equivalent disease kinetics as compared to infected B6 mice. Splenic viral loads of B6 and B6.IDO-/- mice were also equivalent after infection as measured by LP-BM5-specific Def Gag and Eco Gag viral mRNA, determined by qRT-PCR. CONCLUSIONS: Collectively, these results demonstrate IDO neither plays an essential role, nor is required, in LP-BM5-induced disease progression or LP-BM5 viral load.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , Retroviridae/isolamento & purificação , Animais , Progressão da Doença , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Carga Viral
7.
Cytokine ; 61(3): 862-75, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23415673

RESUMO

Interleukin-17 (IL-17), a pro-inflammatory cytokine produced by CD4+ Th17 cells, has been associated with the pathogenesis of several autoimmune diseases including uveitis. The fate of IL-17 during HIV/AIDS, however, remains unclear, and a possible role for IL-17 in the pathogenesis of AIDS-related diseases has not been investigated. Toward these ends, we performed studies using a well-established animal model of experimental murine cytomegalovirus (MCMV) retinitis that develops in C57/BL6 mice with retrovirus-induced immunosuppression (MAIDS). After establishing baseline levels for IL-17 production in whole splenic cells of healthy mice, we observed a significant increase in IL-17 mRNA levels in whole splenic cells of mice with MAIDS of 4-weeks (MAIDS-4), 8-weeks (MAIDS-8), and 10-weeks (MAIDS-10) duration. In contrast, enriched populations of splenic CD4+ T cells, splenic macrophages, and splenic neutrophils exhibited a reproducible decrease in levels of IL-17 mRNA during MAIDS progression. To explore a possible role for IL-17 during the pathogenesis of MAIDS-related MCMV retinitis, we first demonstrated constitutive IL-17 expression in retinal photoreceptor cells of uninfected eyes of healthy mice. Subsequent studies, however, revealed a significant decrease in intraocular levels of IL-17 mRNA and protein in MCMV-infected eyes of MAIDS-10 mice during retinitis development. That MCMV infection might cause a remarkable downregulation of IL-17 production was supported further by the finding that systemic MCMV infection of healthy, MAIDS-4, or MAIDS-10 mice also significantly decreased IL-17 mRNA production by splenic CD4+ T cells. Based on additional studies using IL-10 -/- mice infected systemically with MCMV and IL-10 -/- mice with MAIDS infected intraocularly with MCMV, we propose that MCMV infection downregulates IL-17 production via stimulation of suppressor of cytokine signaling (SOCS)-3 and interleukin-10.


Assuntos
Retinite por Citomegalovirus/patologia , Retinite por Citomegalovirus/virologia , Regulação para Baixo , Interleucina-17/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Muromegalovirus/fisiologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Retinite por Citomegalovirus/genética , Retinite por Citomegalovirus/imunologia , Progressão da Doença , Suscetibilidade a Doenças , Olho/metabolismo , Feminino , Terapia de Imunossupressão , Interleucina-17/biossíntese , Interleucina-17/genética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/genética , Síndrome de Imunodeficiência Adquirida Murina/patologia , Neutrófilos/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retroviridae/fisiologia , Baço/metabolismo , Baço/patologia , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
8.
J Virol ; 86(20): 10961-78, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22837196

RESUMO

AIDS-related human cytomegalovirus (HCMV) retinitis remains a major ophthalmologic problem worldwide. Although this sight-threatening disease is well characterized clinically, many pathogenic issues remain unresolved, among them a basic understanding of the relative roles of cell death pathways during development of retinal tissue destruction. Using an established model of experimental murine cytomegalovirus (MCMV) retinitis in mice with retrovirus-induced immunosuppression (MAIDS), we initially investigated MCMV-infected eyes for evidence of apoptosis-associated molecules in mice with MAIDS of 4 weeks' (MAIDS-4) and 10 weeks' (MAIDS-10) duration, which were resistant and susceptible to retinal disease, respectively, but which harbored equivalent amounts of infectious MCMV. Whereas MCMV-infected eyes of MAIDS-4 mice showed little evidence of apoptosis-associated molecules, MCMV-infected eyes of MAIDS-10 mice showed significant amounts of tumor necrosis factor alpha (TNF-α), TNF receptors 1 and 2, active caspase 8, active caspase 3, TNF-related apoptosis-inducing ligand (TRAIL), TRAIL-R(DR5), Fas, and Fas ligand mRNAs and/or proteins, all detected at peak amounts prior to development of most severe retinal disease. Immunohistochemical staining showed macrophages, granulocytes (neutrophils), Müller cells, and microglial cells as TNF-α sources. Remarkably, quantification of apoptosis by terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) assay suggested that apoptosis contributed minimally to retinal disease in MCMV-infected eyes of MAIDS-10 mice. Subsequent studies demonstrated that MCMV-infected eyes of MAIDS-10 mice, but not MAIDS-4 mice, showed evidence of significant increases in molecules associated with two additional cell death pathways, necroptosis (receptor-interacting protein 1 [RIP1] and RIP3 mRNAs) and pyroptosis (caspase 1, interleukin 1ß [IL-1ß], and IL-18 mRNAs). We conclude that apoptosis, necroptosis, and pyroptosis participate simultaneously during MAIDS-related MCMV retinitis, and all may play a role during AIDS-related HCMV retinitis.


Assuntos
Apoptose , Retinite por Citomegalovirus/patologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Animais , Caspase 1/biossíntese , Caspase 3/biossíntese , Caspase 8/biossíntese , Retinite por Citomegalovirus/complicações , Retinite por Citomegalovirus/imunologia , Proteína Ligante Fas/biossíntese , Proteína Ligante Fas/genética , Feminino , Granulócitos/metabolismo , Interleucina-18/biossíntese , Interleucina-18/genética , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/complicações , Síndrome de Imunodeficiência Adquirida Murina/virologia , Muromegalovirus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/biossíntese , Receptores do Fator de Necrose Tumoral/biossíntese , Retina/patologia , Retina/virologia , Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Fator de Necrose Tumoral alfa/biossíntese
9.
Antimicrob Agents Chemother ; 56(4): 1942-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22271861

RESUMO

The emergence of drug resistance threatens to limit the use of current anti-HIV-1 drugs and highlights the need to expand the number of treatment options available for HIV-1-infected individuals. Our previous studies demonstrated that two clinically approved drugs, decitabine and gemcitabine, potently inhibited HIV-1 replication in cell culture through a mechanism that is distinct from the mechanisms for the drugs currently used to treat HIV-1 infection. We further demonstrated that gemcitabine inhibited replication of a related retrovirus, murine leukemia virus (MuLV), in vivo using the MuLV-based LP-BM5/murine AIDS (MAIDS) mouse model at doses that were not toxic. Since decitabine and gemcitabine inhibited MuLV and HIV-1 replication with similar potency in cell culture, the current study examined the efficacy and toxicity of the drug combination using the MAIDS model. The data demonstrate that the drug combination inhibited disease progression, as detected by histopathology, viral loads, and spleen weights, at doses lower than those that would be required if the drugs were used individually. The combination of decitabine and gemcitabine exerted antiviral activity at doses that were not toxic. These findings indicate that the combination of decitabine and gemcitabine shows potent antiretroviral activity at nontoxic doses and should be further investigated for clinical relevance.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Azacitidina/análogos & derivados , Desoxicitidina/análogos & derivados , HIV-1/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Animais , Fármacos Anti-HIV/efeitos adversos , Azacitidina/uso terapêutico , Peso Corporal/efeitos dos fármacos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/patologia , Decitabina , Desoxicitidina/efeitos adversos , Desoxicitidina/uso terapêutico , Combinação de Medicamentos , Sinergismo Farmacológico , Feminino , Citometria de Fluxo , Humanos , Fígado/patologia , Linfonodos/patologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/patologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Provírus/efeitos dos fármacos , Baço/patologia , Baço/virologia , Linfócitos T/efeitos dos fármacos , Transfecção , Gencitabina
10.
Curr Eye Res ; 29(2-3): 173-80, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15512964

RESUMO

PURPOSE: To correlate tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) synthesis with histopathologic disease and virus replication within murine cytomegalovirus (MCMV)-infected eyes during progression of murine acquired immunodeficiency syndrome (MAIDS). MATERIALS AND METHODS: Groups of normal mice and mice with MAIDS of 2-weeks (MAIDS-2), 4-weeks (MAIDS-4), and 12-weeks (MAIDS-12) duration were infected uniocularly with MCMV by subretinal MCMV injection. MCMV-inoculated eyes from all mice were subjected to histopathologic analysis, quantitative plaque assay, or cytometric bead array analysis for quantification of TNF-alpha and IFN-gamma. RESULTS: Whereas MCMV-inoculated eyes of normal, MAIDS-2, and MAIDS-4 mice were resistant to MCMV retinitis, all MCMV-inoculated eyes of MAIDS-12 mice developed retinitis. Surprisingly, MCMV-inoculated eyes of MAIDS-4 mice without retinitis harbored high amounts of infectious virus at a level equivalent to that of MCMV-inoculated eyes of MAIDS-12 mice that developed retinitis. Intraocular TNF-alpha levels were consistently approximately 50% greater in MCMV-inoculated eyes of MAIDS-12 mice when compared with TNF-alpha levels of normal, MAIDS-2, and MAIDS-4 mice. In contrast, intraocular INF-gamma levels within MCMV-inoculated eyes progressively declined as animals became susceptible to retinitis. CONCLUSIONS: An inverse relationship exists between TNF-alpha and INF-gamma production within MCMV-inoculated eyes during MAIDS evolution that is characterized by an increase in intraocular TNF-alpha levels and a concomitant decrease in intraocular INF-gamma levels. Susceptibility of MCMV-inoculated eyes to virus replication and development of necrotizing retinitis are independent events with susceptibility to MCMV replication preceding susceptibility to MCMV retinitis by several weeks. Time of Th1/Th2 shift in cytokine profile appears to be a crucial event in the pathogenesis of MAIDS-related MCMV retinitis.


Assuntos
Retinite por Citomegalovirus/etiologia , Olho/metabolismo , Interferon gama/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/complicações , Síndrome de Imunodeficiência Adquirida Murina/fisiopatologia , Muromegalovirus , Fator de Necrose Tumoral alfa/metabolismo , Animais , Progressão da Doença , Suscetibilidade a Doenças , Olho/virologia , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/patologia , Necrose , Fator de Necrose Tumoral alfa/biossíntese , Replicação Viral
11.
Biochem J ; 384(Pt 3): 469-76, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15344910

RESUMO

MAIDS (murine AIDS) is caused by infection with the murine leukaemia retrovirus RadLV-Rs and is characterized by a severe immunodeficiency and T-cell anergy combined with a lymphoproliferative disease affecting both B- and T-cells. Hyperactivation of the cAMP-protein kinase A pathway is involved in the T-cell dysfunction of MAIDS and HIV by inhibiting T-cell activation through the T-cell receptor. In the present study, we show that MAIDS involves a strong and selective up-regulation of cyclo-oxygenase type 2 in the CD11b+ subpopulation of T- and B-cells of the lymph nodes, leading to increased levels of PGE2 (prostaglandin E2). PGE2 activates the cAMP pathway through G-protein-coupled receptors. Treatment with cyclo-oxygenase type 2 inhibitors reduces the level of PGE2 and thereby reverses the T-cell anergy, restores the T-cell immune function and ameliorates the lymphoproliferative disease.


Assuntos
Dinoprostona/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Vírus da Leucemia Induzida por Radiação/fisiologia , Linfócitos T/patologia , Linfócitos T/virologia , Animais , Linfócitos B/metabolismo , Linfócitos B/virologia , Antígeno CD11b/análise , Antígeno CD11b/metabolismo , Anergia Clonal , AMP Cíclico/metabolismo , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/antagonistas & inibidores , Dinoprostona/biossíntese , Indução Enzimática , HIV/imunologia , HIV/patogenicidade , Linfonodos/citologia , Linfonodos/enzimologia , Linfonodos/imunologia , Linfonodos/virologia , Ativação Linfocitária , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Vírus da Leucemia Induzida por Radiação/patogenicidade , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo
12.
Antiviral Res ; 62(3): 111-20, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15130534

RESUMO

Inhibition of ribonucleotide reductase (RR) has gained attention as a potential strategy for HIV-1 therapy through the success of hydroxyurea (HU) to potentiate the activity of the nucleoside reverse transcriptase inhibitor (NRTI) didanosine (ddI) in clinical trials. However, the use of HU has been limited by its development of hematopoietic toxicity. In this study, the novel RR inhibitors didox (DX; 3,4-dihydroxybenzohydroxamic acid), and trimidox (TX; 3,4,5-trihydroxybenzamidoxime) were evaluated along with HU for anti-retroviral efficacy in LPBM5-induced retro-viral disease (MAIDS) both as monotherapeutic regimens and in combination with the guanine containing NRTI abacavir (ABC). Anti-retroviral drug efficacy was determined by measuring inhibition of splenomegaly, hypergammaglobulinemia, and splenic levels of proviral DNA. In this study, all RRIs tested showed the ability to improve the efficacy of ABC in the MAIDS model by reducing splenomegaly, hypergammaglobulinemia, and splenic proviral DNA levels.


Assuntos
Antivirais/uso terapêutico , Células da Medula Óssea/efeitos dos fármacos , Hidroxiureia/uso terapêutico , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Ribonucleotídeo Redutases/antagonistas & inibidores , Animais , Antivirais/farmacologia , Benzamidinas/uso terapêutico , Didesoxinucleosídeos/uso terapêutico , Modelos Animais de Doenças , Quimioterapia Combinada , Células-Tronco Hematopoéticas/efeitos dos fármacos , Ácidos Hidroxâmicos/uso terapêutico , Hidroxiureia/efeitos adversos , Hidroxiureia/química , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/sangue , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Baço/patologia , Esplenomegalia
13.
J Immunol ; 172(8): 4917-25, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15067071

RESUMO

We describe successful immunotherapy of murine AIDS (MAIDS) in C57BL/6J mice based on the elimination of replicating CD4(+) regulator T cells. We demonstrate that a single injection of the antimitotic drug vinblastine (Vb) given 14 days postinfection (p.i.) with LP-BM5 can prevent MAIDS progression. Treatment with anti-CD4 mAb at 14 days p.i. is similarly able to prevent MAIDS. Treatment at other time points with Vb or anti-CD4 mAb is ineffective. The effect is based on ablation of a replicating dominantly suppressive CD4(+) T cell population, as indicated by adoptive transfer and in vivo depletion experiments using mAbs against CD4 as well as combinations of mAbs against the known regulatory cell surface markers CD25, GITR, and CTLA-4. Cell surface marker analysis shows a population of CD4(+)CD25(+) cells arising shortly before day 14 p.i. Cytokine analyses show a peak in IL-10 production from day 12 to day 16 p.i. MAIDS-infected mice also have CD4(+) T cells with significantly higher expression levels of CD38 and particularly CD69, which have been demonstrated to be regulator T cell markers in the Friend retroviral model. The immunotherapy appears to prevent disease progression, although no protection against reinfection with LP-BM5 is generated. These data define a new therapy for murine retroviral infection, which has potential for use in other diseases where T regulator cell-mediated immunosuppression plays a role in the disease process.


Assuntos
Linfócitos T CD4-Positivos/patologia , Depleção Linfocítica/métodos , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/prevenção & controle , Animais , Anticorpos Bloqueadores/administração & dosagem , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos T/biossíntese , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/imunologia , Progressão da Doença , Esquema de Medicação , Feminino , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/uso terapêutico , Esquemas de Imunização , Imunização Secundária , Injeções Intraperitoneais , Interleucina-10/antagonistas & inibidores , Interleucina-10/biossíntese , Interleucina-4/antagonistas & inibidores , Interleucina-4/biossíntese , Lectinas Tipo C , Vírus da Leucemia Murina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida Murina/patologia , Receptores de Interleucina-2/biossíntese , Baço/imunologia , Baço/patologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Vimblastina/administração & dosagem , Vimblastina/uso terapêutico , Carga Viral
14.
Comp Immunol Microbiol Infect Dis ; 27(3): 181-9, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15001313

RESUMO

The effect of bovine lactoferrin (bLF) was examined on an AIDS-like disease (ALD) in mice. Induction of disease was achieved by inoculation with infected cell-free plasma from diseased mice to uninfected ones. The effect of treatment with bLF was investigated when administered simultaneously with the virus, 20 days prior to infection, or 20 days after infection. Animals underwent clinical surveillance and enumeration of white blood cells (WBC) and lymphocytes, as well as fluorescent staining of CD4 and CD8 bearing cells. Simultaneous administration of bLF and virus did not affect the pattern of ALD progress along the course of the experiment. Pretreatment with bLF prior to virus inoculation abolished on day 21 the detrimental effect of viral infection that lasted for two months. An opposite outcome was observed when bLF was administered 20 days after the virus. It seems that bLF had played a preventive role for a restricted period of time. However, an adverse response was elicited when bLF was administered 20 days after viral infection.


Assuntos
Lactoferrina/farmacologia , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos , Feminino , Citometria de Fluxo , Hepatomegalia , Imunofenotipagem , Lactoferrina/imunologia , Contagem de Leucócitos , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Estatísticas não Paramétricas
15.
Curr Eye Res ; 26(3-4): 211-7, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12815549

RESUMO

PURPOSE: To determine if murine cytomegalovirus (MCMV)-infected eyes of mice with murine acquired immunodeficiency syndrome (MAIDS) that are destined to develop MCMV retinitis display elevated intraocular levels of interleukin-4 (IL-4) mRNA when compared with uninfected eyes of mice with MAIDS and unmanipulated, uninfected, eyes of normal healthy mice. METHODS: Groups of C57BL/6 mice with MAIDS and normal C57BL/6 mice were infected uniocularly with MCMV by subretinal MCMV injection. IL-4 levels in individual spleens collected five days later from groups of MAIDS mice and normal mice were assessed by quantitative ELISA. MCMV-infected eyes and uninfected contralateral eyes from another group of mice with MAIDS were also collected at five days postinfection and individually subjected to competitive RT-PCR assay and real-time RT-PCR assay for detection and quantification of IL-4 mRNA. Unmanipulated eyes from normal C57BL/6 mice served as controls. RESULTS: IL-4 mRNA was detected at a level of 9.7 +/- 3.4 pg mRNA per 1000 ng total RNA in 100% of MCMV-infected eyes of mice with MAIDS by competitive RT-PCR assay, but could not be detected in any of the uninfected eyes of MAIDS mice. In comparison, the more sensitive technique of real-time RT-PCR assay detected copies of IL-4 cDNA in both MCMV-infected eyes and uninfected eyes of MAIDS mice. MCMV-infected eyes showed a 16-fold increase in the number of IL-4 cDNA copies when compared with uninfected eyes. Neither technique detected IL-4 mRNA in unmanipulated eyes of normal mice. As expected, spleen cells from mice with MAIDS expressed significantly greater levels of IL-4 when compared with spleen cells from normal mice. CONCLUSIONS: MCMV-infected mice with MAIDS exhibited an expected preferential activation of Th2 cells as determined by increased levels of IL-4 in spleen cells when compared with spleen cells of normal mice. MCMV-infected eyes destined to develop retinitis during MAIDS also showed increased levels of detectable IL-4 mRNA when compared with uninfected eyes of mice with MAIDS. It is therefore possible that IL-4 synthesis by Th2 CD4+ T cells during retrovirus-induced immunosuppression serves to inhibit the perforin cytotoxic pathway that subsequently allows susceptibility to MCMV retinitis during MAIDS.


Assuntos
Retinite por Citomegalovirus/complicações , Olho/metabolismo , Infecções por Herpesviridae/complicações , Interleucina-4/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/complicações , Muromegalovirus , Animais , Sistemas Computacionais , Retinite por Citomegalovirus/metabolismo , Retinite por Citomegalovirus/patologia , Suscetibilidade a Doenças , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/patologia , Interleucina-4/genética , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/metabolismo , Baço/patologia
16.
Alcohol Alcohol ; 38(2): 103-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12634255

RESUMO

AIMS: The cardiovascular complications of acquired immunodeficiency syndrome (AIDS) are serious, including the occurrence of pathological heart conditions such as cardiomyopathy. Chronic alcohol consumption accentuates the severity of AIDS and may contribute to the development of cardiomyopathy. The aim of this work was to use a proteomics approach to investigate global alterations in protein expression in a mouse model of AIDS in the presence or absence of chronic alcohol consumption. METHODS: Cardiac proteins were separated by two-dimensional polyacrylamide gel electrophoresis and quantitative computer analysis was used to evaluate the resulting two-dimensional protein profiles. Proteins that were differentially expressed in the hearts of mice from the different experimental groups were identified by peptide mass finger-printing by matrix-assisted laser desorption/ionization mass spectrometry. RESULTS: A number of specific proteins were observed to be differentially expressed in the mouse heart due to the effect of ethanol feeding alone. Differentially expressed proteins were also observed that were due to viral infection alone. Ethanol feeding and viral infection appeared to have similar effects on the expression of a number of proteins. A total of 24 proteins were altered by infection alone. Of these 24 proteins, eight were affected by alcohol, with six alterations being ameliorated and two being exacerbated by alcohol. Two of these proteins have been identified as the 27 kDa heat-shock protein and mitochondrial long-chain acyl-CoA thioesterase 1. CONCLUSIONS: These results suggest that chronic alcohol consumption may exacerbate the effects of viral infection on the heart by lowering the stress response leading to de-protection and further cytotoxic effects.


Assuntos
Alcoolismo/metabolismo , Alcoolismo/patologia , Cardiopatias/metabolismo , Cardiopatias/virologia , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Proteínas/metabolismo , Proteômica/métodos , Administração Oral , Animais , Eletroforese em Gel Bidimensional , Etanol/administração & dosagem , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/patologia
17.
J Cardiovasc Pharmacol ; 41(3): 489-97, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12605029

RESUMO

Infection of people with human immunodeficiency virus (HIV) as well as LP-BM5 infection in mice results in progressive deterioration of the immune system in the majority of untreated hosts. Peptide immunotherapy has been shown to be effective in the stimulation or immunoregulation of T-helper 1 (T(H)1) and T-helper 2 (T(H) 2) response subsets. In murine acquired immunodeficiency syndrome (AIDS), T(H)1 deficiency enables the host to be susceptible to coxsackievirus infection, inducing cardiopathology in a short period. T-cell receptor (TCR) Vbeta8.1 peptide, a 16-mer peptide containing the entire CDR1 segment and part of the FR2 region of human Vbeta8, showed both an immunoregulating and immunostimulating effect in murine AIDS. TCR Vbeta8.1 peptide acts on T cells promoting interleukin-2 production and therefore enhancing a cell-mediated immune response. It retarded development of cardiopathology due to coxsackievirus infection. Retrovirus-infected mice treated with the peptide showed a longer life span than the nontreated, retrovirus-infected animals.


Assuntos
Infecções por Coxsackievirus/terapia , Enterovirus Humano B/imunologia , Síndrome de Imunodeficiência Adquirida Murina/terapia , Miocárdio/patologia , Receptores de Antígenos de Linfócitos T alfa-beta/uso terapêutico , Sequência de Aminoácidos , Animais , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Miocárdio/imunologia
18.
Alcohol Alcohol ; 38(1): 18-24, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12554602

RESUMO

AIMS: The severity of cardiovascular complications in acquired immune deficiency syndrome (AIDS) patients may be associated with acute ischaemia-reperfusion injury. Epidemiological studies suggest that moderate ethanol consumption has myocardial protective effects. However, it is unknown if chronic ethanol consumption benefits acute myocardial ischaemia-reperfusion injury in AIDS. The aim of this study was to determine if chronic ethanol consumption modulates myocardial ischaemia-reperfusion injury in murine AIDS. METHODS: Four groups were studied: control, murine AIDS, ethanol, and ethanol plus murine AIDS. All mice were subjected to 30 min of left anterior descending branch (LAD) occlusion and 120 min of reperfusion. RESULTS: We found that the survival from an acute myocardial infarction was reduced in advanced-stage murine AIDS mice. Although early-stage murine AIDS hearts did survive in acute myocardial infarction, the infarct size was significantly larger. Chronic ethanol consumption significantly decreased infarct size compared to the control group. Chronic ethanol consumption also improved the survival of murine AIDS mice from an acute myocardial infarction. However, chronic ethanol consumption did not significantly reduce infarct size in murine AIDS. CONCLUSIONS: Our results indicate that multiple deleterious effects may enhance acute ischaemia-reperfusion injury in murine AIDS. The beneficial effects of chronic ethanol consumption in myocardial ischaemia-reperfusion injury may be due to modulation of neutrophil adhesion molecule expression and cytokine secretion.


Assuntos
Consumo de Bebidas Alcoólicas/imunologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Infarto do Miocárdio/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , Ativação de Neutrófilo/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/efeitos adversos , Consumo de Bebidas Alcoólicas/patologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Cardiomiopatias/imunologia , Cardiomiopatias/patologia , Feminino , Frequência Cardíaca/efeitos dos fármacos , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Computação Matemática , Camundongos , Camundongos Endogâmicos BALB C , Síndrome de Imunodeficiência Adquirida Murina/patologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/imunologia , Miocárdio/patologia , Ativação de Neutrófilo/imunologia
19.
J Immunol ; 169(10): 5607-14, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12421939

RESUMO

Infection of genetically susceptible mice with the LP-BM5 mixture of murine leukemia viruses including an etiologic defective virus (BM5def) causes an immunodeficiency syndrome called murine AIDS (MAIDS). The disease is characterized by interactions between B cells and CD4(+) T cells resulting in polyclonal activation of both cell types. It is known that BM5def is expressed at highest levels in B cells and that B cells serve as viral APC. The CD19-CD21 complex and CD22 on the surface of B cells play critical roles as regulators of B cell responses to a variety of stimuli, influencing cell activation, differentiation, and survival. CD19 integrates positive signals induced by B cell receptor ligation by interacting with the protooncogene Vav, which leads to subsequent tyrosine phosphorylation of this molecule. In contrast, CD22 negatively regulates Vav phosphorylation. To analyze the role of CD19, CD21, Vav, and CD22 in MAIDS, we infected mice deficient in CD19, CD21 (CR2), Vav-1, or CD22 with LP-BM5 murine leukemia viruses. Infected CR2(-/-) mice developed MAIDS with a time course and severity indistinguishable from that of wild-type mice. In contrast, CD19 as well as Vav-1 deficiency restricted viral replication and suppressed the development of typical signs of MAIDS including splenomegaly, lymphadenopathy, and hypergammaglobulinemia. Finally, CD22 deficiency was found to accelerate MAIDS development. These results provide novel insights into the B cell signaling pathways required for normal induction and progression of MAIDS.


Assuntos
Antígenos CD19/fisiologia , Moléculas de Adesão Celular , Proteínas de Ciclo Celular , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Transdução de Sinais/imunologia , Animais , Antígenos CD/genética , Antígenos CD19/genética , Antígenos de Diferenciação de Linfócitos B/genética , Antivirais/fisiologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/virologia , Linhagem Celular Transformada , Progressão da Doença , Soros Imunes/biossíntese , Switching de Imunoglobulina/genética , Imunoglobulina E/biossíntese , Imunofenotipagem , Lectinas/deficiência , Lectinas/genética , Vírus da Leucemia Murina/imunologia , Vírus da Leucemia Murina/metabolismo , Ativação Linfocitária/genética , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndrome de Imunodeficiência Adquirida Murina/genética , Síndrome de Imunodeficiência Adquirida Murina/patologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-vav , Receptores de Complemento 3d/deficiência , Receptores de Complemento 3d/genética , Índice de Gravidade de Doença , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico , Transdução de Sinais/genética , Baço/citologia , Baço/imunologia , Baço/metabolismo , Baço/patologia , Integração Viral/genética , Integração Viral/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
20.
Antiviral Res ; 56(2): 167-81, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12367722

RESUMO

Recently, the use of the ribonucleotide reductase (RR) inhibitor hydroxyurea (HU) in combination with nucleoside analogs has gained attention as a potential strategy for anti-HIV-1 therapy. However, appeal for the long-term use of HU in HIV-1 infection may be limited by its propensity to induce hematopoietic toxicity. We report a comparison of the efficacy and bone marrow toxicity of HU (400 and 200 mg/kg/day) with the novel RR inhibitors and free radical-scavenging compounds didox (DX; 3,4-dihydroxybenzohydroxamic acid; 350 mg/kg/day) and trimidox (TX; 3,4,5-trihydroxybenzamidoxime; 175 mg/kg/day) in the murine AIDS (LPBM5 MuLV) model of retrovirus infection. Infected mice received daily drug treatment for 8 weeks. Efficacy was determined by measuring drug effects on retroviral-induced disease progression (i.e. development of splenomegaly and hypergammaglobulinemia) and by evaluating splenic levels of proviral DNA. Bone marrow toxicity was evaluated by measuring peripheral blood indices (WBC, hematocrit and reticulocyte counts), femoral cellularity and by determining the numbers of hematopoietic progenitor cells (CFU-GM, BFU-E) per femur and spleen. Compared to infected controls receiving no drug treatment, disease progression was significantly suppressed by TX, DX and HU. However, HU was associated with mortality and induced significant hematopoietic toxicity in a time- and dose-dependent manner. Conversely, TX and DX effectively inhibited retrovirus-induced disease but did not induce hematopoietic toxicity. These results suggest that due to their reduced hematopoietic toxicity and ability to inhibit disease progression in murine AIDS, TX and DX may offer effective alternatives to HU therapy in HIV-1 infection.


Assuntos
Benzamidinas/efeitos adversos , Células da Medula Óssea/efeitos dos fármacos , Sequestradores de Radicais Livres/efeitos adversos , Ácidos Hidroxâmicos/efeitos adversos , Hidroxiureia/efeitos adversos , Vírus da Leucemia Murina/efeitos dos fármacos , Leucemia Experimental/patologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Infecções por Retroviridae/patologia , Ribonucleotídeo Redutases/antagonistas & inibidores , Infecções Tumorais por Vírus/patologia , Animais , Benzamidinas/química , Benzamidinas/uso terapêutico , DNA Viral , Feminino , Fêmur/citologia , Fêmur/efeitos dos fármacos , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/uso terapêutico , Células-Tronco Hematopoéticas/efeitos dos fármacos , Ácidos Hidroxâmicos/química , Ácidos Hidroxâmicos/uso terapêutico , Hidroxiureia/química , Hidroxiureia/uso terapêutico , Hipergamaglobulinemia/tratamento farmacológico , Vírus da Leucemia Murina/genética , Leucemia Experimental/sangue , Leucemia Experimental/tratamento farmacológico , Leucemia Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/sangue , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Provírus/genética , Infecções por Retroviridae/sangue , Infecções por Retroviridae/tratamento farmacológico , Infecções por Retroviridae/imunologia , Baço/patologia , Esplenomegalia , Infecções Tumorais por Vírus/sangue , Infecções Tumorais por Vírus/tratamento farmacológico , Infecções Tumorais por Vírus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...