Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Med Virol ; 92(3): 394-398, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31670405

RESUMO

The mechanisms that contribute to retinal tissue destruction during the onset and progression of AIDS-related human cytomegalovirus (HCMV) retinitis remain unclear. Evidence for the stimulation of multiple cell death pathways including apoptosis, necroptosis, and pyroptosis during the pathogenesis of experimental murine cytomegalovirus (MCMV) retinitis in mice with retrovirus-induced immunosuppression (MAIDS) has been reported. Parthanatos is a caspase-independent cell death pathway mediated by rapid overactivation of poly (ADP-ribose) polymerase-1 (PARP-1) and distinct from other cell death pathways. Using the MAIDS model of MCMV retinitis, studies were performed to test the hypothesis that intraocular MCMV infection of mice with MAIDS stimulates parthanatos-associated messenger RNAs (mRNAs) and proteins within the eye during the development of retinal necrosis that takes place by 10 days after MCMV infection. MCMV-infected eyes of MAIDS mice exhibited significant stimulation of PARP-1 mRNA and proteins at 3 days after infection but declined thereafter at 6 and 10 days after infection. Additional studies showed the intraocular stimulation of mRNAs or proteins before MCMV retinitis development for two additional participants in parthanatos, polymer of ADP-ribose and poly (ADP-ribose) glycohydrolase. These results provide new evidence for a role for parthanatos during MAIDS-related MCMV retinitis that may also extend to AIDS-related HCMV retinitis.


Assuntos
Retinite por Citomegalovirus/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/virologia , Parthanatos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Animais , Morte Celular , Retinite por Citomegalovirus/complicações , Modelos Animais de Doenças , Progressão da Doença , Feminino , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/complicações , Muromegalovirus , Poli(ADP-Ribose) Polimerase-1/genética , Poli Adenosina Difosfato Ribose/genética , Poli Adenosina Difosfato Ribose/metabolismo , RNA Mensageiro/metabolismo , Retina/patologia , Retina/virologia , Retroviridae/imunologia
2.
Virology ; 499: 9-22, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27632561

RESUMO

Monocytic myeloid-derived suppressor cells (M-MDSCs) were increased during LP-BM5 retroviral infection, and were capable of suppressing not only T-cell, but also B-cell responses. In addition to previously demonstrating iNOS- and VISTA-dependent M-MDSC mechanisms, in this paper, we detail how M-MDSCs utilized soluble mediators, including the reactive oxygen and nitrogen species superoxide, peroxynitrite, and nitric oxide, and TGF-ß, to suppress B cells in a predominantly contact-independent manner. Suppression was independent of cysteine-depletion and hydrogen peroxide production. When two major mechanisms of suppression (iNOS and VISTA) were eliminated in double knockout mice, M-MDSCs from LP-BM5-infected mice were able to compensate using other, soluble mechanisms in order to maintain suppression of B cells. The IL-10 producing regulatory B-cell compartment was among the targets of M-MDSC-mediated suppression.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Espécies Reativas de Nitrogênio , Espécies Reativas de Oxigênio , Fator de Crescimento Transformador beta/sangue , Animais , Linfócitos B Reguladores/imunologia , Linfócitos B Reguladores/metabolismo , Comunicação Celular , Tolerância Imunológica , Masculino , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ácido Peroxinitroso/metabolismo
3.
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
4.
J Immunol ; 189(7): 3724-33, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22956581

RESUMO

Distal symmetrical polyneuropathy is the most common form of HIV infection-associated peripheral neuropathy and is often associated with pain. C57BL/6 (B6) mice infected with LP-BM5, a murine retroviral isolate, develop a severe immunodeficiency syndrome similar to that in humans infected with HIV-1, hence the term murine AIDS. We investigated the induction of peripheral neuropathy after LP-BM5 infection in B6 mice. Infected B6 mice, like HIV-infected humans, exhibited behavioral (increased sensitivity to mechanical and heat stimuli) and pathological (transient loss of intraepidermal nerve fibers) signs of peripheral neuropathy. The levels of viral gag RNA were significantly increased in all tissues tested, including spleen, paw skin, lumbar dorsal root ganglia, and lumbar spinal cord, postinfection (p.i.). Correlated with the development of peripheral neuropathy, the tissue levels of several cytokines, including IFN-γ, IL-1ß, IL-6, and IL-12, were significantly elevated p.i. These increases had cytokine-specific and tissue-specific profiles and kinetics. Further, treatment with the antiretroviral agent zidovudine either significantly reduced or completely reversed the aforementioned behavioral, pathologic, and cytokine changes p.i. These data suggest that LP-BM5 infection is a potential mouse model of HIV-associated distal symmetrical polyneuropathy that can be used for investigating the roles of various cytokines in infection-induced neuropathic pain. Further investigation of this model could give a better understanding of, and lead to more effective treatments for, HIV infection-associated painful peripheral neuropathy.


Assuntos
Citocinas/biossíntese , Vírus da Leucemia Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Doenças do Sistema Nervoso Periférico/imunologia , Doenças do Sistema Nervoso Periférico/virologia , Animais , Citocinas/genética , Modelos Animais de Doenças , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Hipersensibilidade/virologia , Vírus da Leucemia Murina/isolamento & purificação , Leucemia Experimental/imunologia , Leucemia Experimental/metabolismo , Leucemia Experimental/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças do Sistema Nervoso Periférico/metabolismo , RNA Mensageiro/biossíntese , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/metabolismo
5.
Retrovirology ; 9: 50, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22691411

RESUMO

BACKGROUND: APOBEC3 proteins are host factors that restrict infection by retroviruses like HIV, MMTV, and MLV and are variably expressed in hematopoietic and non-hematopoietic cells, such as macrophages, lymphocytes, dendritic, and epithelia cells. Previously, we showed that APOBEC3 expressed in mammary epithelia cells function to limit milk-borne transmission of the beta-retrovirus, mouse mammary tumor virus. In this present study, we used APOBEC3 knockout mice and their wild type counterpart to query the role of APOBEC3 in sexual transmission of LP-BM5 MLV - the etiological agent of murine AIDs (mAIDs). RESULTS: We show that mouse APOBEC3 is expressed in murine genital tract tissues and gametes and that genital tract tissue of APOBEC3-deficient mice are more susceptible to infection by LP-BM5 virus. APOBEC3 expressed in genital tract tissues most likely plays a role in decreasing virus transmission via the sexual route, since mice deficient in APOBEC3 gene have higher genitalia and seminal plasma virus load and sexually transmit the virus more efficiently to their partners compared to APOBEC3+ mice. Moreover, we show that female mice sexually infected with LP-BM5 virus transmit the virus to their off-spring in APOBEC3-dependent manner. CONCLUSION: Our data indicate that genital tissue intrinsic APOBEC3 restricts genital tract infection and limits sexual transmission of LP-BM5 virus.


Assuntos
Citidina Desaminase/metabolismo , Genitália/virologia , Vírus da Leucemia Murina/patogenicidade , Síndrome de Imunodeficiência Adquirida Murina/transmissão , Doenças Virais Sexualmente Transmissíveis/transmissão , Animais , Citidina Desaminase/genética , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/virologia , Feminino , Genitália/metabolismo , Células Germinativas/metabolismo , Células Germinativas/virologia , Vírus da Leucemia Murina/metabolismo , Masculino , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/virologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Doenças Virais Sexualmente Transmissíveis/metabolismo , Esplenomegalia/virologia , Carga Viral
6.
Biosci Biotechnol Biochem ; 75(7): 1234-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21737943

RESUMO

The purpose of this study was to determine the effect of isothiocyanates (ITCs) in delaying the progression of the murine immunodeficiency virus to murine AIDS, resulting in increased life span. Furthermore, we investigated the role of ITCs in modulating immune dysfunction caused by LP-BM5 retrovirus infection. Among the tested ITCs, oral administration of sulforaphane (SUL), benzyl isothiocyante (BITC), and phenethyl isothiocyanate (PEITC) showed the inhibition of premature death caused by LP-BM5 retrovirus infection, while indolo[3,2-b] carbazole (ICZ) and indole-3-carbinol (I3C) did not delay the progress of the LP-BM5 retrovirus to murine AIDS. Inhibition of premature death by BITC, PEITC, and SUL could be explained by restoration of the immune system and down regulation of free radicals. Dysfunction of T and B cell mitogenesis caused by retrovirus infection in primary cultured splenocytes has been partially recovered with administration of BITC, PEITC, and SUL. There was a shift from imbalanced cytokine production (increased Th2 and decreased Th1 cell cytokine production) into balanced Th1/Th2 cell secretion of cytokines under administration of these ITCs during the development of murine AIDS. Hepatic vitamin E level was significantly restored by administration of these ITCs, in accordance with reduced hepatic lipid peroxidation levels. This study suggests that certain types of ITCs have beneficial effects in preventing premature death during progression to murine AIDS by restoration of immune dysfunction and removal of excessive free radicals, implying that selective usage of ITCs would be helpful in retarding the progression from HIV infection to AIDS.


Assuntos
Citocinas/efeitos dos fármacos , Isotiocianatos/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Longevidade/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Citocinas/imunologia , Citocinas/metabolismo , Progressão da Doença , Feminino , Longevidade/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Sulfóxidos , Células Th1/efeitos dos fármacos , Células Th1/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Tiocianatos/farmacologia , Vitamina E/metabolismo
7.
J Immunol ; 186(9): 5119-30, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21430226

RESUMO

Type I protein kinase A (PKA) is targeted to the TCR-proximal signaling machinery by the A-kinase anchoring protein ezrin and negatively regulates T cell immune function through activation of the C-terminal Src kinase. RI anchoring disruptor (RIAD) is a high-affinity competitor peptide that specifically displaces type I PKA from A-kinase anchoring proteins. In this study, we disrupted type I PKA anchoring in peripheral T cells by expressing a soluble ezrin fragment with RIAD inserted in place of the endogenous A-kinase binding domain under the lck distal promoter in mice. Peripheral T cells from mice expressing the RIAD fusion protein (RIAD-transgenic mice) displayed augmented basal and TCR-activated signaling, enhanced T cell responsiveness assessed as IL-2 secretion, and reduced sensitivity to PGE(2)- and cAMP-mediated inhibition of T cell function. Hyperactivation of the cAMP-type I PKA pathway is involved in the T cell dysfunction of HIV infection, as well as murine AIDS, a disease model induced by infection of C57BL/6 mice with LP-BM5, a mixture of attenuated murine leukemia viruses. LP-BM5-infected RIAD-transgenic mice resist progression of murine AIDS and have improved viral control. This underscores the cAMP-type I PKA pathway in T cells as a putative target for therapeutic intervention in immunodeficiency diseases.


Assuntos
Proteínas de Ancoragem à Quinase A/imunologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Western Blotting , Separação Celular , Proteínas do Citoesqueleto/imunologia , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Imunofluorescência , Humanos , Immunoblotting , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Transfecção
8.
J Pathol ; 221(1): 106-16, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20217872

RESUMO

Anaplastic plasmacytomas (APCTs) from NFS.V(+) congenic mice and pristane-induced plasmacytic PCTs from BALB/c mice were previously shown to be histologically and molecularly distinct subsets of plasma cell neoplasms (PCNs). Here we extended these comparisons, contrasting primary APCTs and PCTs by gene expression profiling in relation to the expression profiles of normal naïve, germinal centre, and memory B cells and plasma cells. We also sequenced immunoglobulin genes from APCT and APCT-derived cell lines and defined surface phenotypes and chromosomal features of the cell lines by flow cytometry and by spectral karyotyping and fluorescence in situ hybridization. The results indicate that APCTs share many features with normal memory cells and the plasma cell-related neoplasms (PLs) of FASL-deficient mice, suggesting that APCTs and PLs are related and that both derive from memory B cells. Published in 2010 by John Wiley & Sons, Ltd.


Assuntos
Subpopulações de Linfócitos B/imunologia , Memória Imunológica , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Neoplasias de Plasmócitos/imunologia , Plasmocitoma/imunologia , Animais , Sequência de Bases , Sobrevivência Celular/fisiologia , Aberrações Cromossômicas , Perfilação da Expressão Gênica/métodos , Região Variável de Imunoglobulina/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Síndrome de Imunodeficiência Adquirida Murina/complicações , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de Plasmócitos/complicações , Neoplasias de Plasmócitos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Plasmocitoma/complicações , Plasmocitoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
9.
Immunogenetics ; 60(9): 485-94, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18612634

RESUMO

Murine acquired immunodeficiency syndrome (MAIDS) is caused by exposure to murine leukemia virus and serves as a model to study human AIDS. In MAIDS-susceptible C57BL/6 mice, virus exposure leads to progressive immune deficiency, while resistant strains such as BALB/c recover from infection and develop protective immunity. The goal of this study was to identify early gene expression patterns that may be important in establishing this strain-specific differential response. Total RNA was isolated from spleens and pooled lymph nodes of both mouse strains at 3 and 7 days post virus infection. The complementary DNA generated from this RNA was hybridized to mouse oligonucleotide DNA microarrays using a strategy that controlled for inherent variability and highlighted only virus-induced changes. Fluorescent intensities were normalized and analyzed for statistically significant differential expression between strains across both time points and lymphoid organs. The majority of the resistance-associated genes was identified at day 3 post-infection and demonstrated the highest fold differences between strains, while more susceptibility-associated sequences were seen at 7 days post-infection. Among the most highly differentially expressed sequences seen at the earlier time point were genes related to protein metabolism, especially serine proteases. Differential patterns of chemokine-related genes were observed at the later time point. The overall pattern of expression suggests strain-specific differences in proteases and chemokines within secondary lymphoid organs shortly after infection influence the likelihood of disease progression.


Assuntos
Vírus da Leucemia Murina/fisiologia , Sistema Linfático/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Animais , Perfilação da Expressão Gênica , Vírus da Leucemia Murina/imunologia , Linfonodos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade da Espécie , Baço/metabolismo , Fatores de Tempo , Replicação Viral
10.
Cardiovasc Toxicol ; 4(4): 317-25, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15531775

RESUMO

Dilated cardiomyopathy (DCM) is a clinically relevant disease that can occur independently or secondary to other diseases such as HIV infection and AIDS. To study this latter process, we used a model in which mice are infected with the LP-BM5 murine AIDS (MAIDS) retrovirus. Cardiac function of control and infected mice was determined through the in vivo analysis of left ventricular pressure-volume loops. Furthermore, the role of myocarditis was investigated through immunohistochemistry for T-cell, B-cell, and macrophage cardiac infiltrates and Northern blot analysis for tumor necrosis factor (TNF)-alpha and inducible nitric oxide synthase (iNOS). End-systolic and end-diastolic volumes were significantly increased and ventricular stiffness was significantly decreased in infected mice, consistent with DCM; however, no staining for inflammatory cellular infiltrates or TNF-alpha and iNOS was seen. These data support the conclusion that the LP-BM5 HIV model virus causes DCM in the absence of chronic cardiac inflammation. These findings support MAIDS retroviral infection as a new model of idiopathic DCM in which myo-carditis does not occur.


Assuntos
Cardiomiopatia Dilatada/virologia , Modelos Animais de Doenças , Síndrome de Imunodeficiência Adquirida Murina/complicações , Miocardite/virologia , Animais , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/fisiopatologia , DNA Viral/análise , Feminino , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/fisiopatologia , Miocardite/imunologia , Miocardite/metabolismo , Miocárdio/imunologia , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Retroviridae/genética , Retroviridae/isolamento & purificação , Volume Sistólico , Fator de Necrose Tumoral alfa/metabolismo , Disfunção Ventricular Esquerda
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.
Cell Mol Biol (Noisy-le-grand) ; 45(6): 855-63, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10541481

RESUMO

The effect of L. acidophilus supplementation to reduce fecal shedding of Cryptosporidium parvum oocysts was compared to L. reuteri using C57BL/6 female mice immunosuppressed by murine leukemia virus (strain LP-BM5) inoculation. After 12 weeks post LP-BM5 inoculation, 15 immunosuppressed mice each were randomly assinged to one of the following treatment groups: historical control (group A), LP-BM5 control (group B), C. parvum (group C), L. reuteri plus C. parvum (group D) or L. acidophilus plus C. parvum (group E). Mice were pre-fed the L. reuteri or L. acidophilus bacteria strains daily for 13 days, challenged with C. parvum oocysts and thereafter fed the specified Lactobacillus regimens daily during the experimental period. Animals supplemented with L. reuteri shed fewer (p<0.05) oocysts on day-7 post C. parvum challenge compared to controls. Mice supplemented with L. acidophilus also shed fewer (p<0.05) oocysts on days 7 and 14 post-challenge compared to controls. Overall, Lactobacillus supplementation reduced C. parvum shedding in the feces but failed to suppress the production of T-helper type 2 cytokines [interleukin-4 (IL-4), IL-8)] which are associated with immunosuppression. Additionally, Lactobacillus supplementation did not restore T-helper type 1 cytokines (interleukin-2 (IL-2) and gamma interferon (IFN-gamma), which are required for recovery from parasitic infections. Altered T-helper types 1 and 2 cytokine production as a consequence of immunodysfunction permitted the development of persistent cryptosporidiosis while mice with intact immune system were refractory to infection with C. parvum. Reduction in shedding of oocysts observed in the Lactobacillus supplemented mice during deminished IL-2 and IFN-gamma production may be mediated by factors released into the intestinal lumen by the Lactobacillus and possibly other host cellular mechanisms. These observations suggest that L. reuteri or L. acidophilus can reduce C. parvum parasite burdens in the intestinal epithelium during cryptosporidiosis and may serve potential benefits as probiotics for host resistance to intestinal parasitic infections. L. acidophilus was more efficacious in reducing fecal shedding than L. reuteri and therefore may also have implication in the therapy of cryptosporidiosis during immunosuppressive states including human AIDS.


Assuntos
Criptosporidiose/terapia , Cryptosporidium parvum/parasitologia , Lactobacillus , Camundongos Endogâmicos C57BL/parasitologia , Síndrome de Imunodeficiência Adquirida Murina/complicações , Probióticos/uso terapêutico , Infecções Oportunistas Relacionadas com a AIDS/terapia , Animais , Peso Corporal , Criptosporidiose/complicações , Cryptosporidium parvum/crescimento & desenvolvimento , Ingestão de Líquidos , Ingestão de Alimentos , Fezes/parasitologia , Feminino , Intestinos/parasitologia , Lactobacillus acidophilus , Vírus da Leucemia Murina , Camundongos , Camundongos Endogâmicos C57BL/virologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Tamanho do Órgão , Probióticos/farmacologia , Baço/anatomia & histologia , Eliminação de Partículas Virais
13.
Eur J Biochem ; 263(1): 202-11, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10429205

RESUMO

Murine acquired immunodeficiency syndrome (MAIDS) is a complex immunopathology caused by a defective murine leukemia virus (LP-BM5) that mainly targets B-lymphocytes. Lymphadenophathy, splenomegaly, hypergammaglobulinemia and progressive immunodeficiency are prominent features of MAIDS. Previously, we showed that the ubiquitin proteolytic system was upregulated in infected lymph nodes [Crinelli, R., Fraternale, A., Casabianca, A. & Magnani, M. (1997) Eur. J. Biochem. 247, 91-97]. In this report, we demonstrate that increased 26S proteasome activity is responsible for accelerated turnover of the IkappaBalpha inhibitor in lymph node extracts derived from animals with MAIDS. The molecular mechanisms mediating IkappaBalpha proteolysis involved constitutive phosphorylation of IkappaBalpha at Ser32 and Ser36 and subsequent ubiquitination, suggesting persistent activation of an NF-kappaB inducing pathway. Interestingly, enhanced IkappaBalpha degradation did not result in enhanced NF-kappaB DNA binding activity, but rather in a different subunit composition. The modulation of NF-kappaB/IkappaB system may affect multiple immunoregulatory pathways and may in part explain the mechanisms leading to the profound immune dysregulation involved in MAIDS pathogenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Endopeptidases/metabolismo , Proteínas I-kappa B , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Ubiquitinas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cisteína Endopeptidases/metabolismo , DNA/metabolismo , Primers do DNA/genética , Proteínas de Ligação a DNA/genética , Ativação Enzimática , Feminino , Quinase I-kappa B , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multienzimáticos/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/enzimologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma , Proteínas Serina-Treonina Quinases/metabolismo
14.
J Neurochem ; 71(5): 2079-87, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9798933

RESUMO

Mice infected with the LP-BM5 leukemia retrovirus mixture develop a progressive immunodeficiency with associated behavioral, histological, and neurochemical alterations consistent with glutamatergic hyperactivation. To gain insight into the contribution of excitatory amino acids to the neurodegeneration observed in these mice, their concentrations were measured in the CSF and striatal microdialysates. Glutamate concentrations were significantly elevated in CSF but not plasma as early as 4 weeks postinoculation. Steady-state glutamate levels in striatal microdialysates were increased threefold and could be reduced 40% by application of L-alpha-aminoadipate, an inhibitor of microglial glutamate transport. Stimulation of infected mice with KCl/L-trans-2,4-pyrrolidine dicarboxylate further increased glutamate levels 170-270% above those evoked in control mice. Tetrodotoxin suppressed the depolarization-evoked increase in glutamate by 88% in control mice, but it had only negligible effects in 40% of infected mice. Analysis of glutamate transport and catabolism suggests that abnormal astrocytic function does not contribute to the increase in basal extracellular glutamate levels. These findings are the first direct evidence that infection with an immunodeficiency-inducing retrovirus leads to a chronic elevation of extracellular free glutamate levels in the brain, which contributes to the neurodegenerative and cognitive deficits observed in these mice.


Assuntos
Encéfalo/metabolismo , Espaço Extracelular/metabolismo , Ácido Glutâmico/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/complicações , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Complexo Relacionado com a AIDS/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Sistema X-AG de Transporte de Aminoácidos , Aminoácidos/sangue , Aminoácidos/líquido cefalorraquidiano , Aminoácidos/metabolismo , Animais , Corpo Estriado/metabolismo , Glutamato-Amônia Ligase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Concentração Osmolar , Fatores de Tempo
15.
Cell Immunol ; 188(2): 151-7, 1998 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9756645

RESUMO

Murine acquired immunodeficiency syndrome (MAIDS) can be viewed as a lymphoproliferative disease which involves B cells as well as T cells from spleen and lymph nodes while thymus and Peyer's patches do not participate in the process. The 120-kDa protooncogene product c-Cbl was initially cloned from the murine Cas NS-1 B cell lymphoma. It is a main target of immunoreceptor (TCR and BCR)-mediated protein tyrosine kinase activity. Moreover, recent data suggest that c-Cbl might play a crucial role in the regulation of cell proliferation through regulation of GTP-binding proteins. Therefore, the involvement of c-Cbl was evaluated in the lymphoproliferative disease induced by the MAIDS virus. The expression of the c-Cbl protein was dramatically reduced in the lymph node of infected mice while it remained normal in the thymus. In contrast, the expression of actin, TCR-zeta chain, ZAP-70, and p59(fyn) remained similar in controls and infected mice. Identical results were obtained with sorted B cells and T cells. Surprisingly, a B cell lymphoma line derived from late stage MAIDS mice displayed a normal level of c-Cbl.


Assuntos
Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Proteínas Proto-Oncogênicas/análise , Ubiquitina-Proteína Ligases , Animais , Regulação para Baixo , Linfonodos/química , Ativação Linfocitária , Linfócitos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-cbl , RNA Mensageiro/análise , Timo/química
16.
Int Immunol ; 10(10): 1473-80, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9796914

RESUMO

Murine AIDS (MAIDS), caused by a defective murine leukemia virus, is a severe lymphoproliferative disease associated with profound immunodeficiency and increased susceptibility to opportunistic infections. Most subsets of lymphocytes, including CD4+ and CD8+ T cells, are refractory to mitogen stimulation. As a first step to examine proximal signal transduction in the infected mice, Western and Northern blot analyses were performed, and showed that p56lck is dramatically decreased at the protein as well as the mRNA level in the lymph nodes (LN). In contrast, p59(fyn) and its mRNA were slightly increased in the LN of the same mice. Similar results were obtained with purified T cells. Interestingly, the thymus of the infected animals did not show any abnormality regarding p56(lck) or p59(fyn). Tyrosine phosphorylation was constitutively increased in the infected mice and was barely amplified by anti-CD3 mAb stimulation. A similar pattern was observed when tyrosine phosphorylation was selectively examined at the level of ZAP-70. Our results suggest that a reciprocal regulation of p56(lck) and p59(fyn) protein tyrosine kinases, previously described in various models of anergy, could also be involved in the pathogenesis of MAIDS.


Assuntos
Proteína Tirosina Quinase p56(lck) Linfócito-Específica/biossíntese , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Animais , Anticorpos Monoclonais/farmacologia , Complexo CD3/imunologia , Linfa/citologia , Linfa/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/etiologia , Fosforilação , Proteínas Tirosina Quinases/química , Proteínas/química , Proteínas Proto-Oncogênicas c-fyn , RNA Mensageiro/biossíntese , Timo/citologia , Timo/metabolismo , Proteína-Tirosina Quinase ZAP-70
18.
Rom J Physiol ; 35(1-2): 13-24, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-11000861

RESUMO

Lithium gamma linolenic acid (Li-GLA), was evaluated for its possible role as an antiviral agent. Li-GLA 15 micrograms ml-1 was administered to both normal and LP-BM5 MuLV retroviral infected murine bone marrow cultures. After 2 weeks of treatment, numbers of progenitors being produced by infected/treated cultures were reduced to some 10% that of normal cultures. In the remaining 4 weeks, numbers of CFU-GM and BFU-E hematopoietic progenitors returned within normal range. The efficacy of Li-GLA in relieving retroviral hematopoietic bone marrow suppression correlates to a reduction in interleukin-4 (IL-4) secretion, normally elevated in association with LP-BMP5 infection. These data indicate that this reduction in bone marrow suppression of LP-BMP5 infected cells may be due to a killing of infected cells by the Li-GLA, rather than stimulating hematopoiesis as with other lithium compounds. To conclude this may indicate the possible dual effect of administration of LiGLA to virally infected individuals in reducing viral titre and to lower the toxicities associated with long term drug therapy.


Assuntos
Antivirais/farmacologia , Células-Tronco Hematopoéticas/patologia , Síndrome de Imunodeficiência Adquirida Murina/patologia , Ácido gama-Linolênico/farmacologia , Animais , Contagem de Células/efeitos dos fármacos , Células Cultivadas , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/patologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Interleucina-4/metabolismo , Vírus da Leucemia Murina/fisiologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/virologia , Fator de Crescimento Transformador beta/metabolismo , Replicação Viral
19.
Endocrinology ; 138(12): 5505-10, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9389537

RESUMO

This study examines the influence of chronic retroviral infection of mice with a LPBM5 virus mixture on the paracrine system involving immune cells and 1,25-(OH)2D3 in the spleen. Plasma ionized calcium, 25-(OH)D and 1,25-(OH)2D of infected mice were unchanged. In contrast, the specific binding of 1,25-(OH)2D3 to spleen cytosol and the number of monocyte/macrophages expressing 1,25-(OH)2D3 receptors (VDR) were markedly increased. The retroviral infection also influenced the local production of 1,25-(OH)2D3 in the spleen. It did not alter this production in monocyte/macrophages but increased that in isolated T cells. Isolated B cells in control mice did not produce 1,25-(OH)2D3, but they increased the ability of isolated T cells to produce this metabolite during coculture incubations. Infection altered this cell interaction as 1,25-(OH)2D3 production in infected T cells decreased when these cells were cocultured with infected B cells. Thus, chronic retroviral infection alters both the local vitamin D metabolism and VDR expression by immune cells in mice. These findings suggest close local interactions between 1,25-(OH)2D3 and immune system activation during retroviral infection.


Assuntos
Calcitriol/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Receptores de Superfície Celular/metabolismo , Baço/metabolismo , Animais , Calcifediol/metabolismo , Técnicas de Cocultura , Feminino , Linfócitos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , Baço/patologia , Linfócitos T/metabolismo
20.
Proc Soc Exp Biol Med ; 216(3): 386-91, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9402143

RESUMO

The effects of murine leukemia retrovirus infection on production of cytokines was investigated in mice fed different doses of dehydroepiandrosterone (DHEA). Young C57BL/6 female mice were injected with LP-BM5 murine retrovirus or were kept as uninfected controls. Two weeks later, each group was divided into subgroups: fed unsupplemented AIN 93 diet as the control, or diets supplemented with 0.02% DHEA (0.9 mg/mouse/day) or 0.06% DHEA (2.7 mg/mouse/day). The uninfected mice supplemented with 0.06% DHEA showed a significant (P < 0.05) increase in interleukin-2 (IL-2) and gamma-interferon (IFN-gamma) production, and hepatic vitamin E levels. Retroviral infection induced severe oxidative stress that was reduced by DHEAS supplementation in retrovirally infected mice. DHEA supplementation prevented the retrovirus-induced loss of cytokines (IL-2 and IFN-gamma) secretion by mitogen stimulated spleen cells. DHEA also suppressed the production of cytokines interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) by T helper 2 (Th2) cells which were otherwise stimulated by retrovirus infection. Thus, immune dysfunction and increased oxidation induced by murine retrovirus infection were largely prevented by DHEA.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Citocinas/biossíntese , Citocinas/metabolismo , Desidroepiandrosterona/uso terapêutico , Vírus da Leucemia Murina , Linfoma/imunologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Animais , Peso Corporal/efeitos dos fármacos , Colesterol/metabolismo , Citocinas/efeitos dos fármacos , Dieta , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linfoma/tratamento farmacológico , Linfoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Oxirredução/efeitos dos fármacos , Fosfolipídeos/metabolismo , Distribuição Aleatória , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Vitamina A/metabolismo , Vitamina E/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA