Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Aging Cell ; 16(5): 956-965, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28612507

RESUMO

Emerging evidence suggests that cell senescence plays an important role in aging-associated diseases including neurodegenerative diseases. HIV leads to a spectrum of neurologic diseases collectively termed HIV-associated neurocognitive disorders (HAND). Drug abuse, particularly methamphetamine (meth), is a frequently abused psychostimulant among HIV+ individuals and its abuse exacerbates HAND. The mechanism by which HIV and meth lead to brain cell dysregulation is not entirely clear. In this study, we evaluated the impact of HIV and meth on astrocyte senescence using in vitro and several animal models. Astrocytes constitute up to 50% of brain cells and play a pivotal role in marinating brain homeostasis. We show here that HIV and meth induce significant senescence of primary human fetal astrocytes, as evaluated by induction of senescence markers (ß-galactosidase and p16INK4A ), senescence-associated morphologic changes, and cell cycle arrest. HIV- and meth-mediated astrocyte senescence was also demonstrated in three small animal models (humanized mouse model of HIV/NSG-huPBMCs, HIV-transgenic rats, and in a meth administration rat model). Senescent astrocytes in turn mediated neuronal toxicity. Further, we show that ß-catenin, a pro-survival/proliferation transcriptional co-activator, is downregulated by HIV and meth in human astrocytes and this downregulation promotes astrocyte senescence while induction of ß-catenin blocks HIV- and meth-mediated astrocyte senescence. These studies, for the first time, demonstrate that HIV and meth induce astrocyte senescence and implicate the ß-catenin pathway as potential therapeutic target to overcome astrocyte senescence.


Assuntos
Complexo AIDS Demência/genética , Astrócitos/metabolismo , Estimulantes do Sistema Nervoso Central/efeitos adversos , Metanfetamina/efeitos adversos , Neurônios/metabolismo , Transtornos Relacionados ao Uso de Substâncias/genética , beta Catenina/genética , Complexo AIDS Demência/complicações , Complexo AIDS Demência/metabolismo , Complexo AIDS Demência/fisiopatologia , Animais , Animais Geneticamente Modificados , Astrócitos/patologia , Astrócitos/virologia , Proteínas de Ligação ao Cálcio , Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , HIV-1/patogenicidade , HIV-1/fisiologia , Humanos , Masculino , Camundongos , Proteínas dos Microfilamentos , Neurônios/patologia , Neurônios/virologia , Cultura Primária de Células , Ratos , Transdução de Sinais , Transtornos Relacionados ao Uso de Substâncias/complicações , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , beta Catenina/metabolismo
2.
Glia ; 63(3): 441-51, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25331637

RESUMO

HIV-mediated neuropathogenesis is a multifaceted process involving several players, including resident brain cells (neurons, astrocytes, and microglia) and infiltrating cells [peripheral blood mononuclear cells (PBMCs)]. We evaluated the dynamic interaction between astrocytes and infiltrating PBMCs as it impacts HIV in the CNS. We demonstrate that human primary-derived astrocytes (PDAs) predominantly secrete Wnt 1, 2b, 3, 5b, and 10b. Wnts are small secreted glycoproteins that initiate either ß-catenin-dependent or independent signal transduction. The Wnt pathway plays a vital role in the regulation of CNS activities including neurogenesis, neurotransmitter release, synaptic plasticity, and memory consolidation. We show that HIV infection of PDAs altered astrocyte Wnt profile by elevating Wnts 2b and 10b. Astrocyte conditioned media (ACM) inhibited HIV replication in PBMCs by 50%. Removal of Wnts from ACM abrogated its ability to suppress HIV replication in PBMCs. Inversely, PBMCs supernatant activated PDAs, as demonstrated by a 10-fold increase in HLA-DR and a 5-fold increase in IFNγ expression, and enhanced astrocyte susceptibility to HIV by 2-fold, which was mediated by IFNγ in a Stat-3-dependent manner. Collectively, these data demonstrate a dynamic interaction between astrocytes and PBMCs, whereby astrocyte-secreted Wnts exert an anti-HIV effect on infected PBMCs and PBMCs, in turn, secrete IFNγ that enhance astrocyte susceptibility to productive HIV infection and mediate their activation.


Assuntos
Complexo AIDS Demência/fisiopatologia , Astrócitos/fisiologia , Leucócitos Mononucleares/fisiologia , Complexo AIDS Demência/virologia , Astrócitos/virologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados , HIV/fisiologia , Antígenos HLA-DR/metabolismo , Humanos , Interferon gama/metabolismo , Leucócitos Mononucleares/virologia , Fator de Transcrição STAT3/metabolismo , Proteínas Wnt/metabolismo
3.
Curr HIV Res ; 12(3): 164-73, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24862328

RESUMO

Cells of the monocyte/macrophage lineage are an important target for HIV-1 infection. They are often at anatomical sites linked to HIV-1 transmission and are an important vehicle for disseminating HIV-1 throughout the body, including the central nervous system. Monocytes do not support extensive productive HIV-1 replication, but they become more susceptible to HIV-1infection as they differentiate into macrophages. The mechanisms guiding susceptibility of HIV-1 replication in monocytes versus macrophages are not entirely clear. We determined whether endogenous activity of ß-catenin signaling impacts differential susceptibility of monocytes and monocyte-derived macrophages (MDMs) to productive HIV-1 replication. We show that monocytes have an approximately 4-fold higher activity of ß-catenin signaling than MDMs. Inducing ß-catenin in MDMs suppressed HIV-1 replication by 5-fold while inhibiting endogenous ß-catenin signaling in monocytes by transfecting with a dominant negative mutant for the downstream effector of ß- catenin (TCF-4) promoted productive HIV-1 replication by 6-fold. These findings indicate that ß-catenin/TCF-4 is an important pathway for restricted HIV-1 replication in monocytes and plays a significant role in potentiating HIV-1 replication as monocytes differentiate into macrophages. Targeting this pathway may provide a novel strategy to purge the latent reservoir from monocytes/macrophages, especially in sanctuary sites for HIV-1 such as the central nervous system.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , HIV-1/fisiologia , Macrófagos/imunologia , Monócitos/imunologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Replicação Viral , beta Catenina/metabolismo , HIV-1/imunologia , Humanos , Macrófagos/virologia , Monócitos/virologia , Fator de Transcrição 4
4.
AIDS Res Hum Retroviruses ; 30(7): 677-84, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24593860

RESUMO

HIV infection is a risk factor for the tumorigenesis including non-AIDS-defining cancers such as those of the gastrointestinal tract. However, the mechanisms underlying such cancer outgrowth are still unknown. Furthermore, combined HIV/cancer studies are difficult to evaluate using primate models or in the clinical patient setting. To understand the mechanisms of tumor outgrowth in the context of HIV infection, we adopted a humanized mouse model permissive to infection and cancer as well as an in vivo humanized mouse challenge with colon cancer in the context of HIV infection. Immunodeficient NOD SCID IL-2R(-/-) mice were immunologically reconstituted by adoptive transfer of 10(7) HIV-negative donor peripheral blood leukocytes and challenged with 10(6) HCT116 human colon cancer cells. A group of mice was treated with antiretroviral therapy. Tumor microenvironment and epithelial tissues in the context of HIV infection were analyzed using immunohistochemistry. We demonstrate that HIV-infected humanized mice develop significantly larger tumors than uninfected mice (p<0.05). Epithelial cell proliferation in HIV-infected mice is significantly enhanced in comparison to proliferation in uninfected mice (p<0.01). Moreover, the activation of ß-catenin, an important step in intestinal epithelial cell proliferation and tumorigenesis, is elevated in the tumors of HIV-infected mice (p<0.0001). Importantly, antiretroviral therapy reverses these pathological processes independently of CD4(+) T cell return. These findings model the ability of HIV infection to result in tumor outgrowth that is evident in HIV-positive patients and lend insight into previously unrecognized mechanisms that may underlie this pathology.


Assuntos
Neoplasias Gastrointestinais/imunologia , Neoplasias Gastrointestinais/patologia , Infecções por HIV/imunologia , HIV-1/imunologia , Leucócitos/imunologia , Transferência Adotiva , Animais , Antirretrovirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Neoplasias Gastrointestinais/virologia , Células HCT116 , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Humanos , Transfusão de Leucócitos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Receptores de Interleucina-2/genética , beta Catenina/biossíntese
5.
Methods Mol Biol ; 900: 381-401, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22933080

RESUMO

Experimental autoimmune encephalomyelitis (EAE) and Theiler's Murine Encephalitis Virus-Induced Demyelinating Disease (TMEV-IDD) are two clinically relevant murine models of multiple sclerosis (MS). Like MS, both are characterized by mononuclear cell infiltration into the CNS and demyelination. EAE is induced by either the administration of myelin protein or peptide in adjuvant or by the adoptive transfer of encephalitogenic T cell blasts into naïve recipients. The relative merits of each of these protocols are compared. Depending on the type of question being asked, different mouse strains and peptides are used. Different disease courses are observed with different strains and different peptides in active EAE. These variations are also addressed. Additionally, issues relevant to clinical grading of EAE in mice are discussed. In addition to EAE induction, useful references for other disease indicators such as DTH, in vitro proliferation, and immunohistochemistry are provided. TMEV-IDD is a useful model for understanding the possible viral etiology of MS. This section provides detailed information on the preparation of viral stocks and subsequent intracerebral infection of mice. Additionally, virus plaque assay and clinical disease assessment are discussed. Recently, recombinant TMEV strains have been created for the study of molecular mimicry which incorporate various 30 amino acid myelin epitopes within the leader region of TMEV.


Assuntos
Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Técnicas Imunológicas/métodos , Esclerose Múltipla/patologia , Theilovirus/fisiologia , Transferência Adotiva , Sequência de Aminoácidos , Animais , Progressão da Doença , Encefalomielite Autoimune Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Esclerose Múltipla/imunologia , Peptídeos/química , Vacinação
6.
PLoS One ; 6(10): e26143, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22022540

RESUMO

FasL expression on human immune cells and cancer cells plays important roles in immune homeostasis and in cancer development. Our previous study suggests that polymorphisms in the FasL promoter can significantly affect the gene expression in human cells. In addition to the functional FasL SNP -844C>T (rs763110), three other SNPs (SNP -756A>G or rs2021837, SNP -478A>T or rs41309790, and SNP -205 C>G or rs74124371) exist in the proximal FasL promoter. In the current study, we established three major FasL hyplotypes in humans. Interestingly, a transcription motif search revealed that the FasL promoter possessed two consensus T-cell factor (TCF/LEF1) binding elements (TBEs), which is either polymorphic (SNP -205C>G) or close to the functional SNP -844C>T. Subsequently, we demonstrate that both FasL TBEs formed complexes with the TCF-4 and ß-catenin transcription factors in vitro and in vivo. Co-transfection of LEF-1 and ß-catenin transcription factors significantly increased FasL promoter activities, suggesting that FasL is a target gene of the ß-catenin/T-cell factor pathway. More importantly, we found that the rare allele (-205G) of the polymorphic FasL TBE (SNP -205C>G) failed to bind the TCF-4 transcription factor and that SNP -205 C>G significantly affected the promoter activity. Furthermore, promoter reporter assays revealed that FasL SNP haplotypes influenced promoter activities in human colon cancer cells and in human T cells. Finally, ß-catenin knockdown significantly decreased the FasL expression in human SW480 colon cancer cells. Collectively, our data suggest that ß-catenin may be involved in FasL gene regulation and that FasL expression is influenced by FasL SNP haplotypes, which may have significant implications in immune response and tumorigenesis.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proteína Ligante Fas/genética , Haplótipos/genética , Regiões Promotoras Genéticas/genética , Transdução de Sinais , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Proteína Ligante Fas/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único/genética , Ligação Proteica , Linfócitos T/metabolismo , Fator de Transcrição 4 , Regulação para Cima/genética
7.
J Autoimmun ; 36(2): 142-54, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21273044

RESUMO

Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) serves as virus-induced model of chronic progressive multiple sclerosis. Infection of susceptible SJL/J mice leads to life-long CNS virus persistence and a progressive autoimmune demyelinating disease mediated by myelin-specific T cells activated via epitope spreading. In contrast, virus is rapidly cleared by a robust CTL response in TMEV-IDD-resistant C57BL/6 mice. We investigated whether differential induction of regulatory T cells (Tregs) controls susceptibility to TMEV-IDD. Infection of disease-susceptible SJL/J, but not B6 mice, leads to rapid activation and expansion of Tregs resulting in an unfavorable CNS ratio of Treg:Teffector cells. In addition, anti-CD25-induced inactivation of Tregs in susceptible SJL/J, but not resistant B6, mice results in significantly decreased clinical disease concomitant with enhanced anti-viral CD4(+), CD8(+) and antibody responses resulting in decreased CNS viral titers. This is the first demonstration that virus-induced Treg activation regulates susceptibility to autoimmune disease differentially in susceptible and resistant strains of mice and provides a new mechanistic explanation for the etiology of infection-induced autoimmunity.


Assuntos
Doenças Desmielinizantes/imunologia , Modelos Animais de Doenças , Esclerose Múltipla/imunologia , Linfócitos T Reguladores/imunologia , Theilovirus/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/prevenção & controle , Proliferação de Células , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Humanos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Receptores de Fator de Crescimento Neural/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Índice de Gravidade de Doença , Linfócitos T Reguladores/metabolismo , Theilovirus/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA