Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38915632

RESUMO

Cerebral organoids (COs) are a valuable tool to study the intricate interplay between glial cells and neurons in brain development and disease, including HIV-associated neuroinflammation. We developed a novel approach to generate microglia containing COs (CO-iMs) by co-culturing hematopoietic progenitors and induced pluripotent stem cells. This approach allowed for the differentiation of microglia within the organoids concomitantly to the neuronal progenitors. CO- iMs exhibited higher efficiency in generation of CD45 + /CD11b + /Iba-1 + microglia cells compared to conventional COs with physiologically relevant proportion of microglia (∼7%). CO-iMs exhibited substantially higher expression of microglial homeostatic and sensome markers as well as markers for the complement cascade. CO-iMs showed susceptibility to HIV infection resulting in a significant increase in several pro-inflammatory cytokines/chemokines and compromised neuronal function, which were abrogated by addition of antiretrovirals. Thus, CO-iM is a robust model to decipher neuropathogenesis, neurological disorders, and viral infections of brain cells in a 3D culture system.

2.
Antimicrob Agents Chemother ; 66(4): e0194121, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35293780

RESUMO

Combination antiretroviral therapy (cART) dramatically changed the face of the HIV/AIDS pandemic, making it one of the most prominent medical breakthroughs of the past 3 decades. However, as the life span of persons living with HIV (PLWH) continues to approach that of the general population, the same cannot be said regarding their quality of life. PLWH are affected by comorbid conditions such as high blood pressure, diabetes, and neurocognitive impairment at a higher rate and increased severity than their age-matched counterparts. PLWH also have higher levels of inflammation, the drivers of which are not entirely clear. As cART treatment is lifelong, we assessed here the effects of cART, independent of HIV, on primary human monocyte-derived macrophages (MDMs). MDMs were unskewed or skewed to an alternative phenotype and treated with Atripla or Triumeq, two first-line cART treatments. We report that Triumeq skewed alternative MDMs toward an inflammatory nonsenescent phenotype. Both Atripla and Triumeq caused mitochondrial dysfunction, specifically efavirenz and abacavir. Additionally, transcriptome sequencing (RNA-seq) demonstrated that both Atripla and Triumeq caused differential regulation of genes involved in immune regulation and cell cycle and DNA repair. Collectively, our data demonstrate that cART, independent of HIV, alters the MDM phenotype. This suggests that cART may contribute to cell dysregulation in PLWH that subsequently results in increased susceptibility to comorbidities.


Assuntos
Fármacos Anti-HIV , Infecções por HIV , HIV-1 , Fármacos Anti-HIV/uso terapêutico , Combinação Efavirenz, Emtricitabina, Fumarato de Tenofovir Desoproxila/metabolismo , Combinação Efavirenz, Emtricitabina, Fumarato de Tenofovir Desoproxila/farmacologia , Combinação Efavirenz, Emtricitabina, Fumarato de Tenofovir Desoproxila/uso terapêutico , Humanos , Macrófagos , Mitocôndrias , Qualidade de Vida
3.
Cytokine ; 136: 155252, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32818703

RESUMO

Expression of cytokines/chemokines is tightly regulated at the transcription level. This is crucial in the central nervous system to maintain neuroimmune homeostasis. IL-8 a chemoattractant, which recruits neutrophils, T cells, and basophils into the brain in response to inflammation and/or injury is secreted predominantly by neurons, microglia, and astrocytes. Here, we investigated the mechanism by which astrocytes regulate IL-8 expression. We demonstrate that while ß-catenin negatively regulated IL-8 transcription, its canonical transcriptional partners, members of the TCF/LEF transcription factors (TCF1, TCF3, TCF4 and LEF1) and Activating transcription factor 2 (ATF2) positively regulated IL-8 transcription. We further identified a putative TCF/LEF binding site at -175nt close to the minimal transcription region on the IL-8 promoter, mutation of which caused a significant reduction in IL-8 promoter activity. Chromatin immunoprecipitation demonstrated binding of TCF1, TCF4, LEF1 and ATF2 on the IL-8 promoter suggesting that TCFs/LEF partner with ATF2 to induce IL-8 transcription. These findings demonstrate a novel role for ß-catenin in suppression of IL-8 expression and for TCFs/LEF/ATF2 in inducing IL-8. These findings reveal a unique mechanism by which astrocytes tightly regulate IL-8 expression.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Astrócitos/metabolismo , Regulação da Expressão Gênica , Interleucina-8/biossíntese , Fatores de Transcrição TCF/metabolismo , beta Catenina/metabolismo , Linhagem Celular Tumoral , Humanos
4.
Cell Commun Signal ; 18(1): 93, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32546183

RESUMO

BACKGROUND: The Wnt/ß-catenin signaling pathway is a prolific regulator of cell-to-cell communication and gene expression. Canonical Wnt/ß-catenin signaling involves partnering of ß-catenin with members of the TCF/LEF family of transcription factors (TCF1, TCF3, TCF4, LEF1) to regulate gene expression. IL-6 is a key cytokine involved in inflammation and is particularly a hallmark of inflammation in the brain. Astrocytes, specialized glial cells in the brain, secrete IL-6. How astrocytes regulate IL-6 expression is not entirely clear, although in other cells NFκB and C/EBP pathways play a role. We evaluated here the interface between ß-catenin, TCFs/LEF and C/EBP and NF-κB in relation to IL-6 gene regulation in astrocytes. METHODS: We performed molecular loss and/or gain of function studies of ß-catenin, TCF/LEF, NFκB, and C/EBP to assess IL-6 regulation in human astrocytes. Specifically, siRNA mediated target gene knockdown, cDNA over expression of target gene, and pharmacological agents for regulation of target proteins were used. IL-6 levels was evaluated by real time quantitative PCR and ELISA. We also cloned the IL-6 promoter under a firefly luciferase reporter and used bioinformatics, site directed mutagenesis, and chromatin immunoprecipitation to probe the interaction between ß-catenin/TCFs/LEFs and IL-6 promoter activity. RESULTS: ß-catenin binds to TCF/LEF to inhibits IL-6 while TCFs/LEF induce IL-6 transcription through interaction with ATF-2/SMADs. ß-catenin independent of TCFs/LEF positively regulates C/EBP and NF-κB, which in turn activate IL-6 expression. The IL-6 promoter has two putative regions for TCFs/LEF binding, a proximal site located at -91 nt and a distal site at -948 nt from the transcription start site, both required for TCF/LEF induction of IL-6 independent of ß-catenin. CONCLUSION: IL-6 regulation in human astrocytes engages a discordant interaction between ß-catenin and TCF/LEF. These findings are intriguing given that no role for ß-catenin nor TCFs/LEF to date is associated with IL-6 regulation and suggest that ß-catenin expression in astrocytes is a critical regulator of anti-inflammatory responses and its disruption can potentially mediate persistent neuroinflammation. Video Abstract.


Assuntos
Astrócitos/metabolismo , Fatores Nucleares de Hepatócito/metabolismo , Interleucina-6/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Astrócitos/citologia , Linhagem Celular , Humanos
5.
PLoS Pathog ; 16(6): e1008381, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32525948

RESUMO

HIV invades the brain during acute infection. Yet, it is unknown whether long-lived infected brain cells release productive virus that can egress from the brain to re-seed peripheral organs. This understanding has significant implication for the brain as a reservoir for HIV and most importantly HIV interplay between the brain and peripheral organs. Given the sheer number of astrocytes in the human brain and their controversial role in HIV infection, we evaluated their infection in vivo and whether HIV infected astrocytes can support HIV egress to peripheral organs. We developed two novel models of chimeric human astrocyte/human peripheral blood mononuclear cells: NOD/scid-IL-2Rgc null (NSG) mice (huAstro/HuPBMCs) whereby we transplanted HIV (non-pseudotyped or VSVg-pseudotyped) infected or uninfected primary human fetal astrocytes (NHAs) or an astrocytoma cell line (U138MG) into the brain of neonate or adult NSG mice and reconstituted the animals with human peripheral blood mononuclear cells (PBMCs). We also transplanted uninfected astrocytes into the brain of NSG mice and reconstituted with infected PBMCs to mimic a biological infection course. As expected, the xenotransplanted astrocytes did not escape/migrate out of the brain and the blood brain barrier (BBB) was intact in this model. We demonstrate that astrocytes support HIV infection in vivo and egress to peripheral organs, at least in part, through trafficking of infected CD4+ T cells out of the brain. Astrocyte-derived HIV egress persists, albeit at low levels, under combination antiretroviral therapy (cART). Egressed HIV evolved with a pattern and rate typical of acute peripheral infection. Lastly, analysis of human cortical or hippocampal brain regions of donors under cART revealed that astrocytes harbor between 0.4-5.2% integrated HIV gag DNA and 2-7% are HIV gag mRNA positive. These studies establish a paradigm shift in the dynamic interaction between the brain and peripheral organs which can inform eradication of HIV reservoirs.


Assuntos
Astrócitos , Barreira Hematoencefálica , Infecções por HIV , HIV-1/metabolismo , Hipocampo , Liberação de Vírus , Animais , Antirretrovirais/farmacologia , Astrócitos/metabolismo , Astrócitos/patologia , Astrócitos/virologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/virologia , Linhagem Celular Tumoral , Infecções por HIV/genética , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/genética , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/virologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID
6.
Sci Rep ; 10(1): 4746, 2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32179788

RESUMO

Ginkgolic acids (GA) are alkylphenol constituents of the leaves and fruits of Ginkgo biloba. GA has shown pleiotropic effects in vitro, including: antitumor effects through inhibition of lipogenesis; decreased expression of invasion associated proteins through AMPK activation; and potential rescue of amyloid-ß (Aß) induced synaptic impairment. GA was also reported to have activity against Escherichia coli and Staphylococcus aureus. Several mechanisms for this activity have been suggested including: SUMOylation inhibition; blocking formation of the E1-SUMO intermediate; inhibition of fatty acid synthase; non-specific SIRT inhibition; and activation of protein phosphatase type-2C. Here we report that GA inhibits Herpes simplex virus type 1 (HSV-1) by inhibition of both fusion and viral protein synthesis. Additionally, we report that GA inhibits human cytomegalovirus (HCMV) genome replication and Zika virus (ZIKV) infection of normal human astrocytes (NHA). We show a broad spectrum of fusion inhibition by GA of all three classes of fusion proteins including HIV, Ebola virus (EBOV), influenza A virus (IAV) and Epstein Barr virus (EBV). In addition, we show inhibition of a non-enveloped adenovirus. Our experiments suggest that GA inhibits virion entry by blocking the initial fusion event. Data showing inhibition of HSV-1 and CMV replication, when GA is administered post-infection, suggest a possible secondary mechanism targeting protein and DNA synthesis. Thus, in light of the strong effect of GA on viral infection, even after the infection begins, it may potentially be used to treat acute infections (e.g. Coronavirus, EBOV, ZIKV, IAV and measles), and also topically for the successful treatment of active lesions (e.g. HSV-1, HSV-2 and varicella-zoster virus (VZV)).


Assuntos
Antivirais/farmacologia , Infecções por Vírus de DNA/metabolismo , Vírus de DNA/efeitos dos fármacos , Infecções por Vírus de RNA/metabolismo , Vírus de RNA/efeitos dos fármacos , Salicilatos/farmacologia , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Astrócitos/metabolismo , Chlorocebus aethiops , Replicação do DNA/efeitos dos fármacos , Infecções por Vírus de DNA/virologia , Vírus de DNA/genética , DNA Viral/genética , Células HEK293 , Humanos , Infecções por Vírus de RNA/virologia , Vírus de RNA/genética , Células Vero , Proteínas do Envelope Viral/biossíntese , Proteínas Virais de Fusão/biossíntese , Vírion/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
7.
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
8.
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
9.
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
10.
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
11.
Virology ; 443(2): 375-83, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23769242

RESUMO

Human Immunodeficiency virus type 1 (HIV-1) disproportionately affects women, accounting for > 50% of new HIV infections in adults worldwide. While multiple mechanisms may contribute to a greater degree of HIV infection in women than men, we evaluated the direct effect of 17ß-estradiol, the most bioactive form of estrogen in women, on HIV replication in peripheral blood mononuclear cells (PBMCs). We demonstrate that 17ß-estradiol, in an ERα dependent manner, inhibits HIV replication by activating ß-catenin signaling. Specifically, we show for the first time that 17ß-estradiol induces a complex formation between ERα and ß-catenin which tether on the HIV LTR at -143nt site from +1 start site of HIV transcription to repress HIV promoter activity. These studies define a role of 17ß-estradiol in inhibiting HIV replication which may impact HIV pathogenesis in women and add to a growing list of viruses that are inhibited by 17ß-estradiol through ERα engagment.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , HIV-1/efeitos dos fármacos , Adolescente , Adulto , Estradiol/análogos & derivados , Receptor alfa de Estrogênio/genética , Feminino , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Repetição Terminal Longa de HIV/efeitos dos fármacos , Repetição Terminal Longa de HIV/genética , HIV-1/genética , Humanos , Leucócitos Mononucleares/virologia , Replicação Viral/efeitos dos fármacos , Adulto Jovem , beta Catenina/genética , beta Catenina/metabolismo
12.
J Virol ; 86(4): 1911-21, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22156527

RESUMO

The Wnt/ß-catenin pathway is involved in diverse cell functions governing development and disease. ß-Catenin, a central mediator of this pathway, binds to members of the TCF/LEF family of transcription factors to modulate hundreds of genes. Active Wnt/ß-catenin/TCF-4 signaling plays a significant role in repression of HIV-1 replication in multiple cell targets, including astrocytes. To determine the mechanism by which active ß-catenin/TCF-4 leads to inhibition of HIV replication, we knocked down ß-catenin or TCF/LEF members in primary astrocytes and astrocytomas transiently transfected with an HIV long terminal repeat (LTR)-luciferase reporter that contained an integrated copy of the HIV LTR-luciferase construct. Knockdown of either ß-catenin or TCF-4 induced LTR activity by 2- to 3-fold under both the episomal and integrated conditions. This knockdown also increased presence of serine 2-phosphorylated RNA polymerase II (Pol II) on the HIV LTR as well as enhanced its processivity. Knockdown of ß-catenin/TCF-4 also impacted tethering of other transcription factors on the HIV promoter. Specifically, knockdown of TCF-4 enhanced binding of C/EBPß, C/EBPδ, and NF-κB to the HIV LTR, while ß-catenin knockdown increased binding of C/EBPß and C/EBPδ but had no effect on NF-κB. Approximately 150 genes in astrocytes were impacted by ß-catenin knockdown, including genes involved in inflammation/immunity, uptake/transport, vesicular transport/exocytosis, apoptosis/cellular stress, and cytoskeleton/trafficking. These findings indicate that modulation of the ß-catenin/TCF-4 axis impacts the basal level of HIV transcription in astrocytes, which may drive low level/persistent HIV in astrocytes that can contribute to ongoing neuroinflammation, and this axis also has profound effects on astrocyte biology.


Assuntos
Astrócitos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Infecções por HIV/metabolismo , HIV-1/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , beta Catenina/metabolismo , Astrócitos/virologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Linhagem Celular Tumoral , Células Cultivadas , Regulação Viral da Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/virologia , Repetição Terminal Longa de HIV , HIV-1/fisiologia , Humanos , Dados de Sequência Molecular , Transdução de Sinais , Fator de Transcrição 4 , Fatores de Transcrição/genética , beta Catenina/genética
13.
Blood Cells Mol Dis ; 42(2): 136-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19150249

RESUMO

Purified samples of CD45+ hematopoietic cells are a prerequisite for chimerism analysis in transplantation therapies, and are useful in various research and clinical settings such as functional and molecular analysis or disease diagnosis. Recently, we have established a flow-based adhesion molecule-dependent process for the purification of these cells from human bone marrow. However, for practical purposes, it is desirable to apply this approach to process small volumes of human blood. CD45+ cell purities were >94% when PBMNCs and plasma depleted blood were perfused through P-selectin coated microtubes. However, P-selectin surface failed to capture CD45+ cells when fresh blood prior to washing was perfused. The process requires a pre-step of plasma removal which otherwise inhibits interactions of cell surface PSGL-1 with immobilized P-selectin due to the presence of soluble PSGL-1 in plasma. We conclude that P-selectin can be used in a compact flow device to isolate and purify CD45+ cells directly from human peripheral blood. The process is simple, rapid, cost effective and represents a physiologic approach to the capture and purification of CD45+ MNCs from peripheral blood.


Assuntos
Adesão Celular , Separação Celular/instrumentação , Proteínas Imobilizadas/metabolismo , Antígenos Comuns de Leucócito/análise , Leucócitos/química , Glicoproteínas de Membrana/metabolismo , Selectina-P/metabolismo , Separação Celular/métodos , Sobrevivência Celular , Centrifugação com Gradiente de Concentração , Ficoll , Citometria de Fluxo/métodos , Humanos , Perfusão , Plasma , Proteínas Recombinantes/metabolismo
14.
Am J Hematol ; 83(8): 627-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18442063

RESUMO

Recently, a flow-based selectin-dependent method for capture and enrichment of specific type blood cells (CD34+ hematopoietic stem and progenitor cells) from bone marrow and peripheral blood has been described. Using a similar approach, here we show isolation of CD45+ blood cells directly from human bone marrow to a high purity (90%-97%). The process mimics a ubiquitous mechanism of cell trafficking in the body for the recruitment of neutrophils during inflammation. The method is straightforward, rapid, and may represent a practical alternative to CD45+ cell enrichment procedures required for chimerism analysis following allogenic stem-cell transplantation.


Assuntos
Células da Medula Óssea/citologia , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Antígenos Comuns de Leucócito , Adesão Celular , Humanos , Leucócitos/citologia , Métodos , Selectina-P/metabolismo , Perfusão , Ligação Proteica
15.
Br J Haematol ; 140(6): 673-81, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18218048

RESUMO

Clinical infusion of haematopoietic stem and progenitor cells (HSPCs) is vital for restoration of haematopoietic function in many cancer patients. Previously, we have demonstrated an ability to mimic physiological cell trafficking in order to capture CD34-positive (CD34+) HSPCs using monolayers of the cell adhesion protein P-selectin in flow chambers. The current study aimed to determine if HSPCs could be captured directly from circulating blood in vivo. Vascular shunt prototypes, coated internally with P-selectin, were inserted into the femoral artery of rats. Blood flow through the cell capture device resulted in a wall shear stress of 4-6 dynes/cm(2). After 1-h blood perfusion, immunofluorescence microscopy and flow cytometric analysis revealed successful capture of mononuclear cells positive for the HSPC surface marker CD34. Purity of captured CD34+ cells showed sevenfold enrichment over levels found in whole blood, with an average purity of 28%. Robust cell capture and HSPC enrichment were also demonstrated in devices that were implanted in a closed-loop arterio-venous shunt conformation for 2 h. Adherent cells were viable in culture and able to differentiate into burst-forming units. This study demonstrated an ability to mimic the physiological arrest of HSPCs from blood in an implantable device and may represent a practical alternative for adult stem cell capture and enrichment.


Assuntos
Antígenos CD34/sangue , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Implantes Experimentais , Selectina-P/metabolismo , Animais , Adesão Celular , Divisão Celular , Sobrevivência Celular , Ensaio de Unidades Formadoras de Colônias , Artéria Femoral , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
16.
Clin Chem ; 54(1): 77-85, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18024531

RESUMO

BACKGROUND: Enrichment and purification of hematopoietic stem and progenitor cells (HSPCs) is important in transplantation therapies for hematologic disorders and in basic stem cell research. Primitive CD34+ HSPCs have demonstrated stronger rolling adhesion on selectins than mature CD34- mononuclear cells (MNCs). We have exploited this differential rolling behavior to capture and purify HSPCs from bone marrow by perfusing MNCs through selectin-coated microtubes. METHODS: Bone marrow MNCs were perfused through the cell-capture microtubes coated with adhesion molecules. We washed the device lumen and visualized and estimated captured cells by video microscopy. Adherent cells were eluted by high shear, calcium-free buffer, and air embolism. We used immunofluorescence staining followed by flow cytometry to analyze CD34+ HSPCs. RESULTS: CD34+ HSPC purity of cells captured in adhesion molecule-coated devices was significantly higher than the fraction of CD34+ cells found in bone marrow MNCs [mean (SE) 2.5% (0.8%)]. P-selectin-coated surfaces yielded 16% to 20% CD34+ cell purity, whereas antibody-coated surfaces yielded 12% to 18%. Although CD34+ cell purity was comparable between selectin and antibody surfaces, the total number of CD34+ HSPCs captured was significantly higher in P-selectin devices (approximately 5.7 x 10(4) to 7.1 x 10(4)) than antibody devices (approximately 1.74 x 10(4) to 2.61 x 10(4)). CONCLUSIONS: P-selectin can be used in a compact flow device to capture HSPCs. Selectin-mediated capture of CD34+ HSPCs resulted in enrichment approximately 8-fold higher than the CD34+ cell population from bone marrow MNCs. This study supports the hypothesis that flow-based, adhesion molecule-mediated capture may be a viable alternative approach to the capture and purification of HSPCs.


Assuntos
Antígenos CD34/metabolismo , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Selectina-P , Anticorpos Monoclonais , Antígenos CD34/imunologia , Adesão Celular , Movimento Celular , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Selectina-P/genética , Proteínas Recombinantes de Fusão
17.
Mol Microbiol ; 47(6): 1681-94, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12622821

RESUMO

The pathogenic yeast Cryptococcus neoformans (Cn) var. gattii causes meningoencephalitis in healthy individuals, unlike the better known Cn varieties grubii and neoformans, which are common in immunocompromised individuals. The virulence determinants and mechanisms of host predilection are poorly defined for var. gattii. The present study focused on the characterization of a Cu,Zn superoxide dismutase (SOD1) gene knock-out mutant constructed by developing a DNA transformation system. The sod1 mutant was highly sensitive to the redox cycling agent menadione, and showed fragmentation of the large vacuole in the cytoplasm, but no other defects were seen in growth, capsule synthesis, mating, sporulation, stationary phase survival or auxotrophies for sulphur-containing amino acids. The sod1 mutant was markedly attenuated in virulence in a mouse model, and it was significantly susceptible to in vitro killing by human neutrophils (PMNs). The deletion of SOD1 also resulted in defects in the expression of a number of virulence factors, i.e. laccase, urease and phospholipase. Complementation of the sod1 mutant with SOD1 resulted in recovery of virulence factor expression and menadione resistance, and in restoration of virulence. Overall, these results suggest that the antioxidant function of Cu,Zn SOD is critical for the pathogenesis of the fungus, but is dispensable in its saprobic life. This report constitutes the first instance in which superoxide dismutase has been directly implicated in the virulence of a fungal pathogen.


Assuntos
Cryptococcus neoformans/fisiologia , Cryptococcus neoformans/patogenicidade , Proteínas Fúngicas/genética , Superóxido Dismutase/genética , Animais , Encéfalo/microbiologia , Cryptococcus neoformans/efeitos dos fármacos , Modelos Animais de Doenças , Proteínas Fúngicas/efeitos dos fármacos , Proteínas Fúngicas/metabolismo , Teste de Complementação Genética , Engenharia Genética/métodos , Humanos , Masculino , Meningite Criptocócica/microbiologia , Meningoencefalite/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Mutação , Neutrófilos/microbiologia , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Transformação Genética , Vacúolos/genética , Vacúolos/ultraestrutura , Virulência/genética , Virulência/fisiologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Vitamina K 3/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA