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











Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(3)2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36992299

RESUMO

Opioid use disorder (OUD) and HIV are comorbid epidemics that can increase depression. HIV and the viral protein Tat can directly induce neuronal injury within reward and emotionality brain circuitry, including the prefrontal cortex (PFC). Such damage involves both excitotoxic mechanisms and more indirect pathways through neuroinflammation, both of which can be worsened by opioid co-exposure. To assess whether excitotoxicity and/or neuroinflammation might drive depressive behaviors in persons infected with HIV (PWH) and those who use opioids, male mice were exposed to HIV-1 Tat for eight weeks, given escalating doses of morphine during the last two weeks, and assessed for depressive-like behavior. Tat expression decreased sucrose consumption and adaptability, whereas morphine administration increased chow consumption and exacerbated Tat-induced decreases in nesting and burrowing-activities associated with well-being. Across all treatment groups, depressive-like behavior correlated with increased proinflammatory cytokines in the PFC. Nevertheless, supporting the theory that innate immune responses adapt to chronic Tat exposure, most proinflammatory cytokines were unaffected by Tat or morphine. Further, Tat increased PFC levels of the anti-inflammatory cytokine IL-10, which were exacerbated by morphine administration. Tat, but not morphine, decreased dendritic spine density on layer V pyramidal neurons in the anterior cingulate. Together, our findings suggest that HIV-1 Tat and morphine differentially induce depressive-like behaviors associated with increased neuroinflammation, synaptic losses, and immune fatigue within the PFC.


Assuntos
Espinhas Dendríticas , Depressão , Imunidade Inata , Morfina , Córtex Pré-Frontal , Produtos do Gene tat do Vírus da Imunodeficiência Humana , Depressão/induzido quimicamente , Depressão/imunologia , Córtex Pré-Frontal/imunologia , Espinhas Dendríticas/patologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/efeitos adversos , Morfina/efeitos adversos , Masculino , Animais , Camundongos , Comportamento Animal , Citocinas/imunologia , Interleucina-10/imunologia , Doenças Neuroinflamatórias , Camundongos Transgênicos , Transtornos Relacionados ao Uso de Opioides , Infecções por HIV , Analgésicos Opioides/efeitos adversos
2.
Exp Neurol ; 358: 114226, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36096180

RESUMO

HIV-associated sensory neuropathies (HIV-SN) are prevalent in >50% of patients aged over 45 years many of which report moderate to severe chronic pain. Previous preclinical studies have investigated the mechanisms by which HIV-1 causes sensory neuropathies and pain-like behaviors. The aim of the present study is to delineate the role of chronic HIV-1 trans-activator of transcription protein (Tat) exposure in the development of neuropathy in mice. The temporal effects of conditional Tat expression on the development of hypersensitivity to mechanical (von Frey filaments) and thermal (heat or cold) stimuli were tested in male and female mice that transgenically expressed HIV-1 Tat in a doxycycline-inducible manner. Inducing Tat expression produced an allodynic response to mechanical or cold (but not heat) stimuli that respectively persisted for at least 23-weeks (mechanical hypersensitivity) or at least 8-weeks (cold hypersensitivity). Both allodynic states were greater in magnitude among females, compared to males, and mechanical increased hypersensitivity progressively in females over time. Acute morphine or gabapentin treatment partly attenuated allodynia in males, but not females. Irrespective of sex, Tat reduced intraepidermal nerve fiber density, the mean amplitude of sensory nerve action potentials (but not conductance), engagement in some pain-related ethological behaviors (cage-hanging and rearing), and down-regulated PPAR-α gene expression in lumbar spinal cord while upregulating TNF-α expression in dorsal root ganglion. Taken together, these data reveal fundamental sex differences in mechanical and cold hypersensitivity in response to Tat and demonstrate the intractable nature in female mice to current therapeutics. Understanding the role of Tat in these pathologies may aid the design of future therapies aimed at mitigating the peripheral sensory neuropathies that accompany neuroHIV.


Assuntos
Infecções por HIV , HIV-1 , Doenças do Sistema Nervoso Periférico , Animais , Síndromes Periódicas Associadas à Criopirina , Doxiciclina , Feminino , Gabapentina , Produtos do Gene tat , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Morfina/farmacologia , Dor , Receptores Ativados por Proliferador de Peroxissomo , Caracteres Sexuais , Fator de Necrose Tumoral alfa
3.
Neurosci Lett ; 788: 136852, 2022 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-36028004

RESUMO

Despite advances in the treatment of human immunodeficiency virus (HIV), approximately one-half of people infected with HIV (PWH) experience neurocognitive impairment. Opioid use disorder (OUD) can exacerbate the cognitive and pathological changes seen in PWH. HIV increases inflammation and immune cell trafficking into the brain; however, less is known about how opioid use disorder affects the recruitment of immune cells. Accordingly, we examined the temporal consequences of HIV-1 Tat and/or morphine on the recruitment of endocytic cells (predominantly perivascular macrophages and microglia) in the dorsal striatum and hippocampus by infusing multi-colored, fluorescently labeled dextrans before and after exposure. To address this question, transgenic mice that conditionally expressed HIV-1 Tat (Tat+), or their control counterparts (Tat-), received three sequential intracerebroventricular (i.c.v.) infusions of Cascade Blue-, Alexa Fluor 488-, and Alexa Fluor 594-labeled dextrans, respectively infused 1 day before, 1-day after, or 13-days after morphine and/or Tat exposure. At the end of the study, the number of cells labeled with each fluorescent dextran were counted. The data demonstrated a significantly higher influx of newly-labeled cells into the perivascular space than into the parenchyma. In the striatum, Tat or morphine exposure increased the number of endocytic cells in the perivascular space, while only morphine increased the recruitment of endocytic cells into the parenchyma. In the hippocampus, morphine (but not Tat) increased the influx of dextran-labeled cells into the perivascular space, but there were too few labeled cells within the hippocampal parenchyma to analyze. Collectively, these data suggest that HIV-1 Tat and morphine act independently to increase the recruitment of endocytic cells into the brain in a region-specific manner.


Assuntos
Infecções por HIV , HIV-1 , Transtornos Relacionados ao Uso de Opioides , Animais , Corpo Estriado/metabolismo , Dextranos , Fluoresceínas , HIV-1/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Morfina/farmacologia , Ácidos Sulfônicos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
4.
Neurosci Lett ; 640: 136-143, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28057474

RESUMO

HIV-1 infection results in blood-brain barrier (BBB) disruption, which acts as a rate-limiting step for HIV-1 entry into the CNS and for subsequent neuroinflammatory/neurotoxic actions. One mechanism by which HIV may destabilize the BBB involves actions of the HIV-1 regulatory protein, trans-activator of transcription (Tat). We utilized a conditional, Tat-expressing transgenic murine model to examine the influence of Tat1-86 expression on BBB integrity and to assess the relative numbers of phagocytic perivascular macrophages and microglia within the CNS in vivo. The effects of Tat exposure on sodium-fluorescein (Na-F; 0.376kDa), horseradish peroxidase (HRP; 44kDa), and Texas Red-labeled dextran (70kDa) leakage into the brain were assessed in Tat-exposed (Tat+) and control (Tat-) mice. Exposure to HIV-1 Tat significantly increased both Na-F and HRP, but not the larger sized Texas Red-labeled dextran, confirming BBB breakdown and also suggesting the breach was limited to molecules <70kDa. Additionally, at 5 d after Tat induction, Alexa Fluor® 488-labeled dextran was bilaterally infused into the lateral ventricles 5 d before the termination of the experiment. Within the caudate/putamen, Tat induction increased the proportion of dextran-labeled Iba-1+ phagocytic perivascular macrophages (∼5-fold) and microglia (∼3-fold) compared to Tat- mice. These data suggest that HIV-1 Tat exposure is sufficient to destabilize BBB integrity and to increase the presence of activated, phagocytic, perivascular macrophages and microglia in an in vivo model of neuroAIDS.


Assuntos
Barreira Hematoencefálica/metabolismo , Corpo Estriado/citologia , Macrófagos/citologia , Microglia/citologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Animais , Contagem de Células , Corpo Estriado/irrigação sanguínea , Dextranos , Fluoresceína , Corantes Fluorescentes , Peroxidase do Rábano Silvestre , Masculino , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Permeabilidade , Fagocitose , Xantenos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
5.
Brain Behav Immun ; 55: 202-214, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26774528

RESUMO

Human immunodeficiency virus (HIV) is associated with motor and mood disorders, likely influenced by reactive microgliosis and subsequent neural damage. We have recapitulated aspects of this pathology in mice that conditionally express the neurotoxic HIV-1 regulatory protein, trans-activator of transcription (Tat). Progestogens may attenuate Tat-related behavioral impairments and reduce neurotoxicity in vitro, perhaps via progesterone's 5α-reductase-dependent metabolism to the neuroprotective steroid, allopregnanolone. To test this, ovariectomized female mice that conditionally expressed (or did not express) central HIV-1 Tat were administered vehicle or progesterone (4mg/kg), with or without pretreatment of a 5α-reductase inhibitor (finasteride, 50mg/kg). Tat induction significantly increased anxiety-like behavior in an open field, elevated plus maze and a marble burying task concomitant with elevated protein oxidation in striatum. Progesterone administration attenuated anxiety-like effects in the open field and elevated plus maze, but not in conjunction with finasteride pretreatment. Progesterone also attenuated Tat-promoted protein oxidation in striatum, independent of finasteride pretreatment. Concurrent experiments in vitro revealed Tat (50nM)-mediated reductions in neuronal cell survival over 60h, as well as increased neuronal and microglial intracellular calcium, as assessed via fura-2 AM fluorescence. Co-treatment with allopregnanolone (100nM) attenuated neuronal death in time-lapse imaging and blocked the Tat-induced exacerbation of intracellular calcium in neurons and microglia. Lastly, neuronal-glial co-cultures were labeled for Iba-1 to reveal that Tat increased microglial numbers in vitro and co-treatment with allopregnanolone attenuated this effect. Together, these data support the notion that 5α-reduced pregnane steroids exert protection over the neurotoxic effects of HIV-1 Tat.


Assuntos
Inibidores de 5-alfa Redutase/farmacologia , Ansiedade/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Colestenona 5 alfa-Redutase/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Pregnanolona/farmacologia , Progesterona/farmacologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Inibidores de 5-alfa Redutase/administração & dosagem , Animais , Ansiedade/induzido quimicamente , Técnicas de Cocultura , Feminino , Finasterida/administração & dosagem , Finasterida/farmacologia , Camundongos , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ovariectomia , Pregnanolona/administração & dosagem , Progesterona/administração & dosagem , Produtos do Gene tat do Vírus da Imunodeficiência Humana/efeitos dos fármacos
6.
Mol Cell Neurosci ; 65: 11-20, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25616162

RESUMO

Infection of the CNS with HIV-1 occurs rapidly after primary peripheral infection. HIV-1 can induce a wide range of neurological deficits, collectively known as HIV-1-associated neurocognitive disorders. Our previous work has shown that the selected neurotoxic effects induced by individual viral proteins, Tat and gp120, and by HIV(+) supernatant are enhanced by co-exposure to morphine. This mimics co-morbid neurological effects observed in opiate-abusing HIV(+) patients. Although there is a correlation between opiate drug abuse and progression of HIV-1-associated neurocognitive disorders, the mechanisms underlying interactions between HIV-1 and opiates remain obscure. Previous studies have shown that HIV-1 induces neurotoxic effects through abnormal activation of GSK3ß. Interestingly, expression of GSK3ß has shown to be elevated in brains of young opiate abusers indicating that GSK3ß is also linked to neuropathology seen with opiate-abusing patients. Thus, we hypothesize that GSK3ß activation is a point of convergence for HIV- and opiate-mediated interactive neurotoxic effects. Neuronal cultures were treated with supernatant from HIV-1SF162-infected THP-1 cells, in the presence or absence of morphine and GSK3ß inhibitors. Our results show that GSK3ß inhibitors, including valproate and small molecule inhibitors, significantly reduce HIV-1-mediated neurotoxic outcomes, and also negate interactions with morphine that result in cell death, suggesting that GSK3ß-activation is an important point of convergence and a potential therapeutic target for HIV- and opiate-mediated neurocognitive deficits.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , HIV-1/patogenicidade , Morfina/toxicidade , Neurônios/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Células Cultivadas , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Neurônios/virologia , Ácido Valproico/farmacologia
7.
PLoS One ; 9(6): e100196, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24949623

RESUMO

HIV-1 enters the CNS soon after initial systemic infection; within the CNS parenchyma infected and/or activated perivascular macrophages, microglia and astrocytes release viral and cellular toxins that drive secondary toxicity in neurons and other cell types. Our previous work has largely modeled HIV-neuropathology using the individual viral proteins Tat or gp120, with murine striatal neurons as targets. To model disease processes more closely, the current study uses supernatant from HIV-1-infected cells. Supernatant from HIV-1SF162-infected differentiated-U937 cells (HIV+sup) was collected and p24 level was measured by ELISA to assess the infection. Injection drug abuse is a significant risk factor for HIV-infection, and opiate drug abusers show increased HIV-neuropathology, even with anti-retroviral treatments. We therefore assessed HIV+sup effects on neuronal survival and neurite growth/pruning with or without concurrent exposure to morphine, an opiate that preferentially acts through µ-opioid receptors. Effects of HIV+sup ± morphine were assessed on neuronal populations, and also by time-lapse imaging of individual cells. HIV+sup caused dose-dependent toxicity over a range of p24 levels (10-500 pg/ml). Significant interactions occurred with morphine at lower p24 levels (10 and 25 pg/ml), and GSK3ß was implicated as a point of convergence. In the presence of glia, selective neurotoxic measures were significantly enhanced and interactions with morphine were also augmented, perhaps related to a decreased level of BDNF. Importantly, the arrest of neurite growth that occurred with exposure to HIV+sup was reversible unless neurons were continuously exposed to morphine. Thus, while reducing HIV-infection levels may be protective, ongoing exposure to opiates may limit recovery. Opiate interactions observed in this HIV-infective environment were similar, though not entirely concordant, with Tat/gp120 interactions reported previously, suggesting unique interactions with virions or other viral or cellular proteins released by infected and/or activated cells.


Assuntos
HIV-1/fisiologia , Morfina/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/virologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Neuritos/metabolismo , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/virologia , Transdução de Sinais/efeitos dos fármacos , Carga Viral/efeitos dos fármacos
8.
AIDS ; 28(10): 1409-19, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24732776

RESUMO

OBJECTIVE: We explored the antiviral therapeutic potential of ibudilast (AV411, MN-166) and its amino analog, AV1013. METHODS: We analyzed whether Ibudilast, a nonselective cyclic AMP phosphodiesterase inhibitor that has been used clinically in Asia for bronchial asthma, poststroke dizziness, and ocular allergies, and AV1013, attenuate HIV-1 replication and the synergistic interactions seen with opiate abuse-HIV-1 comorbidity in neuronal death and inflammation. RESULTS: AV411 and AV1013 inhibited replication by HIV-1 in microglia and significantly suppressed Tat ± morphine-induced tumor necrosis factor-α and MIF production, the activation of the nuclear factor-kappa B subunit p65, and neuronal death. AV411 and AV1013 prevented HIV-1 replication, and attenuated tumor necrosis factor-α and MIF release at concentrations of 100  nmol/l and 1  µmol/l, which are likely achievable at clinical doses. More importantly, co-exposure with morphine did not negate the inhibitory actions of AV411. CONCLUSION: Collectively, our data suggest that AV411 and its amino analog, AV1013, may be useful neuroprotective agents counteracting neurotoxicity caused by infected and activated glia, and implicate them as potential therapies for the management of HIV-associated neurocognitive disorders in an opioid-abusing population.


Assuntos
Antivirais/farmacologia , HIV-1/fisiologia , Morfina/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Piridinas/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Camundongos , Neurônios/fisiologia
9.
J Neurotrauma ; 29(9): 1785-93, 2012 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-22468884

RESUMO

Traumatic brain injury (TBI) induces a cascade of primary and secondary events resulting in impairment of neuronal networks that eventually determines clinical outcome. The dynorphins, endogenous opioid peptides, have been implicated in secondary injury and neurodegeneration in rodent and human brain. To gain insight into the role of dynorphins in the brain's response to trauma, we analyzed short-term (1-day) and long-term (7-day) changes in dynorphin A (Dyn A) levels in the frontal cortex, hippocampus, and striatum, induced by unilateral left-side or right-side cortical TBI in mice. The effects of TBI were significantly different from those of sham surgery (Sham), while the sham surgery also produced noticeable effects. Both sham and TBI induced short-term changes and long-term changes in all three regions. Two types of responses were generally observed. In the hippocampus, Dyn A levels were predominantly altered ipsilateral to the injury. In the striatum and frontal cortex, injury to the right (R) hemisphere affected Dyn A levels to a greater extent than that seen in the left (L) hemisphere. The R-TBI but not L-TBI produced Dyn A changes in the striatum and frontal cortex at 7 days after injury. Effects of the R-side injury were similar in the two hemispheres. In naive animals, Dyn A was symmetrically distributed between the two hemispheres. Thus, trauma may reveal a lateralization in the mechanism mediating the response of Dyn A-expressing neuronal networks in the brain. These networks may differentially mediate effects of left and right brain injury on lateralized brain functions.


Assuntos
Lesões Encefálicas/metabolismo , Dinorfinas/metabolismo , Análise de Variância , Animais , Corpo Estriado/metabolismo , Lateralidade Funcional/fisiologia , Hipocampo/metabolismo , Masculino , Camundongos , Córtex Pré-Frontal/metabolismo , Radioimunoensaio
10.
J Neurovirol ; 17(1): 41-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21234828

RESUMO

To assess the role of the phosphatase and tensin homologue on chromosome 10 (PTEN) in mediating envelope glycoprotein 120 (gp120)-induced neurotoxicity in the striatum, PTEN was silenced using short interfering RNA (siRNA) vectors. PTEN activity directs multiple downstream pathways implicated in gp120-induced neuronal injury and death. PTEN is a negative regulator of Akt (protein kinase B) phosphorylation, but has also been shown to directly activate extrasynaptic NMDA receptors and dephosphorylate focal adhesion kinase. Rodent striatal neurons were nucleofected with green fluorescent protein (GFP)-expressing siRNA constructs to silence PTEN (PTENsi-GFP) or with negative-control (NCsi-GFP) vectors, and exposed to HIV-1 gp120(IIIB) using rigorously controlled, cell culture conditions including computerized time-lapse microscopy to track the fate of individual neurons following gp120 exposure. Immunofluorescence labeling showed that subpopulations of striatal neurons possess CXCR4 and CCR5 co-receptor immunoreactivity and that gp120(IIIB) was intrinsically neurotoxic to isolated striatal neurons. Importantly, PTENsi-GFP, but not control NCsi-GFP, constructs markedly decreased PTEN mRNA and protein levels and significantly attenuated gp120-induced death. These findings implicate PTEN as a critical factor in mediating the direct neurotoxic effects of HIV-1 gp120, and suggest that effectors downstream of PTEN such as Akt or other targets are potentially affected. The selective abatement of PTEN activity in neurons may represent a potential therapeutic strategy for the CNS complications of HIV-1.


Assuntos
Inativação Gênica , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/patogenicidade , Neostriado/citologia , Neurônios/patologia , PTEN Fosfo-Hidrolase/genética , Análise de Variância , Animais , Apoptose , Células Cultivadas , Camundongos , Camundongos Endogâmicos ICR , Neostriado/metabolismo , Neostriado/virologia , Neurônios/citologia , Neurônios/virologia , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo
11.
Neurosci Lett ; 485(3): 233-6, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20849923

RESUMO

Excitotoxicity and/or microglial reactivity might underlie neurologic dysfunction in HIV patients. The HIV regulatory protein Tat is both neurotoxic and pro-inflammatory, suggesting that Tat might participate in the pathogenesis of HIV-associated neurocognitive disorders (HAND). The present study was undertaken to evaluate if Tat can increase extracellular glutamate, and was specifically designed to determine the degree to which, and the mechanisms by which Tat could drive microglial glutamate release. Data show that application of Tat to cultured primary microglia caused dose-dependent increases in extracellular glutamate that were exacerbated by morphine, which is known to worsen Tat cytotoxicity. Tat-induced glutamate release was decreased by inhibitors of p38 and p42/44 MAPK, and by inhibitors of NADPH oxidase and the x(c)(-) cystine-glutamate antiporter. Furthermore, Tat increased expression of the catalytic subunit of x(c)(-) (xCT), but Tat-induced increases in xCT mRNA were not affected by inhibition of NADPH oxidase or x(c)(-) activity. Together, these data describe a specific and biologically significant signaling component of the microglial response to Tat, and suggest that excitotoxic neuropathology associated with HIV infection might originate in part with Tat-induced activation of microglial glutamate release.


Assuntos
Sistema y+ de Transporte de Aminoácidos/metabolismo , Ácido Glutâmico/metabolismo , Microglia/metabolismo , NADPH Oxidases/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia , Sistema y+ de Transporte de Aminoácidos/antagonistas & inibidores , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Animais Recém-Nascidos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Inflamação/patologia , Microglia/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
J Neurochem ; 108(1): 202-15, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19054280

RESUMO

Opiate abuse alters the progression of human immunodeficiency virus and may increase the risk of neuroAIDS. As neuroAIDS is associated with altered microglial reactivity, the combined effects of human immunodeficiency virus-Tat and morphine were determined in cultured microglia. Specifically, experiments determined the effects of Tat and morphine on microglial-free radical production and oxidative stress, and on cytokine release. Data show that combined Tat and morphine cause early and synergistic increases in reactive oxygen species, with concomitant increases in protein oxidation. Furthermore, combined Tat and morphine, but not Tat or morphine alone, cause reversible decreases in proteasome activity. The effects of morphine on free radical production and oxidative stress are prevented by pre-treatment with naloxone, illustrating the important role of opioid receptor activation in these phenomena. While Tat is well known to induce cytokine release from cultured microglia, morphine decreases Tat-induced release of the cytokines tumor necrosis factor-alpha and interleukin-6, as well as the chemokine monocyte chemoattractant protein-1 (MCP-1). Finally, experiments using the reversible proteasome inhibitor MG115 show that temporary, non-cytotoxic decreases in proteasome activity increase protein oxidation and decrease tumor necrosis factor-alpha, interleukin-6, and MCP-1 release from microglia. Taken together, these data suggest that oxidative stress and proteasome inhibition may be involved in the immunomodulatory properties of opioid receptor activation in microglia.


Assuntos
Analgésicos Opioides/farmacologia , Proteínas Reguladoras de Apoptose/farmacologia , Microglia/efeitos dos fármacos , Morfina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática/métodos , Leupeptinas/farmacologia , Camundongos , Inibidores de Proteases/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ratos , Ratos Sprague-Dawley , Survivina
13.
PLoS One ; 3(12): e4093, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19116667

RESUMO

Astroglia are key cellular sites where opiate drug signals converge with the proinflammatory effects of HIV-1 Tat signals to exacerbate HIV encephalitis. Despite this understanding, the molecular sites of convergence driving opiate-accelerated neuropathogenesis have not been deciphered. We therefore explored potential points of interaction between the signaling pathways initiated by HIV-1 Tat and opioids in striatal astrocytes. Profiling studies screening 152 transcription factors indicated that the nuclear factor-kappa B (NF-kappaB) subunit, c-Rel, was a likely candidate for Tat or Tat plus opiate-induced increases in cytokine and chemokine production by astrocytes. Pretreatment with the NF-kappaB inhibitor parthenolide provided evidence that Tat+/-morphine-induced release of MCP-1, IL-6 and TNF-alpha by astrocytes is NF-kappaB dependent. The nuclear export inhibitor, leptomycin B, blocked the nucleocytoplasmic shuttling of NF-kappaB; causing p65 (RelA) accumulation in the nucleus, and significantly attenuated cytokine production in Tat+/-morphine exposed astrocytes. Similarly, chelating intracellular calcium ([Ca(2+)](i)) blocked Tat+/-morphine-evoked MCP-1 and IL-6 release, while artificially increasing the concentration of extracellular Ca(2+) reversed this effect. Taken together, these results demonstrate that: 1) exposure to Tat+/-morphine is sufficient to activate NF-kappaB and cytokine production, 2) the release of MCP-1 and IL-6 by Tat+/-morphine are highly Ca(2+)-dependent, while TNF-alpha appears to be less affected by the changes in [Ca(2+)](i), and 3) in the presence of Tat, exposure to opiates augments Tat-induced NF-kappaB activation and cytokine release through a Ca(2+)-dependent pathway.


Assuntos
Analgésicos Opioides/farmacologia , Astrócitos/imunologia , Cálcio/metabolismo , Citocinas/biossíntese , HIV-1 , Morfina/farmacologia , NF-kappa B/metabolismo , Transcrição Gênica , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Genes rel , Camundongos , Camundongos Endogâmicos ICR , Modelos Biológicos , NF-kappa B/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
14.
J Neurovirol ; 13(2): 97-106, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17505978

RESUMO

Neurons are targets of toxicity induced by the human immunodeficiency virus (HIV)-1 protein Tat (transactivator of transcription). Exposure to Tat increases [Ca(2+)](i) in striatal neurons and activates multiple cell death pathways. In earlier studies the authors showed that Tat activated both caspase-3 and endonuclease-G, a caspase-independent effector of apoptosis, and that Tat-induced neurotoxicity was not attenuated by a caspase-3 inhibitor. Because Tat activates multiple, parallel death pathways, the authors attempted to reduce Tat-induced neurotoxicity by manipulating signaling pathways upstream of mitochondrial apoptotic events. PTEN (phosphatase and tensin homolog deleted on chromosome 10), a negative regulator of Akt/PKB (protein kinase B) phosphorylation, was chosen as a target for silencing. Akt/PKB activity directs multiple downstream pathways mediated by GSK3beta, BAD, forkhead transcription factors, nuclear factor kappa B (NFkappaB), and others, in a manner that promotes proliferation and survival. Striatal neurons were nucleofected with short interfering RNA (siRNA) vectors targeting PTEN, or a negative-control siRNA. Although Tat(1-86) significantly increased the death of neurons transfected with control construct by 72 h, PTEN-silenced neurons were completely protected. These findings indicate that Akt is a critical intermediary in the direct neurotoxicity induced by HIV-1 Tat, and identify Akt regulation as a possible therapeutic strategy for Tat-induced neurotoxicity in HIV encephalitis (HIVE).


Assuntos
Produtos do Gene tat/toxicidade , Inativação Gênica , Infecções por HIV/virologia , HIV-1 , Neurônios/patologia , PTEN Fosfo-Hidrolase/genética , Animais , Apoptose , Células Cultivadas , Corpo Estriado/citologia , Feminino , Infecções por HIV/fisiopatologia , Infecções por HIV/terapia , HIV-1/química , HIV-1/patogenicidade , Humanos , Camundongos , Neurônios/metabolismo , Neurônios/virologia , PTEN Fosfo-Hidrolase/metabolismo , Transfecção , Virulência , Produtos do Gene tat do Vírus da Imunodeficiência Humana
15.
Neurosci Lett ; 412(1): 34-8, 2007 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-17187929

RESUMO

The health effects of cell phone radiation exposure are a growing public concern. This study investigated whether expression of genes related to cell death pathways are dysregulated in primary cultured neurons and astrocytes by exposure to a working Global System for Mobile Communication (GSM) cell phone rated at a frequency of 1900MHz. Primary cultures were exposed to cell phone emissions for 2h. We used array analysis and real-time RT-PCR to show up-regulation of caspase-2, caspase-6 and Asc (apoptosis associated speck-like protein containing a card) gene expression in neurons and astrocytes. Up-regulation occurred in both "on" and "stand-by" modes in neurons, but only in "on" mode in astrocytes. Additionally, astrocytes showed up-regulation of the Bax gene. The effects are specific since up-regulation was not seen for other genes associated with apoptosis, such as caspase-9 in either neurons or astrocytes, or Bax in neurons. The results show that even relatively short-term exposure to cell phone radiofrequency emissions can up-regulate elements of apoptotic pathways in cells derived from the brain, and that neurons appear to be more sensitive to this effect than astrocytes.


Assuntos
Apoptose/efeitos da radiação , Astrócitos/efeitos da radiação , Telefone Celular , Neurônios/efeitos da radiação , Radiação , Regulação para Cima/efeitos da radiação , Animais , Encéfalo/citologia , Caspases/genética , Caspases/metabolismo , Células Cultivadas , Embrião de Mamíferos , Feminino , Camundongos , Camundongos Endogâmicos ICR , Neurônios/citologia , Gravidez , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo
16.
J Neurochem ; 98(5): 1541-50, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16923165

RESUMO

Although short interfering RNA (siRNA)-induced gene silencing can be transmitted between cells in plants and in Caenorhabditis elegans, this phenomenon has been barely studied in mammalian cells. Both immortalized oligodendrocytes and SNB19 glioblastoma cells were transfected with siRNA constructs for phosphatase and tensin homolog deleted on chromosome 10 (PTEN) or Akt/protein kinase B (Akt). Co-cultures were established between silenced cells and non-silenced cells which were hygromycin resistant and/or expressed green fluorescent protein. After fluorescence sorting or hygromycin selection to remove the silenced cells, the expression of PTEN or Akt genes in the originally unsilenced cells was in all cases significantly decreased. Importantly, silencing did not occur in transwell culture studies, suggesting that transmission of the silencing signal requires a close association between cells. These results provide the first direct demonstration that an siRNA-induced silencing signal can be transmitted between mammalian CNS cells.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Transferência Genética Horizontal/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Northern Blotting/métodos , Western Blotting/métodos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Sistema Nervoso Central/citologia , Cromossomos/metabolismo , Técnicas de Cocultura/métodos , Deleção de Genes , Glioblastoma , Proteínas de Fluorescência Verde/metabolismo , Humanos , Oligodendroglia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transfecção/métodos
17.
J Neuroimmunol ; 178(1-2): 9-16, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16831471

RESUMO

To assess the role of CCL2/MCP-1 in opiate drug abuse and HIV-1 comorbidity, the effects of systemic morphine and intrastriatal HIV-1 Tat on macrophage/microglial and astroglial activation were assessed in wild type and CCR2 null mice. Tat and/or morphine additively increased the proportion of CCL2 immunoreactive astroglia. The effects of morphine were prevented by naltrexone. Glial activation was significantly reduced in CCR2-/- versus wild-type mice following Tat or morphine plus Tat exposure. Thus, CCR2 contributes to local glial activation caused by Tat alone or in the presence of opiates, implicating CCR2 signaling in HIV-1 neuropathogenesis in drug abusers and non-abusers.


Assuntos
Produtos do Gene tat/administração & dosagem , Morfina/farmacologia , Entorpecentes/farmacologia , Neuroglia/efeitos dos fármacos , Receptores de Quimiocinas/efeitos dos fármacos , Complexo AIDS Demência/fisiopatologia , Animais , Proteína Glial Fibrilar Ácida/metabolismo , Infecções por HIV/fisiopatologia , HIV-1 , Imuno-Histoquímica , Injeções Intraventriculares , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Neuroglia/metabolismo , Receptores CCR2 , Receptores de Quimiocinas/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/virologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana
18.
Glia ; 50(2): 91-106, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15630704

RESUMO

Recent evidence suggests that injection drug users who abuse heroin are at increased risk of CNS complications from human immunodeficiency virus (HIV) infection. Opiate drugs may intrinsically alter the pathogenesis of HIV by directly modulating immune function and by directly modifying the CNS response to HIV. Despite this, the mechanisms by which opiates increase the neuropathogenesis of HIV are uncertain. In the present study, we describe the effect of morphine and the HIV-1 protein toxin Tat(1-72) on astroglial function in cultures derived from ICR mice. Astroglia maintain the blood-brain barrier and influence inflammatory signaling in the CNS. Astrocytes can express mu-opioid receptors, and are likely targets for abused opiates, which preferentially activate mu-opioid receptors. While Tat alone disrupts astrocyte function, when combined with morphine, Tat causes synergistic increases in [Ca(2+)](i). Moreover, astrocyte cultures treated with morphine and Tat showed exaggerated increases in chemokine release, including monocyte chemoattractant protein-1 (MCP-1) and regulated on activation, normal T cell expressed and secreted (RANTES), as well as interleukin-6 (IL-6). Morphine-Tat interactions were prevented by the mu-opioid receptor antagonist beta-funaltrexamine, or by immunoneutralizing Tat(1-72) or substituting a nontoxic, deletion mutant (Tat(Delta31-61)). Our findings suggest that opiates may increase the vulnerability of the CNS to viral entry (via recruitment of monocytes/macrophages) and ensuing HIV encephalitis by synergistically increasing MCP-1 and RANTES release by astrocytes. The results further suggest that astrocytes are key intermediaries in opiate-HIV interactions and disruptions in astroglial function and inflammatory signaling may contribute to an accelerated neuropathogenesis in HIV-infected individuals who abuse opiates.


Assuntos
Astrócitos/metabolismo , Cálcio/farmacologia , Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Produtos do Gene tat/metabolismo , HIV-1/metabolismo , Interleucina-6/metabolismo , Entorpecentes/farmacologia , Analgésicos Opioides/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Citocinas/metabolismo , Sinergismo Farmacológico , Homeostase/fisiologia , Humanos , Cinética , Camundongos , Camundongos Endogâmicos ICR , Microglia/metabolismo , Morfina/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Opioides mu/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana
19.
Exp Cell Res ; 296(2): 135-44, 2004 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15149844

RESUMO

Oligodendrocytes (OLs) form myelin within the central nervous system and are targets in numerous demyelinating diseases and injuries. OLs grown in culture maintain the developmental timetable which occurs in vivo and mature into cells with a relatively normal phenotype. In this study, cultured cells are used to test whether EGF can modulate process formation in OLs both before and after transection injury. EGF had no effect on the formation of new processes by OLs at any stage of development. To test the effect of EGF on process outgrowth after injury, mature OLs were selected and injured by laser transection of a single process, then imaged at 24-h intervals for 120 h. EGF promoted the recovery and regrowth of injured processes and also significantly increased outgrowth in uninjured processes. As well, it increased the number of new sprouts formed by OLs after injury. Results suggest that the effects of EGF on process outgrowth are a consequence of EGF interaction with a signaling pathway that is specifically activated within injured OLs. The potent effect of EGF on OL process formation after an injury suggests that modulation of the signaling pathways involved might provide a mechanism to promote remyelination.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Oligodendroglia/ultraestrutura , Regeneração/efeitos dos fármacos , Animais , Axônios , Diferenciação Celular , Tamanho Celular , Células Cultivadas , Córtex Cerebral/citologia , Camundongos , Microscopia de Contraste de Fase , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-fyn , Transdução de Sinais , Fatores de Tempo , Ferimentos e Lesões
20.
Glia ; 38(3): 228-39, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11968060

RESUMO

The biologically active lipid metabolite, platelet-activating factor (PAF), is thought to contribute to inflammatory processes and tissue damage in a variety of central nervous system (CNS) injuries. In previous studies, we found that after contusion spinal cord injury, treatment with a PAF antagonist led to significantly increased white matter tissue sparing as well as decreased mRNA levels for pro-inflammatory cytokines. Some studies suggest that PAF can also have toxic effects on neurons in vitro. Few studies, however, have examined the effects of PAF on glial cells of the CNS. In the present study, the potential for PAF to act as a toxin to cultured astrocytes was examined. Also investigated were the effects of PAF on oligodendrocytes at two different stages of development. Treatment with 0.02-2 microM PAF for 72 h resulted in significant levels of cell death in both cell types (P < 0.05), an effect that was blocked by the PAF receptor antagonists, WEB 2170 and BN 52021. To investigate PAF-induced glial cell death further, we looked for activation of the enzyme, caspase-3, which can be indicative of apoptosis. Immunocytochemistry demonstrated that PAF at all concentrations caused activation of caspase-3 at 24, 48, and 72 h after treatment in both cell types. Caspase-3-dependent cell death was further confirmed using knockout mice (-/-) deficient in the caspase-3 gene. Toxicity was lost when astrocytes (-/-) were exposed to 0.02-2 microM PAF (P < 0.01). Oligodendrocytes (-/-) were not susceptible to toxicity at 2 microM PAF (P < 0.001). The results demonstrate that the pro-inflammatory molecule, PAF, induces cell death in cultured CNS glial cells and that this effect is, in part, dependent on caspase-3 activation.


Assuntos
Astrócitos/metabolismo , Caspases/metabolismo , Morte Celular/fisiologia , Sistema Nervoso Central/metabolismo , Diterpenos , Inflamação/metabolismo , Oligodendroglia/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Traumatismos da Medula Espinal/metabolismo , Animais , Animais Recém-Nascidos , Antígenos de Diferenciação/efeitos dos fármacos , Antígenos de Diferenciação/metabolismo , Astrócitos/efeitos dos fármacos , Azepinas/farmacologia , Caspase 3 , Caspases/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Sistema Nervoso Central/fisiopatologia , Relação Dose-Resposta a Droga , Sequestradores de Radicais Livres/farmacologia , Ginkgolídeos , Proteína Glial Fibrilar Ácida/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/fisiopatologia , Lactonas/farmacologia , Camundongos , Camundongos Endogâmicos CBA , Oligodendroglia/efeitos dos fármacos , Fator de Ativação de Plaquetas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Triazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA