Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Sci Rep ; 10(1): 18828, 2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33110185

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32630025

RESUMO

Despite combined antiretroviral therapy (ART) achieving efficient HIV replication control, HIV-associated neurocognitive disorders (HAND) continue to be highly prevalent in HIV-infected patients. Diabetes mellitus (DM) is a well-known comorbidity of HAND in HIV-infected patients. Blood brain barrier (BBB) dysfunction has been linked recently to dementia development, specifically in DM patients. BBB injury exists both in HIV and DM, likely contributing to cognitive decline. However, its extent, exact cellular targets and mechanisms are largely unknown. In this report, we found a decrease in pericyte coverage and expression of tight junction proteins in human brain tissues from HIV patients with DM and evidence of HAND when compared to HIV-infected patients without DM or seronegative DM patients. Using our in vitro BBB models, we demonstrated diminution of barrier integrity, enhanced monocyte adhesion, changes in cytoskeleton and overexpression of adhesion molecules in primary human brain endothelial cells or human brain pericytes after exposure to HIV and DM-relevant stimuli. Our study demonstrates for the first-time evidence of impaired BBB function in HIV-DM patients and shows potential mechanisms leading to it in brain endothelium and pericytes that may result in poorer cognitive performance compared to individuals without HIV and DM.


Assuntos
Arterite do Sistema Nervoso Central Associada a AIDS/metabolismo , Barreira Hematoencefálica/fisiopatologia , Complicações do Diabetes/metabolismo , Pericitos/metabolismo , Arterite do Sistema Nervoso Central Associada a AIDS/fisiopatologia , Citoesqueleto de Actina/metabolismo , Moléculas de Adesão Celular/metabolismo , Demência Vascular/etiologia , Complicações do Diabetes/fisiopatologia , Humanos , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Microvasos/metabolismo , Cultura Primária de Células
3.
Sci Rep ; 10(1): 7274, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32350344

RESUMO

Cognitive impairment is a well-known complication of diabetes mellitus (DM). Microvascular compromise was described one DM complication. Recently we showed blood brain barrier (BBB) permeability and memory loss are associated with diminution of tight junctions (TJ) in brain endothelium and pericyte coverage and inflammation in cerebral microvessels and brain tissue paralleling hyperglycemia in mice of both DM types. The current study demonstrates that exposure of brain microvessels to hyperglycemic conditions or advanced glycation end products (AGEs) ex vivo resulted in significant abnormalities in membranous distribution of TJ proteins. We found significant increase in the amount of extracellular vesicles (EVs) isolated from DM mice and enhanced presence of TJ proteins, occludin and claudin-5, on EVs. Exposure of BMVECs to high glucose and AGEs led to significant augmentation of ICAM and VCAM expression, elevated leukocyte adhesion to and migration across BMVEC monolayers, and increased BBB permeability in vitro. Pericytes exposed to hyperglycemia and AGEs displayed diminished expression of integrin α1, PDGF-R1ß and connexin-43. Our findings indicate BBB compromise in DM ex vivo, in vitro and in vivo models in association with BMVEC/pericyte dysfunction and inflammation. Prevention of BBB injury may be a new therapeutic approach to avert cognitive demise in DM.


Assuntos
Barreira Hematoencefálica/metabolismo , Claudina-5/metabolismo , Vesículas Extracelulares/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Hiperglicemia/metabolismo , Ocludina/biossíntese , Ocludina/metabolismo , Animais , Barreira Hematoencefálica/patologia , Vesículas Extracelulares/patologia , Regulação da Expressão Gênica , Hiperglicemia/patologia , Masculino , Camundongos , Pericitos/metabolismo , Pericitos/patologia
4.
Brain Behav Immun ; 87: 543-555, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32017988

RESUMO

Stroke is a debilitating disease, accounting for almost 20% of all hospital visits, and 8% of all fatalities in the United States in 2017. Following an ischemic attack, inflammatory processes originating from endothelial cells within the brain microvasculature can induce many toxic effects into the impacted area, from both sides of the blood brain barrier (BBB). In addition to increased BBB permeability, impacted brain microvascular endothelial cells can recruit macrophages and other immune cells from the periphery and can also trigger the activation of microglia and astrocytes within the brain. We have identified a key microRNA, let-7g, which levels were drastically diminished as consequence of transient middle cerebral artery occlusion (tMCAO) in vivo and oxygen-glucose deprivation (OGD) in vitro ischemia/reperfusion conditions, respectively. We have observed that let-7g* liposome-based delivery is capable of attenuating inflammation after stroke, reducing BBB permeability, limiting brain infiltration by CD3+CD4+ T-cells and Ly6G+ neutrophils, lessening microglia activation and neuronal death. These effects consequently improved clinical outcomes, shown by mitigating post-stroke gait asymmetry and extremity motor function. Due to the role of the endothelium in propagating the effects of stroke and other inflammation, treatments which can reduce endothelial inflammation and limit ischemic damage and improving recovery after a stroke are required. Our findings demonstrate a critical link between the CNS inflammation and the immune system reaction and lay important groundwork for future stroke pharmacotherapies.


Assuntos
Isquemia Encefálica , Acidente Vascular Cerebral , Animais , Barreira Hematoencefálica , Células Endoteliais , Infarto da Artéria Cerebral Média , Camundongos , Reperfusão
5.
J Cereb Blood Flow Metab ; 40(10): 1953-1965, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31601141

RESUMO

Most neurological diseases, including stroke, lead to some degree of blood-brain barrier (BBB) dysfunction. A significant portion of BBB injury is caused by inflammation, due to pro-inflammatory factors produced in the brain, and by leukocyte engagement of the brain endothelium. Recently, microRNAs (miRNAs) have appeared as major regulators of inflammation-induced changes to gene expression in the microvascular endothelial cells (BMVEC) that comprise the BBB. However, miRNAs' role during cerebral ischemia/reperfusion is still underexplored. Endothelial levels of miR-98 were significantly altered following ischemia/reperfusion insults, both in vivo and in vitro, transient middle cerebral artery occlusion (tMCAO), and oxygen-glucose deprivation (OGD), respectively. Overexpression of miR-98 reduced the mouse's infarct size after tMCAO. Further, miR-98 lessened infiltration of proinflammatory Ly6CHI leukocytes into the brain following stroke and diminished the prevalence of M1 (activated) microglia within the impacted area. miR-98 attenuated BBB permeability, as demonstrated by changes to fluorescently-labeled dextran penetration in vivo and improved transendothelial electrical resistance (TEER) in vitro. Treatment with miR-98 improved significantly the locomotor impairment. Our study provides identification and functional assessment of miRNAs in brain endothelium and lays the groundwork for improving therapeutic approaches for patients suffering from ischemic attacks.


Assuntos
Barreira Hematoencefálica , Endotélio Vascular , MicroRNAs/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Acidente Vascular Cerebral/prevenção & controle , Animais , Impedância Elétrica , Encefalite/patologia , Glucose/deficiência , Infarto da Artéria Cerebral Média/patologia , Leucócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Microglia/patologia , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/etiologia , Traumatismo por Reperfusão/genética , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/genética , Transfecção
6.
J Neuroinflammation ; 15(1): 25, 2018 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-29373982

RESUMO

BACKGROUND: Secoisolariciresinol diglucoside (SDG), the main lignan in flaxseed, is known for its beneficial effects in inflammation, oxidative stress, heart disease, tumor progression, atherosclerosis, and diabetes. SDG might be an attractive natural compound that protects against neuroinflammation. Yet, there are no comprehensive studies to date investigating the effects of SDG on brain endothelium using relevant in vivo and in vitro models. METHODS: We evaluated the effects of orally administered SDG on neuroinflammatory responses using in vivo imaging of the brain microvasculature during systemic inflammation and aseptic encephalitis. In parallel, the anti-inflammatory actions of SDG on brain endothelium and monocytes were evaluated in vitro blood-brain barrier (BBB) model. Multiple group comparisons were performed by one-way analysis of variance with Dunnet's post hoc tests. RESULTS: We found that SDG diminished leukocyte adhesion to and migration across the BBB in vivo in the setting of aseptic encephalitis (intracerebral TNFα injection) and prevented enhanced BBB permeability during systemic inflammatory response (LPS injection). In vitro SDG pretreatment of primary human brain microvascular endothelial cells (BMVEC) or human monocytes diminished adhesion and migration of monocytes across brain endothelial monolayers in conditions mimicking CNS inflammatory responses. Consistent with our in vivo observations, SDG decreased expression of the adhesion molecule, VCAM1, induced by TNFα, or IL-1ß in BMVEC. SDG diminished expression of the active form of VLA-4 integrin (promoting leukocyte adhesion and migration) and prevented the cytoskeleton changes in primary human monocytes activated by relevant inflammatory stimuli. CONCLUSION: This study indicates that SDG directly inhibits BBB interactions with inflammatory cells and reduces the inflammatory state of leukocytes. Though more work is needed to determine the mechanism by which SDG mediates these effects, the ability of SDG to exert a multi-functional response reducing oxidative stress, inflammation, and BBB permeability makes it an exciting potential therapeutic for neuroinflammatory diseases. SDG can serve as an anti-inflammatory and barrier-protective agent in neuroinflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Butileno Glicóis/farmacologia , Endotélio Vascular/efeitos dos fármacos , Glucosídeos/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Microvasos/efeitos dos fármacos , Animais , Barreira Hematoencefálica/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Microvasos/metabolismo
7.
J Neuroinflammation ; 13(1): 254, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27677851

RESUMO

BACKGROUND: Blood-brain barrier (BBB) dysfunction/disruption followed by leukocyte infiltration into the brain causes neuroinflammation and contributes to morbidity in multiple sclerosis, encephalitis, traumatic brain injury, and stroke. The identification of pathways that decreases the inflammatory potential of leukocytes would prevent such injury. Poly(ADP-ribose) polymerase 1 (PARP) controls various genes via its interaction with myriad transcription factors. Selective PARP inhibitors have appeared lately as potent anti-inflammatory tools. Their effects are outside the recognized PARP functions in DNA repair and transcriptional regulation. In this study, we explored the idea that selective inhibition of PARP in leukocytes would diminish their engagement of the brain endothelium. METHODS: Cerebral vascular changes and leukocyte-endothelium interactions were surveyed by intravital videomicroscopy utilizing a novel in vivo model of localized aseptic meningitis when TNFα was introduced intracerebrally in wild-type (PARP+/+) and PARP-deficient (PARP-/-) mice. The effects of selective PARP inhibition on primary human monocytes ability to adhere to or migrate across the BBB were also tested in vitro, employing primary human brain microvascular endothelial cells (BMVEC) as an in vitro model of the BBB. RESULTS: PARP suppression in monocytes diminished their adhesion to and migration across BBB in vitro models and prevented barrier injury. In monocytes, PARP inactivation decreased conformational activation of integrins that plays a key role in their tissue infiltration. Such changes were mediated by suppression of activation of small Rho GTPases and cytoskeletal rearrangements in monocytes. In vitro observations were confirmed in vivo showing diminished leukocyte-endothelial interaction after selective PARP suppression in leukocytes accompanied by BBB protection. PARP knockout animals demonstrated a substantial diminution of inflammatory responses in brain microvasculature and a decrease in BBB permeability. CONCLUSIONS: These results suggest PARP inhibition in leukocytes as a novel approach to BBB protection in the setting of endothelial dysfunction caused by inflammation-induced leukocyte engagement.

8.
J Cereb Blood Flow Metab ; 36(4): 794-807, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26661157

RESUMO

Brain pericytes are uniquely positioned within the neurovascular unit to provide support to blood brain barrier (BBB) maintenance. Neurologic conditions, such as HIV-1-associated neurocognitive disorder, are associated with BBB compromise due to chronic inflammation. Little is known about pericyte dysfunction during HIV-1 infection. We found decreased expression of pericyte markers in human brains from HIV-1-infected patients (even those on antiretroviral therapy). Using primary human brain pericytes, we assessed expression of pericyte markers (α1-integrin, α-smooth muscle actin, platelet-derived growth factor-B receptor ß, CX-43) and found their downregulation after treatment with tumor necrosis factor-α (TNFα) or interleukin-1 ß (IL-1ß). Pericyte exposure to virus or cytokines resulted in decreased secretion of factors promoting BBB formation (angiopoietin-1, transforming growth factor-ß1) and mRNA for basement membrane components. TNFα and IL-1ß enhanced expression of adhesion molecules in pericytes paralleling increased monocyte adhesion to pericytes. Monocyte migration across BBB models composed of human brain endothelial cells and pericytes demonstrated a diminished rate in baseline migration compared to constructs composed only of brain endothelial cells. However, exposure to the relevant chemokine, CCL2, enhanced the magnitude of monocyte migration when compared to BBB models composed of brain endothelial cells only. These data suggest an important role of pericytes in BBB regulation in neuroinflammation.


Assuntos
Encéfalo/patologia , Neurite (Inflamação)/patologia , Pericitos/patologia , Adulto , Membrana Basal/metabolismo , Barreira Hematoencefálica/patologia , Adesão Celular , Quimiocinas/metabolismo , Doença Crônica , Citocinas/biossíntese , Células Endoteliais/patologia , Feminino , Infecções por HIV/patologia , HIV-1 , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/patologia , Cultura Primária de Células , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
9.
Front Microbiol ; 6: 878, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26379653

RESUMO

Multifactorial mechanisms comprising countless cellular factors and virus-encoded transactivators regulate the transcription of HIV-1 (HIV). Since poly(ADP-ribose) polymerase 1 (PARP-1) regulates numerous genes through its interaction with various transcription factors, inhibition of PARP-1 has surfaced recently as a powerful anti-inflammatory tool. We suggest a novel tactic to diminish HIV replication via PARP-1 inhibition in an in vitro model system, exploiting human primary monocyte-derived macrophages (MDM). PARP-1 inhibition was capable to lessen HIV replication in MDM by 60-80% after 7 days infection. Tat, tumor necrosis factor α (TNFα), and phorbol 12-myristate 13-acetate (PMA) are known triggers of the Long Terminal Repeat (LTR), which can switch virus replication. Tat overexpression in MDM transfected with an LTR reporter plasmid resulted in a 4.2-fold increase in LTR activation; PARP inhibition caused 70% reduction of LTR activity. LTR activity, which increased 3-fold after PMA or TNFα treatment, was reduced by PARP inhibition (by 85-95%). PARP inhibition in MDM exhibited 90% diminution in NFκB activity (known to mediate TNFα- and PMA-induced HIV LTR activation). Cytoskeleton rearrangements are important in effective HIV-1 infection. PARP inactivation reduced actin cytoskeleton rearrangements by affecting Rho GTPase machinery. These discoveries suggest that inactivation of PARP suppresses HIV replication in MDM by via attenuation of LTR activation, NFκB suppression and its effects on the cytoskeleton. PARP appears to be essential for HIV replication and its inhibition may provide an effective approach to management of HIV infection.

10.
J Cereb Blood Flow Metab ; 35(12): 1957-65, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26126865

RESUMO

Pathologic conditions in the central nervous system, regardless of the underlying injury mechanism, show a certain level of blood-brain barrier (BBB) impairment. Endothelial dysfunction is the earliest event in the initiation of vascular damage caused by inflammation due to stroke, atherosclerosis, trauma, or brain infections. Recently, microRNAs (miRNAs) have emerged as a class of gene expression regulators. The relationship between neuroinflammation and miRNA expression in brain endothelium remains unexplored. Previously, we showed the BBB-protective and anti-inflammatory effects of glycogen synthase kinase (GSK) 3ß inhibition in brain endothelium in in vitro and in vivo models of neuroinflammation. Using microarray screening, we identified miRNAs induced in primary human brain microvascular endothelial cells after exposure to the pro-inflammatory cytokine, tumor necrosis factor-α, with/out GSK3ß inhibition. Among the highly modified miRNAs, let-7 and miR-98 were predicted to target the inflammatory molecules, CCL2 and CCL5. Overexpression of let-7 and miR-98 in vitro and in vivo resulted in reduced leukocyte adhesion to and migration across endothelium, diminished expression of pro-inflammatory cytokines, and increased BBB tightness, attenuating barrier 'leakiness' in neuroinflammation conditions. For the first time, we showed that miRNAs could be used as a therapeutic tool to prevent the BBB dysfunction in neuroinflammation.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Inflamação/patologia , MicroRNAs/farmacologia , Animais , Capilares/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/efeitos dos fármacos , Quimiocina CCL5/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Transfecção , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/toxicidade
11.
J Neuroimmune Pharmacol ; 10(2): 302-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25666933

RESUMO

Chronic neuroinflammatory disorders (such as HIV associated neurodegeneration) require treatment that decreases production of inflammatory factors by activated microglia and macrophages and protection of blood brain barrier (BBB) injury secondary to activation of brain endothelium. Cannabioid type 2 receptor (CB2) is highly expressed on macrophages and brain microvasular enndothelial cells (BMVEC) and is upregulated in inflammation and HIV infection. It has been shown that CB2 activation dampened inflammatory responses in macrophages and BMVEC. In this study, we assessed by PCR array the expression of a wide range of genes increased in macrophages and BMVEC in inflammation. TNFα treatment upregulated 33 genes in primary human BMVEC, and two highly selective CB2 agonists diminished expression of 31 and 32 genes. These results were confirmed by functional assays (BBB protection after inflammatory insult and decreased migration of monocytes across BMVEC monolayers after CB2 stimulation). Similarly, CB2 stimulation in primary human macrophages led to the suppression of 35 genes out of the 50 genes upregulated by LPS. Such changes in gene expression paralleled diminished secretion of proinflammatory factors. These results indicate the potential utility of CB2 agonists for the treatment of neuroinflammation.


Assuntos
Encéfalo/metabolismo , Endotélio/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Células Cultivadas , Endotélio/efeitos dos fármacos , Endotélio/imunologia , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Receptor CB2 de Canabinoide/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia
12.
J Cereb Blood Flow Metab ; 35(1): 28-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25248836

RESUMO

Blood-brain barrier (BBB) dysfunction seen in neuroinflammation contributes to mortality and morbidity in multiple sclerosis, encephalitis, traumatic brain injury, and stroke. Identification of molecular targets maintaining barrier function is of clinical relevance. We used a novel in vivo model of localized aseptic meningitis where tumor necrosis factor alpha (TNFα) was introduced intracerebrally and surveyed cerebral vascular changes and leukocyte-endothelium interactions by intravital videomicroscopy. Poly(ADP-ribose) polymerase-1 (PARP) inhibition significantly reduced leukocyte adhesion to and migration across brain endothelium in cortical microvessels. PARP inactivation diminished BBB permeability in an in vivo model of systemic inflammation. PARP suppression in primary human brain microvascular endothelial cells (BMVEC), an in vitro model of BBB, enhanced barrier integrity and augmented expression of tight junction proteins. PARP inhibition in BMVEC diminished human monocyte adhesion to TNFα-activated BMVEC (up to 65%) and migration (80-100%) across BBB models. PARP suppression decreased expression of adhesion molecules and decreased activity of GTPases (controlling BBB integrity and monocyte migration across the BBB). PARP inhibitors down-regulated expression of inflammatory genes and dampened secretion of pro-inflammatory factors increased by TNFα in BMVEC. These results point to PARP suppression as a novel approach to BBB protection in the setting of endothelial dysfunction caused by inflammation.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Meningite Asséptica/fisiopatologia , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiologia , Barreira Hematoencefálica/fisiopatologia , Western Blotting , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/imunologia , Células Endoteliais , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Endotélio Vascular/fisiopatologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Humanos , Leucócitos/citologia , Masculino , Meningite Asséptica/imunologia , Meningite Asséptica/metabolismo , Camundongos Endogâmicos C57BL , Microscopia de Vídeo , Poli(ADP-Ribose) Polimerase-1 , Proteínas de Junções Íntimas/metabolismo , Migração Transendotelial e Transepitelial/efeitos dos fármacos
13.
Am J Pathol ; 183(5): 1548-1558, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24055259

RESUMO

Cannabinoid receptor 2 (CB2) is highly expressed in immune cells and stimulation decreases inflammatory responses. We tested the idea that selective CB2 activation in human monocytes suppresses their ability to engage the brain endothelium and migrate across the blood-brain barrier (BBB), preventing consequent injury. Intravital videomicroscopy was used to quantify adhesion of leukocytes to cortical vessels in lipopolysaccharide-induced neuroinflammation, after injection of ex vivo CB2-activated leukocytes into mice; CB2 agonists markedly decreased adhesion of ex vivo labeled cells in vivo. In an in vitro BBB model, CB2 activation in monocytes largely attenuated adhesion to and migration across monolayers of primary human brain microvascular endothelial cells and diminished BBB damage. CB2 stimulation in monocytes down-regulated active forms of integrins, lymphocyte function-associated antigen 1 (LFA-1), and very late antigen 4 (VLA-4). Cells treated with CB2 agonists exhibited increased phosphorylation levels of inhibitory sites of the actin-binding proteins cofilin and VASP, which are upstream regulators of conformational integrin changes. Up-regulated by relevant stimuli, Rac1 and RhoA were suppressed by CB2 agonists in monocytes. CB2 stimulation decreased formation of lamellipodia, which play a key role in monocyte migration. These results indicate that selective CB2 activation in leukocytes decreases key steps in monocyte-BBB engagement, thus suppressing inflammatory leukocyte responses and preventing neuroinflammation.


Assuntos
Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Endotélio/metabolismo , Leucócitos/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Animais , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/patologia , Encefalite/metabolismo , Encefalite/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio/patologia , Humanos , Integrina alfa4beta1/química , Integrina alfa4beta1/metabolismo , Integrina beta1/metabolismo , Lipopolissacarídeos , Antígeno-1 Associado à Função Linfocitária/química , Antígeno-1 Associado à Função Linfocitária/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , Microvasos/patologia , Monócitos/metabolismo , Monócitos/patologia , Fosfoproteínas/metabolismo , Fosforilação , Pseudópodes/metabolismo , Receptor CB2 de Canabinoide/agonistas , Migração Transendotelial e Transepitelial , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
14.
J Leukoc Biol ; 93(5): 801-10, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23463725

RESUMO

Infiltrating monocytes and macrophages play a crucial role in the progression of HIV-1 infection in the CNS. Previous studies showed that activation of the CB2 can attenuate inflammatory responses and affect HIV-1 infectivity in T cells and microglia. Here, we report that CB2 agonists can also act as immunomodulators on HIV-1-infected macrophages. First, our findings indicated the presence of elevated levels of CB2 expression on monocytes/macrophages in perivascular cuffs of postmortem HIV-1 encephalitic cases. In vitro analysis by FACS of primary human monocytes revealed a step-wise increase in CB2 surface expression in monocytes, MDMs, and HIV-1-infected MDMs. We next tested the notion that up-regulation of CB2 may allow for the use of synthetic CB2 agonist to limit HIV-1 infection. Two commercially available CB2 agonists, JWH133 and GP1a, and a resorcinol-based CB2 agonist, O-1966, were evaluated. Results from measurements of HIV-1 RT activity in the culture media of 7 day-infected cells showed a significant decrease in RT activity when the CB2 agonist was present. Furthermore, CB2 activation also partially inhibited the expression of HIV-1 pol. CB2 agonists did not modulate surface expression of CXCR4 or CCR5 detected by FACS. We speculate that these findings indicate that prevention of viral entry is not a central mechanism for CB2-mediated suppression in viral replication. However, CB2 may affect the HIV-1 replication machinery. Results from a single-round infection with the pseudotyped virus revealed a marked decrease in HIV-1 LTR activation by the CB2 ligands. Together, these results indicate that CB2 may offer a means to limit HIV-1 infection in macrophages.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , HIV-1/efeitos dos fármacos , Macrófagos/virologia , Receptor CB2 de Canabinoide/agonistas , Replicação Viral/efeitos dos fármacos , Anisóis/farmacologia , Canabinoides/farmacologia , Células Cultivadas , Cicloexanóis , Repetição Terminal Longa de HIV/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Indenos/farmacologia , Pirazóis/farmacologia , Receptor CB2 de Canabinoide/análise , Receptor CB2 de Canabinoide/fisiologia , Receptores CCR5/análise , Receptores CXCR4/análise
15.
PLoS One ; 8(2): e55972, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23418486

RESUMO

Neuroinflammatory conditions often involve dysfunction of the Blood-Brain Barrier (BBB). Therefore, identifying molecular targets that can maintain barrier fidelity is of clinical importance. We have previously reported on the anti-inflammatory effects that glycogen synthase kinase 3ß (GSK3ß) inhibition has on primary human brain endothelial cells. Here we show that GSK3ß inhibitors also promote barrier tightness by affecting tight junction (TJ) protein stability. Transendothelial electrical resistance (TEER) was used to evaluate barrier integrity with both pharmacological inhibitors and mutants of GSK3ß. Inhibition of GSK3ß produced a gradual and sustained increase in TEER (as much as 22% over baseline). Analysis of subcellular membrane fractions revealed an increase in the amount of essential tight junction proteins, occludin and claudin-5, but not claudin-3. This phenomenon was attributed to a decrease in TJ protein turnover and not transcriptional regulation. Using a novel cell-based assay, inactivation of GSK3ß significantly increased the half-life of occludin and claudin-5 by 32% and 43%, respectively. A correlation was also established between the enhanced association of ß-catenin with ZO-1 as a function of GSK3ß inhibition. Collectively, our findings suggest the possibility of using GSK3ß inhibitors as a means to extend the half-life of key tight junction proteins to promote re-sealing of the BBB during neuroinflammation.


Assuntos
Barreira Hematoencefálica/metabolismo , Claudina-5/metabolismo , Células Endoteliais/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Ocludina/metabolismo , Junções Íntimas/metabolismo , Benzazepinas/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Impedância Elétrica , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Indóis/farmacologia , Cloreto de Lítio/farmacologia , Junções Íntimas/efeitos dos fármacos , Proteína da Zônula de Oclusão-1/metabolismo
16.
AIDS Res Hum Retroviruses ; 23(1): 123-34, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17263642

RESUMO

Two major interferon (IFN)-mediated antiviral defense enzymes are double-stranded (ds)RNA-dependent 2',5'-oligoadenylate (2-5A) synthetase (2-5OAS) and p68 kinase (PKR). When activated by dsRNA, 2-5OAS synthesizes 2-5A, which binds to and activates RNase L. Activated RNase L hydrolyzes single-stranded viral RNA, thereby inhibiting viral protein synthesis. HIV-1 inhibits the IFN-mediated intracellular antiviral pathways. We have reported the synthesis and characterization of a nuclease-resistant 2-5A agonist (2-5A(N6B)) that overcomes the HIV-1 induced blockades by restoring the 2-5OAS/RNase L antiviral pathway (Homan JW, et al., J Acquir Immune Defic Syndr 2002;30:9-20). The objective of this study was to test the effect of 2-5A(N6B) on chronically infected CD4(+) T lymphocytes and CD14(+) monocytes derived from HIV-1-seropositive individuals. Wild-type HIV-1 replication was effectively inhibited by 2-5A(N6B) in CD4(+) T lymphocytes and CD14(+) monocytes purified from HIV-1 seropositive individuals (n = 18) compared to untreated cells. We also assessed the cytotoxicity of 2-5A(N6B) and report that 2-5A(N6B) exerts its anti-HIV-1 activity with no evidence of cytotoxicity (IC(90) > 100,000 nM). Furthermore, 2-5A(N6B) did not alter the cellular RNA profile, affect CCR5 or CXCR4 coreceptor expression, or activate caspase-dependent apoptosis. Evidence is also provided to show that 2-5A(N6B), and naturally occurring 2-5A(4), act as ligands to activate human Toll-like receptor 4. These results indicate that the 2-5A agonist 2-5A(N6B) has the potential to enhance host cell innate and acquired immune defense mechanisms against HIV-1 infection.


Assuntos
Nucleotídeos de Adenina/farmacologia , Fármacos Anti-HIV/farmacologia , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/metabolismo , HIV-1/efeitos dos fármacos , Receptores de Lipopolissacarídeos , Oligorribonucleotídeos/farmacologia , Nucleotídeos de Adenina/agonistas , Nucleotídeos de Adenina/síntese química , Adulto , Idoso , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Infecções por HIV/tratamento farmacológico , Soropositividade para HIV , HIV-1/fisiologia , Humanos , Fatores Imunológicos/farmacologia , Masculino , Pessoa de Meia-Idade , Monócitos/citologia , Monócitos/imunologia , Oligorribonucleotídeos/agonistas , Oligorribonucleotídeos/síntese química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 4 Toll-Like/metabolismo , Replicação Viral/efeitos dos fármacos
17.
J Interferon Cytokine Res ; 25(6): 345-60, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15957958

RESUMO

Previous studies from this laboratory evaluated the role of p68 kinase (PKR) in the control of HIV-1 replication via retrovirus-mediated gene transfer. PKR was studied because it is a key component of the interferon (IFN)-associated innate antiviral defense pathway in mammalian cells. In this study, CD34(+) hematopoietic stem cells (HSC) were transduced with an HIV-1-based lentiviral vector encoding the PKR transgene (pHIV-PIB) and cultured under conditions that support in vitro differentiation. With high-titer pseudotyped vector stocks, the histogram suggests 100% transduction of the HSC because the cells were blasticidin resistant. Analysis of transduced cells by hybridization revealed an average proviral vector copy number of 1.8 and 2.1 copies of vector sequence per cell. Increased PKR expression and activity (phosphorylation of eukaryotic initiation factor 2alpha [eIF2alpha]) were demonstrated in PKR-transduced, differentiated HSC. There was minimal reduction in cell viability and no induction of apoptosis after transduction of PKR. HSC transduced with the pHIV-PIB lentiviral vector demonstrated normal differentiation into CD34-derived T cell progeny. Two days after HIV-1 infection, lentivirus-mediated transduction of PKR inhibited HIV-1 replication by 72% in T cell progeny compared with cells transduced with the empty vector control (pHIV-IB). By days 5 and 7 post-HIV-1 infection, the surviving PKR-transduced cells were protected from HIV-1 infection, as evidenced by a decrease in p24 antigen expression of at least two orders of magnitude. Our results demonstrate that PKR can be effectively delivered to HSC by a lentiviral vector and can protect CD34-derived T cell progeny from HIV-1 infection. These results provide support for application of the innate antiviral defense pathway in a gene therapy setting to the treatment of HIV-1 infection.


Assuntos
Antígenos CD34 , Infecções por HIV/genética , HIV-1/genética , Células-Tronco Hematopoéticas/fisiologia , Transdução Genética , Replicação Viral/genética , eIF-2 Quinase/genética , Antígenos CD34/metabolismo , Linfócitos T CD4-Positivos/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Vetores Genéticos , Infecções por HIV/metabolismo , Células-Tronco Hematopoéticas/virologia , Humanos , Subpopulações de Linfócitos T/fisiologia , Replicação Viral/fisiologia
18.
J Acquir Immune Defic Syndr ; 30(1): 9-20, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12048358

RESUMO

Opioids potentiate HIV-1 infection in vitro at least partly by suppressing immunoresponsive processes in human lymphocytes and monocytes. For example, it appears that morphine inhibits the interferon (IFN)-alpha, -beta, and -gamma-mediated natural antiviral defense pathways in human peripheral blood mononuclear cells (PBMC). In this study, we show that restoration of a key component of the antiviral pathway reverses morphine-potentiated HIV-1 infection of human PBMC. The data show that HIV-1 replication is potentiated and RNase L activity is inhibited after morphine administration. Because HIV-1 inhibits the antiviral pathway at the level of 2',5'-oligoadenylate (2-5A) synthetase and p68 kinase, antiviral enzymes that require double-stranded RNA, we overcame this blockade by the addition of the nuclease-resistant, nontoxic 2-5A agonist, 2-5A(N6B), to PBMC in culture. Addition of 2-5A(N6B), but not zidovudine or saquinavir, to morphine-treated PBMC completely reversed the morphine-induced potentiation of HIV-1 infection. Further, 2-5A(N6B) significantly enhanced expression of both IFN-alpha and IFN-gamma. Also, increased expression of IFN-gamma was associated with a significant increase in expression of RANTES and monocyte chemotactic protein (MCP)-1, chemokines that may inhibit HIV-1 infection by blocking viral attachment to CCR2 and CCR5 co-receptors. Our results suggest that reactivation of the antiviral pathway by 2-5A agonists may be useful to inhibit opioid-potentiated HIV-1 replication.


Assuntos
Nucleotídeos de Adenina/farmacologia , Antivirais/farmacologia , HIV-1/efeitos dos fármacos , Leucócitos Mononucleares/virologia , Morfina/farmacologia , Entorpecentes/farmacologia , Oligorribonucleotídeos/farmacologia , Replicação Viral/efeitos dos fármacos , Nucleotídeos de Adenina/agonistas , Nucleotídeos de Adenina/síntese química , Células Cultivadas , Quimiocina CCL2/análise , Quimiocina CCL2/biossíntese , Quimiocina CCL5/análise , Quimiocina CCL5/biossíntese , Endorribonucleases/biossíntese , Endorribonucleases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , HIV-1/fisiologia , Humanos , Interferon-alfa/análise , Interferon-alfa/biossíntese , Interferon gama/análise , Interferon gama/biossíntese , Leucócitos Mononucleares/efeitos dos fármacos , Morfina/antagonistas & inibidores , Oligorribonucleotídeos/agonistas , Oligorribonucleotídeos/síntese química , Inibidores da Síntese de Proteínas/agonistas , Inibidores da Transcriptase Reversa/farmacologia , Saquinavir/farmacologia , Zidovudina/farmacologia
19.
J Interferon Cytokine Res ; 22(4): 443-56, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12034027

RESUMO

A 2',5'-oligoadenylate (2-5A)-dependent 37-kDa form of RNase L has been reported in extracts of peripheral blood mononuclear cells (PBMC) from individuals with chronic fatigue syndrome (CFS). In the current study, analytic gel permeation FPLC, azido photoaffinity labeling, two-dimensional (2-D) gel electrophoresis, and matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) have been used to examine the biochemical relationship between the 80-kDa RNase L in healthy control PBMC and the 37-kDa RNase L in PBMC from individuals with CFS. Like the 80-kDa RNase L, the 37-kDa RNase L is present as a catalytically inactive heterodimer complex with the RNase L inhibitor (RLI). Formation of a 37-kDa RNase L-RLI complex indicates that the 37-kDa RNase L is structurally similar to the 80-kDa RNase L at the N-terminus, which contains the 2-5A binding domain. The enzymatically active monomer form of 37-kDa RNase L resolved by 2-D gel electrophoresis has a pI of 6.1. RT-PCR and Southern blot analyses demonstrated that the 37-kDa RNase L is not formed by alternative splicing. In-gel tryptic digestion of the 37-kDa RNase L that was excised from 2-D gels and subsequent MALDI-MS analysis identified three peptide masses that are identical to three predicted peptide masses in the 80-kDa RNase L. The electrophoretic mobility of 2-5A azido photolabeled/immunoprecipitated 37-kDa RNase L was the same under reducing and nonreducing conditions. The results presented show that the 37-kDa form of RNase L in PBMC shares structural and functional features with the native 80-kDa RNase L, in particular in the 2-5A binding and catalytic domains.


Assuntos
Adenosina/análogos & derivados , Endorribonucleases/química , Endorribonucleases/fisiologia , Síndrome de Fadiga Crônica/enzimologia , Adenosina/química , Marcadores de Afinidade/química , Azidas/química , Extratos Celulares/análise , Células Cultivadas , Cromatografia em Gel , Dissulfetos/metabolismo , Eletroforese em Gel Bidimensional , Endorribonucleases/genética , Humanos , Células K562 , Leucócitos Mononucleares/enzimologia , Peso Molecular , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA