Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurovirol ; 26(6): 838-845, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32901392

RESUMO

Despite combined antiretroviral therapy (cART), HIV infection in the CNS persists with reported increases in activation of macrophages (MΦ), microglia, and surrounding astrocytes/neurons, conferring HIV-induced inflammation. Chronic inflammation results in HIV-associated neurocognitive disorders (HAND) with reported occurrence of up to half of individuals with HIV infection. The existing HAND mouse model used by laboratories including ours, and the effect of novel agents on its pathology present with labor-intensive and time-consuming limitations since brain sections and immunohistochemistry assays have to be performed and analyzed. A novel flow cytometry-based system to objectively quantify phenotypic effects of HIV using a SCID mouse HAND model was developed which demonstrated that the HIV-infected mice had significant increases in astrogliosis, loss of neuronal dendritic marker, activation of murine microglia, and human macrophage explants compared to uninfected control mice. HIV p24 could also be quantified in the brains of the infected mice. Correlation of these impairments with HIV-induced brain inflammation and previous behavioral abnormalities studies in mice suggests that this model can be used as a fast and relevant throughput methodology to quantify preclinical testing of novel treatments for HAND.


Assuntos
Encéfalo/metabolismo , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Gliose/genética , Infecções por HIV/genética , HIV-1/genética , Animais , Astrócitos/metabolismo , Astrócitos/virologia , Biomarcadores/metabolismo , Encéfalo/virologia , Disfunção Cognitiva/complicações , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/virologia , Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/complicações , Gliose/metabolismo , Gliose/virologia , Proteína do Núcleo p24 do HIV/genética , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Inflamação , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Masculino , Camundongos , Camundongos SCID , Microglia/metabolismo , Microglia/virologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Fenótipo
2.
J Neuroinflammation ; 16(1): 182, 2019 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-31561750

RESUMO

BACKGROUND: Since HIV-associated neurocognitive disorders (HANDs) occur in up to half of HIV-positive individuals, even with combined antiretroviral therapy (cART), adjunctive therapies are needed. Chronic CNS inflammation contributes to HAND and HIV encephalitis (HIVE). Baricitinib is a JAK 1/2 inhibitor approved in the USA, EU, and Japan for rheumatoid arthritis, demonstrating potent inhibition of IL-6, D-dimer, CRP, TNF-α, IFN-α/ß, and other pro-inflammatory cytokines. METHODS: Our modified murine HAND model was used to evaluate the ability of baricitinib to cross the blood-brain barrier (BBB) and modulate monocyte/macrophage-driven HAND. Severity of HAND was measured by assessing cognitive performance of low- and high-dose baricitinib treated versus untreated HAND mice. The severity of brain neuroinflammation was evaluated in these mouse groups after flow cytometric analyses. We also assessed the ability of baricitinib to block events in myeloid and lymphoid cells in vitro that may undergird the persistence of HIV in the central nervous system (CNS) in primary human macrophages (Mϕ) and lymphocytes including HIV replication, HIV-induced activation, reservoir expansion, and reservoir maintenance. RESULTS: In vivo, both doses of 10 and 50 mg/kg qd baricitinib crossed the BBB and reversed behavioral abnormalities conferred by HIV infection. Moreover, baricitinib significantly reduced HIV-induced neuroinflammation marked by glial activation: activated microglia (MHCII+/CD45+) and astrogliosis (GFAP). Baricitinib also significantly reduced the percentage of p24+ human macrophages in mouse brains (p < 0.05 versus HAND mice; t test). In vitro, baricitinib significantly reduced markers of persistence, reservoir size, and reseeding in Mϕ. CONCLUSION: These results show that blocking the JAK/STAT pathway reverses cognitive deficits and curtails inflammatory markers in HAND in mice. Our group recently reported safety and tolerability of ruxolitinib in HIV-infected individuals (Marconi et al., Safety, tolerability and immunologic activity of ruxolitinib added to suppressive ART, 2019), underscoring potential safety and utility of JAK inhibitors for additional human trials. The data reported herein coupled with our recent human trial with JAK inhibitors provide compelling preclinical data and impetus for considering a trial of baricitinib in HAND individuals treated with cART to reverse cognitive deficits and key events driving viral persistence.


Assuntos
Complexo AIDS Demência/patologia , Complexo AIDS Demência/virologia , Azetidinas/farmacologia , Sulfonamidas/farmacologia , Ativação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos SCID , Purinas , Pirazóis , Latência Viral/efeitos dos fármacos
3.
AIDS ; 32(11): 1403-1411, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29683852

RESUMO

OBJECTIVE: Increased brain interferon-alpha (IFNα) is associated with neurodegenerative disorders, including HIV-associated neurocognitive disorders (HAND). HAND occurs in approximately 50% of individuals with HIV despite combined antiretroviral therapy (cART). Therefore, adjunctive therapies must be developed that prevent progression of mild forms of HAND to HIV-associated dementia. Increased IFNα in the CNS has been associated with HAND in patients and in a HAND mouse model. DESIGN AND METHODS: B18R binds IFNα and ameliorates HAND mouse brain histopathology (HIV encephalitis). The HAND model was used to determine if B18R with cART is superior to cART. Behavioral testing [Object recognition Test (ORT)] was used to show that B18R can reverse behavioral deficits. Rat neuronal cultures were used to investigate mechanisms of IFNα neurotoxicity. RESULTS: Mouse brain immunohistochemistry and densitometry suggests that B18R with a common cART regimen improve histopathological markers better than cART alone. B18R reverses ORT behavioral abnormalities in HAND mice. IFNα-treated rat neurons show decreases in PSD-95, suggesting that dendritic spine architecture is disrupted. Decreases in Arf1, a GTP-binding protein, and AMPA receptors on the surface of rat neurons exposed to IFNα suggest the mechanism of IFNα neurotoxicity may relate to decreased Arf1 resulting in destabilization of dendritic spines, decreased PSD-95 expression, and internalization of AMPA receptors. CONCLUSION: B18R reversal of HAND in the mouse model is further evidence that the treatment of IFNα in individuals with HAND could be a viable adjunctive treatment. Investigating pathways of IFNα neurotoxicity may lead to more specific treatments.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Antirretrovirais/administração & dosagem , Fatores Imunológicos/administração & dosagem , Interferon-alfa/toxicidade , Neurônios/patologia , Proteínas Virais/administração & dosagem , Complexo AIDS Demência/patologia , Animais , Imuno-Histoquímica , Interferon-alfa/antagonistas & inibidores , Masculino , Camundongos , Ratos Sprague-Dawley , Resultado do Tratamento
4.
Neurobiol Dis ; 92(Pt B): 137-43, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26851503

RESUMO

A hallmark of persistent HIV-1 infection in the central nervous system is increased activation of mononuclear phagocytes and surrounding astrogliosis, conferring persistent HIV-induced inflammation. This inflammation is believed to result in neuronal dysfunction and the clinical manifestations of HIV-associated neurocognitive disorders (HAND). The Jak/STAT pathway is activated in macrophages/myeloid cells upon HIV-1 infection, modulating many pro-inflammatory pathways that result in HAND, thereby representing an attractive cellular target. Thus, the impact of ruxolitinib, a Janus Kinase (Jak) 1/2 inhibitor that is FDA approved for myelofibrosis and polycythemia vera, was assessed for its potential to inhibit HIV-1 replication in macrophages and HIV-induced activation in monocytes/macrophages in culture. In addition, a murine model of HIV encephalitis (HIVE) was used to assess the impact of ruxolitinib on histopathological features of HIVE, brain viral load, as well as its ability to penetrate the blood-brain-barrier (BBB). Ruxolitinib was found to inhibit HIV-1 replication in macrophages, HIV-induced activation of monocytes (CD14/CD16) and macrophages (HLA-DR, CCR5, and CD163) without apparent toxicity. In vivo, systemically administered ruxolitinib was detected in the brain during HIVE in SCID mice and markedly inhibited astrogliosis. Together, these data indicate that ruxolitinib reduces HIV-induced activation and infiltration of monocytes/macrophages in vitro, reduces the replication of HIV in vitro, penetrates the BBB when systemically administered in mice and reduces astrogliosis in the brains of mice with HIVE. These data suggest that ruxolitinib will be useful as a novel therapeutic to treat humans with HAND.


Assuntos
Antivirais/farmacologia , Encefalite Viral/tratamento farmacológico , Infecções por HIV/tratamento farmacológico , Inibidores de Janus Quinases/farmacologia , Pirazóis/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Astrócitos/virologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/virologia , Modelos Animais de Doenças , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Gliose/tratamento farmacológico , Gliose/metabolismo , Gliose/patologia , Gliose/virologia , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Masculino , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/virologia , Nitrilas , Pirimidinas , Tenofovir/farmacologia
5.
J Neurosci ; 34(43): 14219-32, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25339736

RESUMO

Spines are dendritic protrusions that receive most of the excitatory input in the brain. Early after the onset of cerebral ischemia dendritic spines in the peri-infarct cortex are replaced by areas of focal swelling, and their re-emergence from these varicosities is associated with neurological recovery after acute ischemic stroke (AIS). Urokinase-type plasminogen activator (uPA) is a serine proteinase that plays a central role in tissue remodeling via binding to the urokinase plasminogen activator receptor (uPAR). We report that cerebral cortical neurons release uPA during the recovery phase from ischemic stroke in vivo or hypoxia in vitro. Although uPA does not have an effect on ischemia- or hypoxia-induced neuronal death, genetic deficiency of uPA (uPA(-/-)) or uPAR (uPAR(-/-)) abrogates functional recovery after AIS. Treatment with recombinant uPA after ischemic stroke induces neurological recovery in wild-type and uPA(-/-) but not in uPAR(-/-) mice. Diffusion tensor imaging studies indicate that uPA(-/-) mice have increased water diffusivity and decreased anisotropy associated with impaired dendritic spine recovery and decreased length of distal neurites in the peri-infarct cortex. We found that the excitotoxic injury induces the clustering of uPAR in dendritic varicosities, and that the binding of uPA to uPAR promotes the reorganization of the actin cytoskeleton and re-emergence of dendritic filopodia from uPAR-enriched varicosities. This effect is independent of uPA's proteolytic properties and instead is mediated by Rac-regulated profilin expression and cofilin phosphorylation. Our data indicate that binding of uPA to uPAR promotes dendritic spine recovery and improves functional outcome following AIS.


Assuntos
Isquemia Encefálica/enzimologia , Espinhas Dendríticas/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/enzimologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Células Cultivadas , Espinhas Dendríticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/enzimologia , Doenças do Sistema Nervoso/patologia , Ligação Proteica/fisiologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Resultado do Tratamento , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/uso terapêutico
6.
J Cereb Blood Flow Metab ; 33(11): 1761-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23881246

RESUMO

Adenosine monophosphate-activated protein kinase (AMPK) is an energy sensor that regulates cellular adaptation to metabolic stress. Tissue-type plasminogen activator (tPA) is a serine proteinase found in the intravascular space, where its main role is as thrombolytic enzyme, and in neurons, where its function is less well understood. Here, we report that glucose deprivation induces the mobilization and package of neuronal tPA into presynaptic vesicles. Mass spectrometry and immunohistochemical studies show that the release of this tPA in the synaptic space induces AMPK activation in the postsynaptic terminal, and an AMPK-mediated increase in neuronal uptake of glucose and neuronal adenosine 5'(tetrahydrogen triphosphate; ATP) synthesis. This effect is independent of tPA's proteolytic properties, and instead requires the presence of functional N-methyl-D-aspartate receptors (NMDARs). In agreement with these observations, positron emission tomography (PET) studies and biochemical analysis with synaptoneurosomes indicate that the intravenous administration of recombinant tPA (rtPA) after transient middle cerebral artery occlusion (tMCAO) induces AMPK activation in the synaptic space and NMDAR-mediated glucose uptake in the ischemic brain. These data indicate that the release of neuronal tPA or treatment with rtPA activate a cell signaling pathway in the synaptic space that promotes the detection and adaptation to metabolic stress.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tecidual/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/biossíntese , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Glucose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Sinaptossomos/patologia , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/uso terapêutico
7.
Mol Cell Neurosci ; 52: 9-19, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23063501

RESUMO

The release of the serine proteinase tissue-type plasminogen activator (tPA) from cerebral cortical neurons has a neuroprotective effect in the ischemic brain. Because excitotoxicity is a basic mechanism of ischemia-induced cell death, here we investigated the effect of tPA on excitotoxin-induced neuronal death. We report that genetic overexpression of neuronal tPA or treatment with recombinant tPA renders neurons resistant to the harmful effects of an excitotoxic injury in vitro and in vivo. We found that at concentrations found in the ischemic brain, tPA interacts with synaptic but not extrasynaptic NMDARs. This effect is independent of tPA's proteolytic properties and leads to a rapid and transient phosphorylation of the extracellular signal regulated kinases1/2 (ERK1/2), with ERK1/2-mediated activation of the cAMP response element binding protein (CREB) and induction of the neuroprotective CREB-regulated activating transcription factor 3 (Atf3). In line with these observations, Atf3 down-regulation abrogates the protective effect of tPA against excitotoxin-induced neuronal death. Our data indicate that tPA preferentially activates synaptic NMDARs via a plasminogen-independent mechanism turning on a cell signaling pathway that protects neurons from the deleterious effects of excitotoxicity.


Assuntos
Neurônios/metabolismo , Transdução de Sinais/fisiologia , Ativador de Plasminogênio Tecidual/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Animais , Western Blotting , Morte Celular/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Metilaspartato/toxicidade , Neurotoxinas/toxicidade , Receptores de N-Metil-D-Aspartato/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativador de Plasminogênio Tecidual/farmacologia
8.
J Neurosci ; 32(29): 9848-58, 2012 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-22815500

RESUMO

The ability to sense and adapt to hypoxic conditions plays a pivotal role in neuronal survival. Hypoxia induces the release of tissue-type plasminogen activator (tPA) from cerebral cortical neurons. We found that the release of neuronal tPA or treatment with recombinant tPA promotes cell survival in cerebral cortical neurons previously exposed to hypoxic conditions in vitro or experimental cerebral ischemia in vivo. Our studies using liquid chromatography and tandem mass spectrometry revealed that tPA activates the mammalian target of rapamycin (mTOR) pathway, which adapts cellular processes to the availability of energy and metabolic resources. We found that mTOR activation leads to accumulation of the hypoxia-inducible factor-1α (HIF-1α) and induction and recruitment to the cell membrane of the HIF-1α-regulated neuronal transporter of glucose GLUT3. Accordingly, in vivo positron emission tomography studies with 18-fluorodeoxyglucose in mice overexpressing tPA in neurons show that neuronal tPA induces the uptake of glucose in the ischemic brain and that this effect is associated with a decrease in the volume of the ischemic lesion and improved neurological outcome following the induction of ischemic stroke. Our data indicate that tPA activates a cell signaling pathway that allows neurons to sense and adapt to oxygen and glucose deprivation.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/efeitos dos fármacos , Fibrinolíticos/farmacologia , Glucose/metabolismo , Neurônios/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/patologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo
9.
J Neuroinflammation ; 9: 45, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22394384

RESUMO

BACKGROUND: Cerebral cortical neurons have a high vulnerability to the harmful effects of hypoxia. However, the brain has the ability to detect and accommodate to hypoxic conditions. This phenomenon, known as preconditioning, is a natural adaptive process highly preserved among species whereby exposure to sub-lethal hypoxia promotes the acquisition of tolerance to a subsequent lethal hypoxic injury. The cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) are found in neurons and their expression is induced by exposure to sub-lethal hypoxia. Accordingly, in this work we tested the hypothesis that the interaction between TWEAK and Fn14 induces tolerance to lethal hypoxic and ischemic conditions. METHODS: Here we used in vitro and in vivo models of hypoxic and ischemic preconditioning, an animal model of transient middle cerebral artery occlusion and mice and neurons genetically deficient in TWEAK, Fn14, or tumor necrosis factor alpha (TNF-α) to investigate whether treatment with recombinant TWEAK or an increase in the expression of endogenous TWEAK renders neurons tolerant to lethal hypoxia. We used enzyme-linked immunosorbent assay to study the effect of TWEAK on the expression of neuronal TNF-α, Western blot analysis to investigate whether the effect of TWEAK was mediated by activation of mitogen-activated protein kinases and immunohistochemical techniques and quantitative real-time polymerase chain reaction analysis to study the effect of TWEAK on apoptotic cell death. RESULTS: We found that either treatment with recombinant TWEAK or an increase in the expression of TWEAK and Fn14 induce hypoxic and ischemic tolerance in vivo and in vitro. This protective effect is mediated by neuronal TNF-α and activation of the extracellular signal-regulated kinases 1 and 2 pathway via phosphorylation and inactivation of the B-cell lymphoma 2-associated death promoter protein. CONCLUSIONS: Our work indicate that the interaction between TWEAK and Fn14 triggers the activation of a cell signaling pathway that results in the induction of tolerance to lethal hypoxia and ischemia. These data indicate that TWEAK may be a potential therapeutic strategy to protect the brain from the devastating effects of an ischemic injury.


Assuntos
Apoptose/efeitos dos fármacos , Infarto da Artéria Cerebral Média/patologia , Precondicionamento Isquêmico/métodos , Fármacos Neuroprotetores/farmacologia , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Necrose Tumoral/farmacologia , Animais , Apoptose/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Citocina TWEAK , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Glucose/deficiência , Hipóxia/tratamento farmacológico , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores do Fator de Necrose Tumoral/deficiência , Receptor de TWEAK , Fatores de Tempo , Fator de Necrose Tumoral alfa/deficiência , Fatores de Necrose Tumoral/deficiência , Fatores de Necrose Tumoral/metabolismo
10.
J Cereb Blood Flow Metab ; 32(1): 57-69, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21792242

RESUMO

Cerebral cortical neurons have a heightened sensitivity to hypoxia and their survival depends on their ability to accommodate to changes in the concentration of oxygen in their environment. Tissue-type plasminogen activator (tPA) is a serine proteinase that activates the zymogen plasminogen into plasmin. Hypoxia induces the release of tPA from cerebral cortical neurons, and it has been proposed that tPA mediates hypoxic and ischemic neuronal death. Here, we show that tPA is devoid of neurotoxic effects and instead is an endogenous neuroprotectant that renders neurons resistant to the effects of lethal hypoxia and ischemia. We present in vitro and in vivo evidence indicating that endogenous tPA and recombinant tPA induce the expression of neuronal tumor necrosis factor-α. This effect, mediated by plasmin and the N-methyl-D-aspartate receptor, leads to increased expression of the cyclin-dependent kinase inhibitor p21 and p21-mediated development of early hypoxic and ischemic tolerance.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Neurônios/metabolismo , Ativador de Plasminogênio Tecidual/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Ensaio de Imunoadsorção Enzimática , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/patologia , Precondicionamento Isquêmico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/farmacologia , Ativador de Plasminogênio Tecidual/uso terapêutico , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
11.
J Clin Invest ; 120(6): 2194-205, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20440070

RESUMO

The best-known function of the serine protease tissue-type plasminogen activator (tPA) is as a thrombolytic enzyme. However, it is also found in structures of the brain that are highly vulnerable to hypoxia-induced cell death, where its association with neuronal survival is poorly understood. Here, we have demonstrated that hippocampal areas of the mouse brain lacking tPA activity are more vulnerable to neuronal death following an ischemic insult. We found that sublethal hypoxia, which elicits tolerance to subsequent lethal hypoxic/ischemic injury in a natural process known as ischemic preconditioning (IPC), induced a rapid release of neuronal tPA. Treatment of hippocampal neurons with tPA induced tolerance against a lethal hypoxic insult applied either immediately following insult (early IPC) or 24 hours later (delayed IPC). tPA-induced early IPC was independent of the proteolytic activity of tPA and required the engagement of a member of the LDL receptor family. In contrast, tPA-induced delayed IPC required the proteolytic activity of tPA and was mediated by plasmin, the NMDA receptor, and PKB phosphorylation. We also found that IPC in vivo increased tPA activity in the cornu ammonis area 1 (CA1) layer and Akt phosphorylation in the hippocampus, as well as ischemic tolerance in wild-type but not tPA- or plasminogen-deficient mice. These data show that tPA can act as an endogenous neuroprotectant in the murine hippocampus.


Assuntos
Hipocampo/metabolismo , Fármacos Neuroprotetores/metabolismo , Ativadores de Plasminogênio/farmacologia , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Encéfalo/metabolismo , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Fibrinolisina , Fibrinolíticos/metabolismo , Hipocampo/citologia , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Ativadores de Plasminogênio/metabolismo , Serina Proteases/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tecidual/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/farmacologia
12.
J Cereb Blood Flow Metab ; 30(6): 1147-56, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20068578

RESUMO

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) are expressed in endothelial cells and perivascular astrocytes. Here, we show that TWEAK induces a dose-dependent increase in the expression of the chemokine monocyte chemoattractant protein-1 (MCP-1) in astrocytes, and that this effect is mediated by its interaction with Fn14 via nuclear factor-kappaB pathway activation. Exposure to oxygen-glucose deprivation (OGD) conditions increases TWEAK and Fn14 mRNA expression in wild-type (Wt) astrocytic cultures. Likewise, incubation under OGD conditions induces the expression of MCP-1 in Wt astrocytes but not in astrocytes deficient on either TWEAK (TWEAK(-/-)) or Fn14 (Fn14(-/-)). We also found that TWEAK induces the passage of neutrophils to the abluminal side of an in vitro model of the blood-brain barrier. Our earlier studies indicate that cerebral ischemia increases the expression of TWEAK and Fn14 in the endothelial cell-basement membrane-astrocyte interface. Here, we report that middle cerebral artery occlusion increases the expression of MCP-1 and the recruitment of neutrophils into the ischemic tissue in Wt but not in TWEAK(-/-) or Fn14(-/-) mice. These novel results indicate that during cerebral ischemia, the interaction between TWEAK and Fn14 leads to the recruitment of leukocytes into the ischemic tissue.


Assuntos
Isquemia Encefálica/metabolismo , Células Endoteliais/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Hipóxia Celular , Células Cultivadas , Quimiocina CCL2/biossíntese , Citocina TWEAK , Células Endoteliais/patologia , Regulação da Expressão Gênica/genética , Glucose/metabolismo , Humanos , Camundongos , Camundongos Knockout , Neutrófilos/patologia , Receptores do Fator de Necrose Tumoral/genética , Receptor de TWEAK , Fatores de Necrose Tumoral/genética
13.
J Cereb Blood Flow Metab ; 29(12): 1946-54, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19672275

RESUMO

Studies in animal models of cerebral ischemia indicate that besides its thrombolytic effect, treatment with tissue-type plasminogen activator (tPA) also induces an increase in matrix metalloproteinase-9 (MMP-9) activity in the ischemic tissue associated with the development of cerebral edema. Earlier, we had shown that the low-density lipoprotein receptor-related protein 1 (LRP1) is a substrate for tPA in the brain. In this study, we investigated the effect of the interaction between tPA and microglial LRP1 on MMP-9 activity after middle cerebral artery occlusion (MCAO). We found that exposure to oxygen-glucose deprivation (OGD) conditions increases MMP-9 activity in wild-type (Wt) and plasminogen-deficient (Plg(-/-)) microglia, but not in tPA (tPA(-/-)) or LRP1-deficient (macLRP-) cells. Treatment with tPA increases MMP-9 expression in tPA(-/-) but not in macLRP- microglia. Middle cerebral artery occlusion increases MMP-9 expression and activity in Wt but not in tPA(-/-) or macLRP- mice, and treatment with tPA increases MMP-9 activity in tPA(-/-) mice but not in macLRP- animals. Finally, MCAO-induced ischemic edema and degradation of the interendothelial right junction protein claudin-5 were significantly attenuated in tPA(-/-) and macLRP- mice. The results of our study indicate that the interaction between tPA and microglial LRP1 increases MMP-9 expression and activity resulting in the degradation of claudin-5 and development of cerebral edema.


Assuntos
Isquemia Encefálica/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Microglia/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Células Cultivadas , Regulação da Expressão Gênica , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/citologia , Ativador de Plasminogênio Tecidual/genética
14.
Cell Microbiol ; 8(8): 1349-60, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16882037

RESUMO

The mammalian plasminogen activation system (PAS) is a complex system involved in multiple physiological and pathological processes. Borrelia burgdorferi interacts with certain components of the PAS. Here we further investigate this interaction to determine its effect on bacterial dissemination and host cell migration in vitro. We show that stimulation of monocytic cells with B. burgdorferi induces the transient production and secretion of urokinase plasminogen activator (uPA), shortly followed by its physiological inhibitor, plasminogen activator inhibitor-2 (PAI-2). Mono Mac 6 (MM6) cells as well as peripheral blood monocytes enhanced transmigration of B. burgdorferi across a barrier coated with fibronectin mediated by uPA. Moreover, the induction of PAI-2 or the addition of recombinant PAI-2 did not have a significant effect on the uPA-potentiated transmigration of B. burgdorferi. In contrast, the induction of PAI-2 by B. burgdorferi resulted in significantly diminished invasion by monocytic cells across a reconstituted basement membrane (matrigel), which could be partially restored by treatment with purified uPA. These results show that the PAS plays a twofold role in the pathogenesis of B. burgdorferi infection, both by enhancing bacterial dissemination and by diminishing host-cell inflammatory migration.


Assuntos
Borrelia burgdorferi/fisiologia , Borrelia burgdorferi/patogenicidade , Inibidor 2 de Ativador de Plasminogênio/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Linhagem Celular , Humanos , Inflamação/etiologia , Monócitos/microbiologia , Transcrição Gênica , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/genética , Virulência/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...