Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nat Metab ; 5(8): 1290-1302, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37460843

RESUMO

Having direct access to brain vasculature, astrocytes can take up available blood nutrients and metabolize them to fulfil their own energy needs and deliver metabolic intermediates to local synapses1,2. These glial cells should be, therefore, metabolically adaptable to swap different substrates. However, in vitro and in vivo studies consistently show that astrocytes are primarily glycolytic3-7, suggesting glucose is their main metabolic precursor. Notably, transcriptomic data8,9 and in vitro10 studies reveal that mouse astrocytes are capable of mitochondrially oxidizing fatty acids and that they can detoxify excess neuronal-derived fatty acids in disease models11,12. Still, the factual metabolic advantage of fatty acid use by astrocytes and its physiological impact on higher-order cerebral functions remain unknown. Here, we show that knockout of carnitine-palmitoyl transferase-1A (CPT1A)-a key enzyme of mitochondrial fatty acid oxidation-in adult mouse astrocytes causes cognitive impairment. Mechanistically, decreased fatty acid oxidation rewired astrocytic pyruvate metabolism to facilitate electron flux through a super-assembled mitochondrial respiratory chain, resulting in attenuation of reactive oxygen species formation. Thus, astrocytes naturally metabolize fatty acids to preserve the mitochondrial respiratory chain in an energetically inefficient disassembled conformation that secures signalling reactive oxygen species and sustains cognitive performance.


Assuntos
Astrócitos , Encéfalo , Camundongos , Animais , Astrócitos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Encéfalo/metabolismo , Cognição , Ácidos Graxos/metabolismo
2.
J Neuroimaging ; 33(2): 289-301, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36536493

RESUMO

BACKGROUND AND PURPOSE: The prognostic significance of postcontrast enhancement of intracranial atheromatous plaque is uncertain. Prospective, long-term follow-up studies in Caucasians, using a multicenter design, are lacking. We aimed to evaluate whether this radiological sign predicts long-term new stroke in symptomatic and asymptomatic intracranial atherosclerotic disease (ICAD) patients. METHODS: This was a prospective, observational, longitudinal, multicenter study. We included a symptomatic and an asymptomatic cohort of ICAD patients that underwent 3T MRI including high-resolution sequences focused on the atheromatous plaque. We evaluated grade of stenosis, plaque characteristics, and gadolinium enhancement ratio (postcontrast plaque signal/postcontrast corpus callosum signal). The occurrence of new events was evaluated at 3, 6, 9, and 12 months and annually thereafter. The association between plaque characteristics and new stroke was studied using Cox multiple regression survival analysis and Kaplan-Meier curves. RESULTS: Forty-eight symptomatic and 13 asymptomatic patients were included. During 56.3 ± 16.9 months, 11 patients (18%) suffered a new event (seven ischemic, two hemorrhagic, and two transient ischemic attacks). A receiver operating characteristic curve identified an enhancement ratio of >1.77 to predict a new event. In a multivariable Cox regression, postcontrast enhancement ratio >1.77 (hazard ratio [HR]= 3.632; 95% confidence interval [CI], 1.082-12.101) and cerebral microbleeds (HR = 5.244; 95% CI, 1.476-18.629) were independent predictors of future strokes. Patients with a plaque enhancement ratio >1.77 had a lower survival free of events (p < .05). CONCLUSIONS: High intracranial postcontrast enhancement is a long-term predictor of new stroke in ICAD patients. Further studies are needed to elucidate whether postcontrast enhancement reflects inflammatory activity of intracranial atheromatous plaque.


Assuntos
Arteriosclerose Intracraniana , Placa Aterosclerótica , Acidente Vascular Cerebral , Humanos , Estudos Prospectivos , Meios de Contraste , Estudos Longitudinais , Gadolínio , Imageamento por Ressonância Magnética/métodos
3.
J Neurochem ; 165(4): 521-535, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36563047

RESUMO

Intracellular Ca2+ concentrations are strictly controlled by plasma membrane transporters, the endoplasmic reticulum, and mitochondria, in which Ca2+ uptake is mediated by the mitochondrial calcium uniporter complex (MCUc), while efflux occurs mainly through the mitochondrial Na+ /Ca2+ exchanger (NCLX). RNAseq database repository searches led us to identify the Nclx transcript as highly enriched in astrocytes when compared with neurons. To assess the role of NCLX in mouse primary culture astrocytes, we inhibited its function both pharmacologically or genetically. This resulted in re-shaping of cytosolic Ca2+ signaling and a metabolic shift that increased glycolytic flux and lactate secretion in a Ca2+ -dependent manner. Interestingly, in vivo genetic deletion of NCLX in hippocampal astrocytes improved cognitive performance in behavioral tasks, whereas hippocampal neuron-specific deletion of NCLX impaired cognitive performance. These results unveil a role for NCLX as a novel modulator of astrocytic glucose metabolism, impacting on cognition.


Assuntos
Astrócitos , Cálcio , Camundongos , Animais , Astrócitos/metabolismo , Cálcio/metabolismo , Trocador de Sódio e Cálcio/genética , Mitocôndrias/metabolismo , Glicólise , Cognição , Sódio/metabolismo , Sinalização do Cálcio/fisiologia
4.
Front Pharmacol ; 13: 884470, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35496276

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline, which is causally related to the accumulation of abnormally folded amyloid-ß (Aß) peptide and hyperphosphorylated tau protein aggregates. The dendritic spine regulator Rho protein kinase 2 (Rock2) accumulates in the brain at the earliest stages of AD and remains increased during disease progression. However, the molecular mechanism that upregulates Rock2 in AD, and its role in the disease progression, are unknown. Here, we found that oligomers of the amyloidogenic fragment 25-35 of the Aß peptide (Aß25-35) trigger Rock2 accumulation and activation in mouse cortical neurons in primary culture and in mouse hippocampus in vivo. Neuronal apoptotic death and memory impairment caused by Aß25-35 administration were rescued by genetic and pharmacological inhibition of Rock2 activity. Mechanistically, Aß25-35 elicited cyclin dependent kinase-5 (Cdk5)-mediated phosphorylation of Cdh1, a cofactor that is essential for the activity of the E3 ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C) in neurons. Notably, phosphorylated Cdh1 was disassembled from the APC/C complex, causing its inactivation and subsequent Rock2 protein stabilization and activation. Moreover, Aß25-35-induced neuronal apoptosis was prevented by expressing a phosphodefective form of Cdh1, but not by a phosphomimetic Cdh1. Finally, Cdh1 inactivation, using both genetic and pharmacological approaches, enhanced Aß25-35-mediated neuronal death through a mechanism that was prevented by inhibition of Rock2 activity. These results indicate that the Cdk5-Cdh1 signaling pathway accounts for the increased Rock2 activity by amyloidogenic Aß peptides and that this mechanism may contribute to neurodegeneration and memory loss in AD.

5.
Front Pharmacol ; 13: 1086540, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36588673

RESUMO

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and the main cause of dementia in the elderly. The disease has a high impact on individuals and their families and represents a growing public health and socio-economic burden. Despite this, there is no effective treatment options to cure or modify the disease progression, highlighting the need to identify new therapeutic targets. Synapse dysfunction and loss are early pathological features of Alzheimer's disease, correlate with cognitive decline and proceed with neuronal death. In the last years, the E3 ubiquitin ligase anaphase promoting complex/cyclosome (APC/C) has emerged as a key regulator of synaptic plasticity and neuronal survival. To this end, the ligase binds Cdh1, its main activator in the brain. However, inactivation of the anaphase promoting complex/cyclosome-Cdh1 complex triggers dendrite disruption, synapse loss and neurodegeneration, leading to memory and learning impairment. Interestingly, oligomerized amyloid-ß (Aß) peptide, which is involved in Alzheimer's disease onset and progression, induces Cdh1 phosphorylation leading to anaphase promoting complex/cyclosome-Cdh1 complex disassembly and inactivation. This causes the aberrant accumulation of several anaphase promoting complex/cyclosome-Cdh1 targets in the damaged areas of Alzheimer's disease brains, including Rock2 and Cyclin B1. Here we review the function of anaphase promoting complex/cyclosome-Cdh1 dysregulation in the pathogenesis of Alzheimer's disease, paying particular attention in the neurotoxicity induced by its molecular targets. Understanding the role of anaphase promoting complex/cyclosome-Cdh1-targeted substrates in Alzheimer's disease may be useful in the development of new effective disease-modifying treatments for this neurological disorder.

6.
Neurochem Res ; 46(1): 51-63, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32189131

RESUMO

A new era for neuroprotective strategies is emerging in ischemia/reperfusion. This has forced to review the studies existing to date based in neuroprotection against oxidative stress, which have undoubtedly contributed to clarify the brain endogenous mechanisms, as well as to identify possible therapeutic targets or biomarkers in stroke and other neurological diseases. The efficacy of exogenous administration of neuroprotective compounds has been shown in different studies so far. However, something must be missing to get these treatments successfully applied in the clinical environment. Here, the mechanisms involved in neuronal protection against physiological level of ROS and the main neuroprotective signaling pathways induced by excitotoxic and ischemic stimuli are reviewed. Also, the endogenous ischemic tolerance in terms of brain self-protection mechanisms against subsequent cerebral ischemia is revisited to highlight how the preconditioning has emerged as a powerful tool to understand these phenomena. A better understanding of endogenous defense against exacerbated ROS and metabolism in nervous cells will therefore aid to design pharmacological antioxidants targeted specifically against oxidative damage induced by ischemic injury, but also might be very valuable for translational medicine.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/terapia , Precondicionamento Isquêmico , Neuroproteção/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Animais , Astrócitos/metabolismo , Expressão Gênica/fisiologia , Humanos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
7.
Sci Rep ; 10(1): 16196, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33004936

RESUMO

We aimed to study the relationship between the ischemic core's (IC) radiological hypodensity and the risk of parenchymal haematoma after endovascular therapy (EVT) in acute ischemic stroke (AIS) presenting > 4.5 h from onset. We studied AIS patients with a proximal anterior circulation occlusion > 4.5 h from symptoms onset treated with primary EVT. The IC regions of interest (ROI) were manually delineated on pretreatment CT within the affected hemisphere and their specular ROIs on the unaffected side. IC hypodensity ratio was calculated by dividing mean Hounsfield Unit (HU) value from all ROIs in affected/unaffected hemisphere. Primary endpoint: parenchymal hematoma (PH) type hemorrhagic transformation. Secondary: poor long-term clinical outcome. From May 2015 to November 2018, 648 consecutive AIS patients received reperfusion therapies and 107 met all inclusion criteria. PH after EVT was diagnosed in 33 (31%) patients. In bivariate analyses, IC hypodensity ratio (p < 0.001) and minimum HU value (p = 0.008) were associated with PH. A lower IC hypodensity ratio [OR < 0.001 (< 0.001-0.116) p 0.016] predicted PH but not poor clinical outcome in multivariable logistic regression models. A lower IC radiological density predicted a higher risk of PH in > 4.5 h-window AIS patients treated with primary EVT, although it was not independently associated with a worse clinical outcome.


Assuntos
Isquemia Encefálica/terapia , Procedimentos Endovasculares/efeitos adversos , Hematoma/etiologia , Acidente Vascular Cerebral/terapia , Trombectomia/efeitos adversos , Terapia Trombolítica/efeitos adversos , Tomografia Computadorizada por Raios X/métodos , Idoso , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/patologia , Feminino , Hematoma/diagnóstico por imagem , Hematoma/patologia , Humanos , Masculino , Prognóstico , Estudos Retrospectivos , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/patologia
8.
Sci Adv ; 6(41)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33028529

RESUMO

Failure of neurons to efficiently repair DNA double-strand breaks (DSBs) contributes to cerebral damage after stroke. However, the molecular machinery that regulates DNA repair in this neurological disorder is unknown. Here, we found that DSBs in oxygen/glucose-deprived (OGD) neurons spatiotemporally correlated with the up-regulation of WRAP53 (WD40-encoding p53-antisense RNA), which translocated to the nucleus to activate the DSB repair response. Mechanistically, OGD triggered a burst in reactive oxygen species that induced both DSBs and translocation of WRAP53 to the nucleus to promote DNA repair, a pathway that was confirmed in an in vivo mouse model of stroke. Noticeably, nuclear translocation of WRAP53 occurred faster in OGD neurons expressing the Wrap53 human nonsynonymous single-nucleotide polymorphism (SNP) rs2287499 (c.202C>G). Patients carrying this SNP showed less infarct volume and better functional outcome after stroke. These results indicate that WRAP53 fosters DNA repair and neuronal survival to promote functional recovery after stroke.


Assuntos
Núcleo Celular , Chaperonas Moleculares/metabolismo , Acidente Vascular Cerebral , Telomerase/metabolismo , Animais , Núcleo Celular/metabolismo , Sobrevivência Celular/genética , Reparo do DNA , Glucose/metabolismo , Humanos , Camundongos , Neurônios/metabolismo , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Fatores de Transcrição/metabolismo
9.
Stroke ; 51(5): 1514-1521, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32188368

RESUMO

Background and Purpose- We aimed to evaluate the impact of brain atrophy on long-term clinical outcome in patients with acute ischemic stroke treated with endovascular therapy, and more specifically, to test whether there are interactions between the degree of atrophy and infarct volume, and between atrophy and age, in determining the risk of futile reperfusion. Methods- We studied consecutive patients with acute ischemic stroke with proximal anterior circulation intracranial arterial occlusions treated with endovascular therapy achieving successful arterial recanalization. Brain atrophy was evaluated on baseline computed tomography with the global cortical atrophy scale, and Evans index was calculated to assess subcortical atrophy. Infarct volume was assessed on control computed tomography at 24 hours using the formula for irregular volumes (A×B×C/2). Main outcome variable was futile recanalization, defined by functional dependence (modified Rankin Scale score >2) at 3 months. The predefined interactions of atrophy with age and infarct volume were studied in regression models. Results- From 361 consecutive patients with anterior circulation acute ischemic stroke treated with endovascular therapy, 295 met all inclusion criteria. Futile reperfusion was observed in 144 out of 295 (48.8%) patients. Cortical atrophy affecting parieto-occipital and temporal regions was associated with futile recanalization. Total global cortical atrophy score and Evans index were independently associated with futile recanalization in an adjusted logistic regression. Multivariable adjusted regression models disclosed significant interactions between global cortical atrophy score and infarct volume (odds ratio, 1.003 [95%CI, 1.002-1.004], P<0.001) and between global cortical atrophy score and age (odds ratio, 1.001 [95% CI, 1.001-1.002], P<0.001) in determining the risk of futile reperfusion. Conclusions- A higher degree of cortical and subcortical brain atrophy is associated with futile endovascular reperfusion in anterior circulation acute ischemic stroke. The impact of brain atrophy on insufficient clinical recovery after endovascular reperfusion appears to be independently amplified by age and by infarct volume.


Assuntos
Isquemia Encefálica/cirurgia , Córtex Cerebral/diagnóstico por imagem , Procedimentos Endovasculares , Acidente Vascular Cerebral/cirurgia , Trombectomia , Idoso , Idoso de 80 Anos ou mais , Atrofia , Córtex Cerebral/patologia , Feminino , Seguimentos , Humanos , Leucoaraiose/diagnóstico por imagem , Masculino , Futilidade Médica , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Índice de Gravidade de Doença , Tomografia Computadorizada por Raios X , Resultado do Tratamento
10.
Neuropharmacology ; 146: 19-27, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30452955

RESUMO

Neurodegeneration in selective brain areas underlies the pathology of Alzheimer's disease (AD). Although oligomeric amyloid-ß (Aß) plays a central role in the AD pathogenesis, the mechanism of neuronal loss in response to Aß remains elusive. The p53 tumor suppressor protein, a key regulator of cell apoptosis, has been described to accumulate in affected brain areas from AD patients. However, whether p53 plays any role in AD pathogenesis remains unknown. To address this issue, here we investigated the involvement of p53 on Aß-induced neuronal apoptosis. We found that exposure of neurons to oligomers of the amyloidogenic fragment 25-35 of the Aß peptide (Aß25-35) promoted p53 protein phosphorylation and stabilization, leading to mitochondrial dysfunction and neuronal apoptosis. To address the underlying mechanism, we focused on cyclin dependent kinase-5 (Cdk5), a known p53-phosphorylating kinase. The results revealed that Aß25-35 treatment activated Cdk5, and that inhibiting Cdk5 activity prevented p53 protein stabilization. Furthermore, Aß25-35-mediated mitochondrial dysfunction and neuronal apoptosis were prevented by both genetic and pharmacological inhibition of either p53 or Cdk5 activities. This effect was mimicked with the full-length peptide Aß1-42. To confirm the mechanism in vivo, Aß25-35 was stereotaxically injected in the cerebral right ventricle of mice, a treatment that caused p53 protein accumulation, dendrite disruption and neuronal death. Furthermore, these effects were prevented in p53 knockout mice or by pharmacologically inhibiting p53. Thus, Aß25-35 triggers Cdk5 activation to induce p53 phosphorylation and stabilization, which leads to neuronal damage. Inhibition of the Cdk5-p53 pathway may therefore represent a novel therapeutic strategy against Aß-induced neurodegeneration.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Quinase 5 Dependente de Ciclina/metabolismo , Fragmentos de Peptídeos/toxicidade , Proteína Supressora de Tumor p53/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose , Dendritos/efeitos dos fármacos , Dendritos/patologia , Infusões Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Fosforilação , Transdução de Sinais , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
11.
Stroke ; 49(10): 2437-2444, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30355102

RESUMO

Background and Purpose- The E3 ubiquitin ligase MDM2 (murine double minute 2) is the main negative regulator of the p53 protein-a key player in neuronal apoptosis after ischemia. A functional single-nucleotide polymorphism in the human MDM2 gene promoter (rs2279744) regulates MDM2 protein expression. We investigated whether the MDM2 SNP309, by controlling p53-mediated apoptosis, determines functional outcome after stroke. Methods- Primary cortical neurons were subjected to oxygen and glucose deprivation. Mice were subjected to ischemic (transient middle cerebral artery occlusion) or hemorrhagic (collagenase injection) stroke models. Protein and mRNA levels of MDM2 and p53 were measured in both neuronal and brain extracts. The interaction of MDM2 with p53 was disrupted by neuronal treatment with nutlin-3a. siRNA was used to knockdown MDM2 expression. We analyzed the link between the MDM2 SNP309 and functional outcome, measured by the modified Rankin Scale scores, in 2 independent hospital-based stroke cohorts: ischemic stroke cohort (408 patients) and intracerebral hemorrhage cohort (128 patients). Results- Experimental stroke and oxygen and glucose deprivation induced the expression of MDM2 in the brain and neurons, respectively. Moreover, oxygen and glucose deprivation promoted MDM2 binding with p53 in neurons. Disruption of the MDM2-p53 interaction with nutlin-3a, or MDM2 knockdown by siRNA, triggered p53 accumulation, which increased neuronal susceptibility to oxygen and glucose deprivation-induced apoptosis. Finally, we showed that patients harboring the G allele in the MDM2 promoter had higher MDM2 protein levels and showed better functional outcome after stroke than those harboring the T/T genotype. The T/T genotype was also associated with large infarct volume in ischemic stroke and increased lesion volume in patients with intracerebral hemorrhage. Conclusions- Our results reveal a novel role for the MDM2-p53 interaction in neuronal apoptosis after ischemia and show that the MDM2 SNP309 determines the functional outcome of patients after stroke.


Assuntos
Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas c-mdm2/genética , Recuperação de Função Fisiológica/genética , Acidente Vascular Cerebral/genética , Alelos , Animais , Genótipo , Humanos , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Acidente Vascular Cerebral/terapia
12.
Sci Rep ; 8(1): 1610, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29371613

RESUMO

Brain preconditioning (PC) refers to a state of transient tolerance against a lethal insult that can be evoked by a prior mild event. It is thought that PC may induce different pathways responsible for neuroprotection, which may involve the attenuation of cell damage pathways, including the apoptotic cell death. In this context, p53 is a stress sensor that accumulates during brain ischemia leading to neuronal death. The murine double minute 2 gene (MDM2), a p53-specific E3 ubiquitin ligase, is the main cellular antagonist of p53, mediating its degradation by the proteasome. Here, we study the role of MDM2-p53 pathway on PC-induced neuroprotection both in cultured neurons (in vitro) and rat brain (in vivo). Our results show that PC increased neuronal MDM2 protein levels, which prevented ischemia-induced p53 stabilization and neuronal death. Indeed, PC attenuated ischemia-induced activation of the p53/PUMA/caspase-3 signaling pathway. Pharmacological inhibition of MDM2-p53 interaction in neurons abrogated PC-induced neuroprotection against ischemia. Finally, the relevance of the MDM2-p53 pathway was confirmed in rat brain using a PC model in vivo. These findings demonstrate the key role of the MDM2-p53 pathway in PC-induced neuroprotection against a subsequent ischemic insult and poses MDM2 as an essential target in ischemic tolerance.


Assuntos
Encéfalo/patologia , Isquemia/patologia , Precondicionamento Isquêmico , Neurônios/patologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Ratos
13.
Cell Death Differ ; 24(1): 144-154, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27768124

RESUMO

Intracerebral hemorrhage (ICH) is a devastating subtype of stroke that lacks effective therapy and reliable prognosis. Neovascularization following ICH is an essential compensatory response that mediates brain repair and modulates the clinical outcome of stroke patients. However, the mechanism that dictates this process is unknown. Bone marrow-derived endothelial progenitor cells (EPCs) promote endothelial repair and contribute to ischemia-induced neovascularization. The human Tp53 gene harbors a common single-nucleotide polymorphism (SNP) at codon 72, which yields an arginine-to-proline amino-acidic substitution (Arg72Pro) that modulates the apoptotic activity of the p53 protein. Previously, we found that this SNP controls neuronal susceptibility to ischemia-induced apoptosis in vitro. Here, we evaluated the impact of the Tp53 Arg72Pro SNP on vascular repair and functional recovery after ICH. We first analyzed EPC mobilization and functional outcome based on the modified Rankin scale scores in a hospital-based cohort of 78 patients with non-traumatic ICH. Patients harboring the Pro allele of the Tp53 Arg72Pro SNP showed higher levels of circulating EPC-containing CD34+ cells, EPC-mobilizing cytokines - vascular endothelial growth factor and stromal cell-derived factor-1α - and good functional outcome following ICH, when compared with the homozygous Arg allele patients, which is compatible with increased neovascularization. To assess directly whether Tp53 Arg72Pro SNP regulated neovascularization after ICH, we used the humanized Tp53 Arg72Pro knock-in mice, which were subjected to the collagenase-induced ICH. The brain endothelial cells of the Pro allele-carrying mice were highly resistant to ICH-mediated apoptosis, which facilitated cytokine-mediated EPC mobilization, cerebrovascular repair and functional recovery. However, these processes were not observed in the Arg allele-carrying mice. These results reveal that the Tp53 Arg72Pro SNP determines neovascularization, brain repair and neurological recovery after ICH. This study is the first in which the Pro allele of Tp53 is linked to vascular repair and ability to functionally recover from stroke.


Assuntos
Hemorragia Cerebral/patologia , Neovascularização Fisiológica , Proteína Supressora de Tumor p53/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose/efeitos dos fármacos , Arginina/genética , Arginina/metabolismo , Células da Medula Óssea/citologia , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Hemorragia Cerebral/etiologia , Hemorragia Cerebral/genética , Quimiocina CXCL12/sangue , Quimiocina CXCL12/metabolismo , Colagenases/metabolismo , Colagenases/toxicidade , Modelos Animais de Doenças , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Polimorfismo de Nucleotídeo Único , Prolina/genética , Prolina/metabolismo , Tomografia Computadorizada por Raios X , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Sci Rep ; 5: 12030, 2015 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-26153530

RESUMO

In this work, we report our study of protein expression in rat peri-infarct tissue, 48 h after the induction of permanent focal cerebral ischemia. Two proteomic approaches, gel electrophoresis with mass spectrometry and combined fractional diagonal chromatography (COFRADIC), were performed using tissue samples from the periphery of the induced cerebral ischemic lesions, using tissue from the contra-lateral hemisphere as a control. Several protein spots (3408) were identified by gel electrophoresis, and 11 showed significant differences in expression between peri-infarct and contra-lateral tissues (at least 3-fold, p < 0.05). Using COFRADIC, 5412 proteins were identified, with 72 showing a difference in expression. Apart from blood-related proteins (such as serum albumin), both techniques showed that the 70 kDa family of heat shock proteins were highly expressed in the peri-infarct tissue. Further studies by 1D and 2D western blotting and immunohistochemistry revealed that only one member of this family (the inducible form, HSP72 or HSP70i) is specifically expressed by the peri-infarct tissue, while the majority of this family (the constitutive form, HSC70 or HSP70c) is expressed in the whole brain. Our data support that HSP72 is a suitable biomarker of peri-infarct tissue in the ischemic brain.


Assuntos
Isquemia Encefálica/patologia , Animais , Western Blotting , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP72/metabolismo , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Proteômica , Ratos , Ratos Sprague-Dawley
15.
J Cell Mol Med ; 18(8): 1571-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24889329

RESUMO

Brain ischaemia (stroke) triggers an intense inflammatory response predominately mediated by the accumulation of inflammatory cells and mediators in the ischaemic brain. In this context, regulatory T (Treg) cells, a subpopulation of CD4(+) T cells with immunosuppressive and anti-inflammatory properties, are activated in the late stages of the disease. To date, the potential therapeutic usefulness of Treg cells has not been tested. In this study, we aimed to investigate whether Treg cells exert protection/repair following stroke. Both the adoptive transfer of Treg cells into ischaemic rats and the stimulation of endogenous T-cell proliferation using a CD28 superagonist reduced the infarct size at 3-28 days following the ischaemic insult. Moreover, T cell-treated animals had higher levels of FoxP3 and lower levels of IL-1ß, CD11b+ and CD68+ cells in the infarcted hemisphere when compared with control animals. However, T-cell treatment did not alter the rate of proliferation of NeuN-, NCAM- or CD31-positive cells, thereby ruling out neurogenesis and angiogenesis in protection. These results suggest that adoptive transfer of T cells is a promising therapeutic strategy against the neurological consequences of stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Infarto da Artéria Cerebral Média/prevenção & controle , Inflamação/imunologia , Neovascularização Patológica/imunologia , Células-Tronco Neurais/imunologia , Acidente Vascular Cerebral/prevenção & controle , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Western Blotting , Isquemia Encefálica/etiologia , Isquemia Encefálica/imunologia , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Técnicas Imunoenzimáticas , Imunossupressores , Infarto da Artéria Cerebral Média/etiologia , Infarto da Artéria Cerebral Média/imunologia , Inflamação/patologia , Ativação Linfocitária , Imageamento por Ressonância Magnética , Masculino , Células-Tronco Neurais/patologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/imunologia , Linfócitos T Reguladores/patologia
16.
Nanomedicine ; 10(4): 851-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24365481

RESUMO

In order to provide sufficient sensibility for detection, selection of an adequate payload of imaging probe is critical, during the design of MRI theranostic nanoplatforms. This fact is particularly crucial for in vivo applications in the brain, where delivery of macromolecules is limited by the blood-brain barrier. Here we report a simple and quick process for the estimation of adequate payloads of gadolinium in liposomes with potential to act as theranostic agents, for in vivo MRI applications in the brain. Our studies show that an excessive payload of gadolinium in liposomes may actually have a negative influence on in vivo T1 contrast. By preparing and characterizing 4 different liposomal compositions of increasing Gadolinium loads, we show that a superior sensitivity for in vivo detection of MRI theranostic molecules can be quickly improved by adjusting the payload of imaging probe in the molecules. FROM THE CLINICAL EDITOR: This team of authors report the development of a simple and quick process for the estimation of adequate payloads of gadolinium in liposomes as theranostic agents for in vivo brain MRI studies, using a rodent model.


Assuntos
Encéfalo/diagnóstico por imagem , Meios de Contraste/farmacologia , Gadolínio/farmacologia , Imageamento por Ressonância Magnética/métodos , Nanopartículas , Animais , Meios de Contraste/química , Gadolínio/química , Lipossomos , Masculino , Radiografia , Ratos Sprague-Dawley
17.
Theranostics ; 4(1): 90-105, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24396517

RESUMO

The use of theranostics in neurosciences has been rare to date because of the limitations imposed on the free delivery of substances to the brain by the blood-brain barrier. Here we report the development of a theranostic system for the treatment of stroke, a leading cause of death and disability in developed countries. We first performed a series of proteomic, immunoblotting and immunohistological studies to characterize the expression of molecular biomarkers for the so-called peri-infarct tissue, a key region of the brain for stroke treatment. We confirmed that the HSP72 protein is a suitable biomarker for the peri-infarct region, as it is selectively expressed by at-risk tissue for up to 7 days following cerebral ischemia. We also describe the development of anti-HSP72 vectorized stealth immunoliposomes containing imaging probes to make them traceable by conventional imaging techniques (fluorescence and MRI) that were used to encapsulate a therapeutic agent (citicoline) for the treatment of cerebral ischemia. We tested the molecular recognition capabilities of these nano-platforms in vitro together with their diagnostic and therapeutic properties in vivo, in an animal model of cerebral ischemia. Using MRI, we found that 80% of vectorized liposomes were located on the periphery of the ischemic lesion, and animals treated with citicoline encapsulated on these liposomes presented lesion volumes up to 30% smaller than animals treated with free (non-encapsulated) drugs. Our results show the potential of nanotechnology for the development of effective tools for the treatment of neurological diseases.


Assuntos
Infarto da Artéria Cerebral Média/tratamento farmacológico , Nanocápsulas/uso terapêutico , Animais , Biomarcadores/metabolismo , Células Cultivadas , Citidina Difosfato Colina/farmacocinética , Citidina Difosfato Colina/uso terapêutico , Proteínas de Choque Térmico HSP72/genética , Proteínas de Choque Térmico HSP72/metabolismo , Infarto da Artéria Cerebral Média/diagnóstico , Infarto da Artéria Cerebral Média/metabolismo , Lipossomos/farmacocinética , Lipossomos/uso terapêutico , Imageamento por Ressonância Magnética/métodos , Masculino , Microscopia de Fluorescência/métodos , Nootrópicos/farmacocinética , Nootrópicos/uso terapêutico , Imagem Óptica/métodos , Proteoma/genética , Proteoma/metabolismo , Ratos , Ratos Sprague-Dawley
18.
Neuropharmacology ; 63(8): 1279-85, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22975409

RESUMO

Endothelin-1 (ET-1) is involved on the development of cerebral edema in acute ischemic stroke. As edema is a therapeutic target in cerebral ischemia, our aim was to study the effect of antagonists for ET-1 receptors (Clazosentan® and BQ-788, specific antagonists for receptors A and B, respectively) on the development of edema, infarct volume and sensorial-motor deficits in rats subjected to ischemia by occlusion of the middle cerebral artery (MCAO). We used Wistar rats (280-320 g) submitted to ischemia by intraluminal transient (90 min) MCAO. After ischemia, rats were randomized into 4 groups (n = 6) treated with; 1) control group (saline), 2) Clazosentan® group (10 mg/kg iv), 3) BQ-788 group (3 mg/kg iv), and 4) combined treatment (Clazosentan® 10 mg/kg plus BQ-788 3 mg/kg iv). We observed that rats treated with Clazosentan® showed a reduction of edema, measured by MRI, at 72 h (hours) and at day 7 (both p < 0.0001), and a decrease in the serum levels of ET-1 at 72 h (p < 0.0001) and at day 7 (p = 0.009). The combined treatment also induced a reduction of edema at 24 h (p = 0.004), 72 h (p < 0.0001) and at day 7 (p < 0.0001), a reduction on infarct volume, measured by MRI, at 24 and 72 h, and at day 7 (all p < 0.01), and a better sensorimotor recovery at 24 and 72 h, and at day 7 (all p < 0.01). Moreover, Clazosentan® induced a decrease in AQP4 expression, while BQ-788 induced an increase in AQP9 expression. These results suggest that antagonists for ET-1 receptors may be a good therapeutic target for cerebral ischemia.


Assuntos
Antagonistas do Receptor de Endotelina A , Fármacos Neuroprotetores , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Aquaporinas/metabolismo , Western Blotting , Edema Encefálico/etiologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Isquemia Encefálica/complicações , Isquemia Encefálica/tratamento farmacológico , Dioxanos/farmacologia , Antagonistas do Receptor de Endotelina B , Endotelina-1/metabolismo , Potenciais Somatossensoriais Evocados/efeitos dos fármacos , Processamento de Imagem Assistida por Computador , Infarto da Artéria Cerebral Média/etiologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/prevenção & controle , Imageamento por Ressonância Magnética , Masculino , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/prevenção & controle , Oligopeptídeos/uso terapêutico , Piperidinas/uso terapêutico , Piridinas/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Wistar , Acidente Vascular Cerebral/patologia , Sulfonamidas/farmacologia , Tetrazóis/farmacologia
19.
PLoS One ; 7(8): e44191, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952923

RESUMO

Glutamate excitotoxicity, metabolic rate and inflammatory response have been associated to the deleterious effects of temperature during the acute phase of stroke. So far, the association of temperature with these mechanisms has been studied individually. However, the simultaneous study of the influence of temperature on these mechanisms is necessary to clarify their contributions to temperature-mediated ischemic damage. We used non-invasive Magnetic Resonance Spectroscopy to simultaneously measure temperature, glutamate excitotoxicity and metabolic rate in the brain in animal models of ischemia. The immune response to ischemia was measured through molecular serum markers in peripheral blood. We submitted groups of animals to different experimental conditions (hypothermia at 33°C, normothermia at 37°C and hyperthermia at 39°C), and combined these conditions with pharmacological modulation of glutamate levels in the brain through systemic injections of glutamate and oxaloacetate. We show that pharmacological modulation of glutamate levels can neutralize the deleterious effects of hyperthermia and the beneficial effects of hypothermia, however the analysis of the inflammatory response and metabolic rate, demonstrated that their effects on ischemic damage are less critical than glutamate excitotoxity. We conclude that glutamate excitotoxicity is the key molecular mechanism which is influenced by body temperature during the acute phase of brain stroke.


Assuntos
Reação de Fase Aguda/fisiopatologia , Temperatura Corporal/efeitos dos fármacos , Encéfalo/fisiopatologia , Ácido Glutâmico/toxicidade , Neurotoxinas/toxicidade , Acidente Vascular Cerebral/fisiopatologia , Reação de Fase Aguda/sangue , Reação de Fase Aguda/patologia , Animais , Metabolismo Basal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/sangue , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Ácido Glutâmico/sangue , Hipotermia Induzida , Inflamação/sangue , Inflamação/complicações , Inflamação/patologia , Interleucina-6/sangue , Imageamento por Ressonância Magnética , Masculino , Neurotoxinas/sangue , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/patologia
20.
Neurochem Int ; 60(5): 495-505, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22361061

RESUMO

In the last decades, the interest in the association between body temperature and stroke outcome has reemerged, and the use of animal models has made it possible to know the underlying pathogenic mechanisms involved, most of them with pending confirmation in human clinics. In this work, we will review the effects of hyperthermia and hypothermia and its pathogenesis on ischemic stroke, and the evidence of the efficacy and safety of anti-hyperthermic and hypothermic treatments. We will describe how treatment of hyperthermia on ischemic stroke patients, improves patient comfort and outcome, both in the short and the long term, but new clinical studies are needed in this field. Despite the theoretical and experimental bases in favor of hypothermia for the treatment of brain ischemia, there is no definitive clinical evidence that has proved its benefits, so far. With current knowledge, an objective of a body temperature between 35.5 and 36.5 °C seems an optimal therapeutic target for both hyperthermic and normothermic patients.


Assuntos
Temperatura Corporal , Isquemia Encefálica/fisiopatologia , Humanos , Hipotermia Induzida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...