Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
1.
J Biol Chem ; 298(3): 101721, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35151685

RESUMO

Hypoxia-inducible factor prolyl 4-hydroxylases (HIF-P4Hs) regulate the hypoxic induction of >300 genes required for survival and adaptation under oxygen deprivation. Inhibition of HIF-P4H-2 has been shown to be protective in focal cerebral ischemia rodent models, while that of HIF-P4H-1 has no effects and inactivation of HIF-P4H-3 has adverse effects. A transmembrane prolyl 4-hydroxylase (P4H-TM) is highly expressed in the brain and contributes to the regulation of HIF, but the outcome of its inhibition on stroke is yet unknown. To study this, we subjected WT and P4htm-/- mice to permanent middle cerebral artery occlusion (pMCAO). Lack of P4H-TM had no effect on lesion size following pMCAO, but increased inflammatory microgliosis and neutrophil infiltration was observed in the P4htm-/- cortex. Furthermore, both the permeability of blood brain barrier and ultrastructure of cerebral tight junctions were compromised in P4htm-/- mice. At the molecular level, P4H-TM deficiency led to increased expression of proinflammatory genes and robust activation of protein kinases in the cortex, while expression of tight junction proteins and the neuroprotective growth factors erythropoietin and vascular endothelial growth factor was reduced. Our data provide the first evidence that P4H-TM inactivation has no protective effect on infarct size and increases inflammatory microgliosis and neutrophil infiltration in the cortex at early stage after pMCAO. When considering HIF-P4H inhibitors as potential therapeutics in stroke, the current data support that isoenzyme-selective inhibitors that do not target P4H-TM or HIF-P4H-3 would be preferred.


Assuntos
Barreira Hematoencefálica , Infarto da Artéria Cerebral Média , Doenças Neuroinflamatórias , Prolil Hidroxilases , Acidente Vascular Cerebral , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/metabolismo , Permeabilidade da Membrana Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/metabolismo , Camundongos , Doenças Neuroinflamatórias/enzimologia , Doenças Neuroinflamatórias/metabolismo , Permeabilidade , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Oxid Med Cell Longev ; 2021: 5173035, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34712383

RESUMO

Cerebral ischemic stroke (IS) is still a difficult problem to be solved; energy metabolism failure is one of the main factors causing mitochondrion dysfunction and oxidation stress damage within the pathogenesis of cerebral ischemia, which produces considerable reactive oxygen species (ROS) and opens the blood-brain barrier. Dichloroacetic acid (DCA) can inhibit pyruvate dehydrogenase kinase (PDK). Moreover, DCA has been indicated with the capability of increasing mitochondrial pyruvate uptake and promoting oxidation of glucose in the course of glycolysis, thereby improving the activity of pyruvate dehydrogenase (PDH). As a result, pyruvate flow is promoted into the tricarboxylic acid cycle to expedite ATP production. DCA has a protective effect on IS and brain ischemia/reperfusion (I/R) injury, but the specific mechanism remains unclear. This study adopted a transient middle cerebral artery occlusion (MCAO) mouse model for simulating IS and I/R injury in mice. We investigated the mechanism by which DCA regulates glycolysis and protects the oxidative damage induced by I/R injury through the PDK2-PDH-Nrf2 axis. As indicated from the results of this study, DCA may improve glycolysis, reduce oxidative stress and neuronal death, damage the blood-brain barrier, and promote the recovery of oxidative metabolism through inhibiting PDK2 and activating PDH. Additionally, DCA noticeably elevated the neurological score and reduced the infarct volume, brain water content, and necrotic neurons. Moreover, as suggested from the results, DCA elevated the content of Nrf2 as well as HO-1, i.e., the downstream antioxidant proteins pertaining to Nrf2, while decreasing the damage of BBB and the degradation of tight junction proteins. To simulate the condition of hypoxia and ischemia in vitro, HBMEC cells received exposure to transient oxygen and glucose deprivation (OGD). The DCA treatment is capable of reducing the oxidative stress and blood-brain barrier of HBMEC cells after in vitro hypoxia and reperfusion (H/R). Furthermore, this study evidenced that HBMEC cells could exhibit higher susceptibility to H/R-induced oxidative stress after ML385 application, the specific inhibitor of Nrf2. Besides, the protection mediated by DCA disappeared after ML385 application. To sum up, as revealed from the mentioned results, DCA could exert the neuroprotective effect on oxidative stress and blood-brain barrier after brain I/R injury via PDK2-PDH-Nrf2 pathway activation. Accordingly, the PDK2-PDH-Nrf2 pathway may play a key role and provide a new pharmacology target in cerebral IS and I/R protection by DCA.


Assuntos
Antioxidantes/farmacologia , Encéfalo/efeitos dos fármacos , Ácido Dicloroacético/farmacologia , Glicólise/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Piruvato Desidrogenase Quinase de Transferência de Acetil/metabolismo , Complexo Piruvato Desidrogenase/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/ultraestrutura , Encéfalo/enzimologia , Encéfalo/fisiopatologia , Encéfalo/ultraestrutura , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/ultraestrutura , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , AVC Isquêmico/enzimologia , AVC Isquêmico/patologia , AVC Isquêmico/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais
3.
Neurochem Int ; 150: 105197, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34592333

RESUMO

Intracerebral hemorrhage (ICH) is a devastating disease with high mortality and morbidity. Soluble epoxide hydrolase (sEH) is the key enzyme in the epoxyeicosatrienoic acids (EETs) signaling. sEH inhibition has been demonstrated to have neuroprotective effects against multiple brain injuries. However, its role in the secondary injuries after ICH has not been fully elucidated. Here we tested the hypothesis that 1-Trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl)urea (TPPU), a potent and highly selective sEH inhibitor, suppresses inflammation and the secondary injuries after ICH. Adult male C57BL/6 mice were subjected to a collagenase-induced ICH model. TPPU alleviated blood-brain barrier damage, inhibited inflammatory response, increased M2 polarization of microglial cells, reduced the infiltration of peripheral neutrophils. In addition, TPPU attenuated neuronal injury and promoted functional recovery. The results suggest that sEH may represent a potential therapeutic target for the treatment of ICH.


Assuntos
Barreira Hematoencefálica/enzimologia , Hemorragia Cerebral/enzimologia , Epóxido Hidrolases/antagonistas & inibidores , Epóxido Hidrolases/metabolismo , Doenças Neuroinflamatórias/enzimologia , Compostos de Fenilureia/uso terapêutico , Piperidinas/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Hemorragia Cerebral/patologia , Hemorragia Cerebral/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neuroinflamatórias/patologia , Doenças Neuroinflamatórias/prevenção & controle , Compostos de Fenilureia/farmacologia , Piperidinas/farmacologia
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(10): 159002, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34197964

RESUMO

DHA (docosahexaenoic acid) is an essential fatty acid that is required for the normal development and function of the brain. Because of its inability to synthesize adequate amounts of DHA from the precursors, the brain has to acquire DHA from plasma through the blood brain barrier (BBB). Recent studies demonstrated the presence of a transporter at the BBB that specifically transports DHA into the brain in the form of lysophosphatidylcholine (LPC-DHA). However, the mechanism by which LPC-DHA is generated in the plasma is not known. Our previous studies showed that there are at least three different enzymes - lecithin cholesterol acyltransferase (LCAT), endothelial lipase (EL), and hepatic lipase (HL), which can generate LPC-DHA from sn-2 DHA phosphatidylcholine. Here we determined the relative contributions of these enzymes in the delivery of DHA to the brain by measuring the brain DHA levels in the mice deficient in each of these enzymes. The results show that the brain DHA levels of LCAT-deficient mice or EL-deficient mice were not significantly lower than those of their littermates. However, brain DHA was significantly decreased in HL deficient mice (13.5% of total fatty acids) compared to their littermates (17.1%) (p < 0.002), and further decreased to 8.3% of total fatty acids in mice deficient in both HL and EL. These results suggest that HL activity may be the major source for the generation of LPC-DHA in the plasma necessary for transport into the brain, and EL might contribute to this process in the absence of HL.


Assuntos
Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Lipase/deficiência , Lipase/metabolismo , Lisofosfatidilcolinas/metabolismo , Animais , Barreira Hematoencefálica/enzimologia , Encéfalo/patologia , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/análise , Humanos , Lipase/genética , Fígado/enzimologia , Lisofosfatidilcolinas/análise , Masculino , Camundongos , Camundongos Knockout , Fosfatidilcolina-Esterol O-Aciltransferase/genética , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo
5.
Oxid Med Cell Longev ; 2021: 6655122, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33859779

RESUMO

BACKGROUND: The blood-brain barrier (BBB) regulates the exchange of molecules between the brain and peripheral blood and is composed primarily of microvascular endothelial cells (BMVECs), which form the lining of cerebral blood vessels and are linked via tight junctions (TJs). The BBB is regulated by components of the extracellular matrix (ECM), and matrix metalloproteinase 3 (MMP3) remodels the ECM's basal lamina, which forms part of the BBB. Oxidative stress is implicated in activation of MMPs and impaired BBB. Thus, we investigated whether MMP3 modulates BBB permeability. METHODS: Experiments included in vivo assessments of isoflurane anesthesia and dye extravasation from brain in wild-type (WT) and MMP3-deficient (MMP3-KO) mice, as well as in vitro assessments of the integrity of monolayers of WT and MMP3-KO BMVECs and the expression of junction proteins. RESULTS: Compared to WT mice, measurements of isoflurane usage and anesthesia induction time were higher in MMP3-KO mice and lower in WT that had been treated with MMP3 (WT+MMP3), while anesthesia emergence times were shorter in MMP3-KO mice and longer in WT+MMP3 mice than in WT. Extravasation of systemically administered dyes was also lower in MMP3-KO mouse brains and higher in WT+MMP3 mouse brains, than in the brains of WT mice. The results from both TEER and Transwell assays indicated that MMP3 deficiency (or inhibition) increased, while MMP3 upregulation reduced barrier integrity in either BMVEC or the coculture. MMP3 deficiency also increased the abundance of TJs and VE-cadherin proteins in BMVECs, and the protein abundance declined when MMP3 activity was upregulated in BMVECs, but not when the cells were treated with an inhibitor of extracellular signal related-kinase (ERK). CONCLUSION: MMP3 increases BBB permeability following the administration of isoflurane by upregulating the ERK signaling pathway, which subsequently reduces TJ and VE-cadherin proteins in BMVECs.


Assuntos
Barreira Hematoencefálica/metabolismo , Sistema de Sinalização das MAP Quinases , Metaloproteinase 3 da Matriz/metabolismo , Animais , Barreira Hematoencefálica/enzimologia , Encéfalo/irrigação sanguínea , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Isoflurano/farmacocinética , Isoflurano/farmacologia , Metaloproteinase 3 da Matriz/deficiência , Metaloproteinase 3 da Matriz/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/farmacologia , Proteínas de Junções Íntimas/metabolismo
6.
Neurochem Int ; 144: 104976, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33524473

RESUMO

Blood-brain barrier (BBB) damage at the early stage of ischemic stroke is a vital cause of brain parenchymal injury. The mechanism of BBB disruption has been intensively investigated, but still not fully understood. ß-1, 3-galactosyltransferase 2 (B3galt2) is expressed in the brain, but its role in the pathogenesis of cerebral ischemia remains unknown. In this study, we investigated the role of B3galt2 in cerebral ischemia in mice. Focal cerebral ischemia was induced in mice by middle cerebral artery occlusion (MCAO). B3galt2 protein levels were determined in microvessels which were isolated from ischemic brain at 12, 24 and 72 h after MCAO. Mice were administered lentiviral vectors encoding B3galt2 (LV- B3galt2) or recombinant transforming growth factor-ß1 (r-TGF-ß1) by intracerebroventricular injection. We assessed infarct volume and neurologic deficits on days 1, 3, and 14 after MCAO, blood-brain barrier (BBB) integrity at 12 and 24 h after MCAO, and the levels of TGF-ß1, TGF-ßR(Ⅱ) and p-Smad2/3 at 24 and 72 h after MCAO. Our results indicated that B3galt2 was expressed in brain microvascular endothelial cells and increased in the ischemic microvessels. Overexpression of B3galt2 by LV- B3galt2 administration reduced infarct volume and improved functional outcome after cerebral ischemia. Moreover, the neuroprotective effects were associated with preventing BBB damage. Compared with wild-type (WT) mice, heterozygous B3galt2 knockout (B3galt2-/+) mice not only showed severe BBB damage, neurologic functional deficits, but also showed reduced expression of TGF-ß1, TGF-ßR(Ⅱ) and p-Smad2/3 in microvessels after cerebral ischemia. Pre-administration of r-TGF-ß1 reduced BBB damage, and improved the neurological outcomes in both WT mice and B3galt2-/+ mice after cerebral ischemia. Our results suggested B3galt2 protected against ischemic stroke in mice, and the underlying mechanism might include TGF-ß signaling pathway in brain microvascular endothelial cells.


Assuntos
Barreira Hematoencefálica/enzimologia , Isquemia Encefálica/enzimologia , Isquemia Encefálica/prevenção & controle , N-Acetilgalactosaminiltransferases/biossíntese , Animais , Barreira Hematoencefálica/patologia , Isquemia Encefálica/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , N-Acetilgalactosaminiltransferases/genética
7.
Int J Mol Sci ; 22(3)2021 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-33573368

RESUMO

Neurodegeneration is the pathological condition, in which the nervous system or neuron loses its structure, function, or both, leading to progressive degeneration or the death of neurons, and well-defined associations of tissue system, resulting in clinical manifestations. Neuroinflammation has been shown to precede neurodegeneration in several neurodegenerative diseases (NDs). No drug is yet known to delay or treat neurodegeneration. Although the etiology and potential causes of NDs remain widely indefinable, matrix metalloproteinases (MMPs) evidently have a crucial role in the progression of NDs. MMPs, a protein family of zinc (Zn2+)-containing endopeptidases, are pivotal agents that are involved in various biological and pathological processes in the central nervous system (CNS). The current review delineates the several emerging evidence demonstrating the effects of MMPs in the progression of NDs, wherein they regulate several processes, such as (neuro)inflammation, microglial activation, amyloid peptide degradation, blood brain barrier (BBB) disruption, dopaminergic apoptosis, and α-synuclein modulation, leading to neurotoxicity and neuron death. Published papers to date were searched via PubMed, MEDLINE, etc., while using selective keywords highlighted in our manuscript. We also aim to shed a light on pathophysiological effect of MMPs in the CNS and focus our attention on its detrimental and beneficial effects in NDs, with a special focus on Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), multiple sclerosis (MS), and Huntington's disease (HD), and discussed various therapeutic strategies targeting MMPs, which could serve as potential modulators in NDs. Over time, several agents have been developed in order to overcome challenges and open up the possibilities for making selective modulators of MMPs to decipher the multifaceted functions of MMPs in NDs. There is still a greater need to explore them in clinics.


Assuntos
Barreira Hematoencefálica/patologia , Inibidores de Metaloproteinases de Matriz/uso terapêutico , Metaloproteinases da Matriz/metabolismo , Doenças Neurodegenerativas/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/imunologia , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Inibidores de Metaloproteinases de Matriz/farmacologia , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/imunologia , Neurônios/imunologia , Neurônios/patologia , Resultado do Tratamento
8.
Am J Physiol Cell Physiol ; 320(4): C619-C634, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33406028

RESUMO

Hyperglycemia exacerbates edema formation and worsens neurological outcome in ischemic stroke. Edema formation in the early hours of stroke involves transport of ions and water across an intact blood-brain barrier (BBB), and swelling of astrocytes. We showed previously that high glucose (HG) exposures of 24 hours to 7 days increase abundance and activity of BBB Na+-K+-2Cl- cotransport (NKCC) and Na+/H+ exchange 1 (NHE1). Further, bumetanide and HOE-642 inhibition of these transporters significantly reduces edema and infarct following middle cerebral artery occlusion in hyperglycemic rats, suggesting that NKCC and NHE1 are effective therapeutic targets for reducing edema in hyperglycemic stroke. The mechanisms underlying hyperglycemia effects on BBB NKCC and NHE1 are not known. In the present study we investigated whether serum-glucocorticoid regulated kinase 1 (SGK1) and protein kinase C beta II (PKCßII) are involved in HG effects on BBB NKCC and NHE1. We found transient increases in phosphorylated SGK1 and PKCßII within the first hour of HG exposure, after 5-60 min for SGK1 and 5 min for PKCßII. However, no changes were observed in cerebral microvascular endothelial cell SGK1 or PKCßII abundance or phosphorylation (activity) after 24 or 48 h HG exposures. Further, we found that HG-induced increases in NKCC and NHE1 abundance were abolished by inhibition of SGK1 but not PKCßII, whereas the increases in NKCC and NHE activity were abolished by inhibition of either kinase. Finally, we found evidence that STE20/SPS1-related proline/alanine-rich kinase and oxidative stress-responsive kinase-1 (SPAK/OSR1) participate in the HG-induced effects on BBB NKCC.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Glucose/toxicidade , Proteínas Imediatamente Precoces/metabolismo , Proteína Quinase C beta/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Trocador 1 de Sódio-Hidrogênio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/patologia , Bovinos , Células Cultivadas , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Ativação Enzimática , Humanos , Fosforilação , Transdução de Sinais , Fatores de Tempo
9.
Microvasc Res ; 135: 104144, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33515567

RESUMO

Ischemic stroke is a leading cause of morbidity and mortality worldwide, with oxidative stress playing a key role in the injury mechanism of thrombolytic therapy. There is increasing evidence that oxidative stress damages endothelial cells (ECs), degrades tight junction proteins (TJs), and contributes to increased blood-brain barrier (BBB) permeability. It has been demonstrated that the breakdown of BBB could increase the risk of intracerebral hemorrhagic transformation in ischemic stroke. And an episode of cerebral ischemia/reperfusion (I/R) also initiates oxidative stress-mediated inflammatory processes in ECs, which further promotes BBB disruption and the progression of brain injury. Previous studies have revealed that antioxidants could inhibit ROS generation and attenuate BBB disruption after cerebral I/R. Peroxiredoxin 4 (Prx4) is a member of the antioxidant enzymes family (Prx1-6) and has been characterized to be an efficient H2O2 scavenger. It should be noted that Prx4 may be directly involved in the protection of ECs from the effects of ROS and function in ECs as a membrane-associated peroxidase. This paper reviewed the implication of Prx4 on vascular integrity and neuroinflammation following a cerebral I/R injury.


Assuntos
Barreira Hematoencefálica/enzimologia , Permeabilidade Capilar , Células Endoteliais/enzimologia , Mediadores da Inflamação/metabolismo , AVC Isquêmico/enzimologia , Neuroimunomodulação , Peroxirredoxinas/metabolismo , Traumatismo por Reperfusão/enzimologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Humanos , AVC Isquêmico/imunologia , AVC Isquêmico/patologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Junções Íntimas/enzimologia , Junções Íntimas/imunologia , Junções Íntimas/patologia
10.
J Neuroinflammation ; 17(1): 292, 2020 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-33028343

RESUMO

BACKGROUND: Sepsis-associated encephalopathy (SAE) is an early and frequent event of infection-induced systemic inflammatory response syndrome. Phosphoinositide 3-kinase γ (PI3Kγ) is linked to neuroinflammation and inflammation-related microglial activity. In homeotherms, variations in ambient temperature (Ta) outside the thermoneutral zone lead to thermoregulatory responses, mainly driven by a gradually increasing sympathetic activity, and may affect disease severity. We hypothesized that thermoregulatory response to hypothermia (reduced Ta) aggravates SAE in PI3Kγ-dependent manner. METHODS: Experiments were performed in wild-type, PI3Kγ knockout, and PI3Kγ kinase-dead mice, which were kept at neutral (30 ± 0.5 °C) or moderately lowered (26 ± 0.5 °C) Ta. Mice were exposed to lipopolysaccharide (LPS, 10 µg/g, from Escherichia coli serotype 055:B5, single intraperitoneal injection)-evoked systemic inflammatory response (SIR) and monitored 24 h for thermoregulatory response and blood-brain barrier integrity. Primary microglial cells and brain tissue derived from treated mice were analyzed for inflammatory responses and related cell functions. Comparisons between groups were made with one-way or two-way analysis of variance, as appropriate. Post hoc comparisons were made with the Holm-Sidak test or t tests with Bonferroni's correction for adjustments of multiple comparisons. Data not following normal distribution was tested with Kruskal-Wallis test followed by Dunn's multiple comparisons test. RESULTS: We show that a moderate reduction of ambient temperature triggers enhanced hypothermia of mice undergoing LPS-induced systemic inflammation by aggravated SAE. PI3Kγ deficiency enhances blood-brain barrier injury and upregulation of matrix metalloproteinases (MMPs) as well as an impaired microglial phagocytic activity. CONCLUSIONS: Thermoregulatory adaptation in response to ambient temperatures below the thermoneutral range exacerbates LPS-induced blood-brain barrier injury and neuroinflammation. PI3Kγ serves a protective role in suppressing release of MMPs, maintaining microglial motility and reinforcing phagocytosis leading to improved brain tissue integrity. Thus, preclinical research targeting severe brain inflammation responses is seriously biased when basic physiological prerequisites of mammal species such as preferred ambient temperature are ignored.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Lipopolissacarídeos/toxicidade , Encefalopatia Associada a Sepse/enzimologia , Encefalopatia Associada a Sepse/fisiopatologia , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/fisiopatologia , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Regulação da Temperatura Corporal/efeitos dos fármacos , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Encefalopatia Associada a Sepse/induzido quimicamente
11.
Clin Exp Immunol ; 202(2): 239-248, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32643149

RESUMO

Peroxiredoxins (PRXs) are intracellular anti-oxidative enzymes but work as inflammatory amplifiers under the extracellular condition. To date, the function of PRXs in the pathogenesis of multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) is not fully understood. The aim of this study was to investigate whether PRXs play a role in the pathogenesis of MS and NMOSD. We analyzed levels of PRXs (PRX1, PRX5 and PRX6) in the cerebrospinal fluid (CSF) and serum of 16 patients with MS, 16 patients with NMOSD and 15 patients with other neurological disorders (ONDs). We identified potential correlations between significantly elevated PRXs levels and the clinical variables in patients with MS and NMOSD. Additionally, pathological analyses of PRXs (PRX1-6) in the central nervous system (CNS) were performed using the experimental autoimmune encephalomyelitis (EAE), animal model of MS. We found that serum levels of PRX5 and PRX6 in patients with MS and NMOSD were higher compared with those in patients with ONDs (P < 0·05). Furthermore, high levels of PRX5 and PRX6 were partly associated with blood-brain barrier dysfunction and disease duration in NMOSD patients. No significant elevation was found in CSF PRXs levels of MS and NMOSD. Spinal cords from EAE mice showed remarkable PRX5 staining, especially in CD45+ infiltrating cells. In conclusion, PRX5 and PRX6 may play a role in the pathogeneses of MS and NMOSD.


Assuntos
Encefalomielite Autoimune Experimental/líquido cefalorraquidiano , Esclerose Múltipla/líquido cefalorraquidiano , Neuromielite Óptica/líquido cefalorraquidiano , Peroxirredoxinas/líquido cefalorraquidiano , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/patologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Esclerose Múltipla/patologia , Neuromielite Óptica/patologia , Medula Espinal/enzimologia , Medula Espinal/patologia
12.
Acta Neuropathol ; 140(4): 549-567, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32651669

RESUMO

The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood-brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation.


Assuntos
Barreira Hematoencefálica/enzimologia , Encefalomielite Autoimune Experimental/imunologia , Células Endoteliais/enzimologia , Heme Oxigenase-1/metabolismo , Inflamação/imunologia , Interleucina-1/imunologia , Animais , Barreira Hematoencefálica/imunologia , Encefalomielite Autoimune Experimental/enzimologia , Regulação da Expressão Gênica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia
13.
Int J Biochem Cell Biol ; 125: 105794, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32562769

RESUMO

Neurological disorders are associated with increased oxidative stress. Reactive oxidants damage tissue and promote cell death, but it is apparent that oxidants can have more subtle effects on cell function through the modulation of redox-sensitive signalling pathways. Cells of the blood-brain barrier regulate the brain microenvironment but become dysfunctional during neurological disease. The blood-brain barrier is maintained by many cell types, and is modulated by redox-sensitive pathways, ranging from the cytoskeletal elements responsible for establishing a barrier, to growth factor and cytokine signalling pathways that influence neurovascular cells. During neurological disease, blood-brain barrier cells are exposed to exogenously generated oxidants from immune cells, as well as increasing endogenously oxidant production. These oxidants impair the function of the blood-brain barrier, leading to increased leakage and reduced blood flow. Reducing the impact of oxidants on the function of blood-brain barrier cells may provide new strategies for delaying the progression of neurological disease.


Assuntos
Barreira Hematoencefálica/citologia , Inflamação/metabolismo , Doenças do Sistema Nervoso/metabolismo , Estresse Oxidativo/fisiologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/metabolismo , Morte Celular/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Humanos , Inflamação/enzimologia , Inflamação/imunologia , Microglia/enzimologia , Microglia/metabolismo , Doenças do Sistema Nervoso/enzimologia , Doenças do Sistema Nervoso/fisiopatologia , Neutrófilos/enzimologia , Neutrófilos/metabolismo , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Pericitos/enzimologia , Pericitos/metabolismo , Pericitos/patologia , Transdução de Sinais/genética
14.
Med Sci Monit ; 26: e922009, 2020 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-32036381

RESUMO

BACKGROUND Intra-abdominal hypertension (IAH) is associated with high morbidity and mortality. IAH leads to intra-abdominal tissue damage and causes dysfunction in distal organs such as the brain. The effect of a combined injury due to IAH and traumatic brain injury (TBI) on the integrity of the blood-brain barrier (BBB) has not been investigated. MATERIAL AND METHODS Intracranial pressure (ICP) monitoring, brain water content, EB permeability detection, immunofluorescence staining, real-time PCR, and Western blot analysis were used to examine the effects of IAH and TBI on the BBB in rats, and to characterize the protective effects of basic fibroblast growth factor (bFGF) on combined injury-induced BBB damage. RESULTS Combined injury from IAH and TBI to the BBB resulted in brain edema and increased intracranial pressure. The effects of bFGF on alleviating the rat BBB injuries were determined, indicating that bFGF regulated the expression levels of the tight junction (TJ), adhesion junction (AJ), matrix metalloproteinase (MMP), and IL-1ß, as well as reduced BBB permeability, brain edema, and intracranial pressure. Moreover, the FGFR1 antagonist PD 173074 and the ERK antagonist PD 98059 decreased the protective effects of bFGF. CONCLUSIONS bFGF effectively protected the BBB from damage caused by combined injury from IAH and TBI, and binding of FGFR1 and activation of the ERK signaling pathway was involved in these effects.


Assuntos
Barreira Hematoencefálica/patologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Hipertensão Intra-Abdominal/tratamento farmacológico , Sistema de Sinalização das MAP Quinases , Substâncias Protetoras/uso terapêutico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/enzimologia , Edema Encefálico/complicações , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/enzimologia , Lesões Encefálicas Traumáticas/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Interleucina-1beta/metabolismo , Hipertensão Intra-Abdominal/complicações , Hipertensão Intra-Abdominal/enzimologia , Hipertensão Intra-Abdominal/fisiopatologia , Pressão Intracraniana/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Metaloproteinases da Matriz/metabolismo , Microvasos/patologia , Permeabilidade , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos Sprague-Dawley , Proteínas de Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
FASEB J ; 33(12): 14281-14295, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31661303

RESUMO

Blood-brain barrier dysfunction in epilepsy contributes to seizures and resistance to antiseizure drugs. Reports show that seizures increase brain glutamate levels, leading to barrier dysfunction. One component of barrier dysfunction is overexpression of the drug efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP). Based on our previous studies, we hypothesized that glutamate released during seizures activates cytosolic phospholipase A2 (cPLA2), resulting in P-gp and BCRP overexpression. We exposed isolated rat brain capillaries to glutamate ex vivo and used an in vivo-ex vivo approach of isolating brain capillaries from rats after status epilepticus (SE) and in chronic epileptic (CE) rats. Glutamate increased cPLA2, P-gp, and BCRP protein and activity levels in isolated brain capillaries. We confirmed the role of cPLA2 in the signaling pathway in brain capillaries from male and female mice lacking cPLA2. We also demonstrated, in vivo, that cPLA2 inhibition prevents overexpression of P-gp and BCRP at the blood-brain barrier in rats after status epilepticus and in CE rats. Our data support the hypothesis that glutamate signals cPLA2 activation, resulting in overexpression of blood-brain barrier P-gp and BCRP.-Hartz, A. M. S., Rempe, R. G., Soldner, E. L. B., Pekcec, A., Schlichtiger, J., Kryscio, R., Bauer, B. Cytosolic phospholipase A2 is a key regulator of blood-brain barrier function in epilepsy.


Assuntos
Barreira Hematoencefálica/enzimologia , Epilepsia/enzimologia , Fosfolipases A2 do Grupo IV/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/enzimologia , Capilares/enzimologia , Epilepsia/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Genótipo , Ácido Glutâmico/farmacologia , Fosfolipases A2 do Grupo IV/genética , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
16.
Mol Neurobiol ; 56(12): 8296-8305, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31209784

RESUMO

Lipoprotein receptor transport across the blood-brain barrier (BBB) mediates beta-amyloid (Aß) accumulation in the brain and may be a contributing factor in Alzheimer's disease (AD) pathogenesis. Lipoprotein receptors are susceptible to proteolytic shedding at the cell surface, which precludes the endocytic transport of ligands. A ligand that closely interacts with the lipoprotein receptors is apolipoprotein E (apoE), which exists as three isoforms (apoE2, apoE3, apoE4). Our prior work showed an inverse relationship between lipoprotein receptor shedding and Aß transport across the BBB, which was apoE-isoform dependent. To interrogate this further, the current studies investigated an enzyme implicated in lipoprotein receptor shedding, matrix metalloproteinase 9 (MMP9). Treatment with MMP9 dose-dependently elevated lipoprotein receptor shedding in brain endothelial cells and freshly isolated mouse cerebrovessels. Furthermore, treatment with a MMP9 inhibitor (SB-3CT) mitigated Aß-induced lipoprotein receptor shedding in brain endothelial cells and the brains of apoE4 animals. In terms of BBB transit, SB-3CT treatment increased the transport of Aß across an in vitro model of the BBB. In vivo, administration of SB-3CT to apoE4 animals significantly enhanced Aß clearance from the brain to the periphery following intracranial administration of Aß. The current studies show that MMP9 impacts lipoprotein receptor shedding and Aß transit across the BBB, in an apoE  isoform-specific manner. In total, MMP9 inhibition can facilitate Aß clearance across the BBB, which could be an effective approach to lowering Aß levels in the brain and mitigating the AD phenotype, particularly in subjects carrying the apoE4 allele.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Peptídeos beta-Amiloides/sangue , Animais , Apolipoproteínas E/metabolismo , Células Endoteliais/enzimologia , Feminino , Proteína Glial Fibrilar Ácida/sangue , Humanos , Masculino , Camundongos Transgênicos , Modelos Biológicos , Receptores de Lipoproteínas/metabolismo , Solubilidade , Transcitose
17.
J Am Heart Assoc ; 8(9): e011801, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-31030600

RESUMO

Background Systemic innate immune priming is a recognized sequela of post-ischemic neuroinflammation and contributor to delayed neurodegeneration. Given mounting evidence linking acute stroke with reactive lung inflammation, we asked whether enhanced expression of the endogenous antioxidant extracellular superoxide dismutase 3 (SOD3) produced by alveolar type II pneumocytes would protect the lung from transient global cerebral ischemia and the brain from the delayed effects of ischemia-reperfusion. Methods and Results Following 15 minutes of global cerebral ischemia or sham conditions, transgenic SOD3 and wild-type mice were followed daily for changes in weight, core temperature, and neurological function. Three days after reperfusion, arterial and venous samples were collected for complete blood counts, flow cytometry, and SOD3 protein blotting, and immunohistochemistry was performed on lung and brain tissue to assess tissue injury, blood-brain barrier permeability, and neutrophil transmigration. Relative to ischemic controls, transgenic SOD3 mice performed better on functional testing and exhibited reduced peripheral neutrophil activation, lung inflammation, and blood-brain barrier leak. Once released from the lung, SOD3 was predominantly not cell associated and depleted in the venous phase of circulation. Conclusions In addition to reducing the local inflammatory response to cerebral ischemia, targeted enrichment of SOD3 within the lung confers distal neuroprotection against ischemia-reperfusion injury. These data suggest that therapies geared toward enhancing adaptive lung-neurovascular coupling may improve outcomes following acute stroke and cardiac arrest.


Assuntos
Isquemia Encefálica/enzimologia , Encéfalo/enzimologia , Pulmão/enzimologia , Acoplamento Neurovascular , Pneumonia/prevenção & controle , Traumatismo por Reperfusão/prevenção & controle , Superóxido Dismutase/metabolismo , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Encéfalo/imunologia , Encéfalo/patologia , Edema Encefálico/enzimologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Permeabilidade Capilar , Modelos Animais de Doenças , Humanos , Imunidade Inata , Pulmão/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infiltração de Neutrófilos , Pneumonia/enzimologia , Pneumonia/imunologia , Pneumonia/patologia , Edema Pulmonar/enzimologia , Edema Pulmonar/patologia , Edema Pulmonar/prevenção & controle , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Superóxido Dismutase/genética
18.
J Clin Invest ; 129(4): 1772-1778, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30882367

RESUMO

Ischemic stroke is a predominant cause of disability worldwide, with thrombolytic or mechanical removal of the occlusion being the only therapeutic option. Reperfusion bears the risk of an acute deleterious calcium-dependent breakdown of the blood-brain barrier. Its mechanism, however, is unknown. Here, we identified type 5 NADPH oxidase (NOX5), a calcium-activated, ROS-forming enzyme, as the missing link. Using a humanized knockin (KI) mouse model and in vitro organotypic cultures, we found that reoxygenation or calcium overload increased brain ROS levels in a NOX5-dependent manner. In vivo, postischemic ROS formation, infarct volume, and functional outcomes were worsened in NOX5-KI mice. Of clinical and therapeutic relevance, in a human blood-barrier model, pharmacological NOX inhibition also prevented acute reoxygenation-induced leakage. Our data support further evaluation of poststroke recanalization in the presence of NOX inhibition for limiting stroke-induced damage.


Assuntos
Barreira Hematoencefálica/enzimologia , Cálcio/metabolismo , NADPH Oxidase 5/metabolismo , Acidente Vascular Cerebral/enzimologia , Animais , Barreira Hematoencefálica/patologia , Humanos , Camundongos , Camundongos Transgênicos , NADPH Oxidase 5/genética , Espécies Reativas de Oxigênio/metabolismo , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
19.
Sci Rep ; 9(1): 2567, 2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30796241

RESUMO

Cocaine use is associated with breach in the blood brain barrier (BBB) and increased HIV-1 neuro-invasion. We show that the cellular enzyme "Prolidase" plays a key role in cocaine-induced disruption of the BBB. We established a barrier model to mimic the BBB by culturing human brain microvascular endothelial cells (HBMECs) in transwell inserts. In this model, cocaine treatment enhanced permeability of FITC-dextran suggesting a breach in the barrier. Interestingly, cocaine treatment increased the activity of matrix metallo-proteinases that initiate degradation of the BBB-associated collagen. Cocaine exposure also induced prolidase expression and activity in HBMECs. Prolidase catalyzes the final and rate-limiting step of collagen degradation during BBB remodeling. Knock-down of prolidase abrogated cocaine-mediated increased permeability suggesting a direct role of prolidase in BBB breach. To decipher the mechanism by which cocaine regulates prolidase, we probed the inducible nitric oxide synthase (iNOS) mediated phosphorylation of prolidase since mRNA levels of the protein were not altered upon cocaine treatment. We observed increased iNOS expression concurrent with increased prolidase phosphorylation in cocaine treated cells. Subsequently, inhibition of iNOS decreased prolidase phosphorylation and reduced cocaine-mediated permeability. Finally, cocaine treatment increased transmigration of monocytic cells through the HBMEC barrier. Knock-down of prolidase reduced cocaine-mediated monocyte transmigration, establishing a key role of prolidase in cocaine-induced breach in endothelial cell barrier.


Assuntos
Barreira Hematoencefálica/enzimologia , Cocaína/efeitos adversos , Dipeptidases/biossíntese , Células Endoteliais/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Microvasos/enzimologia , Barreira Hematoencefálica/lesões , Barreira Hematoencefálica/patologia , Cocaína/farmacologia , Células Endoteliais/patologia , Humanos , Microvasos/lesões , Microvasos/patologia , Monócitos/metabolismo , Monócitos/patologia , Óxido Nítrico Sintase Tipo II/biossíntese , Células THP-1 , Migração Transendotelial e Transepitelial/efeitos dos fármacos
20.
J Appl Toxicol ; 39(7): 966-973, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30784107

RESUMO

Graphene-based nanomaterials hold the potential to be used in a wide variety of applications, including biomedical devices. Pristine graphene (PG) is an un-functionalized, defect-free type of graphene that could be used as a material for neural interfacing. However, the neurotoxic effects of PG, particularly to the blood-brain barrier (BBB), have not been fully studied. The BBB separates the brain tissue from the circulating substances in the blood and is essential to maintain the brain homeostasis. The principal components of the BBB are brain microvascular endothelial cells (BMVECs), which maintain a protectively low permeability due to the expression of tight junction proteins. Here we analyzed the effects of PG on BMVECs in an in vitro model of the BBB. BMVECs were treated with PG at 0, 10, 50 and 100 µg/mL for 24 hours and viability and functional analyses of BBB integrity were performed. PG increased lactate dehydrogenase release at 50 and 100 µg/mL, suggesting the induction of necrosis. Surprisingly, 2,3,-bis(2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino)-carbonyl]-2H-tetrazolium (XTT) conversion was increased at 10 and 50 µg/mL. In contrast, XTT conversion was decreased at 100 µg/mL, suggesting the induction of cell death. In addition, 100 µg/mL PG increased DNA fragmentation, suggesting induction of apoptosis. At the same time, 50 and 100 µg/mL of PG increased the endothelial permeability, which corresponded with a decrease in the expression of the tight junction protein occludin at 100 µg/mL. In conclusion, these results suggest that PG negatively affects the viability and function of the BBB endothelial cells in vitro.


Assuntos
Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Grafite/toxicidade , Microvasos/efeitos dos fármacos , Animais , Apoptose/genética , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/patologia , Encéfalo/irrigação sanguínea , Permeabilidade Capilar/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Grafite/farmacocinética , L-Lactato Desidrogenase/metabolismo , Microvasos/enzimologia , Microvasos/patologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA