Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Obes Metab Syndr ; 33(1): 76-87, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38049179

RESUMO

Background: Remote ischemic postconditioning (RIPostC) may protect the brain from ischemia/reperfusion (I/R) injury. The association between RIPostC and obesity has not yet been extensively studied. Methods: Twelve-week-old male Zucker diabetic fatty (ZDF; n=68) and Zucker diabetic lean (ZDL; n=51) rats were subjected to focal cerebral ischemia for 90 minutes, followed by 24 hours of reperfusion. RIPostC was performed with 5-minute I/R cycles using a tourniquet on the right hind limb. Results: The results showed a negative association between obesity and neurological impairment in ischemic animals. We observed a 70% greater infarct size in ZDF rats compared with their lean counterparts, as evaluated by 2,3,5-triphenyltetrazolium chloride staining. To measure the total fragmented DNA in peripheral lymphocytes, comet assay was performed. Obese rats exhibited higher levels of DNA damage (by approximately 135%) in peripheral blood lymphocytes even before the induction of stroke. RIPostC did not attenuate oxidative stress in the blood in obese rats subjected to ischemia. Focal cerebral ischemia increased core and penumbra tissue glutamate release in the brain and decreased it in the blood of ischemic ZDL rats, and these changes improved following RIPostC treatment. However, changes in blood and tissue glutamate content were not detected in ischemic ZDF rats or after RIPostC intervention. Conclusion: Our findings suggest that obese animals respond more severely to ischemia-reperfusion brain injury. However, obese animals did not achieve neuroprotective benefits of RIPostC treatment.

2.
Neurochem Int ; 173: 105658, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38135159

RESUMO

The successful implementation of remote ischaemic conditioning as a clinical neuroprotective strategy requires a thorough understanding of its basic principles, which can be modified for each patient. The mechanisms of glutamate homeostasis appear to be a key component. In the current study, we focused on the brain-to-blood glutamate shift mediated by glutamate transporters (excitatory amino acid transports [EAATs]) and the effect of remote ischaemic preconditioning (RIPC) as a mediator of ischaemic tolerance. We used model mimicking ischaemia-mediated excitotoxicity (intracerebroventricular administration of glutamate) to avoid the indirect effect of ischaemia-triggered mechanisms. We found quantitative changes in EAAT2 and EAAT3 and altered membrane trafficking of EAAT1 on the cells of the choroid plexus. These changes could underlie the beneficial effects of ischaemic tolerance. There was reduced oxidative stress and increased glutathione level after RIPC treatment. Moreover, we determined the stimulus-specific response on EAATs. While glutamate overdose stimulated EAAT2 and EAAT3 overexpression, RIPC induced membrane trafficking of EAAT1 and EAAT2 rather than a change in their expression. Taken together, mechanisms related to glutamate homeostasis, especially EAAT-mediated transport, represents a powerful tool of ischaemic tolerance and allow a certain amount of flexibility based on the stimulus used.


Assuntos
Proteínas de Transporte de Glutamato da Membrana Plasmática , Precondicionamento Isquêmico , Humanos , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Ácido Glutâmico/toxicidade , Ácido Glutâmico/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 3 de Aminoácido Excitatório/metabolismo , Aminoácidos Excitatórios , Isquemia
3.
Neurochem Res ; 48(12): 3560-3570, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37528283

RESUMO

Remote ischaemic conditioning (RIC) becomes an attractive strategy for the endogenous stimulation of mechanisms protecting neurons against ischaemia. Although the processes underlying the RIC are not clearly understood, the homeostasis of glutamate seems to play an important role. The present study is focused on the investigation of the brain to blood efflux of glutamate in a condition mimicking ischaemia-mediated excitotoxicity and remote ischaemic preconditioning (RIPC). The animals were pre-treated with a hind-limb tourniquet one hour before the intraventricular administration of glutamate and its release was monitored as the concentration of glutamate/glutathione in blood and liquor for up to 1 h. The transport mediated by excitatory amino acid transporters (EAATs) was verified by their inhibition with Evans Blue intraventricular co-administration. RIPC mediated the efflux of glutamate exceeding from CSF to blood in the very early stage of intoxication. As a consequence, the blood level of glutamate rose in a moment. EAATs inhibition confirmed the active role of glutamate transporters in this process. In the blood, elevated levels of glutamate served as a relevant source of antioxidant glutathione for circulating cells in RIPC-treated individuals. All of those RIPC-mediated recoveries in processes of glutamate homeostasis reflect the improvement of oxidative stress, suggesting glutamate-accelerated detoxication to be one of the key mechanisms of RIPC-mediated neuroprotection.


Assuntos
Ácido Glutâmico , Precondicionamento Isquêmico , Humanos , Animais , Encéfalo , Isquemia , Glutationa
4.
Biomolecules ; 12(10)2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36291633

RESUMO

We have recently shown that the blood cell-derived secretome of remote ischaemic (RIC)-conditioned individuals provides an external source of neuroprotection. In this study, we identified the bioactive compounds from the total proteins released by those cells. Our main strategy was to separate protein-protein complexes while maintaining their native structure and testing their bioactive properties. Subsequently, we identified up- and downregulated bioactive proteins. We uncovered two bioactive fractions composed of 18 proteins. Most of the protein peaks were unchanged; however, RIC mediated a decrease in two peaks (comprising seven proteins) and an increase in one peak (identified as haptoglobin). When focussing on the biological activity of these proteins, we found positive impacts on the regulation of cellular metabolic processes and an increase in biological processes related to the acute phase response and inflammation in the RIC-treated samples. Although we have identified the 18 proteins that exert the greatest cytoprotection, additional studies are needed to elucidate their particular function and detailed mechanisms of action.


Assuntos
Fármacos Neuroprotetores , Animais , Ratos , Fármacos Neuroprotetores/farmacologia , Haptoglobinas , Secretoma , Neuroproteção , Células Sanguíneas
5.
Neurochem Int ; 142: 104927, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33259861

RESUMO

Recently, the function of blood cells in remote ischemic conditioning (RIC) mediated neuroprotection was undoubtedly confirmed. In the present paper, we have focused on the role of blood elements in glutamate homeostasis. The blood of remote conditioned (tolerant) animals was incubated ex vivo with 100 µM glutamate, and the quantitative and qualitative changes of excitatory amino acid transporters (EAAT 1, 2, and 3) were determined. We confirmed RIC mediated accelerated sequestration of extracellular glutamate via EAATs and altered distribution of that amino acid between plasma and cell elements compared to non-tolerant counterparts. The activity of EAATs was elevated in erythrocytes and monocytes, while the density of transporters was not affected. Quantitative changes of EAAT1 density were detected solely in platelets where the forced scavenging was independent of EAATs inhibition. Surprisingly, the trafficking of immunovisualised EAAT2 and 3 raised at tolerant erythrocytes and monocytes. We have found that protein synthesis underlined this process. On the other hand, depletion of protein synthesis did not significantly affect the scavenging capacity of those cell populations. Our work has demonstrated that the elevated blood scavenging of glutamate overdose could be one of the potential mechanisms underlying RIC mediated tissue protection.


Assuntos
Proteínas de Transporte de Glutamato da Membrana Plasmática/sangue , Ácido Glutâmico/sangue , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Precondicionamento Isquêmico/métodos , Animais , Transporte Biológico/fisiologia , Masculino , Ratos , Ratos Wistar , Fatores de Tempo
6.
Mol Biol Rep ; 47(7): 5385-5395, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32627140

RESUMO

Glutamate represents the main excitatory neurotransmitter in the mammalian brain; however, its excessive elevation in the extracellular space is cytotoxic and can result in neuronal death. The ischemia initiated brain damage reflects changes in glutamate concentration in peripheral blood. This paper investigated the role of the brain in blood efflux of the glutamate in an improved tolerance of the brain tissue to ischemic conditions. In the rat model of focal brain ischemia, the neuroprotection was initiated by rapid remote ischemic preconditioning (rRIPC). Our results confirmed a strong neuroprotective effect of rRIPC. We observed reduced infarction by about 78% related to improved neuronal survival by about 70% in the ischemic core. The level of tissue glutamate in core and penumbra dropped significantly and decreased to control value also in the core region of the contralateral hemisphere. Despite significant improvement of blood-brain barrier integrity (by about 76%), the additional gain of glutamate content in the peripheral blood was caused by rRIPC. Based on our results, we can assume that neuroprotection mediated by rapid remote ischemic preconditioning could lie in the regulated, whole-brain release of glutamate from nerve tissue to the blood, which preserves neurons from the exposure to glutamate toxicity and results in reduced infarction.


Assuntos
Isquemia Encefálica/metabolismo , Ácido Glutâmico/metabolismo , Animais , Encéfalo/fisiopatologia , Isquemia Encefálica/fisiopatologia , Morte Celular/efeitos dos fármacos , Ácido Glutâmico/sangue , Ácido Glutâmico/toxicidade , Precondicionamento Isquêmico/métodos , Masculino , Neurônios/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
7.
J Neurochem ; 154(1): 99-111, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31600838

RESUMO

The indirect use of the protective potential of stem cells in the form of cell secretomes has become an attractive strategy in regenerative medicine. In the present work, we studied the paracrine activity of blood cells that could be modulated towards a neuroprotective nature using in vivo remote conditioning (i.e. tolerant blood cells). The increased neuronal survival mediated by the tolerant secretome was clearly confirmed in vitro in a model of glutamate toxicity in a primary culture of rat cortical neurons and in vivo in a pre- and post-treatment of rats that were subjected to transient occlusion of the middle cerebral artery. Bioinformatic-based analysis of the protein profile revealed higher amounts of proteins released by the tolerant blood cells; 29 proteins were recognised as secreted. More than half of these secreted proteins were involved in the biological processes of the response to the stimulus (GO:0050896) and the response to chemicals (GO:0042221). The protective phenotype was most likely mediated by the synergistic effect of multiple identified proteins, including unique to the tolerant secretome (ceruloplasmin, D-3-phosphoglycerate dehydrogenase) and was promoted by the co-participation of several reaction pathways. The most probably of these pathways were post-translation protein modification, MAP2K and MAPK activation and platelet activation. Taken together, our results demonstrate that properly stimulated blood cells could serve as a source for cell-free-based therapies of regenerative medicine.


Assuntos
Células Sanguíneas/metabolismo , Isquemia Encefálica , Precondicionamento Isquêmico/métodos , Neurônios/efeitos dos fármacos , Neuroproteção , Proteoma/farmacologia , Animais , Encéfalo , Células Cultivadas , Masculino , Comunicação Parácrina/fisiologia , Plasma/metabolismo , Proteoma/metabolismo , Ratos , Ratos Wistar
8.
Mol Neurobiol ; 57(2): 752-764, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31471878

RESUMO

PPARGC1A encodes a transcriptional co-activator also termed peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1-alpha (PGC-1α) which orchestrates multiple transcriptional programs. We have recently identified CNS-specific transcripts that are initiated far upstream of the reference gene (RG) promoter. The regulation of these isoforms may be relevant, as experimental and genetic studies implicated the PPARGC1A locus in neurodegenerative diseases. We therefore studied cis- and trans-regulatory elements activating the CNS promoter in comparison to the RG promoter in human neuronal cell lines. A naturally occurring variable guanidine thymidine (GT) repeat polymorphism within a microsatellite region in the proximal CNS promoter increases promoter activity in neuronal cell lines. Both the RG and the CNS promoters are activated by ESRRA, and the PGC-1α isoforms co-activate ESRRA on their own promoters suggesting an autoregulatory feedback loop. The proximal CNS, but not the RG, promoter is induced by FOXA2 and co-activated by PGC-1α resulting in robust activation. Furthermore, the CNS, but not the RG, promoter is targeted by the canonical hypoxia response involving HIF1A. Importantly, the transactivation by HIF1A is modulated by the size of the GT polymorphism. Increased expression of CNS-specific transcripts in response to hypoxia was observed in an established rat model, while RG transcripts encoding the full-length reference protein were not increased. These results suggest a role of the CNS region of the PPARGC1A locus in ischemia and warrant further studies in humans as the activity of the CNS promoter as well as its induction by hypoxia is subject to inter-individual variability due to the GT polymorphism.


Assuntos
Sistema Nervoso Central/metabolismo , Regulação da Expressão Gênica , Hipóxia/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Polimorfismo Genético , Sequências Repetitivas de Ácido Nucleico/genética , Animais , Sequência de Bases , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Linhagem Celular , Ciclopirox/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Especificidade de Órgãos/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar
9.
J Chem Neuroanat ; 101: 101666, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31415804

RESUMO

Stroke induces widespread changes in the brain. In this paper, we monitored some markers of early (2 h) and delayed events (1, 3 and 7 days of reperfusion) initiated by middle cerebral artery occlusion in core/penumbra counterparts of the non-ischemic hemisphere (i.e. contra-core and contra-penumbra). Our results showed that a profound transient drop (2 h and 3 days) of protein synthesis was measured in the contra-core, while the contra-penumbra exhibited translation over-activity at the same time. Glutamate release was detected only in the contra-core, with a peak on the first day. Degenerating neurons became visible in the striatum (day 1), followed by cortex (day 3), earlier in contra-penumbra and later in contra-core. Moreover, the loss of NADPH diaphorase-positive neurons in the non-ischemic hemisphere was detected, with the greatest drop at the first day. Total microglia also started to fall, the earliest in the contra-penumbra region of the striatum (day 1), followed by the contra-core of the striatum and both cortex regions at the seventh day. In conclusion, transient focal ischemia affects remote regions of the brain and initiates processes involved in neuronal degeneration in an order which corresponds to the tissue sensitivity to ischemia, namely earlier in the contra-penumbra, and afterwards in the contra-core. The mechanism of secondary damage would influence the progressive neuronal loss of more distant brain regions.


Assuntos
Isquemia Encefálica/patologia , Lateralidade Funcional , Degeneração Neural/patologia , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Wistar
10.
Neurochem Int ; 129: 104461, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31095978

RESUMO

It has been shown that ischemia of remote organs can generate resistance to ischemic conditions within sensitive brain tissues. However, only limited information about its mechanism is available. In the present paper, we used hind-limb ischemia by tourniquet to generate early remote ischemic tolerance in rats. The main objective was to investigate the role of glutamate in the process of neuroprotection and discover parameters that are affected in the blood of ischemia-affected animals. Our results showed that pretreatment with a hind-limb tourniquet caused a decrease in neurodegeneration by about 30%. However, we did not observe neurological deficit recovery. When compared to ischemia, glutamate concentration decreased in all observed brain regions (cortex, CA1 and dentate gyrus of hippocampus), regardless of their sensitivity to blood restrictions. In contrast to this, the blood levels raised significantly from 26% to 29% during the first four days of postischemic reperfusion. Pretreatment of animals reduced systemic oxidative stress-as represented by lymphocytic DNA damage-by about 80%, while changes in blood antioxidant enzymes (catalase, superoxide dismutase) were not detected. With these data we can further hypothesize that hind-limb-tourniquet preconditioning could accelerate brain-to-blood efflux of glutamate which could positively impact neuronal survival of ischemia-affected brain regions. Moreover, remote preconditioning improved systemic oxidative stress and did not seem to be affected by enzymatic antioxidant defenses in the blood.


Assuntos
Encéfalo/metabolismo , Ácido Glutâmico/sangue , Precondicionamento Isquêmico , Estresse Oxidativo/fisiologia , Animais , Isquemia/tratamento farmacológico , Precondicionamento Isquêmico/métodos , Masculino , Neuroproteção/fisiologia , Ratos Wistar , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Superóxido Dismutase/metabolismo
11.
Eur J Neurosci ; 44(11): 2958-2965, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27706857

RESUMO

Ischemic tolerance (IT) has gained attention as an attractive strategy for improving stroke outcome. Recently, it was shown that signal responsible for rapid IT induction (tolerance induction factor - TIF) is transmitted via circulating blood. In this study, we have hypothesized about the role of the blood cell compartment in TIF production. We used hind-limb ischemia to generate TIF as a rapid preconditioning against transient middle cerebral artery occlusion (MCAO). The essential properties of protein synthesis inhibitors actinomycin D and cycloheximide were utilized to obtain the following results: (i) TIF is proteinaceous. Hind-limb ischemia mediates gene expression followed by translation, resulting in the production of TIF. Blocking of each of these two steps in protein synthesis resulted in rapid infarct evolution (281.5 ± 23.37 and 330.4 ± 71.8 mm3 , respectively). (ii) Tourniquet-treated muscle is not a source of TIF. Actinomicine D injected into rat prior to tolerance induction significantly suppressed RNA synthesis in blood cells and muscle tissue. Cross-circulation of those rats (donors) with control animals (recipients) did not mediate significant infarct reduction (272.9 ± 12.45 mm3 ), even when hind-limb ischemia was performed before MCAO in the recipient (223.2 ± 37.51 mm3 ). (iii) Blood cells serve as a source of TIF. Preischemic transfusion of plasma-free, protein-synthesis-inactive blood cells, which were obtained from tolerant animals did not reduce infarct volume in recipients (131 ± 16.1 mm3 ) in a range comparable with their protein-synthesis-active counterparts (17.2 ± 12 mm3 ). We can conclude that blood cells are associated with the induction of rapid IT via production of a bioactive proteinaceous substance.


Assuntos
Células Sanguíneas/metabolismo , Infarto da Artéria Cerebral Média/sangue , Precondicionamento Isquêmico/métodos , Animais , Células Sanguíneas/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Infarto da Artéria Cerebral Média/terapia , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Wistar
12.
Metab Brain Dis ; 31(6): 1391-1403, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27393013

RESUMO

Pyramidal cells in the CA1 brain region exhibit an ischemic tolerance after delayed postconditioning; therefore, this approach seems to be a promising neuroprotective procedure in cerebral postischemic injury improvement. However, little is known about the effect of postconditioning on protein expression patterns in the brain, especially in the affected hippocampal neurons after global cerebral ischemia. This study is focused on the examination of the ischemia-vulnerable CA1 neuronal layer and on the acquisition of protection from delayed neuronal death after ischemia. Ischemic-reperfusion injury was induced in Wistar rats and bradykinin was applied 2 days after the ischemic insult in an attempt to overcome delayed cell death. Analysis of complex peptide CA1 samples was performed by automated two dimensional liquid chromatography (2D-LC) fractionation coupled to tandem matrix assisted laser desorption/ionization time-of-flight (MALDI TOF/TOF) mass spectrometry instrumentation. We devoted our attention to differences in protein expression mapping in ischemic injured CA1 neurons in comparison with equally affected neurons, but with bradykinin application. Proteomic analysis identified several proteins occurring only after postconditioning and control, which could have a potentially neuroprotective influence on ischemic injured neurons. Among them, the prominent position occupies a regulator of glutamate level aspartate transaminase AATC, a scavenger of glutamate in brain neuroprotection after ischemia-reperfusion. We identified this enzyme in controls and after postconditioning, but AATC presence was not detected in the ischemic injured CA1 region. This finding was confirmed by two-dimensional differential electrophoresis followed by MALDI-TOF/TOF MS identification. Results suggest that bradykinin as delayed postconditioning may be associated with modulation of protein expression after ischemic injury and thus this procedure can be involved in neuroprotective metabolic pathways.


Assuntos
Bradicinina/administração & dosagem , Isquemia Encefálica/enzimologia , Isquemia Encefálica/prevenção & controle , Região CA1 Hipocampal/enzimologia , Pós-Condicionamento Isquêmico/métodos , Proteômica/métodos , Animais , Isquemia Encefálica/patologia , Região CA1 Hipocampal/patologia , Regulação Enzimológica da Expressão Gênica , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Fatores de Tempo
13.
Neurochem Res ; 40(8): 1728-38, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26216051

RESUMO

Brain damage caused by ischemia or toxic agents leads in selectively vulnerable regions to apoptosis-like delayed neuronal death and can result in irreversible damage. Selectively vulnerable neurons of the CA1 area of hippocampus are particularly sensitive to ischemic damage. We investigated the effects of bradykinin (BR) postconditioning on cerebral ischemic and toxic injury. Transient forebrain ischemia was induced by four-vessel occlusion for 10 min and toxic injury was induced by trimethyltin (TMT, 8 µg/kg i.p.). BR as a postconditioner at a dose of 150 µg/kg was applied intraperitoneally 48 h after ischemia or TMT intoxication. Experimental animals were divided into groups according to the length of survival (short-3 and 7 days, and long-28 days survival) and according to the applied ischemic or toxic injury. Glutamate concentration was lowered in both CA1 and dentate gyrus areas of hippocampus after the application of BR postconditioning in both ischemic and toxic brain damage. The number of degenerated neurons in the hippocampal CA1 region was significantly lower in BR-treated ischemic and toxic groups compared to vehicle group. The behavioral test used in our experiments confirms also the memory improvement in conditioned animals. The rats' ability to form spatial maps and learn was preserved, which is visible from our Barnes maze results. By using the methods of delayed postconditioning is possible to stimulate the endogenous protective mechanisms of the organism and induce the neuroprotective effect. In this study we demonstrated that BR postconditioning, if applied before the onset of irreversible neurodegenerative changes, induced neuroprotection against ischemic or toxic injury.


Assuntos
Bradicinina/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Hipóxia Encefálica/tratamento farmacológico , Pós-Condicionamento Isquêmico/métodos , Fármacos Neuroprotetores/uso terapêutico , Animais , Bradicinina/farmacologia , Isquemia Encefálica/metabolismo , Hipóxia Encefálica/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar
14.
J Mol Neurosci ; 57(1): 73-82, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25972121

RESUMO

The impact of therapeutic intervention in stroke depends on its appropriate timing during infarct evolution. We have studied markers of brain tissue damage initiated by permanent occlusion of the middle cerebral artery (MCAO) at three time points during which the infarct spread (1, 3 and 6 h). Based on Evans Blue extravasation and immunohistochemical detection of neurons, we confirmed continuous disruption of blood-brain barrier and loss of neurons in the ischaemic hemisphere that peaked at the sixth hour, especially in the core. Glutamate content started to rise dramatically in the entire hemisphere during the first 3 h; the highest level was determined in the core 6 h after MCAO (141 % increase). Moreover, the enzyme antioxidant defence grew by about 42 % since the first hour in the ipsilateral penumbra. Enzymes of the apoptotic pathway as well as mitochondrial enzyme release were detected since the third hour of MCAO in the ischaemic hemisphere; all achieved their maxima in the penumbra during both time periods (except cytochrome C). In conclusion, the preserved integrity of mitochondrial membrane and incompletely developed process of apoptosis may contribute to the better therapeutic outcome after ischaemic attack; however, a whole brain response should not be omitted.


Assuntos
Infarto da Artéria Cerebral Média/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Permeabilidade Capilar , Ácido Glutâmico/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Ratos , Ratos Wistar
15.
J Neurosci Res ; 93(8): 1250-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25787695

RESUMO

This study provides clear evidence that the factor inducing tolerance to ischemia is transmitted via the circulating blood. By using the remote ischemia and the cross-circulation model, the tolerance to ischemia was transmitted from donor to recipient. For this study, the following experimental groups were designed: I, sham control group; II, group of tolerant hindlimb tourniquet-treated rats; III, positive control group; IV, control for cross-circulation influence; preconditioned animals: V, tolerant animals subjected to middle cerebral artery occlusion (MCAO); VI, tolerant animals cross-circulated with SHC, followed by MCAO; VII, SHC animals cross-circulated with tolerant animals and subsequently subjected to MCAO; VIII, tolerant animals cross-circulated with ischemic rats, followed by MCAO; IX, SHC animals cross-circulated with ischemic animals and subjected to MCAO; postconditioned animals: X, ischemic animals treated with a remote limb tourniquet; XI, ischemic animals cross-circulated with SHC control rats; and XII, ischemic animals cross-circulated with tolerant rats. Results confirmed that remote ischemia induced reduction of infarct volume in the preconditioned (V, 60%) as well as in the postconditioned group (X, 52%). Significant diminution was also observed in group XII (56.6%). In the preconditioned group, decreased infarct volume was detected in groups VI and VII (about 65%) and in group IX (about 50%). The greatest infarct reduction (84%) was induced by the presence of ischemic blood in a tolerant rat before ischemia induction. In summary, the factor inducing tolerance to ischemia is generated by remote ischemia and by ischemia itself; from the site of origin to the rest of the body, it is transported by the systemic blood circulation and can be transferred from animal to animal. The effect of conditioning with two different ischemic events (brain and hindlimb ischemia) led to a cumulative, stronger tolerance response.


Assuntos
Isquemia Encefálica/sangue , Isquemia Encefálica/prevenção & controle , Circulação Cruzada/métodos , Precondicionamento Isquêmico/métodos , Animais , Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular/fisiologia , Ratos , Ratos Wistar
16.
PLoS One ; 10(3): e0121958, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25822952

RESUMO

Eukaryotic initiation factor (eIF) 4E-binding proteins (4E-BPs) are translational repressors that bind specifically to eIF4E and are critical in the control of protein translation. 4E-BP2 is the predominant 4E-BP expressed in the brain, but their role is not well known. Here, we characterized four forms of 4E-BP2 detected by two-dimensional gel electrophoresis (2-DGE) in brain. The form with highest electrophoretic mobility was the main form susceptible to phosphorylation at Thr37/Thr46 sites, phosphorylation that was detected in acidic spots. Cerebral ischemia and subsequent reperfusion induced dephosphorylation and phosphorylation of 4E-BP2 at Thr37/Thr46, respectively. The induced phosphorylation was in parallel with the release of 4E-BP2 from eIF4E, although two of the phosphorylated 4E-BP2 forms were bound to eIF4E. Upon long-term reperfusion, there was a decrease in the binding of 4E-BP2 to eIF4E in cerebral cortex, demonstrated by cap binding assays and 4E-BP2-immunoprecipitation experiments. The release of 4E-BP2 from eIF4E was without changes in 4E-BP2 phosphorylation or other post-translational modification recognized by 2-DGE. These findings demonstrated specific changes in 4E-BP2/eIF4E association dependent and independent of 4E-BP2 phosphorylation. The last result supports the notion that phosphorylation may not be the uniquely regulation for the binding of 4E-BP2 to eIF4E under ischemic stress.


Assuntos
Isquemia Encefálica/metabolismo , Fator de Iniciação 4E em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Fatores de Iniciação em Eucariotos/química , Masculino , Estresse Oxidativo , Fosforilação , Ligação Proteica , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Treonina/química
17.
Acta Histochem ; 116(6): 1062-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24935779

RESUMO

To test the appropriateness of using delayed remote ischemic postconditioning against damage caused to the hippocampus by ischemia or apoptosis inducing intoxication, we chose 10-min normothermic ischemia induced by four-vessel occlusion or kainate injection (8 mg/kg i.p.) in rats. Ischemia alone caused the number of degenerated CA1 neurons after 7 days lasting reperfusion to be significantly (p<0.001) increased by 72.77%. Delayed remote ischemic postconditioning lasting 20 min was able to prevent massive increase in the neurodegeneration. The group with 10 min of ischemia and postconditioning after 2 days of reperfusion had only 15.87% increase in the number of apoptotic neurons. Seven days after kainic acid injection the number of surviving neurons was 42.8% (p<0.001), but the portion of surviving pyramidal cells in the postconditioning group is more than 98%. Our data show that remote postconditioning, performed with 20 min of tourniquet ischemia applied to the hind limb, is a simple method able to effectively stop the onset of neurodegeneration and prevent occurrence of massive muscle cell necrosis, even when used 2 days after the end of the adverse event. Surviving neurons retained a substantial part of their learning and memory ability.


Assuntos
Isquemia Encefálica/terapia , Região CA1 Hipocampal/irrigação sanguínea , Pós-Condicionamento Isquêmico , Traumatismo por Reperfusão/terapia , Animais , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Feminino , Ácido Caínico , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/patologia , Ratos Wistar
18.
Neurochem Int ; 72: 22-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24747342

RESUMO

The goal of this study is to investigate the effects of bradykinin (BR) postconditioning on cerebral ischemic injury. Transient focal cerebral ischemia was induced in rats by 60min of middle cerebral artery occlusion (MCAO), followed by 3days of reperfusion. BR as a postconditioner at a dose of 150µg/kg was applied intraperitoneally 3, 6, 24 and 48h after MCAO. BR postconditioning significantly reduced total infarct volumes if applied 3h after MCAO by 95%, 6h after MCAO by 80% and 24h after MCAO by 70% in versus vehicle group. Neurological functions were amarked improvement in the BR groups compared to the ischemia group. The number of degenerated neurons in the hippocampal CA1 region was also significantly lower in BR-treated ischemic groups compared to vehicle group. BR postconditioning prevented the release of MnSOD from the mitochondria and reduced the activity of the total SOD and CAT if it is administrated short time after stroke. Our data proves the ischemic tolerance in the brain induced by BR postconditioning resulted as effective agent against as strong an attack as 60min MCAO even when used many hours after ischemia.


Assuntos
Bradicinina/farmacologia , Isquemia Encefálica/tratamento farmacológico , Pós-Condicionamento Isquêmico/métodos , Animais , Antígenos Nucleares/metabolismo , Isquemia Encefálica/patologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/patologia , Catalase/metabolismo , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
19.
Gen Physiol Biophys ; 32(1): 139-42, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23531843

RESUMO

We monitored possible influence of transient focal and global brain ischemia on BDNF blood level. In both models noticeable fluctuation of BDNF concentration mainly in reperfusion was observed. During the first 90 min, BDNF in total blood and in blood cells continuously decreased in both models but plasma BDNF raised at 40 min and peaked at 90 min of reperfusion. Our data confirm the impact of transient brain ischemia on BDNF levels in the circulatory system, suggest blood cells as a possible source of BDNF and demonstrate the interdependence of blood compartments and physiological state of an affected organism.


Assuntos
Isquemia Encefálica/sangue , Fator Neurotrófico Derivado do Encéfalo/sangue , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/fisiopatologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Neurônios/metabolismo , Ratos , Ratos Wistar , Reperfusão , Traumatismo por Reperfusão , Espectrofotometria/métodos , Fatores de Tempo
20.
Neurochem Int ; 62(6): 854-60, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23454191

RESUMO

In the clinic delayed post-conditioning would represent an attractive strategy for the survival of vulnerable neurons after an ischemic event. In this paper we studied the impact of ischemia and delayed post-conditioning on blood and brain tissue concentrations of glutamate and protein synthesis. We designed two groups of animals for analysis of brain tissues and blood after global ischemia and post-conditioning, and one for analysis of blood glutamate after transient focal ischemia. Our results showed elevated blood glutamate in two models of transient brain ischemia and decreases in blood glutamate to control in the first 20min of post-conditioning recirculation followed by a consecutive drop of about 20.5% on the first day. Similarly, we recorded reduced protein synthesis in hippocampus and cortex 2 and 3days after ischemia. However, increased glutamate was registered only in the hippocampus. Post-conditioning improves protein synthesis in CA1 and dentate gyrus and, surprisingly, leads to 50% reduction in glutamate in whole hippocampus and cortex. In conclusion, ischemia leads to meaningful elevation of blood and tissue glutamate. Post-conditioning activates mechanisms resulting in rapid elimination of glutamate from brain tissue and/or in the circulatory system that could otherwise impede brain-to-blood glutamate efflux mechanisms. Moreover, post-conditioning induces protein synthesis renewing in ischemia affected tissues that could also contribute to elimination of excitotoxicity. In addition, the potential of glutamate for monitoring the progress of ischemia and efficacy of therapy was shown.


Assuntos
Química Encefálica/fisiologia , Isquemia Encefálica/metabolismo , Ácido Glutâmico/metabolismo , Pós-Condicionamento Isquêmico , Biossíntese de Proteínas/fisiologia , Animais , Região CA1 Hipocampal/metabolismo , Córtex Cerebral/metabolismo , Giro Denteado/metabolismo , Ácido Glutâmico/sangue , Hipocampo/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Ataque Isquêmico Transitório/metabolismo , Masculino , Ratos , Ratos Wistar , Reperfusão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...