Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Fortschr Neurol Psychiatr ; 84(S 02): S77-S79, 2016 Oct.
Artigo em Alemão | MEDLINE | ID: mdl-27806419

RESUMO

The circadian rhythms coordinates the internal physiology to increase the homeostatic capacity thereby providing both a survival advantage to the system and an optimization of energy budgeting. Because multiple-oscillator circadian mechanisms are likely to play a role in regulating human health, and may contribute to the aging process, we give an overview of the relationship between the central clock in the hypothalamus and peripheral clocks for psychological health and aging.


Assuntos
Envelhecimento , Transtorno do Deficit de Atenção com Hiperatividade/fisiopatologia , Ritmo Circadiano , Transtorno do Deficit de Atenção com Hiperatividade/genética , Transtorno do Deficit de Atenção com Hiperatividade/psicologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Humanos
2.
J Neural Transm (Vienna) ; 122 Suppl 1: S47-54, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24337666

RESUMO

To a great extent, cognitive health depends on cerebrovascular health and a deeper understanding of the subtle interactions between cerebrovascular function and cognition is needed to protect humans from one of the most devastating affliction, dementia. However, the underlying biological mechanisms are still not completely clear. Many studies demonstrated that the neurovascular unit is compromised in cerebrovascular diseases and also in other types of dementia. The hemodynamic neurovascular coupling ensures a strong increase of the cerebral blood flow (CBF) and an acute increase in neuronal glucose uptake upon increased neural activity. Dysfunction of cerebral autoregulation with increasing age along with age-related structural and functional alterations in cerebral blood vessels including accumulation of amyloid-beta (Aß) in the media of cortical arterioles, neurovascular uncoupling due to astrocyte endfeet retraction, impairs the CBF and increases the neuronal degeneration and susceptibility to hypoxia and ischemia. A decreased cerebral glucose metabolism is an early event in Alzheimer's disease (AD) pathology and may precede the neuropathological Aß deposition associated with AD. Aß accumulation in turn leads to further decreases in the CBF closing the vicious cycle. Alzheimer, aging and diabetes are also influenced by insulin/insulin-like growth factor-1 signaling, and accumulated evidence indicates sporadic AD is associated with disturbed brain insulin metabolism. Understanding how vascular and metabolic factors interfere with progressive loss of functional neuronal networks becomes essential to develop efficient drugs to prevent cognitive decline in elderly.


Assuntos
Envelhecimento , Encéfalo/metabolismo , Transtornos Cerebrovasculares/patologia , Transtornos Cognitivos/patologia , Demência/patologia , Metabolismo Energético/fisiologia , Transtornos Cerebrovasculares/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Demência/fisiopatologia , Humanos
3.
Biogerontology ; 14(6): 651-62, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24057280

RESUMO

Age is the principal nonmodifiable risk factor for stroke. Over the past 10 years, suitable models for stroke in aged rats have been established. At genetic and cellular level there are significant differences in behavioral, cytological and genomics responses to injury in old animals as compared with the young ones. Behaviorally, the aged rats have the capacity to recover after cortical infarcts albeit to a lower extent than the younger counterparts. Similarly, the increased vulnerability of the aged brain to stroke, together with a decreased interhemisphere synchrony after stroke, assessed by different experimental methods (MRI, fMRI, in vivo microscopy, EEG) leads to unfavorable recovery of physical and cognitive functions in aged people and may have a prognostic value for the recovery of stroke patients. Furthermore, in elderly, comorbidities like diabetes or arterial hypertension are associated with higher risk of stroke, increased mortality and disability, and poorer functional status and quality of life. Aging brain reacts strongly to ischemia-reperfusion injury with an early inflammatory response. The process of cellular senescence can be an important additional contributor to chronic post-stroke by creating a "primed" inflammatory environment in the brain. Overall, these pro-inflammatory reactions promote early scar formation associated with tissue fibrosis and reduce functional recovery. A better understanding of molecular factors and signaling pathways underlying the contribution of comorbidities to stroke-induced pathological sequelae, may be translated into successful treatment or prevention therapies for age-associated diseases which would improve lifespan and quality of life.


Assuntos
Envelhecimento , Neurite (Inflamação)/complicações , Acidente Vascular Cerebral/complicações , Fatores Etários , Animais , Comportamento Animal , Encéfalo/patologia , Encéfalo/fisiopatologia , Comorbidade , Modelos Animais de Doenças , Humanos , Neurite (Inflamação)/genética , Neurite (Inflamação)/fisiopatologia , Neurite (Inflamação)/psicologia , Prognóstico , Recuperação de Função Fisiológica , Fatores de Risco , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/psicologia
4.
J Neurooncol ; 92(2): 137-47, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19043776

RESUMO

In this study, we investigated the protein expression of platelet-derived growth factor receptor (PDGFR), insulin like growth factor-1 receptor (IGF-1R), phosphatidylinositol 3-kinase (PI3-K) and extracellular signal-regulated kinase (ERK1/2) in five primary glioblastoma (GB), with a view to their possible use as therapeutic targets. Our results demonstrated that appreciable levels of these proteins could be detected in the analysed GB cell lines, except for a low level of PDGFR and ERK1/2 expression in one GB cell line. The small molecule inhibitors towards IGF-1R, PDGFR, PI3-K and ERK1/2 respectively, have only modest or no anti-tumour activity on GB cells and therefore their combination with other therapy modalities was analysed. The interaction between small inhibitors and radiation was mostly additive or sub-additive; synergistic interaction was found in five of forty analysed combinations. Our results showed that GB cells are in general resistant to treatment and illustrate the difficulties in predicting the treatment response in malignant gliomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais/fisiologia , Antineoplásicos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Citometria de Fluxo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/efeitos da radiação , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/efeitos da radiação , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/efeitos da radiação , Receptor IGF Tipo 1/efeitos dos fármacos , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 1/efeitos da radiação , Receptores de Fatores de Crescimento/efeitos dos fármacos , Receptores de Fatores de Crescimento/efeitos da radiação , Receptores do Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/efeitos da radiação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação
5.
Rom J Morphol Embryol ; 50(4): 559-82, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19942950

RESUMO

UNLABELLED: Old age is associated with an enhanced susceptibility to stroke and poor recovery from brain injury. Therefore, find therapeutic strategies aimed at improving functional recovery after brain ischemia in aged subjects is of considerable clinical interest. While environmental enrichment has been shown to improve the behavioral outcome of stroke in young animals, the effect of an enriched environment, hypothermia and Granulocyte-Colony Stimulating Factor (G-CSF) on behavioral and neuropathological recovery in aged animals is not known. Focal cerebral ischemia was produced by occlusion of the right middle cerebral artery in 3-month- and 20-month-old male Sprague-Dawley rats. The functional outcome was assessed in neurobehavioral tests conducted over a period of 14-28 days following surgery. Brain tissue then was immunostained for proliferating astrocytes and the infarct and scar tissue volumes were measured. Aged rats showed more severe behavioral impairments and diminished functional recovery compared to young rats. Most infarcted animals had disturbances of sensorimotor function, with recovery beginning later, progressing more slowly, and reaching a lower functional endpoint in aged animals. However, the enriched environment significantly improved the rate and extent of recovery in aged animals. Correlation analysis revealed that the beneficial effect of the enriched environment on recovery, both in young and aged rats, correlated highly with a reduction in infarct size, in the number of proliferating astrocytes, and in the volume of the glial scar. These results suggest that temporally modulating astrocytic proliferation and the ensuing scar formation might be a fruitful approach to improving functional recovery after stroke in aged rats. In aged humans, stroke is a major cause of disability for which no neuroprotective measures are available. A viable alternative to conventional drug-based neuroprotective therapies is brain/body cooling, or hypothermia. In animal studies of focal ischemia, short-term hypothermia consistently reduces infarct size. Nevertheless, efficient neuroprotection requires long-term, regulated lowering of whole body temperature. In this study, we show that two days post-stroke exposure of aged rats to a mixture of air and a mild inhibitor of oxidative phosphorylation, H2S, causes deep hypothermia (27.8+/-0.3 degrees C) and a 50% reduction in infarct size without obvious neurological deficits or physiological side effects. G-CSF treatment after stroke exerted a robust and sustained beneficial effect on survival rate and running function. Transient improvement after G-CSF treatment could be observed for coordinative motor function on the inclined plane test and for working memory in the radial maze test. At cellular level, G-CSF treatment increased the number of proliferating cells in the SVZ and the dentate gyrus and increased the number of newborn neurons in the SVZ, ipsilateral to the lesion. These results suggests that the G-CSF treatment in aged rats has a survival enhancing capacity and a beneficial effect on functional outcome most likely via supportive cellular processes such as neurogenesis. CONCLUSIONS: These findings are important for the further clinical development of the drug in elderly stroke patients. Future studies should focus on an optimization of treatment schedule to achieve a maximum of post-stroke recovery enhancement in aged subjects.


Assuntos
Envelhecimento/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Comportamento/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Modelos Animais de Doenças , Retroalimentação Sensorial/fisiologia , Sulfeto de Hidrogênio/uso terapêutico , Masculino , Atividade Motora/efeitos dos fármacos , Neurogênese/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
6.
Rom J Morphol Embryol ; 50(2): 155-67, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19434306

RESUMO

Aging is associated with a decline of locomotor, sensory and cognitive performance in humans and experimental animals. Mechanistically, organismal senescence is caused by a gradual, lifelong accumulation of multifaceted molecular and cellular damage. Further, the rate and pattern of organismal senescence may be regulated in part by changes in multiple genes involved in multiple processes. While this theory is supported by genetic data in lower organisms, a lack of direct experimental evidence in higher organisms has contributed to a broader acceptance of the "stochastic aging" model, in which accumulating, random damaging biological events play an important role. However, these insults alone cannot account for the inexorable deterioration and loss of function that characterizes old biological systems of higher complexity like humans. Recent advances in unbiased gene expression profiling of the entire genome is a valuable tool for the study of complex biological phenomena such as aging. Using this technology, it is now possible to analyze in detail gene expression at the systems level. In the past decade with the advent of high-throughput technologies, biology has migrated from a descriptive science to a predictive. Most importantly, data from animal models has shown that senescent systems do retain some capacity for regeneration and functional recovery after injuries.


Assuntos
Envelhecimento/fisiologia , Lesões Encefálicas/fisiopatologia , Encéfalo/fisiologia , Regeneração , Animais , Encéfalo/patologia , Lesões Encefálicas/patologia , Humanos , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Modelos Animais , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
7.
Rom J Morphol Embryol ; 49(3): 279-302, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18758632

RESUMO

Old age is associated with an enhanced susceptibility to stroke and poor recovery from brain injury, but the cellular processes underlying these phenomena are uncertain. Therefore studying the basic mechanism underlying functional recovery after brain ischemia in aged subjected it is of considerable clinical interest. Potential mechanisms include neuroinflammation, changes in brain plasticity-promoting factors, unregulated expression of neurotoxic factors, or differences in the generation of scar tissue that impedes the formation of new axons and blood vessels in the infarcted region. Available data indicate that behaviorally, aged rats were more severely impaired by ischemia than were young rats, and they also showed diminished functional recovery. Further, as compared to young rats, aged rats develop a larger infarct area, as well as a necrotic zone characterized by a higher rate of cellular degeneration, and a larger number of apoptotic cells. In both old and young rats, the early intense proliferative activity following stroke leads to a precipitous formation of growth-inhibiting scar tissue, a phenomenon amplified by the persistent expression of neurotoxic factors. Reduced transcriptional activity in the healthy, contralateral hemisphere in conjunction with an early upregulation of DNA damage related genes and the early induction of proapoptotic genes in the periinfarct area of aged rats are likely to account for poor neurorehabilitation after stroke in aged rats. Finally, the regenerative potential of the rat brain is largely preserved up to 20 months of age but gene expression is temporally displaced, has lower amplitude, and is sometimes of relatively short duration. Most interestingly, it has recently been shown that the human brain can respond to stroke with increased progenitor proliferation in aged patients opening the possibilities to utilize this intrinsic attempt for neuroregeneration of the human brain as a potential therapy for stroke. Given the heterogeneity of stroke, a universal anti-inflammatory solution may be a distant prospect, but probably neuroprotective drug cocktails targeting inflammatory pathways in combination with thrombolysis may be a possibility for acute stroke treatment in the future.


Assuntos
Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Recuperação de Função Fisiológica/genética , Recuperação de Função Fisiológica/fisiologia , Reabilitação do Acidente Vascular Cerebral , Idoso , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Apoptose/genética , Apoptose/fisiologia , Barreira Hematoencefálica/patologia , Infarto Encefálico/genética , Infarto Encefálico/patologia , Modelos Animais de Doenças , Humanos , Modelos Biológicos , Neurogênese/genética , Neurogênese/fisiologia , Ratos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
8.
Rom J Morphol Embryol ; 49(1): 27-35, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18273499

RESUMO

In an effort to identify new proteins involved in functional recovery after cerebral ischemia, young (3 months) and aged (18 months) male rats were subjected to middle cerebral artery (MCA) occlusion. Brains were harvested at 3- and 14-days post ischemia and proteins from the peri-infarcted and the corresponding contralateral area and total proteins were analyzed by two-dimensional polyacrylamide gel electrophoresis followed by mass spectrometry analysis. Annexin A3 (ANXA3) was identified as one upregulated protein in the post-ischemic rat brain. Using western blotting, real-time PCR and immunohistochemistry, we confirmed that at 3-14 days post-stroke, ANXA3 expression in the peri-infarct area was consistently increased over the corresponding area of control rats. Double staining revealed that ANXA3 is produced by activated microglial cells. We found that aged rats also had more newly proliferating cells expressing ANXA3 than young rats do. Occasionally, ANXA3-immunopositive cells wraped around neurons, suggesting that annexin A3 may be involved in the removal of dying neurons after stroke.


Assuntos
Envelhecimento/fisiologia , Anexina A3/genética , Encéfalo/metabolismo , Acidente Vascular Cerebral/genética , Fatores Etários , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Anexina A3/metabolismo , Proliferação de Células , Regulação da Expressão Gênica , Masculino , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo
9.
Restor Neurol Neurosci ; 25(5-6): 467-84, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18334765

RESUMO

PURPOSE: Aging is associated with a temporally dysregulated cellular response to ischemia as well as poor functional recovery. While environmental enrichment has been shown to improve the behavioral outcome of stroke in young animals, the effect of an enriched environment on behavioral and neuropathological recovery in aged animals is not known. METHODS: Focal cerebral ischemia was produced by electrocoagulation of the right middle cerebral artery in 3 month- and 20 month-old male Sprague-Dawley rats. The functional outcome was assessed in neurobehavioral tests conducted over a period of 28 days following surgery. Brain tissue was then immunostained for proliferating astrocytes and the infarct and scar tissue volumes were measured. RESULTS: Aged rats showed more severe behavioral impairments and diminished functional recovery compared to young rats. Most infarcted animals had disturbances of sensorimotor function, with recovery beginning later, progressing more slowly, and reaching a lower functional endpoint in aged animals. However, the enriched environment significantly improved the rate and extent of recovery in aged animals. Correlation analysis revealed that the beneficial effect of the enriched environment on recovery, both in young and aged rats, correlated highly with a reduction in infarct size, in the number of proliferating astrocytes, and in the volume of the glial scar. CONCLUSIONS: These results suggest that temporally modulating astrocytic proliferation and the ensuing scar formation might be a fruitful approach to improving functional recovery after stroke in aged rats.


Assuntos
Envelhecimento/fisiologia , Meio Ambiente , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Recuperação de Função Fisiológica/fisiologia , Animais , Comportamento Animal , Encéfalo/patologia , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Bromodesoxiuridina/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida , Masculino , Aprendizagem em Labirinto , Atividade Motora , Análise Multivariada , Exame Neurológico , Desempenho Psicomotor , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
10.
Rom J Morphol Embryol ; 48(1): 17-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17502946

RESUMO

Kindled seizures are widely used to model epileptogenesis, but the molecular mechanisms underlying the attainment of kindling status are largely unknown. Recently we showed that achievement of kindling status in the Sprague-Dawley rat is associated with a critical developmental interval of 25 +/- 1 days; the identification of this long, well-defined developmental interval for inducing kindling status makes possible a dissection of the cellular and genetic events underlying this phenomenon and its relationship to normal and pathological brain function. Now we report the identification, by proteomics, of a modified variant of the Rieske iron-sulfur protein, a component of the mitochondrial cytochrome bc1 complex, whose isoelectric point is shifted toward more alkaline values in the hippocampus of kindled rats. By immunohistochemistry the Rieske protein is well-expressed in the hippocampus except in the CA1 subfield, a region of selective vulnerability to seizures in humans and animal models. We also noted an asymmetric, selective expression of the Rieske protein in the subgranular neurons of the dorsal dentate gyrus, a region implicated in neurogenesis. Abnormal changes in Rieske protein immunoreactivity also were found in sections obtained from human epileptic patients. These results suggest that the Rieske protein may play a role in the response of neurons to seizure activity and could give important new insights into the molecular pathogenesis of epilepsy.


Assuntos
Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Epilepsia/metabolismo , Hipocampo/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Proteínas Mitocondriais/metabolismo , Convulsões/metabolismo , Animais , Epilepsia/induzido quimicamente , Hipocampo/patologia , Humanos , Excitação Neurológica/metabolismo , Excitação Neurológica/patologia , Masculino , Pentilenotetrazol , Isoformas de Proteínas/metabolismo , Proteômica , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
11.
Neuroscience ; 142(4): 1071-9, 2006 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-16997484

RESUMO

Members of various transport protein families including ATP-binding cassette transporters and solute carriers were shown to be expressed in brain capillaries, choroid plexus, astrocytes or neurons, controlling drug and metabolite distribution to and from the brain. However, data are currently very limited on how the expression of these transport systems is affected by damage to the brain such as stroke. Therefore we studied the expression of four selected transporters, P-glycoprotein (Mdr1a/b; Abcb1a/b), Mrp5 (Abcc5), Bcrp (Abcg2), and Oatp2 (Slc21a5) in a rat model for stroke. Transporter expression was analyzed by real-time polymerase chain reaction in the periinfarcted region and protein localization and cellular phenotyping were done by immunohistochemistry and confocal immunofluorescence microscopy. After stroke, P-glycoprotein staining was detected in endothelial cells of disintegrated capillaries and by day 14 in newly generated blood vessels. There was no significant difference, however, in the Mdr1a mRNA amount in the periinfarcted region compared with the contralateral site. For Bcrp, a significant mRNA up-regulation was observed from days 3-14. This up-regulation was followed by the protein as confirmed by quantitative immunohistochemistry. Oatp2, located in the vascular endothelium, was also up-regulated at day 14. For Mrp5, an up-regulation was observed in neurons in the periinfarcted region (day 14). In conclusion, after stroke the transport proteins were up-regulated with a maximum at day 14, a time point that coincides with behavioral recuperation. The study further suggests Bcrp as a pronounced marker for the regenerative process and a possible functional role of Mrp5 in surviving neurons.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Infarto Cerebral/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Biomarcadores/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiopatologia , Encéfalo/fisiopatologia , Capilares/metabolismo , Capilares/fisiopatologia , Proteínas de Transporte/genética , Infarto Cerebral/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Regeneração/fisiologia , Fatores de Tempo , Regulação para Cima/fisiologia
12.
Rom J Morphol Embryol ; 47(3): 215-28, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17308679

RESUMO

Old age is associated with an enhanced susceptibility to stroke and poor recovery from brain injury, but the cellular mechanisms underlying such phenomena are not known. Using BrdU-labeling, quantitative immunohistochemistry and 3D-reconstruction of confocal images in a rat model of mild cerebral ischemia, we found that aged rats are highly susceptible to develop an early infarct that is associated with premature cellular proliferation originating from the vascular tree. In aged rats we also found a rapid delimitation of the infarct area by capillary-derived neuroepithelial cells and an early incorporation of these cells into the glial scar. Since most proliferating cells at the infarct site are microglia or nestin-positive cells derived from the vascular wall, we conclude that the vasculature plays a hitherto unrecognized role as a source of proliferating neuroepithelial cells after stroke. Age-associated alterations in the timing and origin of the cytogenic response to cerebral ischemia may underlie the poor functional recovery from stroke. Clarifying the molecular basis of these phenomena could yield novel approaches to enhancing neurorestoration in the elderly. Studies of stroke in experimental animals have demonstrated the neuroprotective efficacy of a variety of interventions, but most of the strategies that have been clinically tested failed to show benefit in aged humans. One possible explanation for this discrepancy between laboratory and clinical investigations is the role that age plays in the recovery of the brain from insult. Although it is well known that aging is a risk factor for stroke (Barnett HJ, 2002), the majority of experimental studies of stroke have been performed on young animals, and therefore may not fully replicate the effects of ischemia on neural tissue in aged subjects (Wang LC et al., 1995; Davies M et al., 1995; Sutherland GR et al., 1996; Popa-Wagner A et al., 1998, 1999a). Hence, the aged post-acute animal model is clinically most relevant to stroke rehabilitation and cellular studies (Lindner MD et al., 2003; Brown AW et al., 2003; Badan I et al., 2003).


Assuntos
Envelhecimento , Proliferação de Células , Infarto Cerebral/patologia , Animais , Apoptose , Vasos Sanguíneos/citologia , Encéfalo/metabolismo , Contagem de Células , Infarto Cerebral/metabolismo , Proteínas de Ligação a DNA , Marcação In Situ das Extremidades Cortadas , Masculino , Monócitos/citologia , Proteínas do Tecido Nervoso/metabolismo , Células Neuroepiteliais/citologia , Proteínas Nucleares/metabolismo , Ratos , Ratos Sprague-Dawley
13.
J Mol Med (Berl) ; 73(9): 465-71, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8528750

RESUMO

The levels and cellular localization of mRNA for complement C1q and C3 were examined by RNA gel blot and nonradioactive in situ hybridization in the frontal cortex of patients with Alzheimer's disease (AD) and age-matched controls. We found that the hybridization signal for C1q mRNA was markedly increased (approx. 3.5-fold) in the frontal cortex of AD patients compared to that in age-matched controls. In contrast to previous reports we also found that the levels of C3 mRNA, although well expressed, did not differ significantly between AD cases and age-matched controls. Nonradioactive in situ hybridization using digoxigenin-labeled ribo-probes revealed that transcripts coding for both C1q and C3 were closely associated with neurons. These results support the hypothesis that complement could play a role in neuronal degeneration which has been observed in the brain of AD patients.


Assuntos
Doença de Alzheimer/metabolismo , Complemento C1q/biossíntese , Complemento C3/biossíntese , Lobo Frontal/metabolismo , Expressão Gênica , RNA Mensageiro/biossíntese , Idoso , Idoso de 80 Anos ou mais , Northern Blotting , Feminino , Humanos , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Valores de Referência
14.
Sci Rep ; 5: 13281, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26335098

RESUMO

Alzheimer's disease (AD) increases dramatically in patients with ischaemic stroke. Monomeric C-reactive protein (mCRP) appears in the ECM of ischaemic tissue after stroke, associating with microvasculature, neurons and AD-plaques, Aß, also, being able to dissociate native-CRP into inflammatory, mCRP in vivo. Here, mCRP injected into the hippocampal region of mice was retained within the retrosplenial tract of the dorsal 3rd ventrical and surrounding major vessels. Mice developed behavioural/cognitive deficits within 1 month, concomitant with mCRP staining within abnormal looking neurons expressing p-tau and in beta-amyloid 1-42-plaque positive regions. mCRP co-localised with CD105 in microvessels suggesting angiogenesis. Phospho-arrays/Western blotting identified signalling activation in endothelial cells and neurons through p-IRS-1, p-Tau and p-ERK1/2-which was blocked following pre-incubation with mCRP-antibody. mCRP increased vascular monolayer permeability and gap junctions, increased NCAM expression and produced haemorrhagic angiogenesis in mouse matrigel implants. mCRP induced tau244-372 aggregation and assembly in vitro. IHC study of human AD/stroke patients revealed co-localization of mCRP with Aß plaques, tau-like fibrils and IRS-1/P-Tau positive neurons and high mCRP-levels spreading from infarcted core regions matched reduced expression of Aß/Tau. mCRP may be responsible for promoting dementia after ischaemia and mCRP clearance could inform therapeutic avenues to reduce the risk of future dementia.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Neurônios/metabolismo , Receptores Imunológicos/metabolismo , Animais , Biomarcadores/metabolismo , Progressão da Doença , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
J Cereb Blood Flow Metab ; 19(4): 425-34, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10197512

RESUMO

Although stroke in humans usually afflicts the elderly, most experimental studies on the nature of cerebral ischemia have used young animals. This is especially important when studying restorative processes that are age dependent. To explore the potential of older animals to initiate regenerative processes after cerebral ischemia, the authors studied the expression of the juvenile-specific cytoskeletal protein, microtubule-associated protein (MAP) 1B, and the adult-specific protein, MAP2, in male Sprague-Dawley rats at 3 months and 20 months of age. The levels of MAP1B and MAP2 transcripts and the corresponding proteins declined with increasing age in the hippocampus. In the cortex, the levels of the transcripts did not change significantly with age, but the morphologic features of immunostained fibers were clearly affected by age; that is, cortical MAP1B fibers became thicker, and MAP2 fibers, more diffuse, in aged rats. Focal cerebral ischemia, produced by reversible occlusion of the right middle cerebral artery, resulted in a large decrease in the expression of both MAP1B and MAP2 in the infarct core at the messenger ribonucleic acid and protein levels. However, at 1 week after the stroke, there was vigorous expression of MAP1B and its messenger ribonucleic acid, as well as MAP2 protein, in the border zone adjacent to the infarct of 3-month-old and 20 month-old male Sprague-Dawley rats. The upregulation of these key cytologic elements generally was diminished in aged rats compared with young animals, although the morphologic features of fibers in the infarct border zone were similar in both age groups. These results suggest that the regenerative potential of the aged rat brain appears to be competent, although attenuated, at least with respect to MAP1B and MAP2 expression up to 20 months of age.


Assuntos
Envelhecimento/metabolismo , Arteriopatias Oclusivas/metabolismo , Isquemia Encefálica/metabolismo , Doenças Arteriais Cerebrais/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Masculino , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Regulação para Cima
16.
J Cereb Blood Flow Metab ; 23(7): 845-54, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12843788

RESUMO

Following cerebral ischemia, perilesional astrocytes and activated microglia form a glial scar that hinders the genesis of new axons and blood vessels in the infarcted region. Since glial reactivity is chronically augmented in the normal aging brain, the authors hypothesized that postischemic gliosis would be temporally abnormal in aged rats compared to young rats. Focal cerebral ischemia was produced by reversible occlusion of the right middle cerebral artery in 3- and 20-month-old male Sprague Dawley rats. The functional outcome was assessed in neurobehavioral tests at 3, 7, 14, and 28 days after surgery. Brain tissue was immunostained for microglia, astrocytes, oligodendrocytes, and endothelial cells. Behaviorally, aged rats were more severely impaired by stroke and showed diminished functional recovery compared with young rats. Histologically, a gradual activation of both microglia and astrocytes that peaked by days 14 to 28 with the formation of a glial scar was observed in young rats, whereas aged rats showed an accelerated astrocytic and microglial reaction that peaked during the first week after stroke. Oligodendrocytes were strongly activated at early stages of infarct development in all rats, but this activation persisted in aged rats. Therefore, the development of the glial scar was abnormally accelerated in aged rats and coincided with the stagnation of recovery in these animals. These results suggest that a temporally anomalous gliotic reaction to cerebral ischemia in aged rats leads to the premature formation of scar tissue that impedes functional recovery after stroke.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiopatologia , Neuroglia/fisiologia , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/fisiopatologia , Animais , Comportamento Animal/fisiologia , Biomarcadores , Encéfalo/patologia , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Infarto da Artéria Cerebral Média , Macrófagos/fisiologia , Masculino , Testes Neuropsicológicos , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia
17.
Neurobiol Aging ; 24(1): 135-45, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12493559

RESUMO

In this study the role of neurofilaments (NFs) in brain plasticity after cerebral infarction in young and middle aged rats was evaluated. Focal cerebral ischemia was produced by reversible occlusion of the right middle cerebral artery in 3- and 20-month-old male Sprague-Dawley rats. After 1 week, brains were removed and in situ hybridization and immunostaining was performed for NF-68 kDa, 160 kDa and 200 kDa in different phosphorylation states. After focal cerebral ischemia the levels of gene and protein expression of neurofilament proteins were increased in the border zone of the infarcted area compared with the unaffected contralateral site. Furthermore, the level of gene expression was significant lower in aged as in young animals. Focal cerebral ischemia resulted in a clearly increased number of immunostained axons in the penumbral region in both young and aged rats. On the other hand the immunostained apical dendrites became thicker and vacuolization appeared. Our results suggest that that neurofilament proteins are involved in response of brain to focal ischemia.


Assuntos
Envelhecimento/metabolismo , Infarto Cerebral/metabolismo , Proteínas de Neurofilamentos/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Animais , Infarto Cerebral/patologia , Expressão Gênica , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Masculino , Proteínas de Neurofilamentos/classificação , Proteínas de Neurofilamentos/genética , Fosforilação , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
18.
Neuroscience ; 94(2): 395-403, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10579203

RESUMO

The aim of the present study was to assess the age-dependent response of microtubule-associated protein 1B, a plasticity-associated protein deriving from a late gene, following administration of an epileptogenic stimulus. The effect of a single administration of the convulsant pentylenetetrazole on microtubule-associated protein 1B expression in the hippocampal formation and cortex of three-, 18- and 28-month-old rats was assessed using northern blot analysis, in situ hybridization and immunohistochemistry. In three-month-old rats, we detected initial increases in microtubule-associated protein 1B messenger RNA at 15 h following pentylenetetrazole administration in the granule cells of the dentate gyrus, in the CA3 region of the hippocampus and in layers II/III of the entorhinal cortex, and these reached a maximum at 44 h. However, in the hippocampus and cortex of 18-month-old rats, the peak occurred at 15 h, and in the brains of 28-month-old rats a blunted peak was reached at 3 h. Pentylenetetrazole treatment in young rats resulted in a robust induction of microtubule-associated protein 1B immunoreactivity in the granule cells of the dentate gyrus and in layers II/III of the entorhinal cortex, but also produced a large decrease in the retrosplenial cortex. However, following pentylenetetrazole treatment in older rats, the granule cells of the dentate gyrus were nearly devoid of microtubule-associated protein 1B immunoreactivity, whereas the retrosplenial cortex showed no changes at all, and the entorhinal cortex had an expression pattern similar to that of young rats. Aberrant immunolabeling of microtubule-associated protein 1B occurred in cortical layer VI of the aged rats where, unlike in young rats, there was heavy staining of neuronal somata. These results suggest that the regulation of the plasticity-associated protein microtubule-associated protein 1B is altered in the ageing rat brain, with the peak of expression shifted to earlier times in 18-month-old rats and blunted, variable increases at even earlier times in 28-month-old rats.


Assuntos
Envelhecimento/metabolismo , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Pentilenotetrazol/farmacologia , Convulsões/metabolismo , Animais , Northern Blotting , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Proteínas Associadas aos Microtúbulos/análise , Especificidade de Órgãos , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
19.
J Gerontol A Biol Sci Med Sci ; 50(4): B218-23, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7614234

RESUMO

Because fibronectin (FN) mRNA has been shown to be increased in the brain of old rats and during in vivo and in vitro aging of both endothelial cells and fibroblasts, it was of interest to determine what effect age and caloric restriction have on the ability of liver cells to express FN mRNA. The prevalence of fibronectin mRNA containing the alternatively spliced variant V95 (FN-V95) was examined by RNA gel blot hybridization in the liver of 10-mo, 18-mo, 30-mo ad lib-fed (AL) and 30-mo food-restricted (FR) rats. The transcripts coding for FN-V95 mRNA were well expressed in the liver of 10-mo and 18-mo-old rats. The hybridization signal then increased (1.3-fold, p < .05) in the liver of 30-mo AL rats vs 10-mo-old rats. However, the prevalence of FN-V95 mRNA was dramatically decreased (approximately 4.5-fold, p < .01) in the liver of 30-mo food-restricted rats as compared to 30-mo-old AL rats. The distribution of FN-V95 mRNA was also investigated by non-radioactive in situ hybridization (ISH) on liver frozen sections. The hybridization signal for FN-V95 mRNA was evenly distributed among liver cells of 10-mo and 18-mo-old rats, while weak hybridization signals were sparsely scattered on liver sections derived from 30-mo-old FR rats. In contrast, strong hybridization signals showing a localized distribution were detected on liver sections obtained from 30-mo-old AL rats. Histologic examination of cryostat sections also revealed a massive accumulation of lipid droplets in the liver of 30-mo-old AL rats.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Envelhecimento/metabolismo , Fibronectinas/metabolismo , Privação de Alimentos/fisiologia , Fígado/metabolismo , RNA Mensageiro/metabolismo , Animais , Northern Blotting , Feminino , Fibronectinas/genética , Hibridização In Situ , Fígado/patologia , Sondas RNA , Ratos , Ratos Sprague-Dawley
20.
J Gerontol A Biol Sci Med Sci ; 55(5): B242-8, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10819311

RESUMO

The ability of the rodent brain to support plasticity-related phenomena declines with increasing age. Here we investigated the extent to which old rats retain the capacity to initiate transcription for immediate early genes, particularly as it relates to brain plasticity, in response to a strong stimulus. The intraperitoneal administration of pentylenetetrazole (PTZ) to rats of various ages evoked tonic-clonic seizures. Using an RNA gel-blot and in situ hybridization analysis, we found that 1 hour after the onset of seizure, messenger RNA (mRNA) for tissue plasminogen activator (TPA) was increased approximately 3.7-fold in the hippocampi of 3-month-old rats. The levels of TPA mRNA in the hippocampi and cortices of 3-month-old rats returned to control levels by 3 hours after PTZ administration. The levels of TPA mRNA increased 2.5-fold in the hippocampi of 18-month-old rats and 1.8-fold in the brains of the 28-month-old-rats at 3 hours and returned to basal levels by 15 hours following PTZ treatment. Quantitatively similar increases were calculated for the cortex. At peak induction the transcripts were localized throughout the cortical layers of the 3-month-old rats, whereas the TPA mRNA expression was restricted to cortical layer V of the older rats. Our results suggest that although the aging brain retains the capacity to respond to chemically induced seizures, the induction of TPA mRNA is temporarily delayed and the levels are diminished with increasing age. Because TPA has been implicated in neuronal plasticity, this finding suggests that immediate early genes are important factors in the limited plasticity of the aging brain.


Assuntos
Envelhecimento/fisiologia , Convulsões/genética , Ativador de Plasminogênio Tecidual/genética , Análise de Variância , Animais , Northern Blotting , Encéfalo/enzimologia , Indução Enzimática , Feminino , Expressão Gênica , Hibridização In Situ , Plasticidade Neuronal/fisiologia , Pentilenotetrazol , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Ativador de Plasminogênio Tecidual/biossíntese , Ativador de Plasminogênio Tecidual/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA