Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
iScience ; 26(7): 107199, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37456841

RESUMO

Maintenance of redox balance plays central roles in a plethora of signaling processes. Although physiological levels of reactive oxygen and nitrogen species are crucial for functioning of certain signaling pathways, excessive production of free radicals and oxidants can damage cell components. The nuclear factor erythroid 2-related factor 2 (Nrf2) signaling cascade is the key pathway that mediates cellular response to oxidative stress. It is controlled at multiple levels, which serve to maintain redox homeostasis within cells. We show here that inositol polyphosphate multikinase (IPMK) is a modulator of Nrf2 signaling. IPMK binds Nrf2 and attenuates activation and expression of Nrf2 target genes. Furthermore, depletion of IPMK leads to elevated glutathione and cysteine levels, resulting in increased resistance to oxidants. Accordingly, targeting IPMK may restore redox balance under conditions of cysteine and glutathione insufficiency.

2.
Br J Pharmacol ; 2023 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-37338307

RESUMO

The gaseous neurotransmitter hydrogen sulfide (H2 S) exerts neuroprotective efficacy in the brain via post-translational modification of cysteine residues by sulfhydration, also known as persulfidation. This process is comparable in biological impact to phosphorylation and mediates a variety of signalling events. Unlike conventional neurotransmitters, H2 S cannot be stored in vesicles due to its gaseous nature. Instead, it is either locally synthesized or released from endogenous stores. Sulfhydration affords both specific and general neuroprotective effects and is critically diminished in several neurodegenerative disorders. Conversely, some forms of neurodegenerative disease are linked to excessive cellular H2 S. Here, we review the signalling roles of H2 S across the spectrum of neurodegenerative diseases, including Huntington's disease, Parkinson's disease, Alzheimer's disease, Down syndrome, traumatic brain injury, the ataxias, and amyotrophic lateral sclerosis, as well as neurodegeneration generally associated with ageing.

3.
Neurochem Int ; 141: 104858, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33010391

RESUMO

Exposure to chronic stress precipitates depression and anxiety. Stress-induced responses are differentially regulated by the prefrontal cortex (PFC) and basolateral amygdala (BLA). For instance, repeated stress leads to hypertrophy of BLA, resulting in the emergence of affective symptoms. Chronic stress-induced changes in the metabolism of monoamines are central in the manifestation of affective symptoms. Interestingly, BLA via its reciprocal connections modulates prefrontal cortical monoaminergic responses to acute stress. However, the effects of BLA inactivation on chronic stress-induced affective behaviors and monoaminergic changes in the PFC are relatively unknown. Thus, we hypothesized that inactivation of BLA might prevent chronic immobilization stress (CIS)-induced depressive-, anxiety-like behaviors, and associated monoaminergic alterations in the prelimbic (PrL) and anterior cingulate cortex (ACC) subregions of PFC. We used two different BLA silencing strategies, namely ibotenic acid lesion and reversible temporary inactivation using lidocaine. We found that CIS precipitates depressive- and anxiety-like behaviors. Further, CIS-induced negative affective behaviors were associated with decreased levels of 5-HT, DA, and NE, and increased 5-HIAA/5-HT, DOPAC + HVA/DA, and MHPG/NE ratio in the PrL and ACC, suggesting enhanced metabolism. Interestingly, BLA lesion prior to CIS blocked the emergence of depressive- and anxiety-like behaviors. Moreover, the lesion of BLA prior to CIS was sufficient to prevent alterations in levels of monoamines and their metabolites in the PrL and ACC. Thereafter, we evaluated whether the effects of BLA lesion could be mirrored by temporary inactivation of BLA, specifically during stress. Remarkably, temporary inactivation of BLA during stress recapitulated the effects of lesion. Our results have implications for understanding the role of BLA in chronic stress-induced metabolic alterations in prefrontal cortical monoaminergic systems, and associated mood and anxiety disorders. The current study supports the hypothesis that combating amygdalar hyperactivity might be a viable strategy for the management of stress and associated affective disorders.


Assuntos
Complexo Nuclear Basolateral da Amígdala/metabolismo , Monoaminas Biogênicas/metabolismo , Imobilização/psicologia , Transtornos do Humor/terapia , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/psicologia , Estresse Psicológico/terapia , Animais , Ansiedade/psicologia , Comportamento Animal , Depressão/etiologia , Depressão/psicologia , Ácido Ibotênico , Lidocaína , Masculino , Transtornos do Humor/metabolismo , Transtornos do Humor/patologia , Atividade Motora , Ratos , Ratos Wistar , Natação/psicologia
4.
Psychopharmacology (Berl) ; 237(10): 2967-2981, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32572589

RESUMO

RATIONALE: Anxiety is one of the most comorbid conditions with major depressive disorder (MDD). Depression-associated anxiety often stems from the dysfunctional hypothalamic-pituitary-adrenal (HPA) axis and its altered regulation by the amygdala. Furthermore, MDD is associated with altered glutamatergic processing leading to anxiety and impaired regulation of the HPA axis. Recent studies have demonstrated that N-acetyl cysteine (NAC), a pleiotropic drug, exerts antidepressant-like effect by modulation of hippocampal functions, periterminal release of glutamate, and/or redox systems. However, the effects of NAC on depression-associated anxiety, HPA axis hyperactivity, and amygdalar dysfunctions are relatively unknown. OBJECTIVES: Accordingly, we evaluated the effect of NAC on neonatal clomipramine (CLI)-induced adulthood anxiety and accompanying changes in plasma corticosterone levels, amygdalar volumes, neuronal/glial densities, levels of monoamines, and their metabolites in the amygdalar complex. RESULTS: We found that chronic treatment with NAC reverses CLI-induced anhedonia and enhanced anxiety. Interestingly, attenuation of CLI-associated anxiety in NAC-treated rats were accompanied by a reversal of adrenal and spleen hypertrophy, and normalization of enhanced plasma corticosterone levels, indicating improved HPA axis functioning. Furthermore, NAC treatment was sufficient to reverse volumetric hypertrophy of basolateral amygdala (BLA), and altered noradrenaline (NA) metabolism in the amygdalar complex. The effects of NAC in the reversal of CLI-induced impairments were similar to that of fluoxetine (FLX). CONCLUSIONS: We suggest that beneficial effects of NAC on antidepressive- and antianxiety-like behaviors are at least in part mediated via restoration of amygdalar and HPA axis functioning. Our results support the hypothesis that NAC might be evolved as a therapeutic strategy for reversal of amygdalar dysfunction in depression.


Assuntos
Acetilcisteína/administração & dosagem , Ansiolíticos/administração & dosagem , Antidepressivos/administração & dosagem , Ansiedade/tratamento farmacológico , Depressão/tratamento farmacológico , Sequestradores de Radicais Livres/administração & dosagem , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Ansiedade/metabolismo , Ansiedade/psicologia , Corticosterona/metabolismo , Depressão/metabolismo , Depressão/psicologia , Esquema de Medicação , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Resultado do Tratamento
5.
Artigo em Inglês | MEDLINE | ID: mdl-32512130

RESUMO

Major depressive disorder (MDD) is associated with enhanced anxiety and reduced reward processing leading to impaired cognitive flexibility. These pathological changes during depression are accompanied by dysfunctional hypothalamic-pituitary-adrenal (HPA) axis and its impaired regulation by the amygdala. Notably, the electrical stimulation of brain reward areas produces an antidepressant effect in both MDD patients and animal models of depression. However, the effects of chronic electrical self-stimulation of lateral hypothalamus - medial forebrain bundle (LH-MFB) on depression-associated anxiety and accompanying changes in plasma corticosterone levels, structural, and neurochemical alterations in the amygdala are unknown. Here, we used the neonatal clomipramine (CLI) model of depression. During adulthood, neonatal CLI and vehicle administered rats were subjected to bilateral electrode implantation at LH-MFB and trained to receive intracranial self-stimulation (ICSS) for 14 days. Rats were then tested for anhedonic and anxiety-like behaviors, followed by estimation of plasma corticosterone levels, assessment of amygdalar volumes and neuronal/glial numbers, levels of monoamines and their metabolites in the amygdala. We found that chronic ICSS of LH-MFB reverses CLI-induced anhedonia and anxiety. Interestingly, amelioration of CLI-induced enhanced anhedonia and anxiety in ICSS rats was associated with partial reversal of enhanced plasma corticosterone levels, hypertrophy of basolateral amygdala (BLA), and altered noradrenaline (NA) metabolism in the amygdalar complex. We suggest that beneficial effects of ICSS on CLI-induced anxiety at least in part mediated by the restoration of amygdalar and HPA axis functioning. Our results support the hypothesis that brain stimulation rewarding experience might be evolved as a therapeutic strategy for reversal of amygdalar dysfunction in depression.


Assuntos
Tonsila do Cerebelo/patologia , Antidepressivos Tricíclicos , Ansiedade/induzido quimicamente , Encéfalo/fisiologia , Clomipramina , Estimulação Elétrica , Recompensa , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Ansiedade/etiologia , Ansiedade/psicologia , Monoaminas Biogênicas/metabolismo , Corticosterona/sangue , Depressão , Modelos Animais de Doenças , Eletrodos Implantados , Feminino , Masculino , Gravidez , Ratos , Ratos Wistar , Autoestimulação
6.
J Chem Neuroanat ; 106: 101797, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32334029

RESUMO

A preliminary observation about resveratrol (RSV) dependent normalization of inflammatory and apoptotic factors in the cortex of hyperammonemic rat model of moderate grade hepatic encephalopathy (MoHE) led us to evaluate whether RSV is ultimately able to confer neuroprotection against MoHE pathogenesis and that it does so by activating its bonafide molecular target SIRT1. The present study compared the profile of relevant neurobehavioral pattern vs neuromorphometry of hippocampal CA1 neurons and SIRT1 activity in the hippocampus of the chronic liver failure (CLF) model of moderate grade HE (MoHE) rats induced by administration of 100 mg/kg body weight of thioacetamide i.p. for 10 days and in the CLF/MoHE rats treated with 10 mg/kg body weight RSV i.p. for 7 days. As compared to the control group rats, the MoHE rats showed significantly deranged pattern of memory and motor functions on MWM and rota rod tests, respectively. These behavioural deficits were associated with a significant reduction in apical dendrite length and number of branching points in the CA1 pyramidal neurons. Interestingly, all these parameters were found to be recovered back to their normal levels in the MoHE rats treated with RSV. Concordantly, MoHE associated declined SIRT1 activity in the hippocampus could be normalized back due to RSV treatment to those MoHE rats. Our findings suggest that RSV is able to normalize MoHE associated memory impairments and motor deficits vis a vis reversal of CA1 dendritic atrophy via SIRT1 activation.


Assuntos
Antioxidantes/farmacologia , Região CA1 Hipocampal/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Encefalopatia Hepática/metabolismo , Células Piramidais/efeitos dos fármacos , Resveratrol/farmacologia , Sirtuína 1/metabolismo , Animais , Antioxidantes/uso terapêutico , Atrofia/tratamento farmacológico , Atrofia/metabolismo , Atrofia/patologia , Comportamento Animal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Dendritos/metabolismo , Dendritos/patologia , Encefalopatia Hepática/patologia , Memória/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Células Piramidais/metabolismo , Células Piramidais/patologia , Ratos , Resveratrol/uso terapêutico
7.
Neurochem Int ; 132: 104605, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31751620

RESUMO

Depression is highly comorbid with anxiety disorders and associated with profound cognitive impairment. Moreover, cognitive deficits associated with hippocampal dysfunction are central in depression and anxiety disorders. Furthermore, depression is accompanied by glutamatergic dysfunction which can further impair the functioning of the hippocampus. Recent studies have shown that N-acetyl cysteine (NAC), a glutamate modulator produces an antidepressant-like effect by normalization of the periterminal release of glutamate and/or antioxidant effects. However, the effects of repeated NAC treatment on depression-induced anxiety, cognitive deficits, and associated neurochemical and structural alterations are relatively unknown. Accordingly, we investigated whether chronic NAC treatment could reverse cognitive deficits, and associated hippocampal volume loss and monoaminergic alterations in the neonatal clomipramine (CLI) model of depression. We found that chronic NAC treatment produces antidepressive and antianhedonic-like effects. NAC treatment also reversed CLI-induced anxiety. Interestingly, repeated NAC treatment improved the performance of CLI rats in rewarded alternation task in T-maze. The antidepressive-like and procognitive effects of NAC was associated with normalization of volume loss in CA1, dentate gyrus (DG) and hilar subfields of the hippocampus. Furthermore, NAC restored CLI-induced decrease in levels of monoamines and normalized enhanced metabolism in the hippocampus. Taken together, chronic NAC treatment ameliorates depressive and anxiety-like behavior, spatial learning deficits, and reverses CLI-induced pathological alterations at structural and neurochemical levels in the hippocampus. Our findings might help in evolving NAC as a viable pharmacotherapy for reversal of cognitive deficits in depression and associated disorders.


Assuntos
Acetilcisteína/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Depressão/tratamento farmacológico , Hipocampo/química , Hipocampo/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Depressão/induzido quimicamente , Depressão/metabolismo , Hipocampo/metabolismo , Masculino , Tamanho do Órgão , Distribuição Aleatória , Ratos , Ratos Wistar , Inibidores Seletivos de Recaptação de Serotonina/toxicidade
8.
Brain Stimul ; 12(3): 752-766, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30765272

RESUMO

BACKGROUND: Major depressive disorder (MDD) is a multifactorial disease which often coexists with cognitive deficits. Depression-induced cognitive deficits are known to be associated with aberrant reward processing, neurochemical and structural alterations. Recent studies have shown that chronic electrical stimulation of brain reward areas induces a robust antidepressant effect. However, the effects of repeated electrical self-stimulation of lateral hypothalamus - medial forebrain bundle (LH-MFB) on depression-induced cognitive deficits and associated neurochemical and structural alterations in the prefrontal cortex (PFC) are unknown. OBJECTIVES: We investigated the effect of chronic rewarding self-stimulation of LH-MFB in neonatal clomipramine (CLI) model of depression. During adulthood, neonatal CLI and saline administered rats were implanted with bilateral electrodes stereotaxically in the LH-MFB and trained to receive intracranial self-stimulation (ICSS) for 14 days. The rats were tested for depressive-like behaviors, learning and memory followed by estimation of PFC volumes, levels of monoamines and its metabolites in the PFC. RESULTS: We found that chronic ICSS of LH-MFB reverses CLI-induced behavioral despair and anhedonia. Interestingly, self-stimulation normalizes the impaired novel object and location recognition memory in CLI rats. The amelioration of learning impairments in CLI rats was associated with the reversal of volume loss and restoration of monoamine metabolism in the PFC. CONCLUSION: We demonstrated that repeated intracranial self-stimulation of LH-MFB ameliorates CLI-induced learning deficits, reverses altered monoamine metabolism and the atrophy of PFC. Our results support the hypothesis that chronic brain stimulation rewarding experience might be evolved as a potential treatment strategy for reversal of learning deficits in depression and associated disorders.


Assuntos
Disfunção Cognitiva/terapia , Estimulação Encefálica Profunda/métodos , Depressão/complicações , Plasticidade Neuronal , Córtex Pré-Frontal/fisiopatologia , Recompensa , Animais , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Masculino , Feixe Prosencefálico Mediano/fisiopatologia , Ratos , Ratos Wistar , Autoestimulação
9.
Mol Neurobiol ; 56(1): 350-366, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29704199

RESUMO

Repeated stress causes cognitive decline and decreases the expression of glial fibrillary acidic protein (GFAP)+ astroglial cells in the prefrontal cortex (PFC). The stress-induced alterations in astroglial density and morphology might significantly contribute to cognitive impairments. Apart from PFC, a key region involved in modulation of repercussions of stress is basolateral amygdala (BLA), which undergoes hypertrophy following chronic immobilization stress (CIS) and has intense reciprocal connections to the PFC. Interestingly, inactivation of BLA precludes stress-induced learning deficits. However, the modulatory role of BLA on CIS-induced alterations in GFAP+ astroglial density and associated learning deficits are presently unknown. Accordingly, we present two sets of experiments evaluating the effects of BLA inactivation either permanently or temporarily on CIS-induced changes in learning and astroglial expression in the PFC. CIS causes impairment in novel object recognition memory and astroglial loss in the PFC. In experiment I, we permanently inactivated the BLA by ibotenate lesion prior to CIS and observed a significant improvement in learning. Surprisingly, BLA lesion also prevented the stress-induced astroglial loss in the PFC. Furthermore, in the experiment II, we analyzed whether the effects of permanent inactivation could be mirrored by the temporary blockage of BLA specifically during stress. Interestingly, temporary inactivation of BLA mimics the effects of lesion. There was a notable prevention of learning impairment and astroglial loss in the PFC following BLA inactivation during stress. The present study emphasizes that stress-induced astroglial loss might contribute to cognitive deficits and modulation of BLA activity might be a viable strategy for management of stress-related PFC dysfunctions.


Assuntos
Astrócitos/patologia , Complexo Nuclear Basolateral da Amígdala/patologia , Córtex Pré-Frontal/patologia , Estresse Psicológico/patologia , Animais , Complexo Nuclear Basolateral da Amígdala/fisiopatologia , Proteína Glial Fibrilar Ácida/metabolismo , Giro do Cíngulo/patologia , Giro do Cíngulo/fisiopatologia , Masculino , Memória , Córtex Pré-Frontal/fisiopatologia , Ratos Wistar , Estresse Psicológico/fisiopatologia , Análise e Desempenho de Tarefas
10.
J Chem Neuroanat ; 95: 134-145, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29277704

RESUMO

Repeated exposure to stress precipitates anxiety, depression and cognitive deficits. Stress-induced activation of the hypothalamus-pituitary-adrenal (HPA) axis is modulated by the prefrontal cortex (PFC) and basolateral amygdala (BLA). It is well established that BLA positively regulates the HPA axis and undergoes hypertrophy following chronic immobilization stress (CIS). However, it is not known whether inactivation of the BLA can modulate the stress-induced changes in the expression of glucocorticoid receptors (GRs) in the PFC. To address this, we stereologically estimated GR+ cell densities in the prelimbic (PrL) and anterior cingulate cortex (ACC). Following ibotenate lesioning of the BLA, rats were subjected to CIS and GR+ cell densities were assessed. CIS increases the GR+ cell densities in PrL and ACC. BLA lesion prior to CIS abolished the CIS-induced increase in GR+ cell densities in both regions. In the second part of experiments, we evaluated whether selective inactivation of BLA during CIS would mimic the effects of BLA lesion. Interestingly, the BLA inactivation specifically during CIS prevented the increase in GR+ cell densities in the PrL and ACC. The findings of our study suggest that BLA regulates the stress-induced increase in prefrontal GR expression, which might be crucial in the emergence of affective and cognitive symptoms following stress. We speculate that modulation of BLA during stress might prevent HPA axis dysfunctions and GR resistance in stress-related disorders, and could assist in the development of novel therapeutic strategies to treat stress and associated disorders like depression. Further, molecular studies are warranted for the understanding of stress-induced GR resistance and its prevention via BLA inactivation.


Assuntos
Complexo Nuclear Basolateral da Amígdala/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores de Glucocorticoides/metabolismo , Estresse Psicológico/metabolismo , Animais , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Sistema Hipófise-Suprarrenal/fisiologia , Ratos , Ratos Wistar , Restrição Física
11.
Artigo em Inglês | MEDLINE | ID: mdl-30036565

RESUMO

Chronic exposure to stress causes cognitive deficits, anxiety and depression. Earlier studies have suggested that the prefrontal cortex (PFC) and basolateral amygdala (BLA) can differentially modulate the stress-induced alterations either by their action on HPA axis or via direct reciprocal connections between them. The PFC dysfunction and BLA hypertrophy following stress are known to cause anxiety and affective symptoms. Recent studies indicate that inactivation of BLA projections to PFC remarkably decreases anxiety. However, the effect of BLA inactivation on stress-induced anxiety and associated volume loss in prelimbic (PrL) and anterior cingulate (ACC) subregions of PFC is not known. Accordingly, we evaluated the effect of BLA lesion or inactivation during chronic immobilization stress (CIS) on an approach-avoidance task and associated volume loss in the PFC. The stressed rats showed a significant volumetric reduction in layer I and II of the PrL and ACC. Interestingly, BLA lesion prior to stress prevented the volume loss in PrL and ACC. Further, BLA lesion blocked the anxiety-like behavior in stressed rats. However, in the absence of stress, BLA lesion increased the number of shocks as compared to controls. As BLA lesion produced an anticonflict effect, we performed temporary inactivation of BLA specifically during stress. Similar to BLA lesion, lidocaine-induced inactivation prevented the stress-induced volume loss and anxiety-like behavior. We demonstrate that inactivation of BLA during stress prevents CIS-induced anxiety and associated structural correlates in the PFC. The present study extends the hypothesis of amygdalar silencing as a possible management strategy for stress and associated disorders.


Assuntos
Ansiedade/etiologia , Complexo Nuclear Basolateral da Amígdala/fisiologia , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/patologia , Análise de Variância , Anestésicos Locais/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/lesões , Doença Crônica , Modelos Animais de Doenças , Eletrochoque/métodos , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Ibotênico/toxicidade , Lidocaína/farmacologia , Masculino , Ratos , Ratos Wistar
12.
Exp Gerontol ; 101: 101-112, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29174497

RESUMO

The present study explored the effects of swimming training and grape seed proanthocyanidin extract (GSPE) on neuronal survival in the hippocampus (HC) of middle-aged rats along with oxidative stress (OS) parameters. Further, the bioavailability of the GSPE, catechin, epicatechin and gallic acid were measured in the HC and plasma. Male Wistar rats were grouped into: sedentary control, SE-C; swimming trained, SW-T; SE-C, supplemented sedentary, SE-C(PA) and swimming trainees, SW-T(PA). The supplement was a daily dose of 400mg GSPE/kg body weight. Swimming training lasted for 2h/day and for 14weeks. Glutathione level was increased in response to single and combined interventions in the middle-aged rats. Adult trainees showed increased glutathione peroxidase activity unlike middle-aged wherein increase was seen in SE-C(PA) alone. Lowered catalase activity with age in the HC increased in response to the combined interventions although single interventions were also effective. HC from both ages showed decrease in lipid peroxidation and hydrogen peroxide levels in response to the interventions. GSPE constituents were seen in the HC of swimming trained middle-aged and adult rats. The study suggests that combined intervention is effective in decreasing LPO and H2O2 generation in the HC. Further, the neuronal numbers and planimetric volumes of CA1 pyramidal layer was significantly reduced in middle-aged rats compared to adults. Interestingly, both interventions enhanced the numbers and volumes in adult and middle-aged rats. Thus, age-associated decrease in CA1 neurons could be restored by both the interventions. The results of the present study will help in developing effective therapies for age-associated degenerative changes and cognitive deficits.


Assuntos
Envelhecimento/fisiologia , Extrato de Sementes de Uva/farmacologia , Hipocampo/patologia , Neurônios , Condicionamento Físico Animal , Proantocianidinas/farmacologia , Natação/fisiologia , Animais , Antioxidantes/farmacologia , Contagem de Células/métodos , Glutationa/metabolismo , Peróxido de Hidrogênio/metabolismo , Peroxidação de Lipídeos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Condicionamento Físico Animal/métodos , Condicionamento Físico Animal/fisiologia , Ratos , Ratos Wistar
13.
Neurobiol Learn Mem ; 142(Pt B): 218-229, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28495605

RESUMO

Chronic stress causes detrimental effects on various forms of learning and memory. The basolateral amygdala (BLA) not only plays a crucial role in mediating certain forms of memory, but also in the modulation of the effects of stress. Chronic immobilization stress (CIS) results in hypertrophy of the BLA, which is believed to be one of the underlying causes for stress' effects on learning. Thus, it is plausible that preventing the effects of CIS on amygdala would preclude its deleterious cognitive effects. Accordingly, in the first part, we evaluated the effect of excitotoxic lesion of the BLA on chronic stress-induced hippocampal-dependent spatial learning using a partially baited radial arm maze task. The BLA was ablated bilaterally using ibotenic acid prior to CIS. Chronically stressed rats showed impairment in spatial learning with decreased percentage correct choice and increased reference memory errors. Excitotoxic lesion of the BLA prevented the impairment in spatial learning and reference memory. In the retention test, lesion of the BLA was able to rescue the chronic stress-induced impairment. Interestingly, stress-induced enhanced plasma corticosterone levels were partially prevented by the lesion of BLA. These results motivated us to evaluate if the same effects can be observed with temporary inactivation of BLA, only during stress. We found that chronic stress-induced spatial learning deficits were also prevented by temporary inactivation of the BLA. Additionally, temporary inactivation of BLA partially precluded the stress-induced increase in plasma corticosterone levels. Thus, inactivation of BLA precludes stress-induced spatial learning deficits, and enhanced plasma corticosterone levels. It is speculated that BLA inactivation-induced reduction in corticosterone levels during stress, might be crucial in restoring spatial learning impairments. Our study provides evidence that amygdalar modulation during stress might be beneficial for strategic management of stress-related cognitive deficits.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/fisiopatologia , Corticosterona/sangue , Aprendizagem Espacial/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Comportamento Animal/fisiologia , Disfunção Cognitiva/etiologia , Modelos Animais de Doenças , Imobilização/fisiologia , Masculino , Ratos , Ratos Wistar , Estresse Psicológico/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA