Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Neurobiol Aging ; 49: 165-182, 2017 01.
Article in English | MEDLINE | ID: mdl-27815990

ABSTRACT

Beta amyloid (Aß) is well accepted to play a central role in the pathogenesis of Alzheimer's disease (AD). The present work evaluated the therapeutic effects of low-level laser irradiation (LLI) on Aß-induced neurotoxicity in rat hippocampus. Aß 1-42 was injected bilaterally to the hippocampus CA1 region of adult male rats, and 2-minute daily LLI treatment was applied transcranially after Aß injection for 5 consecutive days. LLI treatment suppressed Aß-induced hippocampal neurodegeneration and long-term spatial and recognition memory impairments. Molecular studies revealed that LLI treatment: (1) restored mitochondrial dynamics, by altering fission and fusion protein levels thereby suppressing Aß-induced extensive fragmentation; (2) suppressed Aß-induced collapse of mitochondrial membrane potential; (3) reduced oxidized mitochondrial DNA and excessive mitophagy; (4) facilitated mitochondrial homeostasis via modulation of the Bcl-2-associated X protein/B-cell lymphoma 2 ratio and of mitochondrial antioxidant expression; (5) promoted cytochrome c oxidase activity and adenosine triphosphate synthesis; (6) suppressed Aß-induced glucose-6-phosphate dehydrogenase and nicotinamide adenine dinucleotide phosphate oxidase activity; (7) enhanced the total antioxidant capacity of hippocampal CA1 neurons, whereas reduced the oxidative damage; and (8) suppressed Aß-induced reactive gliosis, inflammation, and tau hyperphosphorylation. Although development of AD treatments has focused on reducing cerebral Aß levels, by the time the clinical diagnosis of AD or mild cognitive impairment is made, the brain is likely to have already been exposed to years of elevated Aß levels with dire consequences for multiple cellular pathways. By alleviating a broad spectrum of Aß-induced pathology that includes mitochondrial dysfunction, oxidative stress, neuroinflammation, neuronal apoptosis, and tau pathology, LLI could represent a new promising therapeutic strategy for AD.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/radiotherapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , CA1 Region, Hippocampal/metabolism , Low-Level Light Therapy , Mitochondrial Dynamics , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Alzheimer Disease/psychology , Animals , Apoptosis , CA1 Region, Hippocampal/pathology , Inflammation , Lasers, Semiconductor/therapeutic use , Low-Level Light Therapy/methods , Male , Neurons/pathology , Oxidative Stress , Rats, Sprague-Dawley , Recognition, Psychology , Spatial Memory , Tauopathies/etiology , Tauopathies/radiotherapy
2.
Neurochem Res ; 39(2): 344-52, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24379109

ABSTRACT

Inflammatory process has a fundamental role in the pathogenesis of Alzheimer's disease and insoluble amyloid beta deposits and neurofibrillary tangles provide the obvious stimuli for inflammation. The present study demonstrate the effect of pretreatment of 1,8-cineole (Cin) on inflammation induced by Aß(25-35) in differentiated PC12 cells. The cells were treated with Cin at different doses for 24 h and then replaced by media containing Aß(25-35) for another 24 h. The cell viability was decreased in Aß(25-35) treated cells which was significantly restored by Cin pretreatment. Cin successfully reduced the mitochondrial membrane potential, ROS and NO levels in Aß(25-35) treated cells. Cin also lowered the levels of proinflammatory cytokines TNF-α, IL-1ß and IL-6 in Aß(25-35) treated cells. Moreover, Cin also succeeded in lowering the expression of NOS-2, COX-2 and NF-κB. This study suggests the protective effects of Cin on inflammation and provides additional evidence for its potential beneficial use in therapy as an anti-inflammatory agent in neurodegenerative disease.


Subject(s)
Alzheimer Disease/pathology , Cyclohexanols/pharmacology , Inflammation/prevention & control , Monoterpenes/pharmacology , Amyloid beta-Peptides/physiology , Animals , Cytokines/metabolism , Eucalyptol , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress , PC12 Cells , Peptide Fragments/physiology , Rats , Reactive Oxygen Species/metabolism
3.
Pharmacol Biochem Behav ; 113: 53-62, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24141173

ABSTRACT

The neuronal mitochondria succumb to ischemia-reperfusion injury and release huge amount of reactive oxygen species and ultimately lead the neurons to intrinsic pathway of programmed cell death (iPCD). The present study was undertaken to elucidate the ischemia-reperfusion-induced oxidative stress and molecular events in iPCD 24 h post ischemia-reperfusion injury and plausible mitigation by zingerone, a potent antioxidant of ginger rhizome. The right middle cerebral artery was occluded for 2 h followed by reperfusion for 22 hours. A maximum infarct volume (43.29%) and mitochondrial injury (56.99%) was observed in middle cerebral artery occlusion (MCAO) group. However, zingerone administration (50 and 100 mg/kg b.wt. orally twice) at 5 h and 12 h from initiation of MCAO showed a significant reduction in infarct volume and mitochondrial injury (p<0.001). Zingerone treatment significantly improved behavioral outputs (p<0.05) and histological architecture (p<0.001) by reducing lipid peroxidation (p<0.01), augmenting the reduced glutathione content (p<0.01) and restoring Na(+)-K(+) ATPase and superoxide dismutase activities (p<0.01) in MCAO brain. Zingerone successfully reduced the caspase-3 and -9 activities in MCAO group (p<0.05) and succeeded in lowering the expressions of pro-apoptotic proteins - Apaf-1 and Bax (p<0.001). The present study suggests that zingerone is a potent antioxidant that salvaged the ischemic penumbral zone neurons by inhibiting iPCD and oxidative stress.


Subject(s)
Apoptosis/drug effects , Behavior, Animal/drug effects , Brain Ischemia/physiopathology , Guaiacol/analogs & derivatives , Oxidative Stress/drug effects , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Enzyme-Linked Immunosorbent Assay , Guaiacol/administration & dosage , Guaiacol/pharmacology , Male , Rats , Rats, Wistar
4.
Protoplasma ; 250(5): 1067-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23340606

ABSTRACT

Oxidative stress is a critical contributing factor to age-related neurodegenerative disorders. Therefore, the inhibition of oxidative damage, responsible for chronic detrimental neurodegeneration, is an important strategy for neuroprotective therapy. Withania somnifera (WS) extract has been reported to have potent antioxidant and free radical quenching properties in various disease conditions. The present study evaluated the hypothesis that WS extract would reduce oxidative stress-associated neurodegeneration after intracerebroventricular injection of streptozotocin (ICV-STZ) in rats. To test this hypothesis, male Wistar rats were pretreated with WS extract at doses of 100, 200, and 300 mg/kg body weight once daily for 3 weeks. On day 22nd, the rats were infused bilaterally with ICV-STZ injection (3 mg/kg body weight) in normal saline while sham group received only saline. Two weeks after the lesioning, STZ-infused rats showed cognitive impairment in the Morris water maze test. The rats were sacrificed after 3 weeks of the lesioning for the estimation of the contents of lipid peroxidation, reduced glutathione, and activities of glutathione reductase, glutathione peroxidase, and catalase. Pretreatment with WS extract attenuated behavioral, biochemical, and histological alterations significantly in dose-dependent manner in the hippocampus and cerebral cortex of ICV-STZ-infused rats. These results suggest that WS affords a beneficial effect on cognitive deficit by ameliorating oxidative damage induced by streptozotocin in a model of cognitive impairment.


Subject(s)
Cognition Disorders/drug therapy , Plant Extracts/pharmacology , Withania/chemistry , Animals , Antioxidants/pharmacology , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Maze Learning/drug effects , Oxidative Stress/drug effects , Random Allocation , Rats , Rats, Wistar , Streptozocin
5.
Neurol Sci ; 34(8): 1321-30, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23187787

ABSTRACT

Azadirachta indica Linn. (Meliaceae) has been used from ancient times as a remedy for various ailments. The present study was designed to investigate the antioxidant and anti-apoptotic properties of A. indica seed extract (ASE) in transient middle cerebral artery occlusion (MCAO) rat model. Antioxidant potential of ASE was determined in vitro. Further, ASE was evaluated against neurological deficits, histological alterations (TTC, CV and H&E) and oxidative damage (TBARS, GSH and nitrite) in MCAO rats. Moreover, caspase-3 and -9 were analyzed to evaluate the anti-apoptotic activity of ASE. ASE has shown potent in vitro reducing power (126.2 mg AsAE/g extract) and free radical scavenging activities (DPPH 171.0 and NO 176.0 µg/ml). Furthermore, ASE inhibited oxidative stress and decreased the activities of caspase-3 (26.7 %, p < 0.05) and caspase-9 (31.2 %, p < 0.01) thus, reduced neuronal loss in MCAO rats. Our data revealed that ASE has potent antioxidant and anti-apoptotic properties, and may be explored for its active constituents against neurodegenerative diseases.


Subject(s)
Azadirachta , Brain Ischemia/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Reperfusion Injury/drug therapy , Animals , Apoptosis/drug effects , Brain/drug effects , Brain/pathology , Brain Ischemia/pathology , Disease Models, Animal , Male , Oxidative Stress/drug effects , Psychomotor Disorders/drug therapy , Rats , Rats, Wistar , Reperfusion Injury/pathology
6.
Neurochem Int ; 61(7): 1081-93, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22898296

ABSTRACT

Oxidative stress is involved in Alzheimer's disease (AD)-type neurodegeneration with cognitive impairment (AD-TNDCI) as well as age related cognitive deficit. The present study was designed to investigate the pre-treatment effects of naringenin (NAR), a polyphenolic compound on cognitive dysfunction, oxidative stress in the hippocampus, and hippocampal neuron injury in a rat model of AD-TNDCI. The rats were pre-treated with NAR at a selective dose (50mg/kg, orally) for 2 weeks followed by intracerebroventricular-streptozotocin (ICV-STZ) (3mg/kg; 5µl per site) injection bilaterally. Behavioral alterations were monitored after 2 weeks from the lesion using passive avoidance test and Morris water maze paradigm. Three weeks after the lesion, the rats were sacrificed for measuring non-enzymatic [4-hydroxynonenal (4-HNE), malonaldehyde (MDA), thiobarbituric reactive substances (TBARS), hydrogen peroxide (H(2)O(2)), protein carbonyl (PC), reduced glutathione (GSH)] content and enzymatic [glutathione peroxidase (GPx), glutathione reductase (GR), glutathione-S-transferase (GST), superoxide dismutase (SOD), catalase (CAT) and Na(+)/K(+)-ATPase] activity in the hippocampus, and expression of choline acetyltransferase (ChAT) positive neuron, and histopathology of hippocampal neurons. The non-enzymatic level and enzymatic activity was significantly increased and decreased, respectively, with striking impairments in spatial learning and memory, loss of ChAT positive neuron and severe damage to hippocampal neurons in the rat induced by ICV-STZ. These abnormalities were significantly improved by NAR pre-treatment. The study suggests that NAR can protect against cognitive deficits, neuronal injury and oxidative stress induced by ICV-STZ, and may be used as a potential agent in treatment of neurodegenerative diseases such as AD-TNDCI.


Subject(s)
Alzheimer Disease/prevention & control , Cognition Disorders/prevention & control , Disease Models, Animal , Flavanones/pharmacology , Streptozocin/administration & dosage , Alzheimer Disease/pathology , Animals , Behavior, Animal , Cognition Disorders/pathology , Injections, Intraventricular , Male , Rats , Rats, Wistar
7.
Mol Cell Biochem ; 369(1-2): 55-65, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22752387

ABSTRACT

Beta-amyloid (Aß) peptides are considered to play a major role in the pathogenesis of Alzheimer's disease (AD) and compounds that can prevent pathways of Aß-induced neurotoxicity may be potential therapeutic agents for treatment of AD. This study examined the hypothesis that thymoquinone (TQ) would reduce oxidative stress and mitochondrial dysfunction in differentiated pheochromocytoma (PC 12) cells exposed to Aß fragment 25-35 (Aß(25-35)). To test this hypothesis, Aß was used to induce an in vitro model of AD in differentiated PC 12 cell line of rat. After 24 h of exposure with Aß(25-35), a significant reduction in cell viability and mitochondrial membrane potential (MMP) was observed. In addition, a significant elevation in the TBARS content and nitric oxide (NO) and activity of acetylcholine esterase (AChE) was observed which was restored significantly by TQ pretreatment. Furthermore, TQ also ameliorated glutathione and its dependent enzymes (glutathione peroxidase, glutathione reductase) which were depleted by Aß(25-35) in PC 12 cells. These results were supported by the immunocytochemical finding that has shown protection of cells by TQ from noxious effects of Aß(25-35). These results indicate that TQ holds potential for neuroprotection and may be a promising approach for the treatment of neurodegenerative disorders including AD.


Subject(s)
Amyloid beta-Peptides , Benzoquinones/administration & dosage , Cell Survival/drug effects , Mitochondria , Neuroprotective Agents/administration & dosage , Peptide Fragments , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Animals , Apoptosis/drug effects , Glutathione Peroxidase/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/pathology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , PC12 Cells , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Rats , Thiobarbituric Acid Reactive Substances/metabolism
8.
Mol Cell Biochem ; 367(1-2): 73-84, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22669728

ABSTRACT

The pathophysiological mechanisms leading to neuronal injury in middle cerebral artery occlusion (MCAO) model of cerebral stroke are complex and multifactorial that form the bases of behavioral deficits and inflammation mediated damage. The present study demonstrates the effect of piperine pretreatment (10 mg/kg b wt, once daily p.o. for 15 days) on cerebral ischemia-induced inflammation in male Wistar rats. The right middle cerebral artery was occluded for 2 h followed by reperfusion for 22 h. A maximum infarct volume (57.80 %) was observed in ischemic MCAO group. However, piperine administration prior to ischemia showed a significant reduction in infarct volume (28.29 %; p < 0.05) and neuronal loss (12.72 %; p < 0.01). As a result of piperine pretreatment, a significant improvement in behavioral outputs of MCAO rats (p < 0.05-0.01) was observed. Piperine successfully reduced the level of proinflammatory cytokines IL-1ß, IL-6 and TNF-α, in ischemic group (p < 0.01). Ischemic group brain has shown edematous morphology with vacuolated architecture and pyknotic nuclei in H & E staining which was successfully ameliorated by piperine administration. Moreover, piperine also succeeded in lowering the expression of COX-2, NOS-2, and NF-κB (p < 0.01). Both cytosolic and nuclear NF-κB were down-regulated in ischemic group pre-administered with piperine (p < 0.01). The present study suggests that piperine is able to salvage the ischemic penumbral zone neurons by virtue of its anti-inflammatory property, thereby limiting ischemic cell death.


Subject(s)
Alkaloids/pharmacology , Anti-Inflammatory Agents/pharmacology , Benzodioxoles/pharmacology , Cyclooxygenase 2/metabolism , Infarction, Middle Cerebral Artery/drug therapy , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Reperfusion Injury/prevention & control , Animals , Cyclooxygenase 2/genetics , Cytokines/blood , Cytokines/metabolism , Down-Regulation/drug effects , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Inflammation Mediators/blood , Male , Motor Activity/drug effects , Muscle Strength/drug effects , NF-kappa B/genetics , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Wistar , Reperfusion Injury/etiology , Reperfusion Injury/pathology
9.
Brain Res ; 1389: 133-42, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21376020

ABSTRACT

S-allyl cysteine (SAC), a sulfur containing amino acid derived from garlic, has been reported to have antioxidant, anti-cancer, antihepatotoxic and neurotrophic activity. This study was designed to examine the pre-treatment effects of SAC on cognitive deficits and oxidative damage in the hippocampus of intracerebroventricular streptozotocin (ICV-STZ)-infused mice. Mice pre-treated with SAC (30mg/kg) and vehicle (intraperitoneal; once daily for 15days) were bilaterally injected with ICV-STZ (2.57mg/kg body weight), whereas sham rats received the same volume of vehicle. The pre-treatment of this drug to Swiss albino mice has prevented the cognitive and neurobehavioral impairments. An increased latency and path length were observed in lesion, i.e. streptozotocin (STZ) group as compared to sham group and these were protected significantly in STZ group pre-treated with SAC. Levels of reduced glutathione (GSH) and its dependent enzymes (Glutathione peroxidase [GPx] and glutathione reductase [GR]) were decreased in STZ group as compared to sham group and pre-treatment of STZ group with SAC has protected their activities significantly. Conversely, the elevated level of thiobarbituric acid reactive substances (TBARS) in STZ group was attenuated significantly in SAC pre-treated group when compared with STZ lesioned group. Apoptotic parameters like DNA fragmentation, expression of Bcl2 and p53 were protected by the pre-treatment of SAC against STZ induced cognitive impairment. This study concludes that intervention of SAC could prevent free radicals associated deterioration of cognitive functions and neurobehavioral activities.


Subject(s)
Alzheimer Disease/metabolism , Antioxidants/pharmacology , Cysteine/analogs & derivatives , Nerve Degeneration/prevention & control , Oxidative Stress/drug effects , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Animals , Apoptosis/drug effects , Cysteine/pharmacology , Disease Models, Animal , Immunohistochemistry , Injections, Intraventricular , Maze Learning/drug effects , Mice , Neurotoxins/administration & dosage , Neurotoxins/toxicity , Oxidative Stress/physiology , Streptozocin/administration & dosage , Streptozocin/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...