Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Alzheimers Dement ; 14(7): 913-924, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29601787

RESUMEN

INTRODUCTION: Apolipoprotein E (APOE) ε4 is the major genetic risk factor for Alzheimer's disease (AD), but its prevalence is unclear because earlier studies did not require biomarker evidence of amyloid ß (Aß) pathology. METHODS: We included 3451 Aß+ subjects (853 AD-type dementia, 1810 mild cognitive impairment, and 788 cognitively normal). Generalized estimating equation models were used to assess APOE ε4 prevalence in relation to age, sex, education, and geographical location. RESULTS: The APOE ε4 prevalence was 66% in AD-type dementia, 64% in mild cognitive impairment, and 51% in cognitively normal, and it decreased with advancing age in Aß+ cognitively normal and Aß+ mild cognitive impairment (P < .05) but not in Aß+ AD dementia (P = .66). The prevalence was highest in Northern Europe but did not vary by sex or education. DISCUSSION: The APOE ε4 prevalence in AD was higher than that in previous studies, which did not require presence of Aß pathology. Furthermore, our results highlight disease heterogeneity related to age and geographical location.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Disfunción Cognitiva/metabolismo , Anciano , Alelos , Biomarcadores/líquido cefalorraquídeo , Europa (Continente) , Femenino , Humanos , Masculino , Tomografía de Emisión de Positrones , Prevalencia
2.
Biochim Biophys Acta Mol Basis Dis ; 1863(7): 1858-1866, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27262357

RESUMEN

In the previous study, we demonstrated that dichlorvos induces oxidative stress in dopaminergic neuronal cells and subsequent caspase activation mediates apoptosis. In the present study, we evaluated the effect and mechanism of dichlorvos induced oxidative stress on cell cycle activation in NGF-differentiated PC12 cells. Dichlorvos exposure resulted in oxidative DNA damage along with activation of cell cycle machinery in differentiated PC12 cells. Dichlorvos exposed cells exhibited an increased expression of p53, cyclin-D1, pRb and decreased expression of p21suggesting a re-entry of differentiated cells into the cell cycle. Cell cycle analysis of dichlorvos exposed cells revealed a reduction of cells in the G0/G1 phase of the cell cycle (25%), and a concomitant increase of cells in S phase (30%) and G2/M phase (43.3%) compared to control PC12 cells. Further, immunoblotting of cytochrome c, Bax, Bcl-2 and cleaved caspase-3 revealed that dichlorvos induces a caspase-dependent cell death in PC12 cells. These results suggest that Dichlorvos exposure has the potential to generate oxidative stress which evokes activation of cell cycle machinery leading to apoptotic cell death via cytochrome c release from mitochondria and subsequent caspase-3 activation in differentiated PC12 cells.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Diclorvos/efectos adversos , Neuronas Dopaminérgicas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Intoxicación por Organofosfatos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Ciclo Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Diclorvos/farmacología , Neuronas Dopaminérgicas/patología , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Intoxicación por Organofosfatos/genética , Intoxicación por Organofosfatos/patología , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Ratas
3.
JAMA ; 313(19): 1924-38, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25988462

RESUMEN

IMPORTANCE: Cerebral amyloid-ß aggregation is an early pathological event in Alzheimer disease (AD), starting decades before dementia onset. Estimates of the prevalence of amyloid pathology in persons without dementia are needed to understand the development of AD and to design prevention studies. OBJECTIVE: To use individual participant data meta-analysis to estimate the prevalence of amyloid pathology as measured with biomarkers in participants with normal cognition, subjective cognitive impairment (SCI), or mild cognitive impairment (MCI). DATA SOURCES: Relevant biomarker studies identified by searching studies published before April 2015 using the MEDLINE and Web of Science databases and through personal communication with investigators. STUDY SELECTION: Studies were included if they provided individual participant data for participants without dementia and used an a priori defined cutoff for amyloid positivity. DATA EXTRACTION AND SYNTHESIS: Individual records were provided for 2914 participants with normal cognition, 697 with SCI, and 3972 with MCI aged 18 to 100 years from 55 studies. MAIN OUTCOMES AND MEASURES: Prevalence of amyloid pathology on positron emission tomography or in cerebrospinal fluid according to AD risk factors (age, apolipoprotein E [APOE] genotype, sex, and education) estimated by generalized estimating equations. RESULTS: The prevalence of amyloid pathology increased from age 50 to 90 years from 10% (95% CI, 8%-13%) to 44% (95% CI, 37%-51%) among participants with normal cognition; from 12% (95% CI, 8%-18%) to 43% (95% CI, 32%-55%) among patients with SCI; and from 27% (95% CI, 23%-32%) to 71% (95% CI, 66%-76%) among patients with MCI. APOE-ε4 carriers had 2 to 3 times higher prevalence estimates than noncarriers. The age at which 15% of the participants with normal cognition were amyloid positive was approximately 40 years for APOE ε4ε4 carriers, 50 years for ε2ε4 carriers, 55 years for ε3ε4 carriers, 65 years for ε3ε3 carriers, and 95 years for ε2ε3 carriers. Amyloid positivity was more common in highly educated participants but not associated with sex or biomarker modality. CONCLUSIONS AND RELEVANCE: Among persons without dementia, the prevalence of cerebral amyloid pathology as determined by positron emission tomography or cerebrospinal fluid findings was associated with age, APOE genotype, and presence of cognitive impairment. These findings suggest a 20- to 30-year interval between first development of amyloid positivity and onset of dementia.


Asunto(s)
Péptidos beta-Amiloides/análisis , Apolipoproteína E4/genética , Encéfalo/patología , Disfunción Cognitiva/patología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Biomarcadores/análisis , Líquido Cefalorraquídeo/química , Demencia/patología , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Prevalencia , Factores de Riesgo
4.
Indian J Clin Biochem ; 29(1): 69-73, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24478552

RESUMEN

Dietary and lifestyle factors have been shown to have a profound effect on paraoxonase-1 (PON1) activity. Cigarette smoke has been shown to inhibit its mass and activity where as resveratrol has been shown to enhance it. We exposed hepatoma derived cell line (HepG2) to resveratrol and nicotine in varying doses and measured PON1 enzymatic activity and PON1 gene expression. In addition, total protein content of HepG2 cells was also measured. Resveratrol in a dose of 15 µmol/l or above significantly increased the PON1 enzyme activity (p > 0.001) where as nicotine in a dose of 1 µmol/l or higher significantly reduced it (p < 0.05). The resveratrol in this dose also enhanced the PON1 gene expression whereas nicotine decreased it as compared to controls. However, the protein conent of cells was not changed suggesting that they were not cytotoxic in the doses used. Till date the antioxidant vitamins have shown disappointing results against LDL oxidation and cardiovascular protection. However, the effect of resveratrol on PON1 gene expression and activity was significant, suggesting increase in PON1 activity and enhanced gene expression may be its alternative mechanism for offering protection against cardiovascular disease and may be an potential pharmacological agent which can be used for this.

5.
J Neurosci Res ; 91(3): 444-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23280485

RESUMEN

Dichlorvos, an organophosphate (OP), is known to cause oxidative stress in the central nervous system (CNS). Previously we have shown that dichlorvos treatment promoted the levels of proinflammatory molecules and ultimately induced apoptotic cell death in primary microglial cells. Here we studied the effect of dichlorvos on crucial cell cycle regulatory proteins and the DNA damage sensor ataxia-telangiectasia mutated (ATM). We found a significant increase in p53 and its downstream target, p21, levels in dichlorvos-treated microglial cells compared with control cells. Moreover, dichlorvos exposure promoted the levels of different cell cycle regulatory proteins. These results along with flow cytometry results suggested that primary microglial cells were arrested at G1 and G2/M phase after dichlorvos exposure. We have shown in a previous study that dichlorvos can induce DNA damage in microglia; here we found that microglial cells also tried to repair this damage by inducing a DNA repair enzyme, i.e., ATM. We observed a significant increase in the levels of ATM after dichlorvos treatment compared with control.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Diclorvos/toxicidad , Microglía/efectos de los fármacos , Cultivo Primario de Células , Animales , Animales Recién Nacidos , Puntos de Control del Ciclo Celular/genética , Células Cultivadas , Daño del ADN/genética , Reparación del ADN/genética , Microglía/metabolismo , Organofosfatos/toxicidad , Ratas , Ratas Wistar
6.
Toxicol Appl Pharmacol ; 273(2): 365-80, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24084166

RESUMEN

The present investigation was carried out to elucidate a possible molecular mechanism related to the effects of aluminium-induced oxidative stress on various mitochondrial respiratory complex subunits with special emphasis on the role of Peroxisome proliferator activated receptor gamma co-activator 1α (PGC-1α) and its downstream targets i.e. Nuclear respiratory factor-1(NRF-1), Nuclear respiratory factor-2(NRF-2) and Mitochondrial transcription factor A (Tfam) in mitochondrial biogenesis. Aluminium lactate (10mg/kgb.wt./day) was administered intragastrically to rats for 12 weeks. After 12 weeks of exposure, we found an increase in ROS levels, mitochondrial DNA oxidation and decrease in citrate synthase activity in the Hippocampus (HC) and Corpus striatum (CS) regions of rat brain. On the other hand, there was a decrease in the mRNA levels of the mitochondrial encoded subunits-NADH dehydrogenase (ND) subunits i.e. ND1, ND2, ND3, Cytochrome b (Cytb), Cytochrome oxidase (COX) subunits i.e. COX1, COX3, ATP synthase (ATPase) subunit 6 along with reduced expression of nuclear encoded subunits COX4, COX5A, COX5B of Electron transport chain (ETC). Besides, a decrease in mitochondrial DNA copy number and mitochondrial content in both regions of rat brain was observed. The PGC-1α was down-regulated in aluminium treated rats along with NRF-1, NRF-2 and Tfam, which act downstream from PGC-1α in aluminium treated rats. Electron microscopy results revealed a significant increase in the mitochondrial swelling, loss of cristae, chromatin condensation and decreases in mitochondrial number in case of aluminium treated rats as compared to control. So, PGC-1α seems to be a potent target for aluminium neurotoxicity, which makes it an almost ideal target to control or limit the damage that has been associated with the defective mitochondrial function seen in neurodegenerative diseases.


Asunto(s)
Aluminio/toxicidad , Regulación de la Expresión Génica , Recambio Mitocondrial/fisiología , Estrés Oxidativo/fisiología , Factores de Transcripción/metabolismo , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Recambio Mitocondrial/efectos de los fármacos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Ratas , Ratas Wistar , Factores de Transcripción/biosíntesis
7.
Chem Res Toxicol ; 25(8): 1762-70, 2012 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-22817663

RESUMEN

Dichlorvos [2,2-dichlorovinyl dimethyl phosphate] is one of the most common in-use organophosphate (OP) in developing nations. Previous studies from our lab have shown chronic Dichlorvos exposure leads to neuronal cell death in rats. However, the extent of damage caused by Dichlorvos to other cells of the central nervous system (CNS) is still not clear. Microglial cells are the primary threat sensors of CNS which become activated in many pathological conditions. Activation of microglial cells results in reactive microgliosis, manifested by increased cellular damage in the affected regions. Using rat primary microglial cultures, here we show that Dichlorvos exposure can activate and induce apoptotic cell death in microglia. We observed significant up-regulation of pro-inflammatory molecules like nitric oxide, TNF-α, and IL-1ß when microglia were treated with Dichlorvos (10 µM). Significant up-regulation of CD11b, microglial specific activation marker, was also observed after 24 h of Dichlorvos treatment. The activated microglial cells eventually undergo cell death after 48 h of Dichlorvos treatment. The DNA fragmentation pattern of Dichlorvos treated microglia along with increased expression of Bax in mitochondria, cytochrome c release from mitochondria, and caspase-3 activation led us to assume that microglia were undergoing apoptosis. Thus, the present study showed that Dichlorvos can induce microglial activation and ultimately apoptotic cell death. These findings gave new perspective to the current knowledge of Dichlorvos (OPs) mediated CNS damage and presents microglial activation as a potential therapeutic target for preventing the OP induced neuronal damage.


Asunto(s)
Apoptosis/efectos de los fármacos , Diclorvos/toxicidad , Microglía/citología , Animales , Antígeno CD11b/metabolismo , Caspasa 3/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Fragmentación del ADN/efectos de los fármacos , Diclorvos/química , Interleucina-1beta/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Proteína X Asociada a bcl-2/metabolismo
8.
J Biomed Sci ; 19: 2, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22214483

RESUMEN

BACKGROUND: One of the pathological hallmarks of Alzheimer's disease (AD) is the deposition of the ~4 kDa amyloid ß protein (Aß) within lesions known as senile plaques. Aß is also deposited in the walls of cerebral blood vessels in many cases of AD. A substantial proportion of the Aß that accumulates in the AD brain is deposited as Amyloid, which is highly insoluble, proteinaceous material with a ß-pleated-sheet conformation and deposited extracellularly in the form of 5-10 nm wide straight fibrils. As γ-secretase catalyzes the final cleavage that releases the Aß42 or 40 from amyloid ß -protein precursor (APP), therefore, it is a potential therapeutic target for the treatment of AD. γ-Secretase cleavage is performed by a high molecular weight protein complex containing presenilins (PSs), nicastrin, Aph-1 and Pen-2. Previous studies have demonstrated that the presenilins (PS1 and PS2) are critical components of a large enzyme complex that performs γ-secretase cleavage. METHODS: In this study we used RNA interference (RNAi) technology to examine the effects of small-interfering RNA (siRNA) against PS1 on expression levels of PS1 and Aß42 in IMR-32 Cells using RTPCR, western blotting and immunofluorescence techniques. RESULTS: The results of the present study showed down regulation of PS1 and Aß42 in IMR32 cells transfected with siRNA against PS1. CONCLUSION: Our results substantiate the concept that PS1 is involved in γ-secretase activity and provides the rationale for therapeutic strategies aimed at influencing Aß42 production.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Regulación hacia Abajo , Presenilina-1/metabolismo , ARN Interferente Pequeño/farmacología , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/genética , Western Blotting , Línea Celular Tumoral , Humanos , Microscopía Fluorescente , Presenilina-1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
9.
Pathobiology ; 79(2): 55-71, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22205086

RESUMEN

Alzheimer's disease (AD), the most common cause of dementia, results from the interplay of various deregulated mechanisms triggering a complex pathophysiology. The neurons suffer from and slowly succumb to multiple irreversible damages, resulting in cell death and thus memory deficits that characterize AD. In spite of our vast knowledge, it is still unclear as to when the disease process starts and how long the perturbations continue before the disease manifests. Recent studies provide sufficient evidence to prove amyloid ß (Aß) as the primary cause initiating secondary events, but Aß is also known to be produced under normal conditions and to possess physiological roles, hence, the questions that remain are: What are the factors that lead to abnormal Aß production? When does Aß turn into a pathological molecule? What is the chain of events that follows Aß? The answers are still under debate, and further insight may help us in creating better diagnostic and therapeutic options in AD. The present article attempts to review the current literature regarding AD pathophysiology and proposes a pathophysiologic cascade in AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/fisiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Muerte Celular/fisiología , Humanos , Trastornos de la Memoria/etiología , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/fisiología
10.
Toxicol Appl Pharmacol ; 256(1): 73-82, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21843543

RESUMEN

Numerous epidemiological studies have shown an association between pesticide exposure and increased risk of developing Parkinson's diseases. Oxidative stress generated as a result of mitochondrial dysfunction has been implicated as an important factor in the etiology of Parkinson's disease. Previously, we reported that chronic dichlorvos exposure causes mitochondrial impairments and nigrostriatal neuronal death in rats. The present study was designed to test whether Coenzyme Q(10) (CoQ(10)) administration has any neuroprotective effect against dichlorvos mediated nigrostriatal neuronal death, α-synuclein aggregation, and motor dysfunction. Male albino rats were administered dichlorvos by subcutaneous injection at a dose of 2.5 mg/kg body weight over a period of 12 weeks. Results obtained there after showed that dichlorvos exposure leads to enhanced mitochondrial ROS production, α-synuclein aggregation, decreased dopamine and its metabolite levels resulting in nigrostriatal neurodegeneration. Pretreatment by Coenzyme Q(10) (4.5 mg/kg ip for 12 weeks) to dichlorvos treated animals significantly attenuated the extent of nigrostriatal neuronal damage, in terms of decreased ROS production, increased dopamine and its metabolite levels, and restoration of motor dysfunction when compared to dichlorvos treated animals. Thus, the present study shows that Coenzyme Q(10) administration may attenuate dichlorvos induced nigrostriatal neurodegeneration, α-synuclein aggregation and motor dysfunction by virtue of its antioxidant action.


Asunto(s)
Diclorvos/toxicidad , Neuronas/enzimología , Fármacos Neuroprotectores/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Ubiquinona/análogos & derivados , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Diclorvos/antagonistas & inhibidores , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/administración & dosificación , Ubiquinona/uso terapéutico
11.
Toxicol Appl Pharmacol ; 256(3): 241-8, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21513725

RESUMEN

Arsenicosis, due to contaminated drinking water, is a serious health hazard in terms of morbidity and mortality. Arsenic induced free radicals generated are known to cause cellular apoptosis through mitochondrial driven pathway. In the present study, we investigated the effect of arsenic interactions with various complexes of the electron transport chain and attempted to evaluate if there was any complex preference of arsenic that could trigger apoptosis. We also evaluated if chelation with monoisoamyl dimercaptosuccinic acid (MiADMSA) could reverse these detrimental effects. Our results indicate that arsenic exposure induced free radical generation in rat neuronal cells, which diminished mitochondrial potential and enzyme activities of all the complexes of the electron transport chain. Moreover, these complexes showed differential responses towards arsenic. These early events along with diminished ATP levels could be co-related with the later events of cytosolic migration of cytochrome c, altered bax/bcl(2) ratio, and increased caspase 3 activity. Although MiADMSA could reverse most of these arsenic-induced altered variables to various extents, DNA damage remained unaffected. Our study for the first time demonstrates the differential effect of arsenic on the complexes leading to deficits in bioenergetics leading to apoptosis in rat brain. However, more in depth studies are warranted for better understanding of arsenic interactions with the mitochondria.


Asunto(s)
Apoptosis/efectos de los fármacos , Intoxicación por Arsénico/tratamiento farmacológico , Quelantes/farmacología , Metabolismo Energético/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Succímero/análogos & derivados , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Transporte de Electrón/efectos de los fármacos , Complejo IV de Transporte de Electrones/efectos de los fármacos , Complejo IV de Transporte de Electrones/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , NADH Deshidrogenasa/efectos de los fármacos , NADH Deshidrogenasa/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/análisis , Succímero/farmacología , Succinato Deshidrogenasa/efectos de los fármacos , Succinato Deshidrogenasa/metabolismo
12.
J Appl Toxicol ; 31(6): 499-505, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21607993

RESUMEN

Aluminum phosphide (ALP), a widely used insecticide and rodenticide, is also infamous for the mortality and morbidity it causes in ALP-poisoned individuals. The toxicity of metal phosphides is due to phosphine liberated when ingested phosphides come into contact with gut fluids. ALP poisoning is lethal, having a mortality rate in excess of 70%. Circulatory failure and severe hypotension are common features of ALP poisoning and frequent cause of death. Severe poisoning also has the potential to induce multi-organ failure. The exact site or mechanism of its action has not been proved in humans. Rather than targeting a single organ to cause gross damage, ALP seems to work at the cellular level, resulting in widespread damage leading to multiorgan dysfunction (MOD) and death. There has been proof in vitro that phosphine inhibits cytochrome c oxidase. However, it is unlikely that this interaction is the primary cause of its toxicity. Mitochondria could be the possible site of maximum damage in ALP poisoning, resulting in low ATP production followed by metabolic shutdown and MOD; also, owing to impairment in electron flow, there could be free radical generation and damage, again producing MOD. Evidence of reactive oxygen species-induced toxicity owing to ALP has been observed in insects and rats. A similar mechanism could also play a role in humans and contribute to the missing link in the pathogenesis of ALP toxicity. There is no specific antidote for ALP poisoning and supportive measures are all that are currently available.


Asunto(s)
Compuestos de Aluminio/envenenamiento , Insuficiencia Multiorgánica/inducido químicamente , Fosfinas/envenenamiento , Intoxicación/patología , Animales , Antídotos/farmacología , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/metabolismo , Humanos , Insecticidas/envenenamiento , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Insuficiencia Multiorgánica/patología , Intoxicación/diagnóstico , Intoxicación/epidemiología , Intoxicación/terapia , Especies Reactivas de Oxígeno
13.
Cells ; 10(10)2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34685770

RESUMEN

Alzheimer's disease (AD) is one of the most prominent neurodegenerative diseases, which impairs cognitive function in afflicted individuals. AD results in gradual decay of neuronal function as a consequence of diverse degenerating events. Several neuroimmune players (such as cytokines and growth factors that are key players in maintaining CNS homeostasis) turn aberrant during crosstalk between the innate and adaptive immunities. This aberrance underlies neuroinflammation and drives neuronal cells toward apoptotic decline. Neuroinflammation involves microglial activation and has been shown to exacerbate AD. This review attempted to elucidate the role of cytokines, growth factors, and associated mechanisms implicated in the course of AD, especially with neuroinflammation. We also evaluated the propensities and specific mechanism(s) of cytokines and growth factors impacting neuron upon apoptotic decline and further shed light on the availability and accessibility of cytokines across the blood-brain barrier and choroid plexus in AD pathophysiology. The pathogenic and the protective roles of macrophage migration and inhibitory factors, neurotrophic factors, hematopoietic-related growth factors, TAU phosphorylation, advanced glycation end products, complement system, and glial cells in AD and neuropsychiatric pathology were also discussed. Taken together, the emerging roles of these factors in AD pathology emphasize the importance of building novel strategies for an effective therapeutic/neuropsychiatric management of AD in clinics.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Citocinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Enfermedad de Alzheimer/inmunología , Quimiocinas/metabolismo , Humanos , Inflamación/patología , Modelos Biológicos
14.
Toxicol Appl Pharmacol ; 240(2): 208-18, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19560481

RESUMEN

Recent global events have focused attention on the potential threat of international and domestic chemical terrorism, as well as the possibility of chemical warfare proliferation. Sulphur mustard (SM) is one of the potent chemical warfare agents (CWA), which initiates a cascade of events that converge on the redox mechanisms common to brain injury. The present study was designed to examine the effects of chronic SM exposure on neurobehavioral impairments, mitochondrial oxidative stress in male Swiss Albino mice and its role in inducing apoptotic neuronal cell death. The animals were divided into four groups (control, low, medium and high dose) of 5 animals each. Exposure to SM was given percutaneously daily for 12 weeks. The results demonstrated impairment in neurobehavioral indices viz. rota rod, passive avoidance and water maze tests in a dose dependent manner. There was a significant increase in lipid peroxidation and protein carbonyl content whereas, decrease in the activity of manganese superoxide dismutase (MnSOD), glutathione reductase and glutathione peroxidase suggesting impaired antioxidant defense system. Immunoblotting of cytochrome c, Bcl-2, Bax and activation of caspase-3 suggest induction of apoptosis in a dose dependent manner. Finally, increased p53 expression suggests that it may target the mitochondrial pathway for inducing apoptosis in response to DNA damage signals. In conclusion, chronic SM exposure may have the potential to generate oxidative stress which may trigger the release of cytochrome c as well as caspase-3 activation in neurons leading to cell death by apoptosis in a dose dependent manner which may in the end be responsible for the disruption of cognitive functions in mice.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Sustancias para la Guerra Química/toxicidad , Gas Mostaza/toxicidad , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Encéfalo/ultraestructura , Caspasa 3/metabolismo , Citocromos c/metabolismo , Relación Dosis-Respuesta a Droga , Glutatión Peroxidasa/metabolismo , Glutatión Reductasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Destreza Motora/efectos de los fármacos , Neuronas/metabolismo , Neuronas/ultraestructura , Carbonilación Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Superóxido Dismutasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
15.
Toxicology ; 255(3): 117-23, 2009 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-19010380

RESUMEN

Aluminium has been implicated in various neurodegenerative diseases but exact mechanism of action is still not known. Mitochondria being a major site of reactive oxygen species production are considered to be target of oxidative stress and it seems that the oxidative damage to mitochondrial proteins may underlie the pathogenesis of aluminium induced neurodegeneration. Thus, the present study was undertaken to reveal the effects of chronic aluminium exposure (10mg/kg b.wt, intragastrically for 12 weeks) on the oxidative damage to mitochondrial proteins in male albino Wistar rats. Chronic aluminium exposure resulted in decrease in the activity of mitochondrial superoxide dismutase (MnSOD) and aconitase in different regions of rat brain suggesting increased oxidative stress. This decrease in MnSOD activity in turn might be responsible for the increased protein oxidation as observed in our study. All these processes taken together may cause increased oxidative damage to mitochondrial proteins in general. By taking the advantage of recent immunochemical probe for oxidatively modified proteins, we identified MnSOD to be susceptible to oxidative damage in aluminium treated animals. The quantitative RT-PCR analysis for Lon protease, a protease involved in the removal of oxidatively modified proteins from mitochondria, showed decreased mRNA expression suggesting increased oxidative damage and decreased removal of mitochondrial proteins. The identification of specific proteins as targets of oxidative damage may provide new therapeutic measures to reverse the effects of aluminium induced neurodegeneration.


Asunto(s)
Aluminio/toxicidad , Mitocondrias/efectos de los fármacos , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Aconitato Hidratasa/metabolismo , Animales , Secuencia de Bases , Peso Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Masculino , Mitocondrias/enzimología , Ratas , Ratas Wistar
16.
Toxicology ; 256(3): 175-82, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19100812

RESUMEN

The present study was designed to explore the alternative mechanism (other than AChE inhibition) for chronic, low-level exposure to dichlorvos, an organophosphate, in vivo. Dichlorvos, at a dose of 1.0 and 6.0 mg/kg body weight (b.wt.) for 12 weeks, showed impairment in neurobehavioral indices viz. rota rod, passive avoidance and water maze tests. Though high dose of dichlorvos had a detrimental effect on acetylcholinesterase activity, no significant inhibition was seen with low dose of dichlorvos. Western blot analysis and immunofluorescence studies showed a significant reduction in the expression of M(1), M(2) and M(3) muscarinic receptor subtypes in high dose group animals, whereas in low dose group animals only the M(2) receptor subtype was reduced significantly. Further, the signal transduction cascade linked to these receptor subtypes was affected in high dose group animals whereas in low dose group only adenylyl cyclase-linked signaling pathway was impaired. Finally, the phosphorylation of CREB, a memory enhancing transcription factor, was significantly reduced in both low dose and high dose group animals. Thus, the present study reveals the significance of M(2) muscarinic receptor linked adenylyl cyclase signaling pathway and phosphorylation of CREB in the development of neurobehavioral impairments after chronic low-level exposure to dichlorvos.


Asunto(s)
Acetilcolina/metabolismo , Inhibidores de la Colinesterasa/toxicidad , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Diclorvos/toxicidad , Trastornos de la Memoria/inducido químicamente , Receptores Muscarínicos/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Actividad Motora/efectos de los fármacos , Fosforilación , Ratas , Ratas Wistar , Receptores Muscarínicos/biosíntesis , Transducción de Señal , Membranas Sinápticas/efectos de los fármacos , Membranas Sinápticas/metabolismo
17.
Pharmacol Biochem Behav ; 92(1): 173-81, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19101586

RESUMEN

The present study was designed to explore an alternate mechanism of action other than acetylcholinesterase inhibition for the chronic, low-level exposure to dichlorvos, an organophosphate, in vivo. Dichlorvos, at dose of 1.0 as well as 6.0 mg/kg b. wt. for 12 weeks to rats showed impairment in neurobehavioral indices viz. rota rod, passive avoidance and water maze tests. Though higher dose of dichlorvos had a detrimental effect on acetylcholinesterase activity, no significant inhibition was seen with lower dose of dichlorvos for the same period of exposure i.e. 12 weeks. Muscarinic acetylcholine receptor binding studies revealed a decrease in the number of binding sites (B(max)) in low as well as high dose groups but the dissociation constant (K(d)) value was unaffected with both doses of dichlorvos. Use of selective ligands against M(1), M(2) and M(3) receptor subtypes indicated that M(2) is the major receptor subtype being affected by chronic low-level exposure to dichlorvos. Western blot analysis and immunofluorescence studies also confirmed these biochemical findings. Thus, the present study suggests that M(2) receptors may play a major role in the development of neurobehavioral impairments after chronic exposure to dichlorvos.


Asunto(s)
Acetilcolinesterasa/fisiología , Conducta Animal/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Diclorvos/farmacología , Desempeño Psicomotor/efectos de los fármacos , Animales , Reacción de Prevención/efectos de los fármacos , Western Blotting , Química Encefálica/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Enfermedad Crónica , Relación Dosis-Respuesta a Droga , Técnica del Anticuerpo Fluorescente , Ligandos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Recuerdo Mental/efectos de los fármacos , Ratas , Ratas Wistar , Receptores Muscarínicos/efectos de los fármacos , Percepción Espacial/efectos de los fármacos
18.
Arch Toxicol ; 83(11): 965-78, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19568732

RESUMEN

Aluminium is the most widely distributed metal in the environment and is extensively used in daily life that provides easy exposure to human beings. The exposure to this toxic metal occurs through air, food and water. However, there is no known physiological role for aluminium within the body and hence this metal may produce adverse physiological effects. Chronic exposure of animals to aluminium is associated with behavioural, neuropathological and neurochemical changes. Among them, deficits of learning and behavioural functions are most evident. Some epidemiological studies have shown poor performance in cognitive tests and a higher abundance of neurological symptoms for workers occupationally exposed to aluminium. However, in contrast to well established neurotoxic effects, neurobehavioural studies of aluminium in rodents have generally not produced consistent results. Current researches show that any impairment in mitochondrial functions may play a major role in many human disorders including neurodegenerative disorders. Being involved in the production of reactive oxygen species, aluminium may cause impairments in mitochondrial bioenergetics and may lead to the generation of oxidative stress which may lead to a gradual accumulation of oxidatively modified cellular proteins. In this review, the neuropathologies associated with aluminium exposure in terms of neurobehavioural changes have been discussed. In addition, the impact of aluminium on the mitochondrial functions has also been highlighted.


Asunto(s)
Aluminio/toxicidad , Síndromes de Neurotoxicidad/etiología , Aluminio/farmacocinética , Animales , Conducta Animal/efectos de los fármacos , Exposición a Riesgos Ambientales/efectos adversos , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Síndromes de Neurotoxicidad/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Distribución Tisular
19.
Neurotox Res ; 35(3): 621-634, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30729451

RESUMEN

Alzheimer's disease (AD) is the most common cause of progressive decline of memory function in aged humans. To study about a disease mechanism and progression, animal models for the specific disease are needed. For AD, although highly valid animal models exist, none of the existing models recapitulates all aspects of human AD. The pathogenic mechanisms involved in AD are diverse and thus it is difficult to recapitulate human AD in model organisms. Intracerebroventricular (ICV) injection of okadaic acid (OKA), a protein phosphatase 2A (PP2A) inhibitor, in rats causes neurotoxicity associated with neurofibrillary degeneration. However, this model lacks amyloid pathology as observed in AD. We aimed at combining two different treatments and hence producing a better animal model of AD which may mimic most of the neuropathological, neurobehavioral, and neurochemical changes observed in AD. For this, OKA (200 ng) was microinjected bilaterally into the hippocampus of male Wistar rats followed by exposure of same rats to hypoxic conditions (10%) for 3 days. The result of which, the combination model exhibited tau hyperphosphorylation along with Aß upregulation as evident by western blotting and immunohistochemistry. The observed changes were accompanied with dysfunction of neurotransmitter system, i.e., decreased acetylcholine activity and expression. This combinatorial model also exhibited cognitive deficiency which was assessed by Morris water maze and avoidance tests along with enhanced oxidative stress which is thought to be a major player in AD pathogenesis. Taken together, we established an easily reproducible and reliable rat model for sporadic dementia of Alzheimer's type in rats which allows effective testing of new therapeutic strategies.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Reacción de Prevención , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Hipocampo/metabolismo , Hipocampo/patología , Hipoxia , Masculino , Aprendizaje por Laberinto , Microinyecciones , Neuronas/metabolismo , Neuronas/patología , Ácido Ocadaico , Estrés Oxidativo/fisiología , Ratas Wistar , Técnicas Estereotáxicas
20.
Brain Res ; 1232: 94-103, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18691561

RESUMEN

The present study was designed with an aim to evaluate the effects of chronic aluminium exposure (10 mg/kg b.wt, intragastrically for 12 weeks) on mitochondrial energy metabolism in different regions of rat brain in vivo. Mitochondrial preparations from aluminium treated rats revealed significant decrease in the activity of various electron transport complexes viz. cytochrome oxidase, NADH cytochrome c reductase and succinic dehydrogenase as well, in the hippocampus region. The decrease in the activity of these respiratory complexes was also seen in the other two regions viz. corpus striatum and cerebral cortex, but to a lesser extent. This decrease in the activities of electron transport complexes in turn affected the ATP synthesis and ATP levels adversely in the mitochondria isolated from aluminium treated rat brain regions. We also studied the spectral properties of the mitochondrial cytochromes viz. cyt a, cyt b, cyt c1, and cyt c in both control and treated rat brains. The various cytochrome levels were found to be decreased following 12 weeks of aluminium exposure. Further, these impairments in mitochondrial functions may also be responsible for the production of reactive oxygen species and impaired antioxidant defense system as observed in our study. The electron micrographs of neuronal cells depicted morphological changes in mitochondria as well as nucleus only from hippocampus and corpus striatum regions following 12 weeks exposure to aluminium. The present study thus highlights the significance of altered mitochondrial energy metabolism and increased ROS production as a result of chronic aluminium exposure in different regions of the rat brain.


Asunto(s)
Aluminio/farmacología , Química Encefálica/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Citocromos/metabolismo , Transporte de Electrón/efectos de los fármacos , Complejo IV de Transporte de Electrones/metabolismo , Glutatión/metabolismo , Masculino , Microscopía Electrónica , Mitocondrias/enzimología , NADH Deshidrogenasa/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Fosfatos/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Succinato Deshidrogenasa/metabolismo , Sales de Tetrazolio , Tiazoles
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA