Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Chem Neurosci ; 15(6): 1084-1095, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38462729

RESUMO

People travel to high-altitude regions as tourists, workers, and military personnel on duty. Despite the consistent 21% oxygen content in the atmosphere, ascending to higher altitudes results in a decrease in the partial pressure of oxygen, inducing a state known as hypobaric hypoxia (HH). HH is an environmental stress that is responsible for neuroinflammation and behavioral deficits (anxiety, depression, mood disturbance, etc.), but little is known about its metabolic pathways. The kynurenine pathway (KP) is a promising candidate to uncover the mysteries of HH stress, as it is an important regulator of the immune system and is associated with behavioral deficits. To investigate the role of KP under HH, the levels of KP metabolites in the serum, cerebrospinal fluid (CSF), and brain tissue (prefrontal cortex-PFC, neocortex, and hippocampus) of male Sprague-Dawley rats exposed to HH at 7620 m for 1, 3, and 7 days were estimated utilizing high-performance liquid chromatography (HPLC). The behavioral analogs for anxiety-like and depression-like behavior were assessed using the open field test and forced swim test, respectively. Upon HH exposure, crosstalk between the periphery and central nervous system and KP metabolite region-dependent differential expression in the brain were observed. KP metabolites showed a positive correlation with behavioral parameters. The results of our study are indicative that KP can be proposed as the etiology of behavioral deficits, and KP metabolite levels in serum or CSF can be used as plausible markers for anxiety-like and depression-like behaviors under HH stress with a scope of targeted therapeutic interventions.


Assuntos
Hipóxia , Cinurenina , Humanos , Ratos , Masculino , Animais , Ratos Sprague-Dawley , Hipóxia/metabolismo , Hipocampo/metabolismo , Oxigênio/metabolismo
3.
Toxicol Ind Health ; 39(3): 127-137, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36680355

RESUMO

Zinc oxide nanoparticles (ZnO NP) are commonly used engineered NPs with extensive usage in consumer products, thus leading to direct exposure to humans. The direct route of exposure is through inhalation. Once inhaled, these particles accumulate in the lungs, increasing the chances of respiratory tract illness through cellular organelle damage. Zinc oxide nanoparticle-treated lung cells are reported to display cytotoxicity, increase DNA damage, and induce oxidative stress. The current study focused on the effects of ZnO NPs on mitochondrial dynamics (fission and fusion) in human lung epithelial cells (A549). The lung cells were exposed to ZnO NPs at 50 and 100 µg/ml concentrations, and their mitochondrial dynamics were assessed to understand the effects of the NPs. Treatment with ZnO NPs reduced the activity of mitochondrial complex I and complex III and altered mitochondrial structural and functional characteristics in a concentration-dependent manner. Zinc oxide nanoparticles exposure showed an increase in small and round-shaped mitochondria. The expression of various fission proteins (Drp1 and Fis1) and fusion proteins (Mfn1, Mfn2, and OPA1) was altered upon exposure to ZnO NPs. Our studies showed dysfunction of the mitochondria induced by ZnO NPs. In fibroblast mitochondrial dynamics, fission symbolizes threshold damage. In this paper, we have shown that the mitochondrial fission phenotype increased upon exposure to ZnO NPs. The paper emphasizes that these particles enter mitochondria, triggering a stress response that results in the removal of mitochondria via fission. It provides relevant data for safety guidelines to ensure the safer use of these particles.


Assuntos
Nanopartículas , Óxido de Zinco , Humanos , Óxido de Zinco/toxicidade , Células Epiteliais Alveolares , Espécies Reativas de Oxigênio/metabolismo , Nanopartículas/toxicidade , Mitocôndrias
4.
Chem Biol Interact ; 369: 110284, 2023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-36462549

RESUMO

ZnO nanoparticles (ZnO NPs) are widely used engineered nanomaterials. Due to induced genotoxicity, increased oxidative stress, and teratogenicity, these NPs have been reported to be toxic. In the present study, we emphasise the role of vital proteins in regulating ZnO NP-induced abnormal phenotypes, particularly the deformed thorax and single wing in the Drosophila melanogaster progeny fed on 0.1-10 mM ZnO NPs. To understand how protein expression regulates this particular phenotype on ZnO NPs exposure, toxicoproteomics profile of control and abnormal phenotype flies was generated using LC/MS/MS. Gene ontology enrichment studies of proteomics data were carried out using CLUEGO and STRAP software. The bioinformatics tool STRING was used to generate a protein-protein interaction map of key proteins of enrichment analysis. Following ZnO NP exposure, the differential expression of key proteins of the Wnt pathway was prominent. Altered expression of various proteins of the Wnt pathway (CaMKII), cytoskeleton (Actin), and calponin resulted in developmental defects in drosophila progeny. In addition, immunohistology studies showed a significant deviation in the expression of wingless protein of ZnO NPs treated larvae in comparison to control. According to these findings, the interaction of the wnt pathway and cytoskeletal proteins with ZnO NPs caused developmental abnormalities in the subsequent generation of drosophila, highlighting the transgenerational toxic effects of these nanoparticles.


Assuntos
Óxido de Zinco , Animais , Óxido de Zinco/toxicidade , Drosophila , Via de Sinalização Wnt , Drosophila melanogaster , Espectrometria de Massas em Tandem , Estresse Oxidativo , Proteínas do Citoesqueleto , Citoesqueleto , Calponinas
5.
RSC Adv ; 12(20): 12310-12320, 2022 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-35480352

RESUMO

Graphene quantum dots (GQDs) are a luminescent class of carbon nanomaterials with a graphene-like core structure, possessing quantum confinement and edge effects. They have gained importance in the biological world due to their inherent biocompatibility, good water dispersibility, excellent fluorescence and photostability. The improved properties of GQDs require the logical enactment of functional groups, which can be easily attained through post-synthetic non-covalent routes of modification. In this regard, the present work has for the first time employed a simple one-pot post-modification method utilizing the salt of amino caproic acid, an FDA approved reagent. The adsorption of the modifier on GQDs with varying weight ratios is characterized through DLS, zeta potential, Raman, absorption and fluorescence spectroscopy. A decrease of 20% in the fluorescence intensity with an increase in the modifier ratio from 1 to 1000 and an increased DLS size as well as zeta potential demonstrate the efficient modification as well as higher stability of the modified GQDs. The modified GQDs with a high weight ratio (1 : 100) of the modifier showed superior ability to sense dopamine, a neurotransmitter, as well as competent biofilm degradation ability. The modified GQDs could sense more efficiently than pristine GQDs, with a sensitivity as low as 0.06 µM (limit of detection) and 90% selectivity in the presence of other neurotransmitters. The linear relationship showed a decrease in the fluorescence intensity with increasing dopamine concentration from 0.0625 µM to 50 µM. Furthermore, the efficiency of the modified GQDs was also assessed in terms of their antibiofilm effect against Staphylococcus aureus. The unmodified GQDs showed only 10% disruption of the adhered bacterial colonies, while the modified GQDs (1 : 100) showed significantly more than 60% disruption of the biofilm, presenting the competency of the modified GQDs. The unique modifications of GQDs have thus proven to be an effective method for the proficient utilization of zero-dimensional carbon nanomaterials for biosensing, bioimaging, antibacterial and anti-biofilm applications.

6.
J Psychiatr Res ; 149: 155-161, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35276632

RESUMO

The present study is conducted to understand the association of mood profile with the kynurenine pathway (KP) metabolites, and cerebral hemodynamics in freshly recruited central armed forces personnel. Profile of Mood States questionnaire was utilized to assess mood profile, and Total Mood Disturbance (TMD) score was calculated. Transcranial Doppler was used to record blood flow velocity bilaterally of the middle cerebral artery. Chromatographic profile of the kynurenine metabolites was obtained in serum. Further, personnel were stratified according to sociodemographic variables (gender, age and diet) to observe the changes in their KP metabolic status. An activation of the kynurenic acid branch of the KP and the reduction in the mean blood flow velocity, and an increase in Gosling pulsatility index (PI) were observed in females having high TMD score. On gender comparative analysis, kynurenine metabolites of quinolinic acid branch and serotonin were significantly high in males. In males, with increase in age, a significant increase in the quinolinic acid branch of the KP was observed. Furthermore, a significant difference in level metabolites of the KP among the vegetarian and non-vegetarian groups was also observed. In conclusion we observed that increased TMD score was associated with cerebral hypoperfusion and higher vascular resistance along with activation of the KP. Our findings highlighted the importance of multi-facet brain function to showcase the close interaction of various dimensionalities and true picture of the assessee.


Assuntos
Cinurenina , Militares , Animais , Circulação Cerebrovascular , Feminino , Gansos/metabolismo , Humanos , Masculino , Ácido Quinolínico/metabolismo
7.
Behav Brain Res ; 416: 113568, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34499936

RESUMO

Hypobaric Hypoxia (HH) is known to cause oxidative stress in the brain that leads to spatial memory deficit and neurodegeneration. For decades therapeutic hypothermia is used to treat global and focal ischemia in preserving brain functions that proved to be beneficial in humans and rodents. Considering these previous reports, the present study was designed to establish the therapeutic potential of hypothermia preconditioning on HH induced spatial memory, biochemical and morphological changes in adult rats. Male Sprague Dawley rats were exposed to HH (7620 m, ~ 282 mmHg) for 1, 3 and 7 days with and without hypothermic preconditioning. Spatial learning memory was assessed by Morris water maze (MWM) test along with evaluation of hippocampal pyramidal neuron damage by histological study. Oxidative stress was measured by studying the levels of nitric oxide (NO), reactive oxygen species (ROS), lipid peroxidation (LPO), oxidized and reduced glutathione (GSSG and GSH). Results of MWM test indicated prolonged path length and latency to reach the platform in HH groups that regained to normal in cold pre-treated groups. A likely neurodegeneration was evident in HH groups that lessen in the cold pre-treated groups. Hypothermic preconditioning prevented spatial memory impairment and neurodegeneration in animals subjected to HH via decreasing the NO, ROS and LPO compared to control animals. The GSH level and GSH/GSSG ratio was found to be higher in preconditioned animals as compared to respective HH exposed animals, indicative of redox scavenging and restoration of hippocampal neuronal structure as well as spatial memory. Therefore, hypothermic preconditioning improves spatial memory deficit by reducing HH induced oxidative stress and hippocampal neurodegeneration, hence can be used as a multi-target prophylactic measure to combat HH induced neurodegeneration.


Assuntos
Hipocampo/fisiopatologia , Hipotermia/induzido quimicamente , Hipóxia Encefálica/fisiopatologia , Transtornos da Memória/fisiopatologia , Células Piramidais/patologia , Memória Espacial/fisiologia , Animais , Glutationa/metabolismo , Hipocampo/patologia , Hipóxia Encefálica/patologia , Peroxidação de Lipídeos/fisiologia , Masculino , Teste do Labirinto Aquático de Morris , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio
8.
Int J Radiat Biol ; 97(11): 1606-1616, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34402374

RESUMO

PURPOSE: The model biological organism Drosophila melanogaster has been utilized to assess the effect of extremely low-frequency electromagnetic field (ELF-EMF) on locomotion, longevity, developmental dynamics, cell viability and oxidative stress. MATERIALS AND METHOD: Developmental stages of Drosophila melanogaster (Oregon R strain) individually exposed to ELF-EMF (75 Hz, 550 µT) for 6 h once for acute exposure. For chronic exposure, complete life cycle of fly, that is, egg to adult fly was exposed to ELF-EMF for 6 h daily. The effect of exposure on their crawling and climbing ability, longevity, development dynamics, cellular damage and oxidative stress (generation of reactive oxygen species (ROS)) was evaluated. RESULTS: The crawling ability of larvae was significantly (p < .05) reduced on acute (third stage instar larvae) as well as chronic exposure (F0 and F1 larvae). When locomotion of flies was tested using climbing assay, no alteration was observed in their climbing ability under both acute and chronic exposure; however, when their speed of climbing was compared, a significant decrease in speed of F1 flies was observed (p = .0027) on chronic exposure. The survivability of flies was significantly affected under chronic and acute exposure (at third stage instar larvae). In case of acute exposure of the third stage instar larvae, although all the flies were eclosed by the 17th day, there was a significant decline in the number of flies (p = .007) in comparison to control. While in case of chronic exposure apart from low number of flies eclosed in comparison to control, there was delay in eclosion by one day (p = .0004). Using trypan blue assay, the internal gut damage of third stage instar larvae was observed. Under acute exposure condition at third stage instar larvae, 30% larvae has taken up trypan blue, while only 10% larvae from acute exposure at adult stage. On chronic exposure, 50% larvae of the F1 generation have taken up trypan blue. On evaluation of oxidative stress, there is a significant rise in ROS in case of acute exposure at third stage instar larvae (p = .0004), adult fly stage (p = .0004) and chronic exposure (p = .0001). CONCLUSION: ELF-EMF has maximum effects on acute exposure of third stage instar larvae and chronic exposure (egg to adult fly stage). These results suggest that electromagnetic radiations, though, have become indispensible part of our lives but they plausibly affect our health.


Assuntos
Drosophila melanogaster , Campos Eletromagnéticos , Animais , Campos Eletromagnéticos/efeitos adversos , Larva , Estresse Oxidativo , Espécies Reativas de Oxigênio , Azul Tripano
9.
Front Pharmacol ; 12: 669701, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34326768

RESUMO

Hypobaric hypoxia (HH) is a stressful condition, which is more common at high altitudes and can impair cognitive functions. Ginkgo biloba L. leaf extract (GBE) is widely used as herbal medicine against different disorders. Its ability to improve cognitive functions, reduce oxidative stress, and promote cell survival makes it a putative therapeutic candidate against HH. The present study has been designed to explore the effect of GBE on HH-induced neurodegeneration and memory impairment as well as possible signaling mechanisms involved. 220-250 gm (approximately 6- to 8-week-old) Sprague Dawley rats were randomly divided into different groups. GBE was orally administered to respective groups at a dose of 100 mg/kg/day throughout the HH exposure, i.e., 14 days. Memory testing was performed followed by hippocampus isolation for further processing of different molecular and morphological parameters related to cognition. The results indicated that GBE ameliorates HH-induced memory impairment and oxidative damage and reduces apoptosis. Moreover, GBE modulates the activity of the small conductance calcium-activated potassium channels, which further reduces glutamate excitotoxicity and apoptosis. The exploration of the downstream signaling pathway demonstrated that GBE administration prevents HH-induced small conductance calcium-activated potassium channel activation, and that initiates pro-survival machinery by activating extracellular signal-regulated kinase (ERK)/calmodulin-dependent protein kinase II (CaMKII) and the cAMP response element-binding protein (CREB) signaling pathway. In summary, the current study demonstrates the beneficial effect of GBE on conditions like HH and provides various therapeutic targets involved in the mechanism of action of GBE-mediated neuroprotection.

10.
Exp Brain Res ; 239(6): 1747-1763, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33779792

RESUMO

Professionals and mountaineers often face the problem of reperfusion injury due to re-oxygenation, upon their return to sea-level after sojourn at high altitude. Small conductance calcium-activated potassium channels (SK channels) have a role in regulating hippocampal synaptic plasticity. However, the role of SK channels under hypoxia-reoxygenation (H/R) is unknown. The present study hypothesized that SK channels play a significant role in H/R induced cognitive dysfunction. Sprague-Dawley rats were exposed to simulated HH (25,000 ft) continuously for 7 days followed by reoxygenation periods 3, 6, 24, 48, 72 and 120 h. It was observed that H/R exposure caused impairment in spatial memory as indicated by increased latency (p < 0.001) and pathlength (p < 0.001). The SK1 channel expression increased upon HH exposure (102.89 ± 7.055), which abrogated upon reoxygenation. HH exposure results in an increase in SK2 (CA3, 297.67 ± 6.69) and SK3 (CA1, 246 ± 5.13) channels which continued to increase gradually upon reoxygenation. The number of pyknotic cells (24 ± 2.03) (p < 0.01) and the expression of caspase-3 increased with HH exposure, which continued in the reoxygenation group (177.795 ± 1.264). Similar pattern was observed in lipid peroxidation (p < 0.001), LDH activity (p < 0.001) and ROS production (p < 0.001). A positive correlation of memory, cell death and oxidative stress indicates that H/R exposure increases oxidative stress coupled with SK channel expression, which may play a role in H/R-induced cognitive decline and neurodegeneration.


Assuntos
Hipocampo , Transtornos da Memória , Animais , Hipóxia , Transtornos da Memória/etiologia , Ratos , Ratos Sprague-Dawley , Memória Espacial
11.
J Cell Physiol ; 236(9): 6754-6771, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33788269

RESUMO

Hypobaric hypoxia at higher altitudes usually impairs cognitive function. Previous studies suggested that epigenetic modifications are the culprits for this condition. Here, we set out to determine how hypobaric hypoxia mediates epigenetic modifications and how this condition worsens neurodegeneration and memory loss in rats. In the current study, different duration of hypobaric hypoxia exposure showed a discrete pattern of histone acetyltransferases and histone deacetylases (HDACs) gene expression in the hippocampus when compared with control rat brains. The level of acetylation sites in histone H2A, H3 and H4 was significantly decreased under hypobaric hypoxia exposure compared to the control rat's hippocampus. Additionally, inhibiting the HDAC family with sodium butyrate administration (1.2 g/kg body weight) attenuated neurodegeneration and memory loss in hypobaric hypoxia-exposed rats. Moreover, histone acetylation increased at the promoter regions of brain-derived neurotrophic factor (BDNF); thereby its protein expression was enhanced significantly in hypobaric hypoxia exposed rats treated with HDAC inhibitor compared with hypoxic rats. Thus, BDNF expression upregulated cAMP-response element binding protein (CREB) phosphorylation by stimulation of PI3K/GSK3ß/CREB axis, which counteracts hypobaric hypoxia-induced spatial memory impairment. In conclusion, these results suggested that sodium butyrate is a novel therapeutic agent for the treatment of spatial memory loss associated with hypobaric hypoxia, and also further studies are warranted to explore specific HDAC inhibitors in this condition.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Hipóxia/complicações , Transtornos da Memória/etiologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Memória Espacial , Acetilação/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Hipocampo/metabolismo , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Masculino , Transtornos da Memória/metabolismo , Modelos Biológicos , Degeneração Neural/complicações , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuroproteção/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos
12.
ACS Chem Neurosci ; 11(20): 3194-3203, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33006881

RESUMO

The world is experiencing one of the major viral outbreaks of this millennium, caused by a plus sense single-stranded RNA virus belonging to the Coronaviridae family, COVID-19, declared as pandemic by WHO. The clinical manifestations vary from asymptomatic to mild symptoms like fever, dry cough, and diarrhea, with further increase in severity leading to the development of acute respiratory distress syndrome. Though primary manifestations are respiratory and cardiac, various studies have shown the neuroinvasive capability of this virus resulting in neurological complications, which sometimes can precede common typical symptoms like fever and cough. Common neurological symptoms are headache, dizziness, anosmia, dysgeusia, confusion, and muscle weakening, progressing toward severe complications like cerebrovascular disease, seizures, or paralysis. Older adults and critically ill people are in the high risk group and have shown severe neurological symptoms upon infection. COVID-19 also has a profound impact on the mental health of people across the world. In this review, we briefly discuss the neurological pathologies and psychological impact due to COVID-19, which has not only stressed the physical health of people but has also created social and economic problems resulting in mental health issues.


Assuntos
Infecções por Coronavirus/patologia , Infecções por Coronavirus/psicologia , Transtornos Mentais/virologia , Doenças do Sistema Nervoso/virologia , Pneumonia Viral/patologia , Pneumonia Viral/psicologia , Betacoronavirus , COVID-19 , Humanos , Pandemias , SARS-CoV-2
13.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165769, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32184133

RESUMO

Brain is well known for its disproportionate oxygen consumption and high energy-budget for optimal functioning. The decrease in oxygen supply to brain, thus, necessitates rapid activation of adaptive pathways - the absence of which manifest into vivid pathological conditions. Amongst these, oxygen sensing in glio-vascular milieu and H2S-dependent compensatory increase in cerebral blood flow (CBF) is a major adaptive response. We had recently demonstrated that the levels of H2S were significantly decreased during chronic hypobaric hypoxia (HH)-induced neuro-pathological effects. The mechanistic basis of this phenomenon, however, remained to be deciphered. We, here, describe experimental evidence for marked limitation of cysteine during HH - both in animal model as well as human volunteers ascending to high altitude. We show that the preservation of brain cysteine level, employing cysteine pro-drug (N-acetyl-L-cysteine, NAC), markedly curtailed effects of HH - not only on endogenous H2S levels but also, impairment of spatial reference memory in our animal model. We, further, present multiple lines of experimental evidence that the limitation of cysteine was causally governed by physiological propensity of brain to utilize cysteine, in cystathionine beta synthase (CBS)-dependent manner, past its endogenous replenishment potential. Notably, decrease in the levels of brain cysteine manifested despite positive effect (up-regulation) of HH on endogenous cysteine maintenance pathways and thus, qualifying cysteine as a conditionally essential nutrient (CEN) during HH. In brief, our data supports an adaptive, physiological role of CBS-mediated cysteine-utilization pathway - activated to increase endogenous levels of H2S - for optimal responses of brain to hypobaric hypoxia.


Assuntos
Doença da Altitude/metabolismo , Encéfalo/metabolismo , Cistationina beta-Sintase/genética , Cisteína/metabolismo , Sulfeto de Hidrogênio/metabolismo , Acetilcisteína/farmacologia , Adaptação Fisiológica , Adulto , Doença da Altitude/tratamento farmacológico , Doença da Altitude/genética , Doença da Altitude/patologia , Animais , Encéfalo/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/genética , Cistationina beta-Sintase/metabolismo , Modelos Animais de Doenças , Metabolismo Energético/genética , Humanos , Hipóxia/tratamento farmacológico , Hipóxia/genética , Hipóxia/metabolismo , Masculino , Consumo de Oxigênio/genética , Pró-Fármacos/farmacologia , Ratos , Adulto Jovem
14.
Brain Behav Immun ; 82: 129-144, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31408672

RESUMO

BACKGROUND: An association between neuroinflammation, reduced adult neurogenesis, and cognitive impairment has been established in sleep deprivation (SD). Complement receptors are expressed on neuronal and glial cells, thus, regulate the neuroinflammation, neurogenesis and learning/memory. However, understanding of the effect of SD on the brain-immune system interaction associated with cognitive dysfunction and its mechanisms is obscure. We hypothesized that complement activation induced changes in inflammatory and neurogenesis related proteins might be involved in the cognitive impairment during SD. METHODOLOGY: Adult male Sprague Dawley rats were used. Rats were sleep deprived for 48 h using a novel automated SD apparatus. Dosage of BrdU (50 mg/kg/day, i.p. in 0.07 N NaOH), complement C3a receptor antagonist (C3aRA; SB290157; 1 mg/kg/day, i.p.) in 1.16% v/v PBS and complement C5a receptor antagonist (C5aRA; W-54011; 1 mg/kg/day, i.p.) in normal saline were used. Rats were subjected to spatial memory evaluation following SD. Hippocampal tissue was collected for biochemical, molecular, and immunohistochemical studies. T-test and ANOVA were used for the statistical analysis. RESULTS: An up-regulation in the levels of complement components (C3, C5, C3a, C5a) and receptors (C3aR and C5aR) in hippocampus, displayed the complement activation during SD. Selective antagonism of C3aR/C5aR improved the spatial memory performance of sleep-deprived rats. C3aR antagonist (C3aRA) or C5aR antagonist (C5aRA) treatment inhibited the gliosis, maintained inflammatory cytokines balance in hippocampus during SD. Complement C3aR/C5aR antagonism improved hippocampal adult neurogenesis via up-regulating the BDNF level following SD. Administration of C3aRA and C5aRA significantly maintained synaptic homeostasis in hippocampus after SD. Gene expression analysis showed down-regulation in the mRNA levels of signal transduction pathways (Notch and Wnt), differentiation and axogenous proteins, which were found to be improved after C3aRA/C5aRA treatment. These findings were validated at protein and cellular level. Changes in the corticosterone level and ATP-adenosine-NO pathway were established as the key mechanisms underlying complement activation mediated consequences of SD. CONCLUSION: Our study suggests complement (C3a-C3aR and C5a-C5aR) activation as the novel mechanism underlying spatial memory impairment via promoting neuroinflammation and adult neurogenesis decline in hippocampus during SD, thereby, complement (C3aR/C5aR) antagonist may serve as the novel therapeutics to improve the SD mediated consequences.


Assuntos
Ativação do Complemento/imunologia , Neuroimunomodulação/fisiologia , Privação do Sono/metabolismo , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Compostos Benzidrílicos/farmacologia , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/metabolismo , Ativação do Complemento/fisiologia , Complemento C3a/metabolismo , Hipocampo/metabolismo , Masculino , Neurogênese/imunologia , Neurogênese/fisiologia , Neuroimunomodulação/imunologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Complemento/metabolismo , Transdução de Sinais/fisiologia , Privação do Sono/imunologia , Memória Espacial/fisiologia , Lobo Temporal/metabolismo
15.
Nanotoxicology ; 13(7): 977-989, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31012773

RESUMO

Aluminum oxide (Al2O3) nanoparticles (NPs) have a wide number of applications which cause intentional and unintentional exposure to humans, making it important to understand the nano-bio interaction. In this study, we made an attempt to evaluate the toxic effects of Al2O3 NPs chronic exposure on Drosophila melanogaster. Flies were exposed to Al2O3 NPs at concentration 0.1 and 1 mM via ingestion throughout their lifespan and progeny flies were screened for behavioral and phenotypic abnormalities. Behavioral abnormalities in flies were recorded through larval crawling, climbing in flies and two taste testing. Chronic exposure of Al2O3 NPs resulted in the loss of appendages in flies resulting in five legs flies, four legs flies and absence of haltere. Exposure to Al2O3 NPs caused renal failure in flies as observed by swollen abdomen. Our observations clearly showed that these NPs could cause detrimental health ailments which relate to human birth deformities and kidney failure. Damage at the cellular level was studied through proteomic profiling. Three hundred and seven unique proteins were expressed on exposure to Al2O3 NPs and 51 proteins were differentially expressed. Enrichment analysis of differentially expressed proteins showed significant alteration in striated muscle cell differentiation, digestive tract morphogenesis, phototransduction, regulation of chromatin organization and DNA duplex unwinding.


Assuntos
Óxido de Alumínio/toxicidade , Nanopartículas Metálicas/toxicidade , Animais , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Humanos
16.
Neuroscience ; 388: 418-429, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30048783

RESUMO

Hypobaric Hypoxia (HH) is well-known to cause cognitive impairment and synaptic dysfunction which results in neurodegeneration. Although the role of small conductance calcium-activated potassium channels (SK channels) has been reported in synaptic plasticity, cognition and different neurological disorders; however, the precise role of SK channels in HH-induced memory impairment remains yet to be explored. We, therefore, hypothesized the pivotal role of SK channels in HH-induced cognitive decline and investigated the SK channel expression during different duration of HH exposure (Control, 1, 3, 7 and 14 days) at mRNA and protein level in male Sprague-Dawley rats. Further the role of SK channels in spatial memory and neurodegeneration were explored by inhibiting SK channel through Apamin (a known SK channel blocker). Results from the present study revealed that acute exposure of HH for 3 days leads to significant increase in expression of SK1 and SK3 channels at mRNA and protein levels, which upon chronic exposure restored to normal. Remarkably, SK2 channel expression showed gradual increase from 3 days till 14 days. Immunohistochemical analysis revealed similar pattern in different regions of the hippocampus. Additionally, SK channel inhibition with Apamin prevented HH-induced neurodegeneration and memory impairment as evident from decreased number of Fluoro Jade-positive cells, pyknotic cells, and caspase-3 expression and improved performance in the Morris water maze task. Thus, the present study demonstrates that SK channels play a crucial role in HH-induced cognitive decline and neurodegeneration.


Assuntos
Doença da Altitude/metabolismo , Hipóxia/metabolismo , Deficiências da Aprendizagem/metabolismo , Transtornos da Memória/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Pressão do Ar , Doença da Altitude/patologia , Doença da Altitude/psicologia , Animais , Apamina/farmacologia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Hipóxia/patologia , Hipóxia/psicologia , Deficiências da Aprendizagem/etiologia , Deficiências da Aprendizagem/patologia , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/psicologia , Bloqueadores dos Canais de Potássio/farmacologia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores
17.
Ann Neurosci ; 25(4): 191-200, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31000957

RESUMO

Hypobaric hypoxia (HH) is a major stress factor that is associated with physiological, biochemical, molecular and genomic alterations. Brain is the organ that reacts sensitively to oxygen deprivation, which leads to oxidative stress and cognitive function impairment. Our previous studies have reported that downregulation of brain derived neurotrophic factor (BDNF) leads to neurodegeneration and memory impairment. The aim of the present study was to investigate the effect of HH exposure on DNA methylation and its regulation in BDNF expression, neurodegeneration and spatial memory impairment. For this purpose, Sprague Dawley rats were exposed to HH at a simulated altitude of 25,000 feet for 14 days. Real-time polymerase chain reaction was used for transcriptional expression of DNA Methyltransferases (DNMTs) including DNMT1, DNMT3a and -DNMT3b, and immunoblotting was used for the translational expression of DNMT1, DNMT3a, DNMT3b, Methyl CpG binding protein 2 (MeCP2), pMeCP2 and BDNF in rat hippocampus. Additionally, neuronal morphology alteration and neurodegeneration in CA1 region of hippocampus were investigated though Cresyl violet (CV) staining and Fluoro-Jade C staining respectively. Results obtained suggested that HH exposure increased the expression of DNMT1 DNMT3b at the mRNA as well as protein level, whereas no significant change was observed in the level of DNMT3a. Furthermore, the level of pMeCP2 and BDNF were significantly decreased; however, the expression level of MeCP2 was significantly increased. The CV and Fluoro-Jade C-positive cells were significantly enhanced in the CA1 region of hippocampus in the HH exposed group as compared to unexposed rats. Thus, the present study concluded that HH decreases neuronal activation by the upregulation of DNA methylation and MeCP2 and decreased the expression of pMeCP2, which result in the downregulation of BDNF. The decreased BDNF expression is associated with neuronal loss and spatial memory impairment. This study highlights that DNMT inhibition could be an important therapeutic target for neurodegenerative diseases.

18.
Physiol Behav ; 175: 56-65, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28351559

RESUMO

Hypobaric hypoxia leads to decrease in cellular oxygen content which subsequently damages the hippocampus with an increase in brain oxidative stress and impairs the memory of the individual. In the present study, we have evaluated the cognitive impairment modulating activity of total oligomeric flavonoids fraction of Cyperus rotundus (TOF) in Sprague Dawley rats. The rats were trained for memory activity for a period of 7days followed by 7days exposure to 25,000ft. altitude and the spatial reference memory was evaluated. Behavioral analysis of the rats by Morris water maze experiment showed that TOF supplementation enhanced the spatial reference memory activity of the rats exposed to hypobaric hypoxia. The decrease in antioxidant status of the animals exposed to hypoxia was restored with TOF supplementation. The increase in ROS, lipid peroxidation products and protein carbonyls of the hippocampus was significantly decreased in animals with TOF administration. The histological assessment of the pyramidal cells of the hippocampus of hypoxia-exposed animals showed nuclear damage and TOF supplementation prevented nuclear damage. TOF administration suppressed hypoxia-induced increase in serotonin, dopamine, and norepinephrine. GABA and Ach levels were decreased by hypoxia which was prevented by TOF supplementation. The increase in GFAP, HIF-1α and VEGF expression in CA3 region of the hippocampus in hypoxia-exposed rats was decreased in TOF administered rats. Taken together, TOF extract ameliorates hypobaric hypoxia induced memory impairment and neurodegeneration in hippocampus through its anti-stress effects.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Cyperus/química , Flavonoides/uso terapêutico , Hipóxia/complicações , Extratos Vegetais/uso terapêutico , Acetilcolinesterase/metabolismo , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glutationa/metabolismo , L-Lactato Desidrogenase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Rememoração Mental/efeitos dos fármacos , Neurotransmissores/metabolismo , Nitritos/metabolismo , Carbonilação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
19.
J Hazard Mater ; 327: 180-186, 2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28064146

RESUMO

Zinc oxide nanoparticles (ZnO NPs) are commonly used nanomaterials (NMs) with versatile applications from high-end technologies to household products. This pervasive utilisation has brought human in the close interface with nanoparticles (NPs), hence questioning their safety prior to usage is a must. In this study, we have assessed the effects of chronic exposure to ZnO NPs (<50nm) on the model organism Drosophila melanogaster. Potential toxic effects were studied by evaluating longevity, climbing ability, oxidative stress and DNA fragmentation. Ensuing exposure, the F0 (parent), F1, F2, F3 and F4 generation flies were screened for the aberrant phenotype. Flies exposed to ZnO NPs showed distinctive phenotypic changes, like deformed segmented thorax and single or deformed wing, which were transmitted to the offspring's in subsequent generations. The unique abnormal phenotype is evident of chronic toxicity induced by ZnO NPs, although appalling, it strongly emphasize the importance to understand NPs toxicity for safer use.


Assuntos
Drosophila melanogaster , Nanopartículas Metálicas/toxicidade , Mutagênicos/toxicidade , Óxido de Zinco/toxicidade , Anormalidades Induzidas por Medicamentos/patologia , Animais , Dano ao DNA , Fragmentação do DNA , Hemócitos/efeitos dos fármacos , Longevidade , Atividade Motora/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Medição de Risco
20.
EBioMedicine ; 6: 171-189, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27211559

RESUMO

Hypobaric Hypoxia (HH) is an established risk factor for various neuro-physiological perturbations including cognitive impairment. The origin and mechanistic basis of such responses however remain elusive. We here combined systems level analysis with classical neuro-physiological approaches, in a rat model system, to understand pathological responses of brain to HH. Unbiased 'statistical co-expression networks' generated utilizing temporal, differential transcriptome signatures of hippocampus-centrally involved in regulating cognition-implicated perturbation of Glio-Vascular homeostasis during early responses to HH, with concurrent modulation of vasomodulatory, hemostatic and proteolytic processes. Further, multiple lines of experimental evidence from ultra-structural, immuno-histological, substrate-zymography and barrier function studies unambiguously supported this proposition. Interestingly, we show a significant lowering of H2S levels in the brain, under chronic HH conditions. This phenomenon functionally impacted hypoxia-induced modulation of cerebral blood flow (hypoxic autoregulation) besides perturbing the strength of functional hyperemia responses. The augmentation of H2S levels, during HH conditions, remarkably preserved Glio-Vascular homeostasis and key neuro-physiological functions (cerebral blood flow, functional hyperemia and spatial memory) besides curtailing HH-induced neuronal apoptosis in hippocampus. Our data thus revealed causal role of H2S during HH-induced early Glio-Vascular dysfunction and consequent cognitive impairment.


Assuntos
Doença da Altitude/genética , Disfunção Cognitiva/tratamento farmacológico , Perfilação da Expressão Gênica/métodos , Hipocampo/irrigação sanguínea , Sulfeto de Hidrogênio/administração & dosagem , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Doença da Altitude/complicações , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Hipocampo/química , Hipocampo/efeitos dos fármacos , Humanos , Sulfeto de Hidrogênio/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...