Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Prog Brain Res ; 265: 249-315, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34560923

RESUMEN

Military personnel often exposed to high summer heat are vulnerable to heat stroke (HS) resulting in abnormal brain function and mental anomalies. There are reasons to believe that leakage of the blood-brain barrier (BBB) due to hyperthermia and development of brain edema could result in brain pathology. Thus, exploration of suitable therapeutic strategies is needed to induce neuroprotection in HS. Extracts of Gingko Biloba (EGb-761) is traditionally used in a variety of mental disorders in Chinese traditional medicine since ages. In this chapter, effects of TiO2 nanowired EGb-761 and BN-52021 delivery to treat brain pathologies in HS is discussed based on our own investigations. We observed that TiO2 nanowired delivery of EGb-761 or TiO2 BN-52021 is able to attenuate more that 80% reduction in the brain pathology in HS as compared to conventional drug delivery. The functional outcome after HS is also significantly improved by nanowired delivery of EGb-761 and BN-52021. These observations are the first to suggest that nanowired delivery of EGb-761 and BN-52021 has superior therapeutic effects in HS not reported earlier. The clinical significance in relation to the military medicine is discussed.


Asunto(s)
Bilobálidos , Golpe de Calor , Fármacos Neuroprotectores , China , Ginkgo biloba , Ginkgólidos , Humanos , Lactonas , Fármacos Neuroprotectores/farmacología , Extractos Vegetales
2.
Altern Ther Health Med ; 27(6): 58-66, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34144529

RESUMEN

BACKGROUND: Withania somnifera (Ashwagandha) is an herb with anti-inflammatory properties used in managing arthritis. There is significant clinical data in the public domain on the effects of Ashwagandha and this study was aimed at compiling and analysing these data in a structured manner. The major sources of evidence data were clinical trials and systematic review of extant literature. METHODOLOGY: Retrospective database search was conducted in the Clinical Trial Registry of India for trials registered from April 2008 to March 2020, and published literature related to the anti-arthritic effects of Withania somnifera were reviewed. RESULTS: In all, 77 registered clinical trials were analysed and common among them were interventional, single-centre, randomized, double-blind, two-arm studies with Placebo being the comparator. Similar findings were observed in the 10 published clinical trials on arthritis evaluated for this study. While industry- and government-sponsored trials were identified, government funded sites with approvals from Institutional Ethics Committees were preferred. Most trials were registered as Phase 2 with the highest number of sites in the state of Maharashtra. The solid dosage form was most preferred. CONCLUSION: While the effects of Withania somnifera on various disorders are being investigated by several clinical trials, the ones evaluated for this study provide insight on its potential in managing arthritis when given for a specific duration. Evidence shows a dosage of 6 gm in powder form or extracts in tablets, or 500 -1000 mg capsule consumed for a duration of 8 - 12 weeks may be useful in managing symptoms of arthritis in patients.


Asunto(s)
Withania , Humanos , India , Extractos Vegetales/uso terapéutico , Sistema de Registros , Estudios Retrospectivos
3.
Prog Brain Res ; 258: 1-77, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33223033

RESUMEN

Sleep deprivation (SD) is common in military personnel engaged in combat operations leading to brain dysfunction. Military personnel during acute or chronic SD often prone to traumatic brain injury (TBI) indicating the possibility of further exacerbating brain pathology. Several lines of evidence suggest that in both TBI and SD alpha-melanocyte-stimulating hormone (α-MSH) and brain-derived neurotrophic factor (BDNF) levels decreases in plasma and brain. Thus, a possibility exists that exogenous supplement of α-MSH and/or BDNF induces neuroprotection in SD compounded with TBI. In addition, mesenchymal stem cells (MSCs) are very portent in inducing neuroprotection in TBI. We examined the effects of concussive head injury (CHI) in SD on brain pathology. Furthermore, possible neuroprotective effects of α-MSH, MSCs and neurotrophic factors treatment were explored in a rat model of SD and CHI. Rats subjected to 48h SD with CHI exhibited higher leakage of BBB to Evans blue and radioiodine compared to identical SD or CHI alone. Brain pathology was also exacerbated in SD with CHI group as compared to SD or CHI alone together with a significant reduction in α-MSH and BDNF levels in plasma and brain and enhanced level of tumor necrosis factor-alpha (TNF-α). Exogenous administration of α-MSH (250µg/kg) together with MSCs (1×106) and cerebrolysin (a balanced composition of several neurotrophic factors and active peptide fragments) (5mL/kg) significantly induced neuroprotection in SD with CHI. Interestingly, TiO2 nanowired delivery of α-MSH (100µg), MSCs, and cerebrolysin (2.5mL/kg) induced enhanced neuroprotection with higher levels of α-MSH and BDNF and decreased the TNF-α in SD with CHI. These observations are the first to show that TiO2 nanowired administration of α-MSH, MSCs and cerebrolysin induces superior neuroprotection following SD in CHI, not reported earlier. The clinical significance of our findings in light of the current literature is discussed.


Asunto(s)
Traumatismos Craneocerebrales , Células Madre Mesenquimatosas , Aminoácidos , Animales , Radioisótopos de Yodo , Neuroprotección , Ratas , Privación de Sueño , Titanio , alfa-MSH
4.
Front Pharmacol ; 9: 757, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30127737

RESUMEN

Oxidative stress and neuroinflammation play a key role in dopaminergic (DA) neuronal degeneration, which results in the hindrance of normal ongoing biological processes in the case of Parkinson's disease. As shown in several studies, on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration, different behavioral parameters have suggested motor impairment and damage of antioxidant defence. Thus, some specific biological molecules found in medicinal plants can be used to inhibit the DA neuronal degeneration through their antioxidant and anti-inflammatory activities. With this objective, we studied chlorogenic acid (CGA), a naturally occurring polyphenolic compound, for its antioxidant and anti-inflammatory properties in MPTP-intoxicated mice. We observed significant reoccurrence of motor coordination and antioxidant defence on CGA supplementation, which has been in contrast with MPTP-injected mice. Moreover, in the case of CGA-treated mice, the enhanced expression of tyrosine hydroxylase (TH) within the nigrostriatal region has supported its beneficial effect. The activation of glial cells and oxidative stress levels were also estimated using inducible nitric oxide synthase (iNOS) and glial fibrillary acidic protein (GFAP) immunoreactivity within substantia nigra (SN) and striatum of MPTP-injected mice. Administration of CGA has prevented the neuroinflammation in SN by regulating the nuclear factor-κB expression in the MPTP-induced group. The significant release of certain pro-inflammatory mediators such as tumor necrosis factor-α and interleukin (IL)-1ß has also been inhibited by CGA with the enhanced expression of anti-inflammatory cytokine IL-10. Moreover, reduced GFAP staining within the nigrostriatal region has supported the fact that CGA has significantly helped in the attenuation of astrocyte activation. Hence, our study has shown that CGA supplementation shows its therapeutic ability by reducing the oxidative stress and neuroinflammation in MPTP-intoxicated mice.

5.
Interdiscip Sci ; 10(4): 722-733, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28488219

RESUMEN

A stroke or cerebrovascular accident is a serious, life-threatening medical condition that occurs when the blood supply to part of the brain is severely reduced or cut off, depriving brain tissue of oxygen and nutrients. Studies suggested that level of gelatinases (MMP-2 and MMP-9) usually increases in the brain after stroke. The elevated activity of gelatinases plays the deleterious role in ischemic stroke, hemorrhagic stroke and perinatal hypoxic-ischemic brain injury. Therefore, matrix metalloproteinase (MMP)-2 and MMP-9 inhibition have therapeutic importance in stroke condition. Present in silico study investigates whether Withania somnifera (WS) phytochemicals inhibit the MMP-2 and MMP-9 by binding to the catalytic domain, as similar to their inhibitor or not. For that, we performed molecular docking study to evaluate the gelatinases-inhibitory potential of 36 WS phytochemicals, which compared with gelatinases inhibitors viz. hydroxamic acid, quercetin, doxycycline, minocycline and reverse hydroxamate. The results suggest that 28 out of 36 WS phytochemicals show higher affinity for MMP-2 owing to bind with active site residues of S1'-pocket with lower binding energy and smaller inhibition constant (Ki) than considered inhibitors. As well as, withanolide G and withafastuosin E show higher affinity for MMP-9 than reverse hydroxamate inhibitor. These phytochemicals have neuroprotective potential as an inherently useful oral drug to combat ischemic and hemorrhagic stroke mediated by gelatinases.


Asunto(s)
Simulación por Computador , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Neuroprotección/efectos de los fármacos , Fitoquímicos/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Withania/química , Dominio Catalítico , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 9 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Simulación del Acoplamiento Molecular , Fitoquímicos/uso terapéutico , Accidente Cerebrovascular/enzimología
6.
Int Rev Neurobiol ; 137: 123-165, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29132541

RESUMEN

More than 5.5 million Americans of all ages are suffering from Alzheimer's disease (AD) till today for which no suitable therapy has been developed so far. Thus, there is an urgent need to explore novel therapeutic measures to contain brain pathology in AD. The hallmark of AD includes amyloid-beta peptide (AßP) deposition and phosphorylation of tau in AD brain. Recent evidences also suggest a marked decrease in neurotrophic factors in AD. Thus, exogenous supplement of neurotrophic factors could be one of the possible ways for AD therapy. Human postmortem brain in AD shows alterations in histamine receptors as well, indicating an involvement of the amine in AD-induced brain pathology. In this review, we focused on role of histamine 3 and 4 receptor-modulating drugs in the pathophysiology of AD. Moreover, antibodies to histamine and tau appear to be also beneficial in reducing brain pathology, blood-brain barrier breakdown, and edema formation in AD. Interestingly, TiO2-nanowired delivery of cerebrolysin-a balanced composition of several neurotrophic factors attenuated AßP deposition and reduced tau phosphorylation in AD brain leading to neuroprotection. Coadministration of cerebrolysin with histamine antibodies or tau antibodies has further enhanced neuroprotection in AD. These novel observations strongly suggest a role of nanomedicine in AD that requires further investigation.


Asunto(s)
Enfermedad de Alzheimer/terapia , Aminoácidos/administración & dosificación , Anticuerpos/administración & dosificación , Histamínicos/administración & dosificación , Titanio , Proteínas tau/inmunología , Animales , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Humanos
7.
Mol Neurobiol ; 52(2): 882-98, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26126513

RESUMEN

Increased levels of ubiquitin and heat shock protein (HSP) 72 kD are often seen in spinal cord injury (SCI). However, their roles in cell injury or survival are not well known. Thus, we have investigated the possible relationship between ubiquitin and HSP expressions in relation to cell injury in healthy animals, or following nanoparticle (NP) intoxication in SCI animals. A focal SCI was inflicted on the T10-11 segments over the right dorsal horn; animals were allowed to survive from 5 to 8 h after trauma. Separate groups of rats were exposed to SiO2, Ag, or Cu NPs for 7 days and subjected to SCI on the eighth day. A marked increase in ubiquitin or HSP immunoreactive cells occurred in the T9 to T12 segments 5 h after the injury, which further extended to the T4 and L5 after 8 h of survival. At this time, a marked increase in blood-spinal cord barrier (BSCB) permeability to Evans blue and radioiodine, accompanied by an intense edema formation, was observed. Changes were further exacerbated in NP-treated traumatized rats. The most marked expressions of ubiquitin and HSP in SCI were seen in rats treated with SiO2, followed by Ag, and Cu NPs. Treatment with H-290/51 (50 mg/kg p.o., 30 to 60 min after injury) or carfilzomib (1 mg/kg, i.v., 30 to 60 min after trauma) significantly reduced the ubiquitin or HSP expressions, as well as the BSCB breakdown, the edema formation, and the cell injury in the cord both 5 and 8 h after the injury, in normal animals. However, a double dose of H-290/51 (100 mg/kg) or carfilzomib (2 mg/kg) is needed to reduce cord pathology or ubiquitin and HSP expressions in traumatized animals treated with NPs. H-290/51 showed superior beneficial effects in reducing cord pathology in SCI than carfilzomib. These observations are the first to demonstrate that (i) NP-treated traumatized animals induce a widespread BSCB leakage, edema formation, and cord pathology as well as an overexpression of ubiquitin and HSP, (ii) high doses of antioxidant compounds or proteasome inhibitors are required for neuroprotection in the NP-exposed traumatized group, and (iii) ubiquitin and HSP expressions play a key role in neuronal injury in SCI, not reported earlier.


Asunto(s)
Antioxidantes/uso terapéutico , Cobre/toxicidad , Proteínas del Choque Térmico HSP72/biosíntesis , Indoles/uso terapéutico , Nanopartículas/toxicidad , Proteínas del Tejido Nervioso/biosíntesis , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Inhibidores de Proteasoma/uso terapéutico , Plata/toxicidad , Traumatismos de la Médula Espinal/tratamiento farmacológico , Ubiquitina/biosíntesis , Animales , Antioxidantes/farmacología , Cobre/administración & dosificación , Evaluación Preclínica de Medicamentos , Edema/etiología , Edema/prevención & control , Proteínas del Choque Térmico HSP72/genética , Indoles/farmacología , Masculino , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Inhibidores de Proteasoma/farmacología , Ratas , Ratas Wistar , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/toxicidad , Plata/administración & dosificación , Médula Espinal/metabolismo , Médula Espinal/patología , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/etiología , Traumatismos de la Médula Espinal/metabolismo , Vértebras Torácicas , Ubiquitina/genética , Regulación hacia Arriba
8.
Mol Neurobiol ; 52(2): 837-45, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26126514

RESUMEN

Previous studies from our laboratory showed that topical application of growth hormone (GH) induced neuroprotection 5 h after spinal cord injury (SCI) in a rat model. Since nanodelivery of drugs exerts superior neuroprotective effects, a possibility exists that nanodelivery of GH will induce long-term neuroprotection after a focal SCI. SCI induces GH deficiency that is coupled with insulin-like growth factor-1 (IGF-1) reduction in the plasma. Thus, an exogenous supplement of GH in SCI may enhance the IGF-1 levels in the cord and induce neuroprotection. In the present investigation, we delivered TiO2-nanowired growth hormone (NWGH) after a longitudinal incision of the right dorsal horn at the T10-11 segments in anesthetized rats and compared the results with normal GH therapy on IGF-1 and GH contents in the plasma and in the cord in relation to blood-spinal cord barrier (BSCB) disruption, edema formation, and neuronal injuries. Our results showed a progressive decline in IGF-1 and GH contents in the plasma and the T9 and T12 segments of the cord 12 and 24 h after SCI. Marked increase in the BSCB breakdown, as revealed by extravasation of Evans blue and radioiodine, was seen at these time points after SCI in association with edema and neuronal injuries. Administration of NWGH markedly enhanced the IGF-1 levels and GH contents in plasma and cord after SCI, whereas normal GH was unable to enhance IGF-1 or GH levels 12 or 24 h after SCI. Interestingly, NWGH was also able to reduce BSCB disruption, edema formation, and neuronal injuries after trauma. On the other hand, normal GH was ineffective on these parameters at all time points examined. Taken together, our results are the first to demonstrate that NWGH is quite effective in enhancing IGF-1 and GH levels in the cord and plasma that may be crucial in reducing pathophysiology of SCI.


Asunto(s)
Hormona del Crecimiento/uso terapéutico , Factor I del Crecimiento Similar a la Insulina/análisis , Nanocables , Fármacos Neuroprotectores/uso terapéutico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Administración Tópica , Animales , Sistemas de Liberación de Medicamentos , Implantes de Medicamentos , Edema/etiología , Edema/prevención & control , Azul de Evans/farmacocinética , Hormona del Crecimiento/administración & dosificación , Hormona del Crecimiento/análisis , Hormona del Crecimiento/farmacocinética , Bombas de Infusión , Infusión Espinal , Radioisótopos de Yodo/farmacocinética , Masculino , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacocinética , Permeabilidad , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/análisis , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Médula Espinal/irrigación sanguínea , Médula Espinal/química , Médula Espinal/patología , Traumatismos de la Médula Espinal/sangre , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/fisiopatología , Vértebras Torácicas
9.
Mol Neurobiol ; 52(2): 867-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26133300

RESUMEN

Military personnel are often subjected to sleep deprivation (SD) during combat operations. Since SD is a severe stress and alters neurochemical metabolism in the brain, a possibility exists that acute or long-term SD will influence blood-brain barrier (BBB) function and brain pathology. This hypothesis was examined in young adult rats (age 12 to 14 weeks) using an inverted flowerpot model. Rats were placed over an inverted flowerpot platform (6.5 cm diameter) in a water pool where the water levels are just 3 cm below the surface. In this model, animals can go to sleep for brief periods but cannot achieve deep sleep as they would fall into water and thus experience sleep interruption. These animals showed leakage of Evans blue in the cerebellum, hippocampus, caudate nucleus, parietal, temporal, occipital, cingulate cerebral cortices, and brain stem. The ventricular walls of the lateral and fourth ventricles were also stained blue, indicating disruption of the BBB and the blood-cerebrospinal fluid barrier (BCSFB). Breakdown of the BBB or the BCSFB fluid barrier was progressive in nature from 12 to 48 h but no apparent differences in BBB leakage were seen between 48 and 72 h of SD. Interestingly, rats treated with metal nanoparticles, e.g., Cu or Ag, showed profound exacerbation of BBB disruption by 1.5- to 4-fold, depending on the duration of SD. Measurement of plasma and brain serotonin showed a close correlation between BBB disruption and the amine level. Repeated treatment with the serotonin 5-HT3 receptor antagonist ondansetron (1 mg/kg, s.c.) 4 and 8 h after SD markedly reduced BBB disruption and brain pathology after 12 to 24 h SD but not following 48 or 72 h after SD. However, TiO2-nanowired ondansetron (1 mg/kg, s.c) in an identical manner induced neuroprotection in rats following 48 or 72 h SD. However, plasma and serotonin levels were not affected by ondansetron treatment. Taken together, our observations are the first to show that (i) SD could induce BBB disruption and brain pathology, (ii) nanoparticles exacerbate SD-induced brain damage, and (iii) serotonin 5-HT3 receptor antagonist ondansetron is neuroprotective in SD that is further potentiated byTiO2-nanowired delivery, not reported earlier.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/prevención & control , Cobre/toxicidad , Nanopartículas/toxicidad , Fármacos Neuroprotectores/farmacología , Ondansetrón/farmacología , Agonistas del Receptor de Serotonina 5-HT3/farmacología , Plata/toxicidad , Privación de Sueño/fisiopatología , Animales , Proteínas Sanguíneas/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/patología , Encéfalo/fisiopatología , Química Encefálica/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/fisiopatología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Trastornos del Conocimiento/prevención & control , Colorantes/farmacocinética , Cobre/administración & dosificación , Evaluación Preclínica de Medicamentos , Implantes de Medicamentos , Azul de Evans/farmacocinética , Fatiga/etiología , Fatiga/fisiopatología , Fatiga/prevención & control , Radioisótopos de Yodo/farmacocinética , Masculino , Nanocables , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Ondansetrón/administración & dosificación , Ondansetrón/uso terapéutico , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante , Trastornos de la Sensación/etiología , Trastornos de la Sensación/fisiopatología , Trastornos de la Sensación/prevención & control , Serotonina/análisis , Agonistas del Receptor de Serotonina 5-HT3/administración & dosificación , Agonistas del Receptor de Serotonina 5-HT3/uso terapéutico , Plata/administración & dosificación , Privación de Sueño/complicaciones , Factores de Tiempo
10.
Mol Neurobiol ; 52(2): 1043-53, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26108181

RESUMEN

The possibility that ubiquitin expression is altered in cardiac arrest-associated neuropathology was examined in a porcine model using immunohistochemical and biochemical methods. Our observations show that cardiac arrest induces progressive increase in ubiquitin expression in the cortex and hippocampus in a selective and specific manner as compared to corresponding control brains using enzyme-linked immunoassay technique (enzyme-linked immunosorbent assay (ELISA)). Furthermore, immunohistochemical studies showed ubiquitin expression in the neurons exhibiting immunoreaction in the cytoplasm and karyoplasm of distorted or damaged cells. Separate Nissl and ubiquitin staining showed damaged and distorted neurons and in the same cortical region ubiquitin expression indicating that ubiquitin expression after cardiac arrest represents dying neurons. The finding that methylene blue treatment markedly induced neuroprotection following identical cardiac arrest and reduced ubiquitin expression strengthens this view. Taken together, our observations are the first to show that cardiac arrest enhanced ubiquitin expression in the brain that is related to the magnitude of neuronal injury and the finding that methylene blue reduced ubiquitin expression points to its role in cell damage, not reported earlier.


Asunto(s)
Barrera Hematoencefálica , Corteza Cerebral/patología , Paro Cardíaco/metabolismo , Hipocampo/patología , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/metabolismo , Ubiquitina/biosíntesis , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Reanimación Cardiopulmonar , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica , Paro Cardíaco/tratamiento farmacológico , Paro Cardíaco/patología , Paro Cardíaco/terapia , Hipocampo/metabolismo , Azul de Metileno/farmacología , Azul de Metileno/uso terapéutico , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Distribución Aleatoria , Albúmina Sérica/análisis , Sus scrofa , Porcinos , Tálamo/metabolismo , Tálamo/patología , Ubiquitina/genética , Regulación hacia Arriba
11.
J Physiol Biochem ; 70(4): 901-13, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25292423

RESUMEN

Neurotransmitter imbalance is an inevitable outcome in cerebral ischemia that leads to neuronal death. In the present study, we evaluated the effects of piroxicam, a nonsteroidal anti-inflammatory drug (NSAID), on extracellular brain glutamate and γ-aminobutyric acid (GABA) release, survival time, and neuronal cell death. Transient focal cerebral ischemia in male Charles Foster rat led to neuronal infarction and compromised intrinsic antioxidant status. Thirty-minute preadministration of piroxicam (10 mg/kg b.w.) showed a significant (P < 0.01) reduction in cerebral infarct volume and potentiation of the intrinsic antioxidant status. High-performance liquid chromatography of brain cortex and striatum revealed changes in extracellular concentrations of neurotransmitters which were found to be 0.519 ± 0.44 pmole/mg (GABA); 1.18 ± 0.28 pmole/mg (glutamate), and 0.63 ± 0.21 pmole/mg (serotonin), respectively. Hydroxyl radical (·OH) adduct of salicylate in the frontal cortex and striatum in control, untreated, and treated groups was found to be 0.261 ± 0.06, 0.68 ± 0.52, and 0.401 ± 0.68 pmole/mg, respectively. After stroke, the extracellular level of glutamate in rat brain increases continuously as compared to that of control group. However, piroxicam administration in stroke rat significantly reduced (P < 0.05) elevated extracellular cerebral glutamate. This indicates that piroxicam attenuates extracellular glutamate release and also reduces neuronal cell death due to reduction in oxidative stress in cerebral ischemia. Our results also indicate a consequent increase of extracellular GABA in brain regions administered with piroxicam, which hints that piroxicam alleviates glutamate excitotoxicity possibly by GABA agonism.


Asunto(s)
Agonistas del GABA/farmacología , Ácido Glutámico/fisiología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Piroxicam/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Catalasa/metabolismo , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Evaluación Preclínica de Medicamentos , Agonistas del GABA/uso terapéutico , Infarto de la Arteria Cerebral Media/patología , Masculino , Fármacos Neuroprotectores/uso terapéutico , Piroxicam/uso terapéutico , Ratas , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Ácido gamma-Aminobutírico/metabolismo
12.
J Nanosci Nanotechnol ; 14(1): 577-95, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24730284

RESUMEN

Functionalized Magnetic Iron Oxide Nanoparticles (FMIONPs) are being explored for the development of various biomedical applications, e.g., cancer chemotherapy and/or several other radiological or diagnostic purposes. However, the effects of these NPs per se on the central nervous system (CNS) injury or repair are not well known. This review deals with different aspects of FMIONPs in relation to brain function based on the current literature as well as our own investigation in animal models of CNS injuries. It appears that FMIONPs are innocuous when administered intravenously within the CNS under normal conditions. However, abnormal reactions to FMIONPs in the brain or spinal cord could be seen if they are combined with CNS injuries e.g., hyperthermia or traumatic insults to the brain or spinal cord. Thus, administration of FMIONPs in vivo following whole body hyperthermia (WBH) or a focal spinal cord injury (SCI) exacerbates cellular damage. Since FMIONPs could help in diagnostic purposes or enhance the biological effects of radiotherapy/chemotherapy it is likely that these NPs may have some adverse reaction as well under disease condition. Thus, under such situation, adjuvant therapy e.g., Cerebrolysin (Ever NeuroPharma, Austria), a suitable combination of several neurotrophic factors and active peptide fragments are the need of the hour to contain such cellular damages caused by the FMIONPs in vivo. Our observations show that co-administration of Cerebrolysin prevents the FMIONPs induced pathologies associated with CNS injuries. These observations support the idea that FMIONPs are safe for the CNS in disease conditions when co-administered with cerebrolysin. This indicates that cerebrolysin could be used as an adjunct therapy to prevent cellular damages in disease conditions where the use of FMIONPs is required for better efficacy e.g., cancer treatment.


Asunto(s)
Aminoácidos/administración & dosificación , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/prevención & control , Nanopartículas de Magnetita/efectos adversos , Nanocápsulas/efectos adversos , Nanocápsulas/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Aminoácidos/química , Animales , Interacciones Farmacológicas , Humanos , Nanopartículas de Magnetita/uso terapéutico , Nanocápsulas/ultraestructura , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química
13.
Int Rev Neurobiol ; 102: 107-46, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22748828

RESUMEN

Oxidative stress in the central nervous system is one of the key players for neurodegeneration. Thus, antioxidants could play important roles in treating several neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and aging-related brain disorders. This review is focused on the new developments in oxidative stress-induced neurodegeneration. Further, based on our own investigations, new roles of quercetin, an antioxidant compound in hypoxia and ischemia induced neuroprotection in relation to suppression of oxidative stress, improvement in behavioral function, reduction in infarct volume, brain swelling, and cellular injury in both in vivo and in vitro models are discussed. Our new findings clearly suggest that antioxidant compounds have potential role in therapeutic strategies to treat neurodegenerative diseases in clinical settings.


Asunto(s)
Antioxidantes/farmacología , Hipoxia/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Quercetina/farmacología , Animales , Antioxidantes/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Radicales Libres/metabolismo , Hipoxia/fisiopatología , Modelos Biológicos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Estrés Oxidativo/fisiología , Quercetina/uso terapéutico , Especies Reactivas de Oxígeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA