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










Database
Language
Publication year range
1.
J Ayurveda Integr Med ; 14(1): 100539, 2023.
Article in English | MEDLINE | ID: mdl-35078695

ABSTRACT

STUDY BACKGROUND: In a pluralistic health care delivery model, it is important to assess whether the individual's health care choices are based upon evidences of efficacy and safety. Since the essence of medical pluralism lies in the fact that all such systems are equally accessible to a seeker, in such situation, it is highly relevant to check what defines such choices in real life. OBJECTIVE: To identify the factors influencing the health care choices in a subpopulation seeking Ayurveda health care in an Ayurvedic teaching hospital. MATERIALS AND METHOD: The study was an all-inclusive cross sectional survey, done on randomly selected out patients visiting an Ayurveda teaching hospital. The data was collected using a 21 items questionnaire refined through pilot testing from 7.9.2017 to 30.9.2017. RESULTS: The data of 289 respondents who have given their consent were included in statistical analysis. Out of 21 variables studied for their agreement or disagreement in the study population 8 were found to have a significant proportion in favour of agreement. Among these relative safety (Item 9); disease eradicating potential (Item 14); belief (Item 3) and indirect evidences of efficacy (Item 4) were found to have high significance (p < 0.001). CONCLUSION: Participants chose Ayurveda treatment due to its perceived safety and probability of helping in a particular clinical condition. Contrary to the common perception, enabling factors like availability, accessibility and affordability were given less importance by the participants in making health care choices related to Ayurveda.

2.
Eur J Pharmacol ; 891: 173685, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33127363

ABSTRACT

α7 nicotinic acetylcholine receptor (α7 nAChR) is an extensively validated target for several neurological and psychiatric conditions namely, dementia and schizophrenia, owing to its vital roles in cognition and sensorimotor gating. Positive allosteric modulation (PAM) of α7 nAChR represents an innovative approach to amplify endogenous cholinergic signaling in a temporally restricted manner in learning and memory centers of brain. α7 nAChR PAMs are anticipated to side-step burgeoning issues observed with several clinical-stage orthosteric α7 nAChR agonists, related to selectivity, tolerance/tachyphylaxis, thus providing a novel dimension in therapeutic strategy and pharmacology of α7 nAChR ion-channel. Here we describe a novel α7 nAChR PAM, LL-00066471, which potently amplified agonist-induced Ca2+ fluxes in neuronal IMR-32 neuroblastoma cells in a α-bungarotoxin (α-BTX) sensitive manner. LL-00066471 showed excellent oral bioavailability across species (mouse, rat and dog), low clearance and good brain penetration (B/P ratio > 1). In vivo, LL-00066471 robustly attenuated cognitive deficits in both procognitive and antiamnesic paradigms of short-term episodic and recognition memory in novel object recognition task (NORT) and social recognition task (SRT), respectively. Additionally, LL-00066471 mitigated apomorphine-induced sensorimotor gating deficits in acoustic startle reflex (ASR) and enhanced antipsychotic efficacy of olanzapine in conditioned avoidance response (CAR) task. Further, LL-00066471 corrected redox-imbalances and reduced cortico-striatal infarcts in stroke model. These finding together suggest that LL-00066471 has potential to symptomatically alleviate cognitive deficits associated with dementias, attenuate sensorimotor gating deficits in schizophrenia and correct redox-imbalances in cerebrovascular disorders. Therefore, LL-00066471 presents potential for management of cognitive impairments associated with neurological and psychiatric conditions.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cholinergic Agents/pharmacology , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Gait Disorders, Neurologic/prevention & control , Sensory Gating/drug effects , alpha7 Nicotinic Acetylcholine Receptor/drug effects , Animals , Brain/metabolism , Brain/physiopathology , Cell Line, Tumor , Cholinergic Agents/pharmacokinetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Dogs , Exploratory Behavior/drug effects , Gait Disorders, Neurologic/metabolism , Gait Disorders, Neurologic/physiopathology , Gait Disorders, Neurologic/psychology , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Ischemic Stroke/physiopathology , Male , Mice, Inbred BALB C , Open Field Test/drug effects , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Rats, Wistar , Reflex, Startle/drug effects , Signal Transduction , Social Behavior , alpha7 Nicotinic Acetylcholine Receptor/metabolism
3.
BMC Neurosci ; 16: 48, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26232001

ABSTRACT

BACKGROUND: Inflammation following intracerebral hemorrhage (ICH) significantly contributes to secondary brain damage and poor outcomes. Prostaglandin E2 (PGE2) is known to modulate neuroinflammatory responses and is upregulated in response to brain injury as a result of changes in inducible cyclooxygenase 2 (COX-2) and the membrane-bound type of PGE synthase. Inhibition of COX-2 activity has been reported to attenuate ICH-induced brain injury; however, the clinical utility of such drugs is limited due to the potential for severe side effects. Therefore, it is now important to search for downstream targets capable of preferentially modulating PGE2 signaling, and the four E prostanoid receptors, EP1-4, which are the main targets of PGE2, remain a viable therapeutic option. We have previously shown that EP1 receptor deletion aggravates ICH-induced brain injury and impairs functional recovery, thus the current study aimed to elaborate on these results by including a pharmacologic approach targeting the EP1 receptor. RESULTS: Chronic post-treatment with the selective EP1 receptor antagonist, SC-51089, increased lesion volume by 30.1 ± 14.5% (p < 0.05) and treatment with the EP1 agonist, 17-pt-PGE2, improved neuromuscular functional recovery on grip strength (p < 0.01) and hanging wire (p < 0.05) behavioral testing. To begin identifying the mechanisms involved in EP1-mediated neuroprotection after ICH, histology was performed to assess ferric iron content, neuroinflammation, leukocyte transendothelial migratory potential, and peripheral neutrophil and immunoglobulin infiltration. Following ICH, mice treated with the antagonist displayed increased ferric iron (p < 0.05) and cortical microgliosis (p < 0.05), whereas treatment with the agonist decreased cortical (p < 0.01) and striatal (p < 0.001) astrogliosis, leukocyte transendothelial migratory potential (p < 0.01), neutrophil infiltration (p < 0.05), and blood brain barrier breakdown (p < 0.05). CONCLUSIONS: In agreement with our previous results, selective antagonism of the EP1 receptor aggravated ICH-induced brain injury. Furthermore, EP1 receptor agonism improved anatomical outcomes and functional recovery. Thus, the present data continues to reinforce a putative role for EP1 as a new and more selective therapeutic target for the treatment of ICH that could reduce the side effects associated with COX-2 inhibition while still exploiting the beneficial effects.


Subject(s)
Brain/drug effects , Cerebral Hemorrhage/drug therapy , Receptors, Prostaglandin E, EP1 Subtype/agonists , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/pathology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Collagenases , Disease Models, Animal , Gliosis/drug therapy , Gliosis/immunology , Gliosis/pathology , Hydrazines/pharmacology , Iron/metabolism , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/pathology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Neuroprotective Agents/pharmacology , Oxazepines/pharmacology , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Recovery of Function/drug effects
4.
Int J Stroke ; 9(5): 560-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23013350

ABSTRACT

BACKGROUND: Cerebral ischaemia results in enhanced expression of type 1 angiotensin receptor and oxidative stress. Free radicals due to oxidative stress lead to excessive DNA damage causing overactivation of poly (ADP-ribose) polymerase-1 resulting in neuronal death. Activation of both type 1 angiotensin receptors and poly (ADP-ribose) polymerase-1 following cerebral ischaemia takes place simultaneously, but until now, no study has explored the effect of combined blockade of both angiotensin type 1 angiotensin receptor and poly (ADP-ribose) polymerase-1 in cerebral ischaemia. AIM: Our purpose was to compare the effect of single and combined treatment with angiotensin type 1 angiotensin receptor blocker, candesartan, and the poly (ADP-ribose) polymerase-1 inhibitor, 1, 5 isoquinolinediol, on brain damage and oxidative stress in transient focal cerebral ischaemia in rats. METHOD: Transient focal cerebral ischaemia was induced in Sprague-Dawley rats by an intraluminal technique for two-hours following 48 h of reperfusion. Candesartan (0·05 mg/kg) was administered just after initiation of ischaemia followed by a repeat administration at 24 h while 1, 5 isoquinolinediol (0·1 mg/kg) was given one-hour after of ischaemia. After 24 h of reperfusion, neurological deficit was evaluated in the different treatment groups. After 48 h of reperfusion, the rats were sacrificed and the brain was isolated. Ischaemic brain damage by 2,3,5 triphenyl tetrazolium chloride staining, oxidative stress markers, and levels of reactive oxygen species were determined biochemically. RESULT: Single treatment with candesartan and 1, 5 isoquinolinediol significantly reduced neurological deficit, infarct, and oedema volume as compared to ischaemic control and different vehicle groups for each of the drugs. However, treatment with candesartan + 1, 5 isoquinolinediol offered greater reduction in neurological deficit, cerebral infarct volume, and oedema as compared to single-drug treatments. Furthermore, treatment with candesartan + 1, 5 isoquinolinediol significantly decreased oxidative stress as compared to single treatments with each drug. CONCLUSION: The study suggests that blockade of either type 1 angiotensin receptor or poly (ADP-ribose) polymerase-1 alone provides neuroprotection, but the better result was achieved when both type 1 angiotensin receptor and poly (ADP-ribose) polymerase-1 were blocked together by the combined use of their pharmacological inhibitor in transient cerebral ischaemia in rat.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Benzimidazoles/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Isoquinolines/administration & dosage , Neuroprotective Agents/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors , Tetrazoles/administration & dosage , Animals , Biphenyl Compounds , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Edema/drug therapy , Brain Edema/pathology , Brain Edema/physiopathology , Coloring Agents , Drug Therapy, Combination , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Neuropsychological Tests , Oxidative Stress/drug effects , Oxidative Stress/physiology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tetrazolium Salts
5.
Eur J Neurosci ; 38(11): 3659-68, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24112193

ABSTRACT

Consumption of flavan-3-ols, notably (-)-epicatechin (EC), has been highly recommended in complementary and alternative medicine (CAM) due to reports that flavan-3-ols boost antioxidant activity, support vascular function, and prevent cardiovascular disease. To date, in vivo efficacy and mechanisms of action for many CAM therapies, including EC, remain elusive in brain ischemia. In contrast to its purported direct antioxidant role, we hypothesized protection through activation of the endogenous transcriptional factor Nrf2. To screen cellular protection and investigate Nrf2 activation, we adopted a pretreatment paradigm using enriched primary neuronal cultures from mice and washed out EC prior to oxygen glucose deprivation to attenuate direct antioxidant effects. EC protected primary neurons from oxygen glucose deprivation by increasing neuronal viability (40.2 ± 14.1%) and reducing protein oxidation, effects that occurred concomitantly with increased Nrf2-responsive antioxidant protein expression. We also utilized wildtype and Nrf2 C57BL/6 knockout mice in a permanent model of focal brain ischemia to evaluate glial cell regulation and complex sensorimotor functioning. EC-treated wildtype mice displayed a reduction or absence of forelimb motor coordination impairments that were evident in vehicle-treated mice. This protection was associated with reduced anatomical injury (54.5 ± 8.3%) and microglia/macrophage activation/recruitment (56.4 ± 13.0%). The protective effects elicited by EC in both model systems were abolished in tissues and neuronal cultures from Nrf2 knockout mice. Together, these data demonstrate EC protection through Nrf2 and extend the benefits to improved performance on a complex sensorimotor task, highlighting the potential of flavan-3-ols in CAM approaches in minimizing subsequent stroke injury.


Subject(s)
Catechin/therapeutic use , Infarction, Middle Cerebral Artery/prevention & control , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Administration, Oral , Animals , Catechin/administration & dosage , Catechin/pharmacology , Cell Survival , Cells, Cultured , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL , Movement , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Neurons/physiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Oxidative Stress
6.
Neurotox Res ; 24(4): 549-59, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23824501

ABSTRACT

Prostaglandin E2 (PGE2) has been described to exert beneficial and detrimental effects in various neurologic disorders. These conflicting roles of PGE2 could be attributed to its diverse receptor subtypes, EP1-EP4. At present, the precise role of EP1 in intracerebral hemorrhage (ICH) is unknown. Therefore, to elucidate its possible role in ICH, intrastriatal injection of collagenase was given in randomized groups of adult male wildtype (WT) and EP1 receptor knockout (EP1⁻/⁻)C57BL/6 mice. Functional outcomes including neurologic deficits, rotarod performance, open field activity, and adhesive removal performance were evaluated at 24, 48, and 72 h post-ICH. Lesion volume, cell survival and death, were assessed using Cresyl Violet, and Fluoro-Jade staining, respectively. Microglial activation and phagocytosis were estimated using Iba1 immunoreactivity and fluorescently-labeled microspheres. Following 72 h post-ICH, EP1⁻/⁻ mice showed deteriorated outcomes compared to the WT control mice. These outcomes were demonstrated by elevated neurological deficits, exacerbated lesion volume, and significantly worsened sensorimotor functions. Fluoro-Jade staining showed significantly increased numbers of degenerating neurons and reduced neuronal survival in EP1⁻/⁻ compared to WT mice. To assess in vivo phagocytosis, the number of microspheres phagocytosed by Iba1-positive cells was 145.4 ± 15.4 % greater in WT compared to EP1⁻/⁻ mice. These data demonstrate that EP1 deletion exacerbates neuro-behavioral impairments following ICH, potentially by slowing down/impairing microglial phagocytosis. A better understanding of this EP1 mechanism could lead to improved intervention strategies for hemorrhagic stroke.


Subject(s)
Brain Injuries/physiopathology , Cerebral Hemorrhage/physiopathology , Microglia/metabolism , Receptors, Prostaglandin E, EP1 Subtype/physiology , Animals , Brain Injuries/etiology , Cell Death , Cerebral Hemorrhage/complications , Corpus Striatum/metabolism , Corpus Striatum/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, Prostaglandin E, EP1 Subtype/deficiency , Receptors, Prostaglandin E, EP1 Subtype/genetics
7.
ACS Chem Neurosci ; 4(8): 1151-62, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23758534

ABSTRACT

Resveratrol, a natural stilbene present at relatively high concentrations in grape skin and seeds and red wine, is known for its purported antioxidant activity in the vascular and nervous systems. In contrast to its direct antioxidant role within the central nervous system, recent research supports a protective mechanism through increasing endogenous cellular antioxidant defenses, which triggers a cascade of parallel neuroprotective pathways. A growing body of in vitro and in vivo evidence indicates that resveratrol acts through multiple pathways and reduces ischemic damage in vital organs, such as the heart and the brain, in various rodent models. Most of the protective biological actions of resveratrol have been associated with its antioxidative, anti-inflammatory, and antiapoptotic properties and other indirect pathways. Continued public interest and increasing resveratrol supplements on the market warrant a review of the available in vitro and in vivo science reported in the stroke-related literature. Rigorous clinical trials evaluating the effects of resveratrol in stroke are absent, though the general population consumption appears to be relatively safe. Resveratrol has shown potential for treating stroke in laboratory animals and in vitro human cell studies, yet there is still a need for human research in preclinical settings. This review summarizes many of the findings on the neuroprotective potential of resveratrol in cerebral stroke, focusing on both the in vitro and in vivo experimental models and some proposed mechanisms of action.


Subject(s)
Brain/drug effects , Neuroprotective Agents/metabolism , Stilbenes/metabolism , Stroke/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Apoptosis/drug effects , Biological Availability , Disease Models, Animal , Drug Evaluation, Preclinical , Gerbillinae , Humans , Mice , Neuroprotective Agents/pharmacokinetics , Oxidative Stress/drug effects , Rats , Resveratrol , Stilbenes/pharmacokinetics , Stroke/physiopathology
8.
Neurochem Int ; 57(8): 876-83, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20868715

ABSTRACT

Inflammation plays an important role in ischemic pathology. NF-κB is a transcription factor that has a crucial role in inflammation and cell survival, but its precise role in the ischemic aftermath is still uncertain. Therefore, we evaluated the effect of intracerebroventricular administration of a highly specific NF-κB inhibitor peptide, IKK-NBD, on transient focal cerebral ischemic injury in rat using middle cerebral artery occlusion model. The assessment of ischemia-induced neurological deficits, alterations in the proinflammatory cytokine IL-1ß level, OX-42 immunoreactivity, changes in blood-brain barrier (BBB) permeability, reactive oxygen species (ROS) production and DNA fragmentation by terminal dUTP nick end labelling (TUNEL) were monitored at 24h post reperfusion following 1h of ischemia after pre-treatment with either 40µg of IKK-NBD or the inactive IKK-NBD peptide, which served as control. Pre-treatment with IKK-NBD peptide significantly ameliorated the cerebral ischemia-induced neurological deficits. Quantification of IL-1ß by ELISA revealed significantly reduced striatal IL-1ß level in IKK-NBD peptide treated rats. The treatment also resulted in reduced staining of microglial OX-42 and significantly reduced extravasation of Evans blue dye, indicating protection of BBB from ischemic insult. These results indicate that specific NF-κB inhibition downplays post-ischemic inflammation. Furthermore, reduction in DNA fragmentation as assessed by TUNEL staining also confirms the neuroprotective effect of IKK-NBD peptide. Thus, it may be inferred that IKK-NBD peptide reduces ischemic brain damage and this can, at least partly, be attributed to reduction in inflammation following ischemic injury.


Subject(s)
Carrier Proteins/physiology , I-kappa B Kinase/physiology , Inflammation Mediators/physiology , Ischemic Attack, Transient/prevention & control , NF-kappa B/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Peptide Fragments/physiology , Peptides/physiology , Reperfusion Injury/prevention & control , Animals , Carrier Proteins/therapeutic use , Cell Death/physiology , I-kappa B Kinase/therapeutic use , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Inflammation Mediators/therapeutic use , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , NF-kappa B/physiology , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Peptides/therapeutic use , RNA-Binding Proteins , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
9.
Behav Brain Res ; 199(2): 235-40, 2009 May 16.
Article in English | MEDLINE | ID: mdl-19103228

ABSTRACT

The Renin-angiotensin system, besides blood pressure regulation, affects learning and memory as evidenced by improvement of cognition in hypertensive patients being treated with AT1 receptor blockers like candesartan. The present study examined the influence of candesartan on memory impairment induced by intracerebral streptozotocin (IC STZ 0.5 mg/kg) in mice. Candesartan (0.05 mg/kg and 0.1 mg/kg, i.p.) was given for 14 days following IC STZ administration. The dose of 0.1 mg/kg significantly improved latency period in passive avoidance test. Further, treatment with 0.1 mg/kg candesartan for 14 days significantly improved spatial memory in mice in water maze test also. In another group, after memory impairment in mice following IC STZ administration, memory improving effect of a 7 days treatment with 0.1 mg/kg candesartan lasted only for 3 subsequent days in water maze task. IC STZ increased oxidative stress but pretreatment with 0.1 mg/kg candesartan decreased oxidative stress as indicated by a decrease in MDA and increase in GSH. Further, candesartan decreased free radicals as evidenced by flow cytometry. IC STZ affected cholinergic system also by increasing acetylcholine esterase activity that was restored by pretreatment with 0.1 mg/kg candesartan. Locomotor activity and serum glucose level remained unaffected by candesartan treatment. These results suggest that AT1 receptors play a facilitatory role in STZ induced memory deficit and corroborate number of human studies that AT1 receptor blockers can be used therapeutically against cognitive decline in hypertensive patients.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Memory Disorders/drug therapy , Receptor, Angiotensin, Type 1/physiology , Tetrazoles/pharmacology , Acetylcholinesterase/metabolism , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Benzimidazoles/therapeutic use , Biphenyl Compounds , Blood Glucose , Brain/drug effects , Brain/metabolism , Malondialdehyde/metabolism , Memory Disorders/chemically induced , Mice , Motor Activity/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/drug effects , Streptozocin/administration & dosage , Streptozocin/pharmacology , Tetrazoles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...