Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Nutrients ; 15(14)2023 Jul 09.
Article in English | MEDLINE | ID: mdl-37513501

ABSTRACT

This study investigated the effect of nicotinamide (NAM) supplementation on the development of brain inflammation and microglial activation in a mouse model of type 1 diabetes mellitus. C57BL/6J male mice, which were made diabetic with five consecutive, low-dose (55 mg/kg i.p.) streptozotocin (STZ) injections. Diabetic mice were randomly distributed in different experimental groups and challenged to different doses of NAM (untreated, NAM low-dose, LD, 0.1%; NAM high-dose, HD, 0.25%) for 25 days. A control, non-diabetic group of mice was used as a reference. The NAD+ content was increased in the brains of NAM-treated mice compared with untreated diabetic mice (NAM LD: 3-fold; NAM HD: 3-fold, p-value < 0.05). Immunohistochemical staining revealed that markers of inflammation (TNFα: NAM LD: -35%; NAM HD: -46%; p-value < 0.05) and microglial activation (IBA-1: NAM LD: -29%; NAM HD: -50%; p-value < 0.05; BDKRB1: NAM LD: -36%; NAM HD: -37%; p-value < 0.05) in brains from NAM-treated diabetic mice were significantly decreased compared with non-treated T1D mice. This finding was accompanied by a concomitant alleviation of nuclear NFκB (p65) signaling in treated diabetic mice (NFκB (p65): NAM LD: -38%; NAM HD: -53%, p-value < 0.05). Notably, the acetylated form of the nuclear NFκB (p65) was significantly decreased in the brains of NAM-treated, diabetic mice (NAM LD: -48%; NAM HD: -63%, p-value < 0.05) and inversely correlated with NAD+ content (r = -0.50, p-value = 0.03), suggesting increased activity of NAD+-dependent deacetylases in the brains of treated mice. Thus, dietary NAM supplementation in diabetic T1D mice prevented brain inflammation via NAD+-dependent deacetylation mechanisms, suggesting an increased action of sirtuin signaling.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Encephalitis , Mice , Male , Animals , Niacinamide/pharmacology , NAD , Mice, Inbred C57BL , Encephalitis/prevention & control
2.
Life Sci ; 286: 119989, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34597609

ABSTRACT

AIMS: Isoformononetin (IFN), a methoxyl isoflavone present in most of human dietary supplements. However, being a highly potent antioxidant and anti-inflammatory molecule, its activity against neuronal oxidative stress and neuroinflammation has not been explored till now. The present study was inquested to assess the antioxidant, anti-apoptotic and anti-inflammatory activity of IFN against streptozotocin induced neuroinflammation in different brain regions of rat. MAIN METHODS: Four groups of animals were subjected to treatment as control, toxic control (STZ; single intracerebrovascular injection), third group (STZ + IFN; 20 mg/kg p.o.), fourth group (IFN) for 14 days. The different brain regions of rats were evaluated for inflammatory, apoptotic and biochemical antioxidant markers. The brain tissues were further assessed for gene expression, immunohistochemical and western blotting examination for localization of inflammasome cascade expression that plays a pivotal role in neuroinflammation. KEY FINDINGS: The modulation in oxidant/antioxidant status after exposure of STZ was significantly balanced after administration of IFN to rats. Further, IFN was also found to be an apoptotic agent as it modulates the apoptotic gene (Bax) and anti-apoptotic gene (BcL2) expression. IFN significantly curtailed the augmented protein expression of NLRP3, NLRP2, ASC, NFκBP65, IL-1ß and caspase-1 due to STZ administration in cortex and hippocampus rat brain regions. SIGNIFICANCE: The aforementioned results proclaim the neuroprotective functioning of IFN against STZ induced inflammation. IFN significantly prevents the neuroinflammation by decreasing the generation of ROS that reduces the activation of NLRP3/ASC/IL-1 axis thereby exerting neuroprotection as evidenced in rat model of STZ induced neuroninflammation.


Subject(s)
Antioxidants/pharmacology , CARD Signaling Adaptor Proteins/metabolism , Encephalitis/prevention & control , Interleukin-1/metabolism , Isoflavones/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Streptozocin/toxicity , Animals , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/pathology , Gene Expression/physiology , Interferons/physiology , Lipid Peroxidation/drug effects , Nitric Oxide/biosynthesis , Oxidative Stress/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
3.
Am J Respir Crit Care Med ; 204(12): 1391-1402, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34491883

ABSTRACT

Rationale: Mechanical ventilation (MV) is associated with hippocampal apoptosis and inflammation, and it is important to study strategies to mitigate them. Objectives: To explore whether temporary transvenous diaphragm neurostimulation (TTDN) in association with MV mitigates hippocampal apoptosis and inflammation after 50 hours of MV. Methods: Normal-lung porcine study comparing apoptotic index, inflammatory markers, and neurological-damage serum markers between never-ventilated subjects, subjects undergoing 50 hours of MV plus either TTDN every other breath or every breath, and subjects undergoing 50 hours of MV (MV group). MV settings in volume control were Vt of 8 ml/kg, and positive end-expiratory pressure of 5 cm H2O. Measurements and Main Results: Apoptotic indices, microglia percentages, and reactive astrocyte percentages were greater in the MV group in comparison with the other groups (P < 0.05). Transpulmonary pressure at baseline and at study end were both lower in the group receiving TTDN every breath, but lung injury scores and systemic inflammatory markers were not different between the groups. Serum concentrations of four neurological-damage markers were lower in the group receiving TTDN every breath than in the MV group (P < 0.05). Heart rate variability declined significantly in the MV group and increased significantly in both TTDN groups over the course of the experiments. Conclusions: Our study found that mechanical ventilation is associated with hippocampal apoptosis and inflammation, independent of lung injury and systemic inflammation. Also, in a porcine model, TTDN results in neuroprotection after 50 hours, and the degree of neuroprotection increases with greater exposure to TTDN.


Subject(s)
Apoptosis , Brain Injuries/prevention & control , Diaphragm/innervation , Electric Stimulation Therapy/methods , Encephalitis/prevention & control , Hippocampus/pathology , Respiration, Artificial/adverse effects , Animals , Brain Injuries/diagnosis , Brain Injuries/etiology , Brain Injuries/pathology , Encephalitis/diagnosis , Encephalitis/etiology , Encephalitis/pathology , Female , Phrenic Nerve , Respiration, Artificial/methods , Swine , Treatment Outcome
4.
Biomed Pharmacother ; 139: 111579, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33845375

ABSTRACT

Alzheimer's disease (AD) is the most common type of dementia worldwide, characterized by the deposition of neurofibrillary tangles and amyloid-ß (Aß) peptides in the brain. Additionally, increasing evidence demonstrates that a neuroinflammatory state and oxidative stress, iron-dependent, play a crucial role in the onset and disease progression. Besides conventional therapies, the use of natural-based products represents a future medical option for AD treatment and/or prevention. We, therefore, evaluated the effects of a ribonucleotides-based ingredient (Ribodiet®) in a non-genetic mouse model of AD. To this aim, mice were injected intracerebroventricularly (i.c.v.) with Aß1-42 peptide (3 µg/3 µl) and after with Ribodiet® (0.1-10 mg/mouse) orally (p.o.) 3 times weekly for 21 days following the induction of experimental AD. The mnemonic and cognitive decline was then evaluated, and, successively, we have assessed ex vivo the modulation of different cyto-chemokines on mice brain homogenates. Finally, the level of GFAP, S100ß, and iron-related metabolic proteins were monitored as markers of reactive gliosis, neuro-inflammation, and oxidative stress. Results indicate that Ribodiet® lessens oxidative stress, brain inflammation, and amyloid pathology via modulation of iron-related metabolic proteins paving the way for its rationale use for the treatment of AD and other age-related diseases.


Subject(s)
Alzheimer Disease/prevention & control , Cerebral Amyloid Angiopathy/prevention & control , Dietary Supplements , Encephalitis/prevention & control , Oxidative Stress/drug effects , Ribonucleotides/therapeutic use , Alzheimer Disease/psychology , Amyloid beta-Peptides , Animals , Behavior, Animal/drug effects , Biomarkers , Cerebral Amyloid Angiopathy/psychology , Diet , Encephalitis/psychology , Gliosis/prevention & control , Injections, Intraventricular , Male , Mice , Nonheme Iron Proteins/metabolism , Peptide Fragments , Psychomotor Performance/drug effects , Ribonucleotides/pharmacology
5.
Neurotoxicology ; 83: 89-105, 2021 03.
Article in English | MEDLINE | ID: mdl-33412218

ABSTRACT

The aggregation of Aß plays a major role in the progression of Alzheimer's disease (AD) and induces neuroinflammation, neurodegeneration and cognitive decline. Recent studies have shown that the soluble aggregates of Aß are the major culprits in the development of these aberrations inside the brain. In this study, we investigated the neuroprotective potential of carbenoxolone (Cbx), which has been found to possess anti-inflammatory and nootropic properties. Male SD rats (250-300 g) were divided into the four groups (n = 8 per group): (1) sham control rats injected with vehicles, (2) Aß 1-42 group rats injected i.c.v. with Aß 42 oligomers (10 µl/rat), (3) Aß 1-42+Cbx group rats injected i.c.v. with Aß 42 oligomers (10 µl/rat) and i.p. with carbenoxolone disodium (20 mg/kg body weight) for six-weeks and (4) Cbx group rats injected i.p. with carbenoxolone disodium (20 mg/kg body weight) for six-weeks. Progressive learning and memory deficits were seen through a battery of behavioral tests and a significant increase in the expressions of GFAP and Iba-1 was observed which resulted in the release of pro-inflammatory cytokines post Aß oligomer injection. The levels of BDNF, Bcl-2 and pCREB were decreased while Bax, caspase-3, caspase-9 and cytochrome c levels were induced. Also, neurotransmitter levels were altered and neuronal damage was observed through histopathological studies. After Cbx supplementation, the expressions of GFAP, IBA-1, pro-inflammatory cytokines, iNOS, nNOS and nitric oxide levels were normalized. The expression levels of pro-apoptotic markers were decreased and neurotrophin levels were restored. Also, neurotransmitter levels and neuronal profile were improved and progressive improvements in behavioural performance were observed. Our results demonstrated that Cbx might have prevented the Aß induced neurodegeneration and cognitive decline by inhibiting the neuroinflammation and inducing BDNF/CREB signalling. These findings suggest that Cbx can be explored as a potential therapeutic agent against the progression of AD.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Carbenoxolone/pharmacology , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Encephalitis/prevention & control , Neuroprotective Agents/pharmacology , Acetylcholinesterase/metabolism , Amyloid beta-Peptides , Animals , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/physiopathology , GPI-Linked Proteins/metabolism , Inflammation Mediators/metabolism , Male , Memory/drug effects , Monoamine Oxidase/metabolism , NF-kappa B/metabolism , Peptide Fragments , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction
6.
Neural Plast ; 2020: 8841332, 2020.
Article in English | MEDLINE | ID: mdl-33014035

ABSTRACT

Lipopolysaccharides (LPS) are proinflammation mediators that can induce the inflammatory model of the hippocampal neuron, and neuroinflammation participates in the pathophysiology of depression. Xiaoyao Pill is a classical Chinese medicine formula that has been used for the treatment of mental disorders such as depression in China since the Song dynasty. We established a hippocampal neuronal cell inflammation model by LPS and investigate the intervention effect and mechanism of Xiaoyao Pills. The expression levels of IL-6, TNF-α, IDO, 5-HT, brain-derived neurotrophic factor, and ß-nerve growth factor were detected by enzyme-linked immunosorbent assay. mRNA levels of IL-6, TNF-α, 5-HT1A, IDO-1, brain-derived neurotrophic factor, nerve growth factor, tropomyosin receptor kinase B, tropomyosin receptor kinase A, and cAMP response element-binding protein were detected by reverse transcription-polymerase chain reaction. To further validate, protein expression was determined by western blot and immunofluorescence. Lipopolysaccharide-induced neuroinflammatory state resulted in the release of IL-6, TNF-α, and IDO and a decrease of BDNF, NGF, TrkB, TrkA, CREB, p-CREB, p-CREB/CREB, and SYP and inhibited hippocampal neurogenesis in the hippocampal neuron. Xiaoyao Pills significantly decreased the levels of IL-6, TNF-α, and IDO in cell supernatant and increased the expression of BDNF, NGF, TrkB, TrkA, CREB, p-CREB, p-CREB/CREB, and SYP as well as the average optical density of BrdU/NeuN double-labelled positive cells. Our study shows that lipopolysaccharides induce inflammation and nerve damage in hippocampal neurons, which are closely related to the pathological mechanism of depression. Xiaoyao Pills (XYW) play an important neuroprotective effect, which is related to its inhibition of neuronal inflammation and promoting the recovery of nerve injury. These results provide a pharmacologic basis for the treatment of depression of XYW in clinical application.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Encephalitis/metabolism , Hippocampus/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Animals , Encephalitis/chemically induced , Encephalitis/prevention & control , Hippocampus/metabolism , Lipopolysaccharides/administration & dosage , Male , Neurons/metabolism , Rats, Sprague-Dawley
7.
Nutr Neurosci ; 23(8): 600-612, 2020 Aug.
Article in English | MEDLINE | ID: mdl-30353787

ABSTRACT

Lingonberries (LB) have been shown to have beneficial metabolic effects, which is associated with an altered gut microbiota. This study investigated whether the LB-induced improvements were associated with altered gut- and neuroinflammatory markers, as well as cognitive performance in ApoE-/- mice fed high-fat (HF) diets. Whole LB, as well as two separated fractions of LB were investigated. Eight-week-old male ApoE-/- mice were fed HF diets (38% kcal) containing whole LB (wLB), or the insoluble (insLB) and soluble fractions (solLB) of LB for 8 weeks. Inclusion of wLB and insLB fraction reduced weight gain, reduced fat deposition and improved glucose response. Both wLB and insLB fraction also changed the caecal microbiota composition and reduced intestinal S100B protein levels. The solLB fraction mainly induced weight loss in the mice. There were no significant changes in spatial memory, but significant increases in synaptic density in the hippocampus were observed in the brain of mice-fed wLB and insLB. Thus, this study shows that all lingonberry fractions counteracted negative effects of HF feedings on metabolic parameters. Also, wLB and insLB fraction showed to potentially improve brain function in the mice.


Subject(s)
Brain/drug effects , Encephalitis/prevention & control , Gastritis/prevention & control , Gastrointestinal Microbiome/drug effects , Plant Extracts/administration & dosage , Vaccinium vitis-idaea , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , Diet, High-Fat , Fatty Acids, Volatile , Lipid Metabolism , Male , Mice, Knockout, ApoE , S100 Calcium Binding Protein beta Subunit/metabolism , Synapses/drug effects
8.
Brain Res ; 1726: 146488, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31586625

ABSTRACT

Acute ischemic stroke is a leading cause of disability with limited therapeutic options. Continuous theta burst stimulation (cTBS) has recently been shown to be a promising noninvasive therapeutic strategy for neuroprotection in ischemic stroke patients. Here, we investigated the protective effects of cTBS following acute infarction using a photothrombotic stroke (PTS) model in the right posterior parietal cortex (PPC) of C57BL/6 mice. Treatment with cTBS resulted in a reduction in the volume of the infarct region and significantly increased vascular diameter and blood flow velocity in peri-infarct region, as well as decreased the numbers of calcium binding adapter molecule 1 (Iba-1)-positive microglia and glial fibrillary acidic protein (GFAP)-positive astrocytes. Moreover, the number of CD16/32 positive microglia was decreased, whereas the number of CD206 positive microglia was increased. In addition, performance in a water maze task was significantly improved. These results indicated that cTBS protected against PPC infarct region, leading to an improvement in spatial cognitive function, possibly as a result of changes to cerebral microvascular function and inflammatory responses.


Subject(s)
Brain/blood supply , Brain/physiopathology , Electric Stimulation Therapy/methods , Encephalitis/prevention & control , Ischemic Stroke/prevention & control , Neuroprotection , Animals , Capillaries/physiopathology , Cerebrovascular Circulation , Disease Models, Animal , Encephalitis/complications , Ischemic Stroke/complications , Ischemic Stroke/psychology , Male , Mice, Inbred C57BL , Microglia/physiology , Spatial Memory , Vasodilation
9.
Nutr Neurosci ; 22(3): 185-195, 2019 Mar.
Article in English | MEDLINE | ID: mdl-28784051

ABSTRACT

OBJECTIVES: Urolithins, ellagitannin-gut microbial-derived metabolites, have been reported to mediate pomegranate's neuroprotective effects against Alzheimer's disease (AD), but there are limited data on their effects against neuroinflammation. Herein, we: (1) evaluated whether urolithins (urolithins A and B and their methylated derivatives) attenuate neuroinflammation in murine BV-2 microglia and human SH-SY5Y neurons, and (2) evaluated hippocampus of transgenic AD (R1.40) mice administered a pomegranate extract (PE; 100 or 200 mg/kg/day for 3 weeks) for inflammatory biomarkers. METHODS: Effects of urolithins (10 µM) on inflammatory biomarkers were evaluated in lipopolysaccharide (LPS)-stimulated BV-2 microglia. In a non-contact co-culture cell model, SH-SY5Y cell viability was assessed after exposure to media collected from LPS-BV-2 cells treated with or without urolithins. Effects of urolithins on apoptosis and caspase 3/7 and 9 release from H2O2-induced oxidative stress of BV-2 and SH-SY5Y cells were assessed. Hippocampal tissues of vehicle and PE-treated transgenic R1.40 mice were evaluated for gene expression of inflammatory biomarkers by qRT-PCR. RESULTS: Urolithins decreased media levels of nitric oxide, interleukin 6 (IL-6), prostaglandin E2, and tumor necrosis factor alpha from LPS-BV-2 microglia. In the co-culture cell model, media from LPS-BV-2 cells treated with urolithins preserved SH-SY5Y cell viability greater than media from cells treated without urolithins. Urolithins mitigated apoptosis and caspase 3/7 and 9 release from H2O2-induced oxidative stress of BV-2 and SH-SY5Y cells. While not statistically significant, inflammatory biomarkers (TNF-α, COX-2, IL-1, and IL-6) appeared to follow a decreasing trend in the hippocampus of high-dose PE-treated animals compared to controls. DISCUSSION: The attenuation of neuroinflammation by urolithins may contribute, in part, toward pomegranate's neuroprotective effects against AD.


Subject(s)
Coumarins/administration & dosage , Encephalitis/metabolism , Gastrointestinal Microbiome , Hydrolyzable Tannins/metabolism , Lythraceae/metabolism , Neuroprotective Agents/administration & dosage , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Encephalitis/chemically induced , Encephalitis/prevention & control , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Inflammation Mediators , Lipopolysaccharides/administration & dosage , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Plant Extracts/administration & dosage
10.
Nutr Neurosci ; 22(4): 264-272, 2019 Apr.
Article in English | MEDLINE | ID: mdl-28946820

ABSTRACT

OBJECTIVES: Coenzyme Q10 (CoQ10, ubiquinone) stands among the safest supplements in the elderly to protect against cardiovascular disorders. Noteworthy, CoQ10 deficiency is common in many surviving stroke patients as they are mostly prescribed statins for the secondary prevention of stroke incidence lifelong. Accordingly, the current study aims to experimentally examine whether CoQ10 supplementation in animals receiving atorvastatin may affect acute stroke-induced injury. METHODS: Adult rats underwent transient middle cerebral artery occlusion after atorvastatin pretreatment (5 or 10 mg/ kg/day; po; 30 days) with or without CoQ10 (200 mg/kg/day). After 24 hours ischemic/reperfusion injury, animals were subjected to functional assessments followed by cerebral molecular and histological to detect inflammation, apoptosis and oxidative stress. RESULTS: Animals dosed with 10 mg/kg presented the worst neurological function and brain damage in the acute phase of stroke injury. CoQ10 supplementation efficiently improved functional deficit and cerebral infarction in all stroke animals, particularly those exhibiting statin toxicity. Such benefits were associated with remarkable anti-inflammatory and anti-apoptotic effects, based on the analyzed tumor necrosis factor-α, interleukin-6, Bax/Bcl2 and cleaved caspase 3/9 immunoblots. Importantly, our fluoro-jade staining data indicated CoQ10 may revert the stroke-induced neurodegeneration. No parallel alteration was detected in stroke-induced oxidative stress as determined by malondialdehyde and 8-oxo-2'-deoxyguanosine levels. DISCUSSION: These data suggest that all stroke animals may benefit from CoQ10 administration through modulating inflammatory and degenerative pathways. This study provides empirical evidence for potential advantages of CoQ10 supplementation in atorvastatin-receiving patients which may not shadow its antioxidant properties.


Subject(s)
Atorvastatin/administration & dosage , Brain Ischemia/prevention & control , Neuroprotective Agents/administration & dosage , Stroke/prevention & control , Ubiquinone/analogs & derivatives , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/pathology , Encephalitis/etiology , Encephalitis/prevention & control , Male , Oxidative Stress/drug effects , Rats, Wistar , Stroke/complications , Stroke/metabolism , Stroke/pathology , Treatment Outcome , Ubiquinone/administration & dosage
11.
Phytother Res ; 32(12): 2531-2540, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30277284

ABSTRACT

The acute exposure of trimethyltin (TMT) develops clinical syndrome characterized by amnesia, aggressive behavior, and complex seizures. This neurotoxicant selectively induces hippocampal neuronal injury and glial activation accompanied with resultant neuroinflammation. Here we report two candidates ginsenosides Rg3 and Rh2 as neuroprotection agents using a mouse model of TMT intoxication via a single injection (2 mg/kg) and primary neuronal culture systems. Four-week administration of Rg3 or Rh2 significantly reduced TMT-induced seizures and behavioral changes. Rg3 and Rh2 significantly attenuated the oxidative stress evidenced by improvement on antioxidant enzymes and neuronal loss and astrocytic activation in mouse brain. In primary cultures, TMT induced significant neuronal death after 24-h intoxication and vigorous secretion of inflammatory cytokines (IL-1α/ß, IL-6, TNF-α, and MCP-1) in astrocytes. Pretreatment with Rg3 or Rh2 not only reduced cell death but efficiently suppressed above mentioned inflammatory cytokines confirmed by antibody array test. The underlying protective mechanism by Rg3 and Rh2 was delineated through selective upregulation of PI3K/Akt and suppression of ERK activation. Intriguingly, Rg3 and Rh2 protected oligodendrocyte progenitor cells (O-2A) from TMT intoxication via promoting type 2 astrocytic differentiation without further inflammatory activation. Collectively, Rg3 and Rh2 interventions aimed at reducing oxidative stress and neuroinflammation neurotoxicity therefore are of therapeutic benefit in TMT-induced neurodegeneration.


Subject(s)
Encephalitis/prevention & control , Ginsenosides/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Embryo, Mammalian , Encephalitis/chemically induced , Encephalitis/pathology , Neurons/physiology , Neuroprotection/drug effects , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Trimethyltin Compounds
12.
Biomed Pharmacother ; 106: 1153-1160, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119182

ABSTRACT

Gypenosides, a saponins extract isolated from the Gynostemma pentaphyllum plant, produces neuroprotective effects in the brain. Our previous studies have shown that hippocampal glucocorticoid receptor (GR)-brain-derived neurotrophic factor (BDNF)-TrkB signaling was involved in the antidepressant-like effects of gypenosides. It remains unknown whether gypenosides could alleviate neuroinflammation in depressive-like animals. The aim of the present study was to address this issue in chronic unpredictable mild stress (CUMS). Gypenosides was administrated for four weeks, followed by sucrose preference test and tail suspension test, which were performed to evaluate the effects of gypenosides. The results showed that gypenosides reversed both the decreased sucrose preference and increased immobility time in CUMS mice. In addition, gypenosides also attenuated the increase of pro-inflammatory cytokine levels in the hippocampus of CUMS animals. Furthermore, the activation of NF-κB, as well as its upstream mediators IKKα and IKKß were inhibited by gypenosides. Last but not the least, CUMS promoted the activation of microglia, while gypenosides suppressed it according to the reduced number of iba1 positive cells. In conclusion, this study demonstrates that gypenosides exhibits the antidepressant-like effects in mice, which may be mediated by the inhibition of microglia and NF-κB signaling in the hippocampus.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Depression/drug therapy , Encephalitis/prevention & control , Hippocampus/drug effects , Stress, Psychological/drug therapy , Animals , Calcium-Binding Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Depression/metabolism , Depression/pathology , Depression/psychology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Encephalitis/psychology , Feeding Behavior/drug effects , Gynostemma , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , I-kappa B Kinase/metabolism , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , NF-kappa B/metabolism , Phosphorylation , Plant Extracts/pharmacology , Signal Transduction/drug effects , Stress, Psychological/metabolism , Stress, Psychological/pathology , Stress, Psychological/psychology
13.
Brain Res ; 1682: 61-70, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29291393

ABSTRACT

Almost all of the candidate drugs for ischemic stroke failed to be translated from bench to beside. One important reason is that animals used in experimental studies cannot mimic ischemic patients due to lack of comorbidities like hypertension, diabetes and obesity. Therefore, it is better to test candidate drugs not only in normal animals but also in animals with comorbidities. Patchouli alcohol (PA), a natural tricyclic sesquiterpene in the traditional Chinese herb Pogostemonisherba, is well recognized for its anti-inflammation function in various inflammatory diseases. And as inflammation plays a very important role in cerebral ischemia/reperfusion (I/R) injury process and determines the ultimate brain damage, we hypothesized that PA could protect against cerebral I/R injury through its anti-inflammation ability. In this study, the effects of PA on cerebral I/R injury were evaluated in normal mice and obese mice. In normal mice with cerebral I/R injury, PA treatment reduced the infarct volume and neurological deficits in a dose- and time-dependent manner. PA treatment alleviated BBB dysfunction, inhibited mRNA and protein levels of TNF-α and IL-1ß and modulated the activation of MAPKs signaling pathways. Moreover, PA also reduced infarct volume, alleviated the BBB dysfunction and inhibited inflammation in ob/ob mice with cerebral I/R injury. In conclusion, we demonstrated for the first time that PA could protect against cerebral I/R injury not only in normal mice but also in obese mice via inhibiting inflammation, suggesting that PA can be a potential drug for clinical treatment of ischemic stroke.


Subject(s)
Brain Injuries/prevention & control , Encephalitis/etiology , Encephalitis/prevention & control , Mitogen-Activated Protein Kinase Kinases/metabolism , Neuroprotective Agents/therapeutic use , Sesquiterpenes/therapeutic use , Animals , Brain Injuries/etiology , Brain Ischemia/complications , Disease Models, Animal , Dose-Response Relationship, Drug , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Reperfusion Injury/complications , Signal Transduction/drug effects , Time Factors
14.
Nutr Neurosci ; 21(8): 539-545, 2018 Oct.
Article in English | MEDLINE | ID: mdl-28504078

ABSTRACT

OBJECTIVES: The growing rate of neurological disorders is a major concern in today's scenario. Today's research is focusing on therapeutic interventions providing benefits in these disorders. Presently, drugs of natural origin have gained more interest for the treatment of central nervous system disorders for their efficacy and less/ no side effects. This review is emphasizing the cited roles of Trigonella foenum graecum (fenugreek) and its constituents in different neurological manifestations. METHOD: A review of the literature, relevant to the role of fenugreek and its major constituents including saponins and alkaloids in different neurological aspects and in delineating the health benefits, was conducted. RESULTS: The cited research acknowledged that fenugreek and its constituents exert positive influence on neurological health. Few studies have reported the beneficial role of fenugreek and its constituents like trigonelline in pathological symptoms of Alzheimer's disease. Similarly, other studies evidenced the neuroprotective, antidepressant, antianxiety as well as modulatory effect on cognitive functions and Parkinson's disease. DISCUSSION: Large populations are the sufferers of the neurological disorders, pointing the need for investigation of such therapeutic interventions which target and delay the underlying pathological hallmarks and exert positive influence on different neurological health problems. Hypolipidemic, hypoglycemic, antioxidant, and immunomodulatory effects of fenugreek and its constituents with their potential role in various neurological disorders were already reported. In future, it would be of even greater interest to further develop more effective dosage, supplementation period, and to evaluate the therapeutic potentials of fenugreek and its constituents in neurological disorders by exploring underlying cellular and molecular mechanisms.


Subject(s)
Dietary Supplements , Evidence-Based Medicine , Neurodegenerative Diseases/therapy , Nootropic Agents/therapeutic use , Plant Extracts/therapeutic use , Seeds/chemistry , Trigonella/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/adverse effects , Antioxidants/therapeutic use , Dietary Supplements/adverse effects , Encephalitis/immunology , Encephalitis/prevention & control , Encephalitis/therapy , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/therapeutic use , Nervous System Diseases/immunology , Nervous System Diseases/prevention & control , Nervous System Diseases/therapy , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/adverse effects , Neuroprotective Agents/therapeutic use , Nootropic Agents/adverse effects , Plant Extracts/adverse effects
15.
J Nutr Biochem ; 49: 133-140, 2017 11.
Article in English | MEDLINE | ID: mdl-28950154

ABSTRACT

It has been recently shown that xanthohumol, a flavonoid present in hops, possesses antioxidant, anti-inflammatory and chemopreventive properties. However, its role in the aging brain has not been addressed so far. Therefore, this study aimed to investigate the possible neuroprotective activity of xanthohumol against age-related inflammatory and apoptotic brain damage in male senescence-accelerated prone mice (SAMP8). Animals were divided into 4 groups: Untreated young mice, untreated old mice and old mice treated either with 1 mg kg-1 day-1 or 5 mg kg-1 day-1 xanthohumol. Young and old senescence accelerated resistant mice (SAMR1) were used as controls. After 30 days of treatment, animals were sacrificed and their brains were collected and immediately frozen in liquid nitrogen. mRNA (GFAP, TNF-α, IL-1ß, AIF, BAD, BAX, XIAP, NAIP and Bcl-2) and protein (GFAP, TNF-α, IL-1ß, AIF, BAD, BAX, BDNF, synaptophysin and synapsin) expressions were measured by RT-PCR and Western blotting, respectively. Significant increased levels of pro-inflammatory (TNF-α, IL-1ß) and pro-apoptotic (AIF, BAD, BAX) markers were observed in both SAMP8 and SAMR1 old mice compared to young animals (P<.05) and also in SAMP8 untreated old mice compared to SAMR1 (P<.05). These alterations were significantly less evident in animals treated with both doses of xanthohumol (P<.05). Also, a reduced expression of synaptic markers was observed in old mice compared to young ones (P<.05) but it significantly recovered with 5 mg kg-1 day-1 xanthohumol treatment (P<.05). In conclusion, xanthohumol treatment modulated the inflammation and apoptosis of aged brains, exerting a protective effect on damage induced by aging.


Subject(s)
Aging , Brain/metabolism , Cognitive Dysfunction/prevention & control , Dietary Supplements , Flavonoids/therapeutic use , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Propiophenones/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biomarkers/metabolism , Brain/immunology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/prevention & control , Flavonoids/administration & dosage , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Inflammation Mediators/metabolism , Male , Mice, Inbred Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/immunology , Neuroprotective Agents/administration & dosage , Propiophenones/administration & dosage , Synaptophysin/genetics , Synaptophysin/metabolism
16.
Nutrients ; 9(10)2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28961195

ABSTRACT

Choline is involved in relevant neurochemical processes. In particular, it is the precursor and metabolite of acetylcholine (ACh). Choline is an essential component of different membrane phospholipids that are involved in intraneuronal signal transduction. On the other hand, cholinergic precursors are involved in ACh release and carry out a neuroprotective effect based on an anti-inflammatory action. Based on these findings, the present study was designed to evaluate the effects of choline and choline precursor (Choline alphoscerate, GPC) in the modulation of inflammatory processes in the rat brain. Male Wistar rats were intraperitoneally treated with 87 mg of choline chloride/kg/day (65 mg/kg/day of choline), and at choline-equivalent doses of GPC (150 mg/kg/day) and vehicle for two weeks. The brains were dissected and used for immunochemical and immunohistochemical analysis. Inflammatory cytokines (Interleukin-1ß, IL-1ß; Interleukin-6 , IL-6 and Tumor Necrosis Factor-α, TNF-α) and endothelial adhesion molecules (Intercellular Adhesion Molecule, ICAM-1 and Vascular cell Adhesion Molecule, VCAM-1) were studied in the frontal cortex, hippocampus, and cerebellum. The results clearly demonstrated that treatment with choline or GPC did not affect the expression of the inflammatory markers in the different cerebral areas evaluated. Therefore, choline and GPC did not stimulate the inflammatory processes that we assessed in this study.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cerebral Cortex/drug effects , Choline/therapeutic use , Encephalitis/prevention & control , Glycerylphosphorylcholine/therapeutic use , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Biomarkers/metabolism , Cerebellum/drug effects , Cerebellum/immunology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Choline/administration & dosage , Choline/adverse effects , Cytokines/metabolism , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/pathology , Frontal Lobe/drug effects , Frontal Lobe/immunology , Frontal Lobe/metabolism , Frontal Lobe/pathology , Glycerylphosphorylcholine/administration & dosage , Glycerylphosphorylcholine/adverse effects , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/pathology , Injections, Intraperitoneal , Intercellular Adhesion Molecule-1/metabolism , Male , Nerve Tissue Proteins/metabolism , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/cerebrospinal fluid , Rats, Wistar , Vascular Cell Adhesion Molecule-1/metabolism
17.
Neurosci Lett ; 651: 43-51, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28458018

ABSTRACT

The incorporation of Polygonum multiflorum into the diet can result in anti-aging effects owing to its wide range of biological and pharmaceutical properties. We investigated the anti-neuroinflammatory properties of CRPE56IGIH isolated from P. multiflorum by focusing on its role in the induction of phase II antioxidant enzymes and the modulation of upstream signaling pathways. In microglia, CRPE56IGIH significantly inhibited lipopolysaccharide (LPS)-stimulated nitric oxide and prostaglandin E2 production with nonspecific cytotoxicity. CRPE56IGIH also markedly inhibited LPS-inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 protein and mRNA expression in the same manner as it inhibited nitric oxide and prostaglandin E2 production. In the control cells, NF-κB transactivation and nuclear translocation occurred at a baseline level, which was significantly increased in response to LPS. However, pretreatment with CRPE56IGIH concentration-dependently inhibited the LPS-induced NF-κB transactivation and nuclear translocation. The phosphorylation of Janus kinase-signal transducers and activators of transcription and mitogen-activated protein kinases was markedly upregulated by LPS, but considerably and dose-dependently inhibited by pretreatment with CRPE56IGIH. Furthermore, CRPE56IGIH induced the expression of phase II antioxidant enzymes, including heme oxygenase-1 (HO-1) and NADPH dehydrogenase quinone-1 (NQO-1). The activation of upstream signaling pathways, such as the Nrf2 pathway, was significantly increased following CRPE56IGIH treatment. Furthermore, the anti-neuroinflammatory effect of CRPE56IGIH was reversed by transfection of Nrf2, HO-1, and NQO-1 siRNA. Our results indicated that CRPE56IGIH isolated from P. multiflorum could be used as a natural anti-neuroinflammatory agent that induces phase II antioxidant enzymes via Nrf2 signaling.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Encephalitis/metabolism , Fallopia multiflora/chemistry , Microglia/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , NF-kappa B/antagonists & inhibitors , Plant Extracts/administration & dosage , Animals , Cells, Cultured , Encephalitis/chemically induced , Encephalitis/prevention & control , Lipopolysaccharides , Mice, Inbred ICR , Microglia/metabolism , Plant Extracts/chemistry , Signal Transduction , Up-Regulation
18.
Neuropharmacology ; 113(Pt A): 137-147, 2017 02.
Article in English | MEDLINE | ID: mdl-27712995

ABSTRACT

Neuropathic pain, a debilitating pain condition and the underlying pathogenic mechanisms are complex and interwoven amongst each other and still there is scant information available regarding therapies which promise to treat the condition. Evidence indicate that oxidative/nitrosative stress induced poly (ADP-ribose) polymerase (PARP) overactivation initiate neuroinflammation and bioenergetic crisis culminating into neurodegenerative changes following nerve injury. Hence, we investigated the therapeutic effect of combining an antioxidant, quercetin and a PARP inhibitor, 4-amino 1, 8-naphthalimide (4-ANI) on the hallmark deficits induced by chronic constriction injury (CCI) of sciatic nerve in rats. Quercetin (25 mg/kg, p.o.) and 4-ANI (3 mg/kg, p.o.) were administered either alone or in combination for 14 days to examine sciatic functional index, allodynia and hyperalgesia using walking track analysis, Von Frey, acetone spray and hot plate tests respectively. Malondialdehyde, nitrite and glutathione levels were estimated to detect oxidative/nitrosative stress; mitochondrial membrane potential and cytochrome c oxidase activity to assess mitochondrial function; NAD & ATP levels to examine the bioenergetic status and levels of inflammatory markers were evaluated in ipsilateral sciatic nerve. Quercetin and 4-ANI alone improved the pain behaviour and biochemical alterations but the combination therapy demonstrated an appreciable reversal of CCI-induced changes. Nitrotyrosine and Poly ADP-Ribose (PAR) immunopositivity was decreased and nuclear factor erythroid 2-related factor (Nrf-2) levels were increased significantly in micro-sections of the sciatic nerve and dorsal root ganglion (DRG) of treatment group. These results suggest that simultaneous inhibition of oxidative stress-PARP activation cascade may potentially be useful strategies for management of trauma induced neuropathic pain.


Subject(s)
1-Naphthylamine/analogs & derivatives , Antioxidants/administration & dosage , Encephalitis/prevention & control , Naphthalimides/administration & dosage , Neuralgia/prevention & control , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerases/metabolism , Quercetin/administration & dosage , Quinolones/administration & dosage , 1-Naphthylamine/administration & dosage , 1-Naphthylamine/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Antioxidants/therapeutic use , Encephalitis/complications , Encephalitis/enzymology , Hyperalgesia/prevention & control , Male , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Naphthalimides/therapeutic use , Neuralgia/complications , Neuralgia/enzymology , Oxidative Stress/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Quercetin/therapeutic use , Quinolones/therapeutic use , Rats , Rats, Sprague-Dawley , Sciatic Nerve/injuries
19.
Nutr Neurosci ; 20(9): 497-504, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27256583

ABSTRACT

OBJECTIVE: There is an urge to identify new molecules which can modulate process of epileptogenesis, since currently available drugs act symptomatically and one-third of the patients remain refractory to the disease. Hence, the present study was conducted to evaluate the effects of Resveratrol (RESV) on epileptogenesis in pentylenetetrazole (PTZ)-induced kindling in mice. METHOD: Swiss albino mice were administered RESV (10, 20 and 40 mg/kg,p.o) in acute study. On the seventh day animals were subjected to various neurological and neurobehavioral tests viz, Increasing Current Electroshock Test (ICES), PTZ-induced seizures, passive avoidance response, and elevated plus maze test. For the development of kindling PTZ was administered in a dose of 25 mg/kg, i.p. on every alternate day and RESV in all the three doses was administered daily. Seizure score was continuously monitored till the development of kindling and cognition tests were performed in the end of the study. The animals were sacrificed and levels of inflammatory biomarkers viz., IL-1ß, interleukin-1 receptor antagonist (IL1-Ra), IL-6, and TNF-α were assessed in the hippocampus and cortex of the kindled animals. RESULTS: RESV in all three doses increased the seizure threshold to hind limb extension in the ICES test. RESV in all the tested doses suppressed the development of kindling and reduced the levels of IL-1ß, IL1-Ra, IL-6, and TNF-α in kindled mice. CONCLUSION: RESV suppressed the development of kindling in mice and decreased the levels of inflammatory biomarkers in their hippocampus. RESV modified brain inflammation during epileptogenesis and found to possess nootropic activity in the kindled mice.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticonvulsants/therapeutic use , Cerebral Cortex/metabolism , Dietary Supplements , Hippocampus/metabolism , Seizures/prevention & control , Stilbenes/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anticonvulsants/administration & dosage , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Avoidance Learning/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/immunology , Convulsants/antagonists & inhibitors , Convulsants/toxicity , Encephalitis/chemically induced , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/prevention & control , Female , Hippocampus/drug effects , Hippocampus/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Kindling, Neurologic/drug effects , Kindling, Neurologic/immunology , Kindling, Neurologic/metabolism , Male , Maze Learning/drug effects , Mice , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Pentylenetetrazole/antagonists & inhibitors , Pentylenetetrazole/toxicity , Random Allocation , Resveratrol , Seizures/chemically induced , Seizures/immunology , Seizures/metabolism , Stilbenes/administration & dosage
20.
Neurotherapeutics ; 14(2): 447-462, 2017 04.
Article in English | MEDLINE | ID: mdl-27928708

ABSTRACT

Abnormal cytoplasmic mislocalization of transactive response DNA binding protein 43 (TARDBP or TDP-43) in degenerating neurons is a hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U). Our previous work suggested that nuclear factor kappa B (NF-κB) may constitute a therapeutic target for TDP-43-mediated disease. Here, we investigated the effects of root extract of Withania somnifera (Ashwagandha), an herbal medicine with anti-inflammatory properties, in transgenic mice expressing a genomic fragment encoding human TDP-43A315T mutant. Ashwagandha extract was administered orally to hTDP-43A315T mice for a period of 8 weeks starting at 64 and 48 weeks of age for males and females, respectively. The treatment of hTDP-43A315T mice ameliorated their motor performance on rotarod test and cognitive function assessed by the passive avoidance test. Microscopy examination of tissue samples revealed that Ashwagandha treatment of hTDP-43A315T mice improved innervation at neuromuscular junctions, attenuated neuroinflammation, and reduced NF-κB activation. Remarkably, Ashwagandha treatment reversed the cytoplasmic mislocalization of hTDP-43 in spinal motor neurons and in brain cortical neurons of hTDP-43A315T mice and it reduced hTDP-43 aggregation. In vitro evidence is presented that the neuronal rescue of TDP-43 mislocalization may be due to the indirect effect of factors released from microglial cells exposed to Ashwagandha. These results suggest that Ashwagandha and its constituents might represent promising therapeutics for TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/physiopathology , Plant Extracts/administration & dosage , TDP-43 Proteinopathies/physiopathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/prevention & control , Animals , Avoidance Learning/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Encephalitis/prevention & control , Female , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/prevention & control , Male , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , NF-kappa B/metabolism , Neuromuscular Junction/drug effects , Neurons/drug effects , Neurons/metabolism , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Rotarod Performance Test , Spinal Cord/drug effects , Spinal Cord/metabolism , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/prevention & control , Withania
SELECTION OF CITATIONS
SEARCH DETAIL