Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
2.
Int J Mol Sci ; 22(24)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34948033

ABSTRACT

This study was designed to determine the effect of acute caffeine (CAF) administration, which exerts a broad spectrum of anti-inflammatory activity, on the synthesis of pro-inflammatory cytokines and their receptors in the hypothalamus and choroid plexus (ChP) during acute inflammation caused by the injection of bacterial endotoxin-lipopolysaccharide (LPS). The experiment was performed on 24 female sheep randomly divided into four groups: control; LPS treated (iv.; 400 ng/kg of body mass (bm.)); CAF treated (iv.; 30 mg/kg of bm.); and LPS and CAF treated. The animals were euthanized 3 h after the treatment. It was found that acute administration of CAF suppressed the synthesis of interleukin (IL-1ß) and tumor necrosis factor (TNF)α, but did not influence IL-6, in the hypothalamus during LPS-induced inflammation. The injection of CAF reduced the LPS-induced expression of TNF mRNA in the ChP. CAF lowered the gene expression of IL-6 cytokine family signal transducer (IL6ST) and TNF receptor superfamily member 1A (TNFRSF1) in the hypothalamus and IL-1 type II receptor (IL1R2) in the ChP. Our study on the sheep model suggests that CAF may attenuate the inflammatory response at the hypothalamic level and partly influence the inflammatory signal generated by the ChP cells. This suggests the potential of CAF to suppress neuroinflammatory processes induced by peripheral immune/inflammatory challenges.


Subject(s)
Caffeine/administration & dosage , Choroid Plexus/immunology , Cytokines/genetics , Encephalitis/drug therapy , Hypothalamus/immunology , Lipopolysaccharides/adverse effects , Administration, Intravenous , Animals , Caffeine/pharmacology , Choroid Plexus/drug effects , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/genetics , Female , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Interleukin-1beta/genetics , Interleukin-6/metabolism , Sheep , Tumor Necrosis Factor-alpha/genetics
3.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34948457

ABSTRACT

High-fat diet (HFD)-induced comorbid cognitive and behavioural impairments are thought to be the result of persistent low-grade neuroinflammation. Metformin, a first-line medication for the treatment of type-2 diabetes, seems to ameliorate these comorbidities, but the underlying mechanism(s) are not clear. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP) are neuroprotective peptides endowed with anti-inflammatory properties. Alterations to the PACAP/VIP system could be pivotal during the development of HFD-induced neuroinflammation. To unveil the pathogenic mechanisms underlying HFD-induced neuroinflammation and assess metformin's therapeutic activities, (1) we determined if HFD-induced proinflammatory activity was present in vulnerable brain regions associated with the development of comorbid behaviors, (2) investigated if the PACAP/VIP system is altered by HFD, and (3) assessed if metformin rescues such diet-induced neurochemical alterations. C57BL/6J male mice were divided into two groups to receive either standard chow (SC) or HFD for 16 weeks. A further HFD group received metformin (HFD + M) (300 mg/kg BW daily for 5 weeks) via oral gavage. Body weight, fasting glucose, and insulin levels were measured. After 16 weeks, the proinflammatory profile, glial activation markers, and changes within the PI3K/AKT intracellular pathway and the PACAP/VIP system were evaluated by real-time qPCR and/or Western blot in the hypothalamus, hippocampus, prefrontal cortex, and amygdala. Our data showed that HFD causes widespread low-grade neuroinflammation and gliosis, with regional-specific differences across brain regions. HFD also diminished phospho-AKT(Ser473) expression and caused significant disruptions to the PACAP/VIP system. Treatment with metformin attenuated these neuroinflammatory signatures and reversed PI3K/AKT and PACAP/VIP alterations caused by HFD. Altogether, our findings demonstrate that metformin treatment rescues HFD-induced neuroinflammation in vulnerable brain regions, most likely by a mechanism involving the reinstatement of PACAP/VIP system homeostasis. Data also suggests that the PI3K/AKT pathway, at least in part, mediates some of metformin's beneficial effects.


Subject(s)
Diet, High-Fat/adverse effects , Encephalitis/drug therapy , Metformin/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Vasoactive Intestinal Peptide/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Case-Control Studies , Down-Regulation , Encephalitis/chemically induced , Encephalitis/genetics , Encephalitis/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Signal Transduction/drug effects , Vasoactive Intestinal Peptide/genetics
4.
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
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
7.
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
8.
Endocrinology ; 161(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32603429

ABSTRACT

The hypothalamus plays a critical role in controlling energy balance. High-fat diet (HFD) feeding increases the gene expression of proinflammatory mediators and decreases insulin actions in the hypothalamus. Here, we show that a gut-derived hormone, glucose-dependent insulinotropic polypeptide (GIP), whose levels are elevated during diet-induced obesity, promotes and mediates hypothalamic inflammation and insulin resistance during HFD-induced obesity. Unbiased ribonucleic acid sequencing of GIP-stimulated hypothalami revealed that hypothalamic pathways most affected by intracerebroventricular (ICV) GIP stimulation were related to inflammatory-related responses. Subsequent analysis demonstrated that GIP administered either peripherally or centrally, increased proinflammatory-related factors such as Il-6 and Socs3 in the hypothalamus, but not in the cortex of C57BL/6J male mice. Consistently, hypothalamic activation of IκB kinase-ß inflammatory signaling was induced by ICV GIP. Further, hypothalamic levels of proinflammatory cytokines and Socs3 were significantly reduced by an antagonistic GIP receptor (GIPR) antibody and by GIPR deficiency. Additionally, centrally administered GIP reduced anorectic actions of insulin in the brain and diminished insulin-induced phosphorylation of Protein kinase B and Glycogen synthase kinase 3ß in the hypothalamus. Collectively, these findings reveal a previously unrecognized role for brain GIP signaling in diet-induced inflammation and insulin resistance in the hypothalamus.


Subject(s)
Encephalitis/chemically induced , Gastric Inhibitory Polypeptide/pharmacology , Hypothalamus/drug effects , Inflammation/chemically induced , Insulin Resistance , Receptors, Gastrointestinal Hormone/physiology , Animals , Diet, High-Fat , Encephalitis/genetics , Gastric Inhibitory Polypeptide/administration & dosage , Gastric Inhibitory Polypeptide/physiology , Hypothalamus/immunology , Hypothalamus/pathology , Inflammation/genetics , Infusions, Intraventricular , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Receptors, Gastrointestinal Hormone/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Biomed Res Int ; 2020: 4259316, 2020.
Article in English | MEDLINE | ID: mdl-32596307

ABSTRACT

The systemic administration of lipopolysaccharide (LPS) has been recognized to induce neuroinflammation which plays a significant role in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. In this study, we aimed to determine the protective effect of Zingiber cassumunar (Z. cassumunar) or Phlai (in Thai) against LPS-induced neuronal cell loss and the upregulation of glial fibrillary acidic protein (GFAP) of astrocytes in the hippocampus. Adult male Wistar rats were orally administered with Z. cassumunar extract at various doses (50, 100, and 200 mg/kg body weight) for 14 days before a single injection of LPS (250 µg/kg/i.p.). The results indicated that LPS-treated animals exhibited neuronal cell loss and the activation of astrocytes and also increased proinflammatory cytokine interleukin- (IL-) 1ß in the hippocampus. Pretreatment with Z. cassumunar markedly reduced neuronal cell loss in the hippocampus. In addition, Z. cassumunar extract at a dose of 200 mg/kg BW significantly suppressed the inflammatory response by reducing the expression of GFAP and IL-1ß in the hippocampus. Therefore, the results suggested that Z. cassumunar extract might be valuable as a neuroprotective agent in neuroinflammation-induced brain damage. However, further investigations are essential to validate the possible active ingredients and mechanisms of its neuroprotective effect.


Subject(s)
Astrocytes/drug effects , Encephalitis/physiopathology , Hippocampus/drug effects , Neuroprotective Agents/administration & dosage , Plant Extracts/administration & dosage , Zingiber officinale , Animals , Encephalitis/chemically induced , Hippocampus/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/administration & dosage , Male , Rats, Wistar
10.
Int Immunopharmacol ; 80: 106181, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31926446

ABSTRACT

Saikosaponin-d (SSd), a triterpenoid saponins compound extracted from Radix Bupleuri, has been demonstrated to effectively alleviate chronic mild stress-induced depressive behaviors in rats, but the underlying molecular mechanisms are still uncertain. Increasing evidence indicated that microglia activation and inflammatory responses were involved in the pathogenesis of depression. Thus, we desired to induce inflammation-related depressive-like behaviors in mice by injecting lipopolysaccharide (LPS) to investigate whether the antidepressant effect of SSd is related to inhibiting inflammation. The results of behavioral tests showed that SSd administration ameliorated LPS-induced depressive-like behaviors, as shown by increased sucrose consumption in the sucrose preference test and decreased immobility time in the tail suspension test and forced swimming test. Furthermore, immunostaining results showed that SSd pretreatment inhibited LPS-induced microglia activation in the hippocampus of mice and primary microglia cells. Enzyme-linked immunosorbent assay (ELISA) results showed that SSd pretreatment suppressed LPS-induced overexpression of inflammatory factors such as interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α both in vivo and in vitro. Immunostaining and western blot analysis results demonstrated that SSd pretreatment also inhibited LPS-induced HMGB1 translocation from nuclear to extracellular and decreased the protein levels of TLR4, p-IκB-α, NF-κBp65. These results suggested that SSd effectively improved LPS-induced inflammation-related depressive-like behaviors by inhibiting LPS-induced microglia activation and neuroinflammation, and the possible mechanism might associate with the regulation of the HMGB1/TLR4/NF-κB signaling pathway.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Encephalitis/drug therapy , Microglia/drug effects , Oleanolic Acid/analogs & derivatives , Saponins/therapeutic use , Animals , Antidepressive Agents/pharmacology , Cell Survival/drug effects , Cells, Cultured , Depression/chemically induced , Depression/metabolism , Encephalitis/chemically induced , Encephalitis/metabolism , HMGB1 Protein/metabolism , Lipopolysaccharides , Male , Mice, Inbred ICR , Microglia/metabolism , NF-kappa B/metabolism , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Saponins/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
11.
Neurotoxicology ; 76: 99-110, 2020 01.
Article in English | MEDLINE | ID: mdl-31678059

ABSTRACT

Environmental pollution due to arsenic is associated with several adverse health effects including neurotoxicity in animals and humans. Selenium is a nutritionally essential trace metalloid well documented to elicit compelling pharmacological activities in vitro and in vivo. Report on the influence of selenium on arsenic-mediated behavioral derangement is lacking in literature. Hence, to fill this knowledge gap, rats were either exposed to arsenic per se in drinking water at 60 µg AsO2Na/L or co-administered with inorganic selenium at 0.25 mg/kg or organic selenium diphenyl diselenide (DPDS) at 2.5 mg/kg body weight for 45 successive days. Neurobehavioural data from rats in a new environment using video-tracking software evinced that inorganic and organic forms of selenium significantly (p < 0.05) abrogated arsenic-induced motor and locomotor insufficiencies such as increased negative geotaxis and fecal pellets numbers as well as the diminution in grip strength, body rotation, maximum speed, absolute turn angle and total distance travelled. The augmentation in the behavioral activities in rats co-administered with arsenic and both forms of selenium was substantiated using track and occupancy plots analyses. Selenium mitigated arsenic-induced decreases in glutathione level and acetylcholinesterase activity as well as the increase in oxidative stress and reactive oxygen and nitrogen species. Moreover, selenium diminished inflammatory parameters (myeloperoxidase activity, nitric oxide, tumour necrosis factor alpha and interleukin-1 beta levels), caspase-3 activity and ameliorated histological lesions in the cerebellum, cerebrum and liver of the rats. Collectively, selenium abated arsenic-induced behavioral derangements via anti-inflammation, antioxidant and anti-apoptotic mechanisms in rats.


Subject(s)
Arsenic/toxicity , Behavior, Animal/drug effects , Environmental Pollutants/toxicity , Neuroprotective Agents/administration & dosage , Selenium/administration & dosage , Animals , Brain/drug effects , Brain/pathology , Brain Chemistry/drug effects , Encephalitis/chemically induced , Encephalitis/metabolism , Liver/drug effects , Liver/metabolism , Locomotion/drug effects , Male , Motor Activity/drug effects , Rats, Wistar
12.
Metab Brain Dis ; 35(2): 295-303, 2020 02.
Article in English | MEDLINE | ID: mdl-31828693

ABSTRACT

Tyrosinemia type II is an autosomal recessive inborn error of metabolism caused by hepatic cytosolic tyrosine aminotransferase deficiency. Importantly, this disease is associated with neurological and developmental abnormalities in many patients. Considering that the mechanisms underlying neurological dysfunction in hypertyrosinemic patients are poorly understood, in the present work we investigated the levels of cytokines - tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), IL-6 and IL-10 - in cerebellum, hippocampus, striatum of young rats exposed to chronic administration of L-tyrosine. In addition, we also investigated the impact of the supplementation with Omega-3 fatty acids (n-3 PUFA) on the rodent model of Tyrosinemia. Notably, previous study demonstrated an association between L-tyrosine toxicity and n-3 PUFA deficiency. Our results showed a significant increase in the levels of pro- and anti-inflammatory cytokines in brain structures when animals were administered with L-tyrosine. Cerebral cortex and striatum seem to be more susceptible to the inflammation induced by tyrosine toxicity. Importantly, n-3 PUFA supplementation attenuated the alterations on cytokines levels induced by tyrosine exposure in brain regions of infant rats. In conclusion, the brain inflammation is also an important process related to tyrosine neurotoxicity observed in the experimental model of Tyrosinemia. Finally, n-3 PUFA supplementation could be considered as a potential neuroprotective adjunctive therapy for Tyrosinemias, especially type II.


Subject(s)
Dietary Supplements , Encephalitis/chemically induced , Encephalitis/drug therapy , Fatty Acids, Omega-3/administration & dosage , Inflammation Mediators/antagonists & inhibitors , Tyrosine/toxicity , Animals , Animals, Newborn , Drug Administration Schedule , Encephalitis/metabolism , Inflammation Mediators/metabolism , Male , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Tyrosine/administration & dosage
13.
Nutr Neurosci ; 23(4): 321-334, 2020 Apr.
Article in English | MEDLINE | ID: mdl-30032721

ABSTRACT

A high-fat diet induces hypothalamic inflammation in rodents which, in turn, contributes to the development of obesity by eliciting both insulin and leptin resistance. However, the mechanism by which long-chain saturated fatty acids trigger inflammation is still contentious. To elucidate this mechanism, the effect of fatty acids on the expression of the pro-inflammatory cytokines IL-6 and TNFα was investigated in the mHypoE-N42 hypothalamic cell line (N42). N42 cells were treated with lauric acid (LA) and palmitic acid (PA). PA challenge was carried out in the presence of either a TLR4 inhibitor, a ceramide synthesis inhibitor (L-cycloserine), oleic acid (OA) or eicosapentaenoic acid (EPA). Intracellular ceramide accumulation was quantified using LC-ESI-MS/MS. PA but not LA upregulated IL-6 and TNFα. L-cycloserine, OA and EPA all counteracted PA-induced intracellular ceramide accumulation leading to a downregulation of IL-6 and TNFα. However, a TLR4 inhibitor failed to inhibit PA-induced upregulation of pro-inflammatory cytokines.In conclusion, PA induced the expression of IL-6 and TNFα in N42 neuronal cells independently of TLR4 but, partially, via ceramide synthesis with OA and EPA being anti-inflammatory by decreasing PA-induced intracellular ceramide build-up. Thus, ceramide accumulation represents one on the mechanisms by which PA induces inflammation in neurons.


Subject(s)
Ceramides/biosynthesis , Encephalitis/metabolism , Hypothalamus/metabolism , Palmitic Acid/administration & dosage , Palmitic Acid/metabolism , Toll-Like Receptor 4/metabolism , Animals , Apoptosis/drug effects , Cell Line , Encephalitis/chemically induced , Hypothalamus/drug effects , Inflammation Mediators/metabolism , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley
14.
Cells ; 8(12)2019 12 11.
Article in English | MEDLINE | ID: mdl-31835795

ABSTRACT

Beta-aminoisobutyric acid (BAIBA), a natural thymine catabolite, is involved in the beneficial effects of exercise on metabolic disorders. In particular, it has been reported to reverse the inflammatory processes observed in the peripheral organs of animal models of obesity. Therefore, this study aimed to investigate whether BAIBA improves hypothalamic inflammation, which is also tightly coupled with the development of obesity. We observed that treatment with BAIBA effectively reversed palmitic acid-induced hypothalamic inflammation and microglial activation in vivo. Consistent with these findings, we confirmed that BAIBA reversed body weight gain and increased adiposity observed in mice fed with a high-fat diet. Collectively, the current findings evidence the beneficial impacts of BAIBA on the imbalance of energy metabolism linked to hypothalamic inflammation.


Subject(s)
Aminoisobutyric Acids/administration & dosage , Encephalitis/drug therapy , Hypothalamus/drug effects , Microglia/immunology , Obesity/drug therapy , Palmitic Acid/adverse effects , Aminoisobutyric Acids/pharmacology , Animals , Cell Line , Cytokines/genetics , Cytokines/immunology , Diet, High-Fat/adverse effects , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/immunology , Energy Metabolism/drug effects , Humans , Hypothalamus/immunology , Male , Mice , Microglia/drug effects , Obesity/chemically induced , Obesity/complications
15.
Neurochem Int ; 131: 104563, 2019 12.
Article in English | MEDLINE | ID: mdl-31589911

ABSTRACT

Peripheral lipopolysaccharide (LPS) injection induces systemic inflammation through the activation of the inhibitor of nuclear factor kappa B (NF-κB) kinase (IKK)/NF-κB signaling pathway, which promotes brain dysfunction resulting in conditions including anorexia. LPS-mediated reduction of food intake is associated with activation of NF-κB signaling and phosphorylation of the transcription factor signal transducer and activator of transcription 3 (STAT3) in the hypothalamus. We recently reported phospholipase C-related catalytically inactive protein (PRIP) as a new negative regulator of phosphatidylinositol 3-kinase/AKT signaling. AKT regulates the IKK/NF-κB signaling pathway; therefore, this study aimed to investigate the role of PRIP/AKT signaling in LPS-mediated neuroinflammation-induced anorexia. PRIP gene (Prip1 and Prip2) knockout (Prip-KO) mice intraperitoneally (ip) administered with LPS exhibited increased anorexia responses compared with wild-type (WT) controls. Although few differences were observed between WT and Prip-KO mice in LPS-elicited plasma pro-inflammatory cytokine elevation, hypothalamic pro-inflammatory cytokines were significantly upregulated in Prip-KO rather than WT mice. Hypothalamic AKT and IKK phosphorylation and IκB degradation were significantly increased in Prip-KO rather than WT mice, indicating further promotion of AKT-mediated NF-κB signaling. Consistently, hypothalamic STAT3 was further phosphorylated in Prip-KO rather than WT mice. Furthermore, suppressor of cytokine signaling 3 (Socs3), a negative feedback regulator for STAT3 signaling, and cyclooxogenase-2 (Cox2), a candidate molecule in LPS-induced anorexigenic responses, were upregulated in the hypothalamus in Prip-KO rather than WT mice. Pro-inflammatory cytokines were upregulated in hypothalamic microglia isolated from Prip-KO rather than WT mice. Together, these findings indicate that PRIP negatively regulates LPS-induced anorexia caused by pro-inflammatory cytokine expression in the hypothalamus, which is mediated by AKT-activated NF-κB signaling. Importantly, hypothalamic microglia participate in this PRIP-mediated process. Elucidation of PRIP-mediated neuroinflammatory responses may provide novel insights into the pathophysiology of many brain dysfunctions.


Subject(s)
Anorexia/enzymology , Encephalitis/enzymology , Hypothalamus/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Anorexia/chemically induced , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Eating , Encephalitis/chemically induced , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
16.
Sci Rep ; 9(1): 7026, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31065039

ABSTRACT

Continuous exposure to preservatives such as nitrite salts has deleterious effects on different organs. Meanwhile, Nigella sativa oil can remediate such organ dysfunction. Here, we studied the effect of consumption of thymoquinone (TQ); the main component of Nigella sativa oil on the brain damage induced by sodium nitrite. Forty adult male rats were daily given oral gavage of sodium nitrite (80 mg/kg) with or without thymoquinone (50 mg/kg). Oxidative stress, cytokines of inflammation, fibrotic elements and apoptotic markers in brain tissue were measured. Exposure to sodium nitrite (SN) resulted in increased levels of malondialdehyde, TGF-ß, c-reactive protein, NF-κB, TNF-α, IL-1ß and caspase-3 associated with reduced levels of glutathione, cytochrome c oxidase, Nrf2 and IL-10. However, exposure of rats' brain tissues to thymoquinone resulted ameliorated all these effects. In conclusion, thymoquinone remediates sodium nitrite-induced brain impairment through several mechanisms including attenuation of oxidative stress, retrieving the reduced concentration of glutathione, blocks elevated levels of pro-inflammatory cytokines, restores cytochrome c oxidase activity, and reducing the apoptosis markers in the brain tissues of rats.


Subject(s)
Benzoquinones/administration & dosage , Encephalitis/drug therapy , Food Preservatives/adverse effects , Plant Oils/chemistry , Sodium Nitrite/adverse effects , Animals , Benzoquinones/pharmacology , Cytokines/metabolism , Disease Models, Animal , Electron Transport Complex IV/metabolism , Encephalitis/chemically induced , Encephalitis/metabolism , Gene Expression Regulation/drug effects , Glutathione/metabolism , Male , Mice , Oxidative Stress/drug effects
17.
J Neuroendocrinol ; 31(1): e12678, 2019 01.
Article in English | MEDLINE | ID: mdl-30582235

ABSTRACT

The links between obesity, inflammation and insulin resistance, which are all key characteristics of type 2 diabetes mellitus, are yet to be delineated in the brain. One of the key neuroinflammatory proteins detected in the hypothalamus with over-nutrition is tumour necrosis factor (TNF)α. Using immortalised embryonic rat and mouse hypothalamic cell lines (rHypoE-7 and mHypoE-46) that express orexigenic neuropeptide Y and agouti-related peptide, we investigated changes in insulin signalling and inflammatory gene marker mRNA expression after TNFα exposure. A quantitative polymerase chain reaction array of 84 inflammatory markers (cytokines, chemokines and receptors) demonstrated an increase in the expression of multiple genes encoding inflammatory markers upon exposure to 100 ng mL-1 TNFα for 4 hours. Furthermore, neurones pre-exposed to TNFα (50 ng mL-1 ) for 6 or 16 hours exhibited a significant reduction in phosphorylated Akt compared to control after insulin treatment, indicating the attenuation of insulin signalling. mRNA expression of insulin signalling-related genes was also decreased with exposure to TNFα. TNFα significantly increased mRNA expression of IκBα, Tnfrsf1a and IL6 at 4 and 24 hours, activating a pro-inflammatory state. An inhibitor study using an inhibitor of nuclear factor kappa B kinase subunit ß (IKK-ß) inhibitor, PS1145, demonstrated that TNFα-induced neuroinflammatory marker expression occurs through the IKK-ß/nuclear factor-kappa B pathway, whereas oleate, a monounsaturated fatty acid, had no effect on inflammatory markers. To test the efficacy of anti-inflammatory treatment to reverse insulin resistance, neurones were treated with TNFα and PS1145, which did not significantly restore the TNFα-induced changes in cellular insulin sensitivity, indicating that an alternative pathway may be involved. In conclusion, exposure to the inflammatory cytokine TNFα causes cellular insulin resistance and inflammation marker expression in the rHypoE-7 and mHypoE-46 neurones, consistent with effects seen with TNFα in peripheral tissues. It also mimics insulin- and palmitate-induced insulin resistance in hypothalamic neurones. The present study provides further evidence that altered central energy metabolism may be caused by obesity-induced cytokine expression.


Subject(s)
Encephalitis/metabolism , Hypothalamus/metabolism , Insulin Resistance/physiology , Neurons/metabolism , Tumor Necrosis Factor-alpha/metabolism , Agouti-Related Protein/metabolism , Animals , Cell Line , Encephalitis/chemically induced , Gene Expression Regulation , Hypothalamus/drug effects , Inflammation Mediators/metabolism , Mice , Neurons/drug effects , Neuropeptide Y/metabolism , RNA, Messenger/metabolism , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/administration & dosage
18.
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
19.
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
20.
Neuromolecular Med ; 20(3): 343-362, 2018 09.
Article in English | MEDLINE | ID: mdl-29846872

ABSTRACT

Reactive gliosis, microgliosis, and subsequent secretion of various inflammatory mediators like cytokines, proteases, reactive oxygen, and nitrogen species are the suggested key players associated with systemic inflammation-driven neuroinflammation and cognitive impairments in various neurological disorders. Conventionally, non-steroidal anti-inflammatory drugs are prescribed to suppress inflammation but due to their adverse effects, their usage is not well accepted. Natural products are emerging better therapeutic agents due to their affordability and inherent pleiotropic biological activities. In Ayurveda, Ashwagandha (Withania somnifera) is well known for its immunomodulatory properties. The current study is an extension of our previous report on in vitro model system and was aimed to investigate anti-neuroinflammatory potential of water extract from the Ashwagandha leaves (ASH-WEX) against systemic LPS-induced neuroinflammation and associated behavioral impairments using in vivo rat model system. Oral feeding of ASH-WEX for 8 weeks significantly ameliorated the anxiety-like behavior as evident from Elevated plus maze test. Suppression of reactive gliosis, inflammatory cytokines production like TNF-α, IL-1ß, IL-6, and expression of nitro-oxidative stress enzymes like iNOS, COX2, NOX2 etc were observed in ASH-WEX-treated animals. NFκB, P38, and JNK MAPKs pathways analysis showed their involvement in inflammation suppression which was further confirmed by inhibitor studies. The current study provides first ever preclinical evidence and scientific validation that ASH-WEX exhibits the anti-neuroinflammatory potential against systemic LPS-induced neuroinflammation and ameliorates associated behavioral abnormalities. Aqueous extract from Ashwagandha leaves and its active phytochemicals may prove to be promising candidates to prevent neuroinflammation associated with various neuropathologies.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Encephalitis/therapy , Phytotherapy , Plant Extracts/therapeutic use , Withania/chemistry , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anxiety/drug therapy , Cell Line , Cytokines/biosynthesis , Encephalitis/chemically induced , Gliosis/drug therapy , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Male , Microglia/drug effects , Microglia/metabolism , NF-kappa B/metabolism , Plant Extracts/administration & dosage , Plant Leaves/chemistry , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL