Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Phytomedicine ; 118: 154930, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37348246

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid-ß (Aß) and excessive neuroinflammation, resulting in neuronal cell death and cognitive impairments. Eugenol, a phenylpropene, is the main component of Syzygium aromaticum L. (Myrtaceae) and has multiple therapeutic effects, including neuroprotective and anti-inflammatory effects, through multimodal mechanisms. PURPOSE: We aimed to investigate the effect of eugenol on AD pathologies using a 5× familiar AD (5×FAD) mouse model. METHODS: Eight-month-old 5×FAD and wild-type mice were administered with eugenol (10 or 30 mg/kg/day, p.o) for 2 months. Y-maze and Morris water maze tests were performed to assess the cognitive function of mice. After the behavioral test, molecular analysis was conducted to investigate the therapeutic mechanism of eugenol. RESULTS: Our findings indicate that eugenol treatment effectively mitigated cognitive impairments in 5×FAD mice. This beneficial effect was associated with a decrease in AD pathologies, including neuronal cell loss and Aß deposition. Specifically, eugenol inhibited necroptosis activation and increased microglial phagocytosis, which were the underlying mechanisms for the observed reductions in neuronal cell loss and Aß deposition, respectively. CONCLUSION: Overall, our data suggest that eugenol would be a potential therapeutic candidate for AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Mice , Animals , Alzheimer Disease/metabolism , Eugenol/pharmacology , Eugenol/therapeutic use , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Disease Models, Animal
2.
Phytother Res ; 37(7): 2854-2863, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36814130

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease characterized by amyloid-ß (Aß) deposition, accompanied by neuroinflammation and memory dysfunction. Houttuyniae Herba (aerial parts of Houttuynia cordata, also known as fish mint; HH), an herbal medicine traditionally used to treat fever, urinary disorders, and pus, is revealed to protect neurons from Aß toxicity and regulate cholinergic dysfunction in AD models. In this study, we aimed to investigate the effects of HH on excessive accumulation of Aß followed by neuroinflammation, synaptic degeneration, and memory impairment. Two-month-old 5xFAD transgenic mice were administered HH at 100 mg/kg for 4 months. We observed that HH treatment ameliorated memory impairment and reduced Aß deposits in the brains of the mice. HH directly inhibited Aß aggregation in vitro using the Thioflavin T assay and indirectly suppressed the amyloidogenic pathway by increasing alpha-secretase expression in the mice brain. In addition, HH exerted antineuroinflammatory effects by reducing of glial activation and p38 phosphorylation. Moreover, HH treatment increased the expression of synaptophysin, a presynaptic marker protein. Overall, HH alleviates memory impairment in AD by facilitating nonamyloidogenic pathway and inhibiting neuroinflammation. Therefore, we suggest that HH can be a promising herbal drug for patients with AD requiring multifaceted improvement.


Subject(s)
Alzheimer Disease , Houttuynia , Neurodegenerative Diseases , Mice , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Houttuynia/metabolism , Neuroinflammatory Diseases , Mice, Transgenic , Plant Components, Aerial , Disease Models, Animal
3.
Phytomedicine ; 109: 154553, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36610153

ABSTRACT

BACKGROUND: We previously reported the potential inhibitory activity of 3',4'-dihydroxyflavone (DHF) on nitric oxide (NO) and prostaglandin E2 (PGE2) production in lipopolysaccharide (LPS)-stimulated macrophages. PURPOSE: We investigated the underlying molecular mechanisms of DHF in LPS-activated macrophages and evaluated its effect on LPS-induced septic shock in mice. METHODS: To explore the anti-inflammatory effect of DHF, nitrite, PGE2, and cytokines were measured in vitro and in vivo experiments. In addition, to verify the molecular signaling pathway, quantitative real time-PCR, luciferase assay, nuclear extraction, electrophoretic mobility shift assay, immunocytochemistry, immunoprecipitation, molecular docking analysis, and myeloid differentiation 2 (MD2)-LPS binding assay were conducted. RESULTS: DHF suppressed the LPS-induced expression of proinflammatory mediators through nuclear factor-κB (NF-κB), activator protein-1 (AP-1), and interferon regulatory factor 3 (IRF3) inactivation pathways in RAW 264.7 macrophages. Importantly, molecular docking analysis and in vitro binding assays showed that DHF interacts with the hydrophobic pocket of MD2 and then interferes with the interaction between LPS and toll-like receptor 4 (TLR4). DHF inhibited LPS-induced oxidative stress by upregulating nuclear factor erythroid 2-related factor 2 (Nrf2). Treatment of LPS-induced endotoxemia mice with DHF reduced the expression levels of pro-inflammatory mediators via the inactivation of NF-κB, AP-1, and signal transducer and activator of transcription 1 (STAT1) in the lung tissue, thus increasing the survival rate. CONCLUSION: Taken together, our data first time revealed the underlying mechanism of the DHF-dependent anti-inflammatory effect by preventing LPS from binding to the TLR4/MD2 complex. Therefore, DHF may be a possible anti-inflammatory agent for the treatment of LPS-mediated inflammatory diseases.


Subject(s)
Lipopolysaccharides , NF-kappa B , Animals , Mice , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factor AP-1/metabolism , Molecular Docking Simulation , Anti-Inflammatory Agents/pharmacology
4.
Nutrients ; 14(3)2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35277054

ABSTRACT

The hepatic adiponectin and farnesoid X receptor (FXR) signaling pathways play multiple roles in modulating lipid and glucose metabolism, reducing hepatic inflammation and fibrosis, and altering various metabolic targets for the management of non-alcoholic fatty liver disease (NAFLD). Alisma orientale (AO, Ze xie in Chinese and Taeksa in Korean) is an herbal plant whose tubers are enriched with triterpenoids, which have been reported to exhibit various bioactive properties associated with NAFLD. Here, the present study provides a preclinical evaluation of the biological functions and related signaling pathways of AO extract for the treatment of NAFLD in a Western diet (WD)-induced mouse model. The findings showed that AO extract significantly reversed serum markers (liver function, lipid profile, and glucose) and improved histological features in the liver sections of mice fed WD for 52 weeks. In addition, it also reduced hepatic expression of fibrogenic markers in liver tissue and decreased the extent of collagen-positive areas, as well as inhibited F4/80 macrophage aggregation and inflammatory cytokine secretion. The activation of adiponectin and FXR expression in hepatic tissue may be a major mechanistic signaling cascade supporting the promising role of AO in NAFLD pharmacotherapy. Collectively, our results demonstrated that AO extract improves non-alcoholic steatohepatitis (NASH) resolution, particularly with respect to NASH-related fibrosis, along with the regulation of liver enzymes, postprandial hyperglycemia, hyperlipidemia, and weight loss, probably through the modulation of the hepatic adiponectin and FXR pathways.


Subject(s)
Alisma , Diet, Western , Non-alcoholic Fatty Liver Disease , Adiponectin/metabolism , Alisma/chemistry , Animals , Diet, Western/adverse effects , Fibrosis , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/etiology , Plant Extracts/therapeutic use
5.
Nutr Neurosci ; 25(9): 1940-1947, 2022 Sep.
Article in English | MEDLINE | ID: mdl-33877009

ABSTRACT

OBJECTIVES: Amyloid beta (Aß)-induced abnormal neuroinflammation is recognized as a major pathological factor of Alzheimer's disease (AD), which results in memory impairment. Inhibition of excessive neuroinflammation mediated by Aß is considered a promising strategy to ameliorate AD symptoms. To regulate the inflammatory response, nutritional and dietary supplements have been used for centuries. Based on this idea, we investigated whether MBN, a novel nutritional mixture including cassia bark, turmeric root, and ginkgo leaf, can prevent AD progression through neuroinflammatory regulation. METHODS: MBN (10, 30, or 100 µg/ml) and Aß1-42 monomer were incubated together, and the degree of Aß aggregation was measured using Thioflavin T assay. The effects of MBN on Aß pathology in vivo were evaluated by orally administering MBN (40 mg/kg/day for 16 weeks) to five familial AD (5xFAD) mice. RESULTS: We found that treatment with MBN inhibited Aß aggregation in vitro. Next, MBN treatment significantly inhibited the activation of microglia induced by aggregated Aß in 5xFAD mice. Caspase-1 activation, which plays an important role in the maturation of interleukin-1ß, was markedly reduced by MBN. We also found that oral administration of MBN in 5xFAD mice alleviated memory decline. Taken together, our findings demonstrate that MBN suppresses neuroinflammation by downregulating the caspase-1 expression, thereby ameliorating memory impairment in 5xFAD mice. DISCUSSION: Based on these results, we suggest that MBN may be a preventive and therapeutic supplement for AD through the regulation of neuroinflammation.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Caspases/therapeutic use , Disease Models, Animal , Inflammasomes/therapeutic use , Interleukin-1beta , Memory Disorders/pathology , Memory Disorders/prevention & control , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
6.
Food Funct ; 12(6): 2672-2685, 2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33656018

ABSTRACT

Obesity is an increasing health problem worldwide as it is the major risk factor for metabolic diseases. In the present study, we investigated the anti-obesity effects of WHS by examining its effects on high fat diet (HFD)-induced obese mice. Male C57BL/6 mice were fed either a normal diet (ND) or a high fat diet (HFD) with or without WHS. At the end of the experiment, we observed the changes in their body weight and white adipose tissue (WAT) weight and lipid profiles in plasma. We performed western blot and histological analyses of WAT and liver to elucidate the molecular mechanisms of action. We also conducted fecal 16S rRNA analysis for investigating the gut microbiota. Our results indicated that pre- and post-oral administration of WHS significantly prevented body weight gain and reduced body fat weight in HFD-induced obese mice. In addition, WHS was found to improve adipocyte hypertrophy and liver fat accumulation by regulating the AMPK and AKT/mTOR pathways. WHS ameliorated hyperlipidemia by reducing total cholesterol and low-density lipoprotein (LDL) and decreased the energy metabolism-related hormones, leptin and insulin, in mouse plasma. Furthermore, we found that WHS modulated gut dysbiosis by normalizing HFD-induced changes. Taken together, our in vivo data implicate that WHS can be considered as a potential dietary supplement for alleviating obesity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Gastrointestinal Microbiome/drug effects , Hydrangea/chemistry , Obesity/metabolism , Plant Extracts/pharmacology , Animals , Body Weight/drug effects , Diet, High-Fat , Lipids/blood , Mice , Mice, Obese , Plant Leaves/chemistry , Signal Transduction/drug effects
7.
Nutrients ; 12(10)2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33023237

ABSTRACT

Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder that occurs in children characterized by inattention and hyperactivity. Prenatal alcohol exposure (PAE) can disrupt fetal neuronal development and cause an ADHD-like hyperactive behavior in the offspring. In this study, we hypothesized that metabolic disturbance would involve in ADHD neuropathology and aimed to investigate the changes in metabolite profile in PAE-induced ADHD-like model and the effects of HX106, a nutraceutical, on ADHD-like pathophysiology and metabolite changes. To this end, we administered HX106 to the mouse offspring affected by PAE (OPAE) and assessed the hyperactivity using the open field test. We observed that HX106-treated OPAE showed less hyperactive behavior than vehicle-treated OPAE. The effects of HX106 were found to be related to the regulation of dopamine transporter and D2 dopamine receptor expression. Furthermore, using gas chromatography time-of-flight mass spectrometry-based metabolomics, we explored the metabolite changes among the experimental groups. The metabolite profile, particularly related with the amino acids, linoleic acid and amino sugar pathways, was altered by PAE and reversed by HX106 treatment partially similar to that observed in the control group. Overall, this study suggest that metabolite alteration would be involved in ADHD pathology and that HX106 can be an efficient supplement to overcome ADHD by regulating dopamine signaling-related protein expression and metabolite changes.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Dietary Supplements , Fetal Alcohol Spectrum Disorders/drug therapy , Plant Extracts/pharmacology , Prenatal Exposure Delayed Effects/drug therapy , Animals , Attention Deficit Disorder with Hyperactivity/chemically induced , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/drug effects , Female , Fetal Alcohol Spectrum Disorders/psychology , Gas Chromatography-Mass Spectrometry/methods , Metabolomics , Mice , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/psychology
8.
Int J Mol Sci ; 21(12)2020 Jun 24.
Article in English | MEDLINE | ID: mdl-32599846

ABSTRACT

Abnormal amyloid-ß (Aß) accumulation is the most significant feature of Alzheimer's disease (AD). Among the several secretases involved in the generation of Aß, ß-secretase (BACE1) is the first rate-limiting enzyme in Aß production that can be utilized to prevent the development of Aß-related pathologies. Cinnamon extract, used in traditional medicine, was shown to inhibit the aggregation of tau protein and Aß aggregation. However, the effect of trans-cinnamaldehyde (TCA), the main component of cinnamon, on Aß deposition is unknown. Five-month-old 5XFAD mice were treated with TCA for eight weeks. Seven-month-old 5XFAD mice were evaluated for cognitive and spatial memory function. Brain samples collected at the conclusion of the treatment were assessed by immunofluorescence and biochemical analyses. Additional in vivo experiments were conducted to elucidate the mechanisms underlying the effect of TCA in the role of Aß deposition. TCA treatment led to improvements in cognitive impairment and reduced Aß deposition in the brains of 5XFAD mice. Interestingly, the levels of BACE1 were decreased, whereas the mRNA and protein levels of three well-known regulators of BACE1, silent information regulator 1 (SIRT1), peroxisome proliferator-activated receptor γ (PPARγ) coactivator 1α (PGC1α), and PPARγ, were increased in TCA-treated 5XFAD mice. TCA led to an improvement in AD pathology by reducing BACE1 levels through the activation of the SIRT1-PGC1α-PPARγ pathway, suggesting that TCA might be a useful therapeutic approach in AD.


Subject(s)
Acrolein/analogs & derivatives , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/toxicity , Gene Expression Regulation/drug effects , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/metabolism , Acrolein/pharmacology , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Animals , Antimutagenic Agents/pharmacology , Female , Humans , Male , Mice , Mice, Transgenic , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Sirtuin 1/genetics
9.
Nutrients ; 12(2)2020 Feb 12.
Article in English | MEDLINE | ID: mdl-32059355

ABSTRACT

Inflammatory bowel disease (IBD) is a major risk factor of colorectal cancer. Drugs currently used for IBD exhibit adverse effects including vomiting, nausea, and diarrhea. Naturally derived novel alternative therapies are required to overcome these limitations. In this study, we investigated the protective effects of ethanol extract of Cicer arietinum (CEE) in a dextran sodium sulfate (DSS)-induced mouse model of colitis. CEE markedly improved DSS-induced clinical symptoms and histological status, such as the disease activity index, spleen weight, and colon length. Moreover, CEE-treated mice showed significant recovery of DSS-induced crypt damage and cell death. CEE suppressed myeloperoxidase (MPO) activity and macrophage marker F4/80 mRNA expression in colonic tissue of mice with DSS-induced colitis, indicating neutrophil infiltration and macrophage accumulation, respectively. Although DSS upregulated pro-inflammatory mediators and activated transcription factors, CEE downregulated the mRNA expression of cytokines including interleukin-6, interleukin-1ß, and tumor necrosis factor-α, protein expression of cyclooxygenase-2 and inducible nitric oxide synthase, as well as activation of nuclear factor-kappa B (NF-кB) and signal transducer and activator of transcription 3 (STAT3). Hence, our findings reveal that the anti-inflammatory properties of CEE, involving the downregulation of the expression of pro-inflammatory mediators by inactivating NF-кB and STAT3 in DSS-induced colitis mice.


Subject(s)
Anti-Inflammatory Agents , Cicer/chemistry , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Dextran Sulfate/adverse effects , Ethanol , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Animals , Colitis, Ulcerative/etiology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Inflammation Mediators/metabolism , Male , Mice, Inbred ICR , NF-kappa B/genetics , NF-kappa B/metabolism , Plant Extracts/isolation & purification , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
10.
Neurotherapeutics ; 17(1): 189-199, 2020 01.
Article in English | MEDLINE | ID: mdl-31741224

ABSTRACT

One of the most significant pathologies of Alzheimer's disease (AD), an irreversible and progressive neurodegenerative disease that causes cognitive impairment, is the neuroinflammation facilitating the accumulation of amyloid-ß (Aß) peptide. Hence, the inhibition of abnormal neuroinflammatory response is considered a promising therapeutic approach for AD. Picrorhiza kurroa Bentham, Scrophulariae (PK) is a medicinal herb that has been traditionally used for the treatment of various diseases, including inflammation. This study aims to report the significance of PK treatment in markedly improving spatial learning memory and dramatically decreasing Aß levels in Tg6799 mice, also known 5xFAD mice, which have five familial AD (FAD) mutations. Remarkably, these effects correlated with reversal of disease-related microglial neuroinflammation, as evidenced by shifting microglia phenotypes from the inflammatory form to the anti-inflammatory form and inhibiting the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3 inflammasome activity. Moreover, PK administration induced silent information regulator type1/peroxisome proliferator-activated receptor-γ signaling, resulting in a decrease of ß-secretase 1 (BACE1) expression, which involved in Aß production. Overall, this study suggests that PK exhibits a neuroprotective effect by inducing alternative activation of microglia and downregulating the BACE1 expression, thereby ameliorating the disease pathophysiology and reversing the cognitive decline related to Aß deposition in AD mice.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Inflammasomes/drug effects , Inflammasomes/metabolism , Maze Learning/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Picrorhiza , Spatial Memory/drug effects , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Peptide Fragments/metabolism , Signal Transduction/drug effects
11.
Nutrients ; 11(11)2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31661844

ABSTRACT

Alzheimer's disease (AD) is the most common type of dementia in the elderly. It is characterized by the accumulation of amyloid-beta (Aß) and progressive cognitive impairment. To alleviate the symptoms of AD, functional foods and nutrients have been used for centuries. In this study, we investigated whether Cuscutae Japonicae Semen (CJS), a medicinal food traditionally used in East Asia, has effects on memory improvement and synapse protection in AD. We orally administered CJS to 5x familiar AD (5xFAD) transgenic mice and performed the Morris water maze test. The results showed that CJS treatment ameliorated the decline of memory function. Then, we demonstrated that CJS attenuated the degeneration of pre- and post-synaptic proteins in the hippocampi of 5xFAD mice. To demonstrate the effects of CJS in vitro, we treated Aß in primary neuronal culture with CJS and observed that CJS rescued the loss of functional synapses. The protective effects of CJS on the synapse were due to the inhibition of activated caspase-3 expression. Additionally, CJS inhibited the phosphorylation of glycogen synthase kinase-3ß and tau proteins, which contribute to synaptic dysfunction. Taken together, our results suggest that CJS is efficient in alleviating memory loss by rescuing caspase-3-mediated synaptic damage in AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Cuscuta/chemistry , Plant Extracts/pharmacology , Plants, Medicinal , Animals , Cognitive Dysfunction/drug therapy , Female , Maze Learning/drug effects , Memory/drug effects , Mice, Transgenic , Plant Extracts/chemistry
12.
Biomed Pharmacother ; 111: 1359-1366, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30841450

ABSTRACT

Abnormal inflammatory response in the central nervous system plays a critical role in various neurological disorders such as Parkinson's disease, Alzheimer's disease and Huntington's disease. Therefore, modulation of abnormal neuroinflammation is thought to be a promising therapeutic strategy for these diseases. Based on this idea, we focused on finding a potential candidate material that would regulate excessive neuroinflammation. Iresine celosia has long been used as a traditional Mexican medicine to treat fever and oral disorders. In the present study, we evaluated the anti-neuroinflammatory effects of Iresine celosia extract (ICE) in lipopolysaccharide (LPS)-stimulated BV2 microglia cells and mice models. In BV2 microglia cells, ICE markedly inhibited production of nitric oxide and proinflammatory cytokines such as tumor necrosis factor-α, interleukin-1ß, and interleukin-6 without causing cytotoxicity. ICE also ameliorated translocation of nuclear factor-κB from cytosol to nucleus by LPS. Moreover, ICE attenuated behavioral disturbances by inhibiting activation of microglia and astrocytes in LPS-treated mice. Collectively, these data indicate that ICE is a potential therapeutic agent for treating inflammation-related diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Celosia/chemistry , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Microglia/drug effects , Plant Extracts/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line , Cytokines/metabolism , Inflammation/metabolism , Medicine, Traditional/methods , Mice , Mice, Inbred C57BL , Microglia/metabolism , Nitric Oxide/metabolism
13.
Neurosci Lett ; 686: 161-167, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30213621

ABSTRACT

Neuroinflammation is an inflammatory process within the central nervous system that is mediated by microglial activation, which releases pro-inflammatory mediators leading to neurodegeneration. In this study, we investigated the effects of Peucedani Japonici Radix (PJR), a medicinal herb traditionally used in East Asia to treat neuroinflammation both in vitro and in vivo. First, we examined the effects of PJR on pro-inflammatory mediators in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells. The results showed that PJR suppressed the LPS-induced increase of several inflammatory factors, such as nitric oxide, inducible nitric oxide synthase, cyclooxygenase-2, prostaglandin E2, interleukin-1ß, and tumor necrosis factor-α. We also revealed that PJR inhibited the nuclear factor kappa B (NF-κB) pathway, which is the upstream modulator of inflammatory processes. Furthermore, to confirm the regulatory effects of PJR on microglia in vivo, we measured the number of ionized calcium-binding adapter molecule 1-positive cells in mouse brains and found that PJR treatment reduced microglial activation. Taken together, these results suggest that PJR inhibits microglia-mediated neuroinflammation through the modulation of NF-κB signaling and has the therapeutic potential to prevent inflammation-related neurodegenerative diseases.


Subject(s)
Inflammation Mediators/pharmacology , Inflammation/drug therapy , Microglia/drug effects , Nitric Oxide Synthase Type II/drug effects , Signal Transduction/drug effects , Animals , Cell Line , Cyclooxygenase 2/metabolism , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Microglia/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
Int J Mol Sci ; 19(6)2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29865187

ABSTRACT

Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, and is characterized by the accumulation of amyloid beta (Aß) as a pathological hallmark. Aß plays a central role in neuronal degeneration and synaptic dysfunction through the generation of excessive oxidative stress. In the present study, we explored whether leaves of Petasites japonicus (Siebold & Zucc.) Maxim. (PL), called butterbur and traditionally used in folk medicine, show neuroprotective action against Aß25⁻35 plaque neurotoxicity in vitro and in vivo. We found that PL protected Aß25⁻35 plaque-induced neuronal cell death and intracellular reactive oxygen species generation in HT22 cells by elevating expression levels of phosphorylated cyclic AMP response element-binding protein, heme oxygenase-1, and NAD(P)H quinine dehydrogenase 1. These neuroprotective effects of PL were also observed in Aß25⁻35 plaque-injected AD mouse models. Moreover, administration of PL diminished Aß25⁻35 plaque-induced synaptic dysfunction and memory impairment in mice. These findings lead us to suggest that PL can protect neurons against Aß25⁻35 plaque-induced neurotoxicity and thus may be a potential candidate to regulate the progression of AD.


Subject(s)
Alzheimer Disease/complications , Disease Models, Animal , Memory Disorders/drug therapy , Plant Extracts/pharmacology , Amyloid beta-Peptides , Animals , Apoptosis , Cells, Cultured , Male , Memory Disorders/etiology , Memory Disorders/prevention & control , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Peptide Fragments , Petasites/chemistry , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Plaque, Amyloid
15.
J Vet Med Sci ; 77(6): 693-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25715651

ABSTRACT

Niemann-Pick type C disease (NP-C) is a rare and ultimately fatal lysosomal storage disorder with variable neurologic symptoms. Loss of neuronal function and neuronal cell death occur in the NP-C brain, similar to the findings for other neurodegenerative diseases. Targeting of neuronal cells in the brain therefore represents a potential clinical intervention strategy to reduce the rate of disease progression and improve the quality of life. We previously reported that bone marrow stem cells show a neurogenic effect through CCL2 (also known as monocyte chemoattractant protein-1, MCP-1) secretion in the brains of NP-C mice. However, the direct effect of CCL2 on neurogenesis has not been ascertained. Here, to define neurogenic effects of CCL2 in NP-C, we applied human recombinant CCL2 to neural stem cells (NSCs) derived from NP-C mice. CCL2-treated NSCs showed significantly increased capacity for self-renewal, proliferation and neuronal differentiation. Similar results were observed in the subventricular zone of NP-C mice after CCL2 treatment. Furthermore, infusion of CCL2 into the NP-C mouse brain resulted in reduction of neuroinflammation. Taken together, our results demonstrate that CCL2 is a potential new therapeutic agent for NP-C.


Subject(s)
Chemokine CCL2/pharmacology , Drug Evaluation, Preclinical/methods , Neural Stem Cells/physiology , Neurogenesis/drug effects , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/metabolism , Analysis of Variance , Animals , Bromodeoxyuridine , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chemokine CCL2/metabolism , DNA Primers/genetics , Immunohistochemistry , Mice , Mice, Inbred BALB C , Neural Stem Cells/drug effects , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Brain Res ; 1529: 113-24, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23831521

ABSTRACT

Accumulation of amyloid-ß (Aß) is thought to be a central pathology in the brain of patients with Alzheimer's disease (AD). Neprilysin (NEP), a plasma membrane glycoprotein of the neutral zinc metalloendopeptidase family, is known as a major Aß-degrading enzyme in the brain. The level of NEP is reduced in the brains of patients with AD; therefore, NEP is under intense investigation as a potential therapeutic source for degradation of deposited Aß in AD. Previous studies have utilized viral vectors expressing NEP for reduction of Aß deposition in the brain. However, viral vectors have disadvantages regarding difficulty in control of insert size, expression desired (short- or long-term), and target cell type. Here, in order to overcome these disadvantages, we produced recombinant soluble NEP from insect cells using an NEP expression vector, which was administered by intracerebral injection into AD mice, resulting in significantly reduced accumulation of Aß. In addition, AD mice treated with NEP showed improved behavioral performance on the water maze test. These data support a role of recombinant soluble NEP in improving memory impairment by regulation of Aß deposition and suggest the possibility that approaches using protein therapy might have potential for development of alternative therapies for treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Antipsychotic Agents/therapeutic use , Memory Disorders/metabolism , Memory Disorders/therapy , Neprilysin/therapeutic use , Peptide Fragments/metabolism , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Hippocampus/cytology , Humans , In Situ Nick-End Labeling , Maze Learning/drug effects , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Presenilin-1/genetics , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL