Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
2.
Food Funct ; 9(1): 171-178, 2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29171599

ABSTRACT

Ginger, which has been widely used for dietary condiment, has been reported to improve memory dysfunction in an animal model of Alzheimer's disease (AD). Recently, a few trials have been carried out to enhance the effects of ginger by improving the bioavailability of its relevant components via fermentation. Some reports have suggested that the fermented ginger has the ability to affect the AD in vitro systems; however, its anti-amnesic effects on an in vivo model still remain to be investigated. In the present study, we aimed to investigate the neuroprotective effects of ginger fermented with Schizosaccharomyces pombe (FG) in the in vivo models of AD. The neuroprotective effects were investigated by employing behavioral, western blotting, and immunohistochemical assays. The administration of FG improved recognition memory, impaired by scopolamine injection, than that of non-fermented ginger. In addition, FG ameliorated memory impairment in amyloid beta1-42 (Aß1-42) plaque-injected mice via protecting neuronal cells in the CA3 area of the mouse hippocampus. Moreover, FG reinstated the pre- and postsynaptic protein levels decreased by Aß1-42 plaque-toxicity. Overall, these data suggest that FG attenuates memory impairment in Aß1-42 plaque-induced AD mice through inhibition of neuronal cell loss and synaptic disruption.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Amyloid beta-Peptides/toxicity , Hippocampus/drug effects , Neuroprotective Agents/administration & dosage , Plant Extracts/administration & dosage , Schizosaccharomyces/metabolism , Zingiber officinale/microbiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Fermentation , Zingiber officinale/chemistry , Hippocampus/cytology , Hippocampus/physiopathology , Humans , Male , Memory/drug effects , Mice , Mice, Inbred ICR , Neurons/drug effects , Neuroprotective Agents/metabolism , Plant Extracts/metabolism
3.
BMC Complement Altern Med ; 17(1): 501, 2017 Nov 25.
Article in English | MEDLINE | ID: mdl-29178947

ABSTRACT

BACKGROUND: Dangguijakyak-san (DJS) is an herbal formulation that has been clinically applicable for treating postmenopausal symptoms and neurological disorders. It is reported that hippocampal estrogen attenuates memory impairment via neuroprotection and synaptogenesis. However, the effect of DJS on hippocampal estrogen synthesis remains unknown. In this study, we explored the effect of DJS and its neuroprotective mechanism against memory impairment in ovariectomized (OVX) mice, with respect to hippocampal estrogen stimulation. METHODS: Cell cultures were prepared from the hippocampi of 18-day-old embryos from timed pregnant Sprague-Dawley rats. The hippocampi were dissected, collected, dissociated, and plated in 60-mm dishes. The cells were treated with DJS for 48 h and the supernatant was collected to determine estrogen levels. Female ICR mice (8-weeks-old) were housed for 1 week and ovariectomy was performed to remove the influence of ovary-synthesized estrogens. Following a 2-week post-surgical recovery period, the mice were administrated with DJS (50 and 100 mg/kg/day, p.o.) or 17ß-estradiol (200 µg/kg/day, i.p.) once daily for 21 days. Hippocampal and serum estrogen levels were determined using enzyme-linked immunosorbent assay kit. Memory behavioral tests, western blot, and immunohistochemical analyses were performed to evaluate the neuroprotective effects of DJS in this model. RESULTS: DJS treatment promoted estrogen synthesis in primary hippocampal cells and the hippocampus of OVX mice, resulting in the amelioration of OVX-induced memory impairment. Hippocampal estrogen stimulated by DJS treatment contributed to the activation of cAMP response element-binding protein and synaptic protein in OVX mice. CONCLUSION: DJS may attenuate memory deficits in postmenopausal women via hippocampal estrogen synthesis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Estrogens/metabolism , Hippocampus , Memory/drug effects , Neuroprotective Agents/pharmacology , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Drugs, Chinese Herbal/chemistry , Female , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Inbred ICR , Neuroprotective Agents/chemistry , Ovariectomy , Phosphorylation/drug effects , Pregnancy , Rats, Sprague-Dawley
4.
Int J Mol Sci ; 17(1)2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26784178

ABSTRACT

Monoamine oxidase (MAO) catalyzes the oxidative deamination of monoamines including dopamine (DA). MAO expression is elevated in Parkinson's disease (PD). An increase in MAO activity is closely related to age, and this may induce neuronal degeneration in the brain due to oxidative stress. MAO (and particularly monoamine oxidase B (MAO-B)) participates in the generation of reactive oxygen species (ROS), such as hydrogen peroxide that are toxic to dopaminergic cells and their surroundings. Although the polyphenol-rich aqueous walnut extract (JSE; an extract of Juglandis Semen) has been shown to have various beneficial bioactivities, no study has been dedicated to see if JSE is capable to protect dopaminergic neurons against neurotoxic insults in models of PD. In the present study we investigated the neuroprotective potential of JSE against 1-methyl-4-phenylpyridinium (MPP⁺)- or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicities in primary mesencephalic cells and in a mouse model of PD. Here we show that JSE treatment suppressed ROS and nitric oxide productions triggered by MPP⁺ in primary mesencephalic cells. JSE also inhibited depletion of striatal DA and its metabolites in vivo that resulted in significant improvement in PD-like movement impairment. Altogether our results indicate that JSE has neuroprotective effects in PD models and may have potential for the prevention or treatment of PD.


Subject(s)
Antioxidants/therapeutic use , MPTP Poisoning/drug therapy , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Animals , Antioxidants/pharmacology , Cells, Cultured , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Juglans/chemistry , Male , Mesencephalon/drug effects , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley
5.
J Alzheimers Dis ; 50(1): 189-200, 2016.
Article in English | MEDLINE | ID: mdl-26639976

ABSTRACT

The progressive aggregation of amyloid-ß protein (Aß) into senile plaques is a major pathological factor of Alzheimer's disease (AD) and is believed to result in memory impairment. We aimed to investigate the effect of an optimized combination of ginger and peony root (OCGP), a standardized herbal mixture of ginger and peony root, on Aß accumulation and memory impairment in amyloid-ß protein precursor (AßPP)/presenilin 1 (PS1) double-transgenic mice. In an in vitro thioflavin T fluorescence assay, 100 µg/ml OCGP inhibited Aß accumulation to the same extent as did 10 µM curcumin. Furthermore, AßPP/PS1 double-transgenic mice treated with OCGP (50 or 100 mg/kg/day given orally for 14 weeks) exhibited reduced Aß plaque accumulation in the hippocampus and lower levels of glial fibrillary acid protein and cyclooxygease-2 expression compared with vehicle-treated controls. These results suggest that OCGP may prevent memory impairment in AD by inhibiting Aß accumulation and inflammation in the brain.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Paeonia , Phytotherapy/methods , Plaque, Amyloid/pathology , Zingiber officinale , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cyclooxygenase 2/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Zingiber officinale/chemistry , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Mice , Mice, Transgenic , Mutation/genetics , Paeonia/chemistry , Plant Preparations/therapeutic use , Plaque, Amyloid/drug therapy , Presenilin-1/genetics
6.
Phytother Res ; 30(2): 208-13, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26563629

ABSTRACT

The flower bud of Zingiber mioga Roscoe, known as 'myoga' or Japanese ginger, has a pungent aroma and is commonly consumed as a spice, with pickles, or as a health supplement in Eastern Asia. Here, we evaluated the activity of myoga in the brain, focusing especially on nerve growth factor (NGF), which is believed to mediate synaptic plasticity, supporting learning and memory. In a rat primary hippocampal astrocyte culture system, treatment with myoga extract for 24 h significantly stimulated the production of NGF. In mice administered myoga extract for 14 days, 200 and 400 mg/kg/day treatment resulted in increased NGF levels in the hippocampus. Myoga extract treatment also regulated the phosphorylation of extracellular signal-regulated kinases and cAMP response element-binding protein in the mouse hippocampus, leading to increased synaptic plasticity. In addition, it significantly increased novel object recognition time and spontaneous alternation, indicating improvement in learning and memory. These results suggest that myoga helps regulate NGF and synaptic plasticity, increasing memory ability.


Subject(s)
Memory/drug effects , Nerve Growth Factor/metabolism , Neuronal Plasticity/drug effects , Plant Extracts/pharmacology , Signal Transduction , Zingiberaceae/chemistry , Animals , Astrocytes/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Flowers/chemistry , Hippocampus/drug effects , Learning/drug effects , Male , Mice , Mice, Inbred ICR , Phosphorylation , Primary Cell Culture , Rats , Rats, Sprague-Dawley
7.
Article in English | MEDLINE | ID: mdl-26539239

ABSTRACT

Kyung-Ok-Ko (KOK), a well-known traditional Korean medicinal formula, has long been used to invigorate the essential qi. This use of KOK may be associated with reproductive ability as a more modern concept. The protective effect of KOK was evaluated against deterioration of testicular function induced by heat exposure in male mice. Male fertility was disrupted by scrotal heat stress at 43°C for 5 weeks. KOK (0.25, 0.50, and 2.00 g/kg/day) was administered orally at 3 h after the stress. To evaluate the protective effect of KOK, body weight, testicular weight, sperm count, sperm motility, and histopathological changes in the testes were evaluated. KOK-treated mice significantly recovered their general health, as evidenced by body weight. KOK-treated mice also showed significantly higher testes weights, sperm counts, and sperm motility than did the heat stress group. KOK-treated mice significantly recovered the morphological appearance of the seminiferous tubules and seminiferous epithelium. Furthermore, KOK-treated mice significantly increased antioxidant enzyme activities and reduced the protein expressions of apoptosis in the testes. KOK significantly protects against heat-induced damage to testicular function in male mice by inhibiting oxidative stress and apoptosis, indicating that KOK may be an effective agent for treatment of heat-induced male infertility.

8.
BMC Complement Altern Med ; 15: 388, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26506837

ABSTRACT

BACKGROUND: Rhei Rhizoma (RR) has been widely used as laxative and processed to alter its therapeutic actions or reduce its side effects. In this study, we evaluated experimentally the clinical application guideline that RR should be alcohol-steamed seven times before being used in elderly patients, as described in Dongeuibogam, the most famous book on Korean traditional medicine. METHODS: Unprocessed RR (RR-U) was soaked in rice wine, steamed and then fully dried (RR-P1). The process was repeated four (RR-P4) or seven times (RR-P7). Reversed-phase high-performance liquid chromatography was used to determine the RR-U, RR-P1, RR-P4 and RR-P7 (RRs) constituents. To evaluate the effect of RRs on liver toxicity, human hepatoma cells (HepG2) were treated with RRs at 100 µg/mL for 4 h and then cell viabilities were measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method. To confirm the effects in vivo, 5-week-old male Sprague-Dawley rats were treated with RRs at 3 g/kg/day for 21 days. Body weight and serum biochemical parameters were measured and liver histology was assessed. RESULTS: The levels of sennosides decreased in processed RRs in an iteration-dependent manner, while the emodin level was unaffected. In HepG2 cells, cell viability was reduced with RR-U, while the toxicity decreased according to the number of processing cycles. The changes in body weight, relative liver weight and liver enzymes of RR-U-treated rats were reduced in processed RRs-treated rats. Histopathological analysis indicated swelling and cholestasis improved following seven times alcohol-steaming cycles. CONCLUSIONS: These results provide experimental evidence that RR-P7 almost completely reduces RR hepatotoxicity.


Subject(s)
Drug Compounding/methods , Drugs, Chinese Herbal , Liver/drug effects , Rheum , Analysis of Variance , Animals , Body Weight/drug effects , Cell Survival/drug effects , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/toxicity , Emodin/analysis , Hep G2 Cells , Humans , Male , Rats , Rheum/chemistry , Rheum/toxicity , Rhizome/chemistry , Rhizome/toxicity , Senna Extract/analysis , Sennosides
9.
Planta Med ; 81(18): 1697-704, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26366753

ABSTRACT

Epilepsy is a complex neurological disorder characterized by the repeated occurrence of electrical activity known as seizures. This activity induces increased intracellular calcium, which ultimately leads to neuronal damage. Houttuyniae Herba, the aerial part of Houttuynia cordata, has various pharmacological effects and is widely used as a traditional herb. In the present study, we evaluated the protective effects of Houttuyniae Herba water extract on kainic acid-induced neurotoxicity. Kainic acid directly acts on calcium release, resulting in seizure behavior, neuronal damage, and cognitive impairment. In a rat primary hippocampal culture system, Houttuyniae Herba water extract significantly protected neuronal cells from kainic acid toxicity. In a seizure model where mice received intracerebellar kainic acid injections, Houttuyniae Herba water extract treatment resulted in a lower seizure stage score, ameliorated cognitive impairment, protected neuronal cells against kainic acid-induced toxicity, and suppressed neuronal degeneration in the hippocampus. In addition, Houttuyniae Herba water extract regulated increases in the intracellular calcium level, its related downstream pathways (reactive oxygen species production and mitochondrial dysfunction), and calcium/calmodulin complex kinase type II immunoreactivity in the mouse hippocampus, which resulted from calcium influx stimulation induced by kainic acid. These results demonstrate the neuroprotective effects of Houttuyniae Herba water extract through inhibition of calcium generation in a kainic acid-induced epileptic model.


Subject(s)
Calcium/metabolism , Hippocampus/drug effects , Houttuynia/chemistry , Neuroprotective Agents/pharmacology , Seizures/prevention & control , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Hippocampus/metabolism , Kainic Acid/antagonists & inhibitors , Male , Mice , Mice, Inbred ICR , Neurons/drug effects , Neurotoxins/antagonists & inhibitors , Rats , Seizures/chemically induced
10.
J Ethnopharmacol ; 171: 196-204, 2015 Aug 02.
Article in English | MEDLINE | ID: mdl-26068423

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: A growing body of literature supports the concept that antiaging herbs may be potential candidates for use in treating age-related neurodegeneration, including Alzheimer׳s disease (AD). Mori Fructus is a well-known traditional herbal medicine, food, and dietary supplement. This study employed models of amyloid beta (Aß)-induced AD to investigate the protective effects of Mori Fructus ethanol extract (ME) against age-related disease and cognitive deficits. MATERIALS AND METHODS: To examine the protective effect of ME, we measured cell viability, cytotoxicity, and survival in rat primary hippocampal cultures. We performed behavioral tests and histological analysis in mouse models of AD induced by Aß(25-35) toxicity. To investigate the mechanism underlying the protective effect, we performed western blotting using antibodies against apoptotic markers as well as the nonphosphorylated and phosphorylated forms of Akt, glycogen synthase kinase-3ß (GSK-3ß), and tau. We also measured apoptotic marker fluorescence intensity. RESULTS: ME significantly attenuated Aß-induced cell damage, enhanced Akt and GSK-3ß phosphorylation, and reduced tau phosphorylation. ME reduced apoptotic markers that were activated by GSK-3ß, and reduced reactive oxygen species production. Further, ME decreased the B-cell lymphoma 2 (Bcl-2)/Bcl-2-associated X expression ratio, mitochondria depolarization, cytochrome c release from mitochondria, and caspase-3 activation. We confirmed that ME treatment improved cognitive impairment and neuronal cell death induced by Aß(25-35) toxicity in the mouse hippocampus via its antiapoptotic activity. CONCLUSIONS: These results indicate that ME protects cognition and neurons in AD-like models induced by Aß via reduction of tau phosphorylation and apoptosis through GSK-3ß inactivation.


Subject(s)
Alzheimer Disease/drug therapy , Morus , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Death/drug effects , Disease Models, Animal , Fruit , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/cytology , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred ICR , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments , Phytotherapy , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
11.
Article in English | MEDLINE | ID: mdl-25945108

ABSTRACT

Obesity has become a global health problem, contributing to various diseases including diabetes, hypertension, cancer, and dementia. Increasing evidence suggests that obesity can also cause neuronal damage, long-term memory loss, and cognitive impairment. The leaves and the fruits of Morus alba L., containing active phytochemicals, have been shown to possess antiobesity and hypolipidemic properties. Thus, in the present study, we assessed their effects on cognitive functioning in mice fed a high-fat diet by performing immunohistochemistry, using antibodies against c-Fos, synaptophysin, and postsynaptic density protein 95 and a behavioral test. C57BL/6 mice fed a high-fat diet for 21 weeks exhibited increased body weight, but mice coadministered an optimized Mori Folium and Mori Fructus extract mixture (2 : 1; MFE) for the final 12 weeks exhibited significant body weight loss. Additionally, obese mice exhibited not only reduced neural activity, but also decreased presynaptic and postsynaptic activities, while MFE-treated mice exhibited recovery of these activities. Finally, cognitive deficits induced by the high-fat diet were recovered by cotreatment with MFE in the novel object recognition test. Our findings suggest that the antiobesity effects of MFE resulted in recovery of the cognitive deficits induced by the high-fat diet by regulation of neural and synaptic activities.

12.
J Neuroinflammation ; 12: 102, 2015 May 23.
Article in English | MEDLINE | ID: mdl-26001832

ABSTRACT

BACKGROUND: Heat stress induces many pathophysiological responses and has a profound impact on brain structure. It has been demonstrated that exposure to high temperature induces cognitive impairment in experimental animals and humans. Although the effects of heat stress have long been studied, the mechanisms by which heat stress affects brain structure and cognition not well understood. METHODS: In our longitudinal study of mice exposed to heat over 7, 14, or 42 days, we found that heat stress time dependently impaired cognitive function as determined by Y-maze, passive avoidance, and novel object recognition tests. To elucidate the histological mechanism by which thermal stress inhibited cognitive abilities, we examined heat stress-induced inflammation in the hippocampus. RESULTS: In mice subjected to heat exposure, we found: 1) an increased number of glial fibrillary acid protein (GFAP)- and macrophage-1 antigen (Mac-1)-positive cells, 2) up-regulated nuclear factor (NF)-κB, a master regulator of inflammation, and 3) marked increases in cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and cytokine interleukin (IL)-1ß and tumor necrosis factor (TNF)-α in the mouse hippocampus. We also observed that neuronal and synaptic densities were degenerated significantly in hippocampal regions after heat exposure, as determined by histological analysis of neuronal nuclei (NeuN), postsynaptic density protein 95 (PSD-95), and synaptophysin expression. Moreover, in heat-exposed mice, we found that the number of cells positive for doublecortin (DCX), a marker of neurogenesis, was significantly decreased compared with control mice. Finally, anti-inflammatory agent minocycline inhibited the heat stress-induced cognitive deficits and astogliosis in mice. CONCLUSIONS: Together, these findings suggest that heat stress can lead to activation of glial cells and induction of inflammatory molecules in the hippocampus, which may act as causative factors for memory loss, neuronal death, and impaired adult neurogenesis.


Subject(s)
Heat Stress Disorders/complications , Inflammation/etiology , Inflammation/physiopathology , Memory Disorders/etiology , Memory Disorders/physiopathology , Neuritis/etiology , Neuritis/physiopathology , Animals , Cognition Disorders , Cyclooxygenase 2/metabolism , Disease Models, Animal , Doublecortin Protein , Hippocampus/pathology , Hippocampus/physiopathology , Hot Temperature/adverse effects , Inflammation/metabolism , Longitudinal Studies , Male , Memory Disorders/metabolism , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/physiopathology , Neurogenesis/physiology , Nitric Oxide Synthase Type II/metabolism
13.
J Nutr Biochem ; 25(10): 1058-65, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25049196

ABSTRACT

Ginger (the rhizome of Zingiber officinale Roscoe) has been used worldwide for many centuries in cooking and for treatment of several diseases. The main pharmacological properties of ginger include anti-inflammatory, antihyperglycemic, antiarthritic, antiemetic and neuroprotective actions. Recent studies demonstrated that ginger significantly enhances cognitive function in various cognitive disorders as well as in healthy brain. However, the biochemical mechanisms underlying the ginger-mediated enhancement of cognition have not yet been studied in normal or diseased brain. In the present study, we assessed the memory-enhancing effects of dried ginger extract (GE) in a model of scopolamine-induced memory deficits and in normal animals by performing a novel object recognition test. We found that GE administration significantly improved the ability of mice to recognize novel objects, indicating improvements in learning and memory. Furthermore, to elucidate the mechanisms of GE-mediated cognitive enhancement, we focused on nerve growth factor (NGF)-induced signaling pathways. NGF enzyme-linked immunosorbent assay analysis revealed that GE administration led to elevated NGF levels in both the mouse hippocampus and rat glioma C6 cells. GE administration also resulted in phosphorylation of extracellular-signal-regulated kinase (ERK) and cyclic AMP response element-binding protein (CREB), as revealed by Western blotting analysis. Neutralization of NGF with a specific NGF antibody inhibited GE-triggered activation of ERK and CREB in the hippocampus. Also, GE treatment significantly increased pre- and postsynaptic markers, synaptophysin and PSD-95, which are related to synapse formation in the brain. These data suggest that GE has a synaptogenic effect via NGF-induced ERK/CREB activation, resulting in memory enhancement.


Subject(s)
Cognition , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/drug effects , Nerve Growth Factor/metabolism , Plant Extracts/pharmacology , Zingiber officinale/chemistry , Animals , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Hippocampus/metabolism , Male , Memory/drug effects , Mice , Mice, Inbred ICR , Nerve Growth Factor/genetics , Neurogenesis/drug effects , Phosphorylation , Rats , Signal Transduction
14.
Biomol Ther (Seoul) ; 22(3): 176-83, 2014 May.
Article in English | MEDLINE | ID: mdl-25009697

ABSTRACT

Cognitive impairment is a result of dementia of diverse causes, such as cholinergic dysfunction and Alzheimer's disease (AD). Houttuynia cordata Thunb. (Saururaceae) has long been used as a traditional herbal medicine. It has biological activities including protective effects against amyloid beta (Aß) toxicity, via regulation of calcium homeostasis, in rat hippocampal cells. To extend previous reports, we investigated the effects of water extracts of H. cordata herb (HCW) on tauopathies, also involving calcium influx. We then confirmed the effects of HCW in improving memory impairment and neuronal damage in mice with Aß-induced neurotoxicity. We also investigated the effects of HCW against scopolamine-induced cholinergic dysfunction in mice. In primary neuronal cells, HCW inhibited the phosphorylation of tau by regulating p25/p35 expression in Aß-induced neurotoxicity. In mice with Aß-induced neurotoxicity, HCW improved cognitive impairment, as assessed with behavioral tasks, such as novel object recognition, Y-maze, and passive avoidance tasks. HCW also inhibited the degeneration of neurons in the CA3 region of the hippocampus in Aß-induced neurotoxicity. Moreover, HCW, which had an IC50 value of 79.7 µg/ml for acetylcholinesterase inhibition, ameliorated scopolamine-induced cognitive impairment significantly in Y-maze and passive avoidance tasks. These results indicate that HCW improved cognitive impairment, due to cholinergic dysfunction, with inhibitory effects against tauopathies and cholinergic antagonists, suggesting that HCW may be an interesting candidate to investigate for the treatment of AD.

15.
J Med Food ; 17(8): 862-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24963872

ABSTRACT

Contact dermatitis (CD) is a pattern of inflammatory responses in the skin that occurs through contact with external factors. The clinical picture is a polymorphic pattern of skin inflammation characterized by a wide range of clinical features, including itching, redness, scaling, and erythema. Coriandrum sativum L. (CS), commonly known as coriander, is a member of the Apiaceae family and is cultivated throughout the world for its nutritional and culinary values. Linoleic acid and linolenic acid in CS have various pharmacological activities. However, no study of the inhibitory effects of CS on CD has been reported. In this study, we demonstrated the protective effect of CS against 2,4-dinitrochlorobenzene-induced CD-like skin lesions. CS, at doses of 0.5-1%, applied to the dorsal skin inhibited the development of CD-like skin lesions. Moreover, the Th2-mediated inflammatory cytokines, immunoglobulin E, tumor necrosis factor-α, interferon-γ, interleukin (IL)-1, IL-4, and IL-13, were significantly reduced. In addition, CS increased the levels of total glutathione and heme oxygenase-1 protein. Thus, CS can inhibit the development of CD-like skin lesions in mice by regulating immune mediators and may be an effective alternative therapy for contact diseases.


Subject(s)
Coriandrum/chemistry , Dermatitis, Contact/drug therapy , Dinitrochlorobenzene/toxicity , Plant Extracts/administration & dosage , Animals , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Humans , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Male , Mice , Mice, Inbred ICR , Skin/drug effects , Skin/immunology , Skin/pathology
16.
Can J Physiol Pharmacol ; 92(6): 429-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24820962

ABSTRACT

Microglia-mediated inflammation is a major pathological mechanism contributing to Alzheimer's disease (AD), and has been proposed as a potential therapeutic target. Chunghyuldan (CHD; Qingxue-dan in Chinese and Daio-Orengedokuto in Japanese) possesses wide-ranging biological effects, including anti-hyperlipidemic, anti-stroke, anti-inflammatory, and antioxidant activities that could affect neurological functions. In this study, we examined the effects of CHD in in-vitro and in-vivo models of AD induced by the oligomeric form of amyloid-beta (Aß oligomer), which acts directly on microglia-mediated neuroinflammation to result in neuronal damage and cognitive impairment. CHD at 0.1-100 µg·mL(-1) significantly protected PC12 cells and rat primary hippocampal cells from Aß oligomer1-42 toxicity. In addition, CHD at 1-10 µg·mL(-1) inhibited Aß oligomer1-42 induced production of nitric oxide, tumor necrosis factor-α, and interleukin-1ß in microglial cells. In an in-vivo AD model, administration of CHD (50 mg·(kg body mass)(-1), for 5 days, per oral) inhibited the activation of astrocytes and microglia in the dentate gyrus and neuronal damage in the CA1 of the ipsilateral hippocampus. Moreover, CHD ameliorated cognitive impairment induced by Aß oligomer1-42 toxicity. These results demonstrate the neuroprotective effects of CHD through inhibition of microglia-mediated neuroinflammation in in-vitro and in-vivo AD-like models induced by Aß oligomer1-42 toxicity.


Subject(s)
Amyloid beta-Peptides/toxicity , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peptide Fragments/toxicity , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Animals , Avoidance Learning/drug effects , Cell Survival/drug effects , Cells, Cultured , Cognition/drug effects , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/prevention & control , Interleukin-1beta/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Primary Cell Culture , Rats , Tumor Necrosis Factor-alpha/metabolism
17.
Biochem Biophys Res Commun ; 449(1): 8-13, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24796668

ABSTRACT

Recently, increased attention has been directed towards medicinal extracts as potential new drug candidates for dementia. Ginger has long been used as an important ingredient in cooking and traditional herbal medicine. In particular, ginger has been known to have disease-modifying effects in Alzheimer's disease (AD). However, there is no evidence of which constituents of ginger exhibit therapeutic effects against AD. A growing number of experimental studies suggest that 6-shogaol, a bioactive component of ginger, may play an important role as a memory-enhancing and anti-oxidant agent against neurological diseases. 6-Shogaol has also recently been shown to have anti-neuroinflammatory effects in lipopolysaccharide (LPS)-treated astrocytes and animal models of Parkinson's disease, LPS-induced inflammation and transient global ischemia. However, it is still unknown whether 6-shogaol has anti-inflammatory effects against oligomeric forms of the Aß (AßO) in animal brains. Furthermore, the effects of 6-shogaol against memory impairment in dementia models are also yet to be investigated. In this study, we found that administration of 6-shogaol significantly reduced microgliosis and astrogliosis in intrahippocampal AßO-injected mice, ameliorated AßO and scopolamine-induced memory impairment, and elevated NGF levels and pre- and post-synaptic marker in the hippocampus. All these results suggest that 6-shogaol may play a role in inhibiting glial cell activation and reducing memory impairment in animal models of dementia.


Subject(s)
Catechols/administration & dosage , Cognition Disorders/drug therapy , Dementia/drug therapy , Encephalitis/drug therapy , Animals , Cognition/drug effects , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Dementia/complications , Dementia/physiopathology , Dose-Response Relationship, Drug , Encephalitis/complications , Encephalitis/physiopathology , Zingiber officinale/chemistry , Male , Mice , Mice, Inbred ICR , Neuroprotective Agents/administration & dosage , Plant Extracts/administration & dosage , Treatment Outcome
18.
Food Chem Toxicol ; 65: 293-300, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24389454

ABSTRACT

Parkinson's disease (PD) is generally characterized by the progressive loss of dopaminergic neurons projecting from the substantia nigra pars compacta (SNpc) to the striatum that results in movement dysfunction, but also entails mitochondrial dysfunction. The purpose of this study is to evaluate the protective effects of Moutan Cortex Radicis (MCE, Moutan peony) on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD-like symptoms and to elucidate the underlying mechanisms of action, with a focus on mitochondrial function. In a rat primary mesencephalic culture system, MCE significantly protected dopaminergic neurons from the neurotoxic effects of 1-methyl-4-phenylpyridinium (MPP(+)), an active form of MPTP. Additionally, in a subacute mouse model of MPTP-induced PD, MCE resulted in enhanced recovery from PD-like motor symptoms, including increased locomotor activity and reduced bradykinesia. MCE increased dopamine availability and protected against MPTP-induced dopaminergic neuronal damage. Moreover, MCE inhibited MPTP-induced mitochondrial dysfunction and resulted in increased expression of phosphorylated Akt, ND9, mitochondrial transcription factor A, and H2AX in the SNpc. Mitochondria-mediated apoptosis was also inhibited, via the regulation of B-cell lymphoma family proteins and the inhibition of cytochrome C release and caspase-3 activation. These results indicate that MCE has neuroprotective effects in PD models and may be useful for preventing or treating PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Neuroprotective Agents/pharmacology , Paeonia/chemistry , Parkinsonian Disorders/chemically induced , Plant Extracts/pharmacology , Plant Roots/chemistry , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy , Rats , Rats, Sprague-Dawley
19.
Neurotoxicology ; 40: 23-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24189446

ABSTRACT

Recent studies on Alzheimer's disease (AD) have focused on soluble oligomeric forms of amyloid-beta (Aß oligomer, AßO) that are directly associated with AD-related pathologies, such as cognitive decline, neurodegeneration, and neuroinflammation. Donepezil is a well-known anti-dementia agent that increases acetylcholine levels through inhibition of acetylcholinesterase. However, a growing body of experimental and clinical studies indicates that donepezil may also provide neuroprotective and disease-modifying effects in AD. Additionally, donepezil has recently been demonstrated to have anti-inflammatory effects against lipopolysaccharides and tau pathology. However, it remains unknown whether donepezil has anti-inflammatory effects against AßO in cultured microglial cells and the brain in animals. Further, the effects of donepezil against AßO-mediated neuronal death, astrogliosis, and memory impairment have also not yet been investigated. Thus, in the present study, we examined the anti-inflammatory effect of donepezil against AßO and its neuroinflammatory mechanisms. Donepezil significantly attenuated the release of inflammatory mediators (prostaglandin E2, interleukin-1 beta, tumor necrosis factor-α, and nitric oxide) from microglia. Donepezil also decreased AßO-induced up-regulation of inducible nitric oxide synthase and cyclooxygenase-2 protein and phosphorylation of p38 mitogen-activated protein kinase as well as translocation of nuclear factor-kappa B. We next showed that donepezil suppresses activated microglia-mediated toxicity in primary hippocampal cells using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. In intrahippocampal AßO-injected mice, donepezil significantly inhibited microgliosis and astrogliosis. Furthermore, behavioral tests revealed that donepezil (2 mg/kg/day, 5 days, p.o.) significantly ameliorated AßO-induced memory impairment. These results suggest that donepezil directly inhibits microglial activation induced by AßO through blocking MAPK and NF-κB signaling and, in part, contributing to the amelioration of neurodegeneration and memory impairment.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents/pharmacology , Indans/pharmacology , Microglia/drug effects , Microglia/metabolism , Piperidines/pharmacology , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Donepezil , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation Mediators/antagonists & inhibitors , Male , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Mice , Mice, Inbred ICR , Rats, Sprague-Dawley , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Article in English | MEDLINE | ID: mdl-24204389

ABSTRACT

Ampelopsis Radix, the root of Ampelopsis japonica (Thunb.) Makino (Vitaceae), is a herbal medicine which has been widely used in East Asia. The present study was done to explore whether the standardized extract of Ampelopsis Radix (AJW) protects dopaminergic neurons via antioxidant mechanisms in Parkinson's disease (PD) models. The effects of AJW on primary mesencephalic cultures stressed with 1-methyl-4-phenylpyridinium were investigated using tyrosine hydroxylase (TH) immunohistochemistry and reactive oxygen species measurement. The eliminative effects of AJW on the 2,2-diphenyl-1-picrylhydrazyl and 2,2'-azino-bis-(3-ethylbenzthiazoline-6-sulphonic acid) radicals were explored using colorimetric methods. The effects of AJW on the mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) were determined by pole test as well as TH and 8-hydroxydeoxyguanosine immunohistochemistry. AJW protected dopaminergic neurons by inhibiting reactive oxygen species generation in vitro. Moreover, AJW showed potent radical scavenging activities in vitro. In the mouse PD model, AJW protected the dopaminergic neurons in the brain, leading to motor improvements. AJW inhibited the MPTP-evoked accumulation of 8-hydroxydeoxyguanosine in the brain. These data suggest that AJW has neuroprotective effects with antioxidant mechanisms in PD models.

SELECTION OF CITATIONS
SEARCH DETAIL
...