Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37100105

ABSTRACT

Globally around 24 million elderly population are dealing with dementia, and this pathological characteristic is commonly seen in people suffering from Alzheimer's disease (AD). Despite having multiple treatment options that can mitigate AD symptoms, there is an imperative call to advance our understanding of the disease pathogenesis to unfold disease-modifying treatments/therapies. To explore the driving mechanisms of AD development, we stretch out further to study time-dependant changes after Okadaic acid (OKA)-induced AD-like conditions in zebrafish. We evaluated the pharmacodynamics of OKA at two-time points, i.e., after 4-days and 10-days exposure to zebrafish. T-Maze was utilized to observe the learning and cognitive behaviour, and inflammatory gene expressions such as 5-Lox, Gfap, Actin, APP, and Mapt were performed in zebrafish brains. To scoop everything out from the brain tissue, protein profiling was performed using LCMS/MS. Both time course OKA-induced AD models have shown significant memory impairment, as evident from T-Maze. Gene expression studies of both groups have reported an overexpression of 5-Lox, GFAP, Actin, APP, and OKA 10D group has shown remarkable upregulation of Mapt in zebrafish brains. In the case of protein expression, the heatmap suggested an important role of some common proteins identified in both groups, which can be explored further to investigate their mechanism in OKA-induced AD pathology. Presently, the preclinical models available to understand AD-like conditions are not completely understood. Hence, utilizing OKA in the zebrafish model can be of great importance in understanding the pathology of AD progression and as a screening tool for drug discovery.


Subject(s)
Alzheimer Disease , Aged , Animals , Humans , Alzheimer Disease/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Zebrafish/metabolism , Proteomics , Actins/metabolism , Brain/metabolism , Okadaic Acid/adverse effects , Okadaic Acid/metabolism , Genomics , Disease Models, Animal
2.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069531

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most common neurodegenerative disease, and it manifests as progressive memory loss and cognitive decline. However, there are no effective therapies for AD, which is an urgent problem to solve. Evodiamine, one of the main bioactive ingredients of Evodia rutaecarpa, has been reported to ameliorate blood-brain barrier (BBB) permeability and improve cognitive impairment in ischemia and AD mouse models. However, whether evodiamine alleviates tauopathy remains unclear. This study aimed to examine whether evodiamine ameliorates tau phosphorylation and cognitive deficits in AD models. METHODS: A protein phosphatase 2A inhibitor, okadaic acid (OA), was used to induce tau phosphorylation to mimic AD-like models in neuronal cells. Protein expression and cell apoptosis were detected using Western blotting and flow cytometry, respectively. Spatial memory/cognition was assessed using water maze, passive avoidance tests, and magnetic resonance imaging assay in OA-induced mice models, and brain slices were evaluated further by immunohistochemistry. RESULTS: The results showed that evodiamine significantly reduced the expression of phosphor-tau, and further decreased tau aggregation and neuronal cell death in response to OA treatment. This inhibition was found to be via the inhibition of glycogen synthase kinase 3ß, cyclin-dependent kinase 5, and mitogen-activated protein kinase pathways. In vivo results indicated that evodiamine treatment ameliorated learning and memory impairments in mice, whereas Western blotting and immunohistochemical analysis of the mouse brain also confirmed the neuroprotective effects of evodiamine. CONCLUSIONS: Evodiamine can decrease the neurotoxicity of tau aggregation and exhibit a neuroprotective effect. Our results demonstrate that evodiamine has a therapeutic potential for AD treatment.


Subject(s)
Quinazolines/pharmacology , Tauopathies/drug therapy , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Apoptosis/drug effects , Brain/metabolism , Cell Line , Cognition/drug effects , Cognition/physiology , Cognition Disorders/metabolism , Disease Models, Animal , Humans , Male , Maze Learning/drug effects , Mice , Mice, Inbred ICR , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Okadaic Acid/adverse effects , Okadaic Acid/pharmacology , Phosphorylation , Quinazolines/metabolism , Spatial Memory/drug effects , tau Proteins/drug effects , tau Proteins/metabolism
3.
Exp Anim ; 70(3): 412-418, 2021 Aug 06.
Article in English | MEDLINE | ID: mdl-33952794

ABSTRACT

The mouse bioassay for diarrhetic shellfish poisoning (DSP) toxins had been used as the official method in Japan and also used in the world. In this study, hypothermia, one of the symptoms observed in mice after inoculation with DSP toxins, were characterized. Lethal and sublethal doses of okadaic acid (OA), a representative component of DSP toxins, were inoculated intraperitoneally into mice. Body-temperature changes over time were measured by an electronic thermometer or monitored by an infrared camera. Drastic hypothermia (<30°C in some mice) was observed in a few hours after administration of a lethal dose of OA. Dose-dependency was clearly seen between doses of OA inoculated and body-temperature decrease. Drastic hypothermia was also detected by using an infrared camera. These results suggest that hypothermia could be used as an index for the humane endpoint in experimental animal toxicological studies.


Subject(s)
Hypothermia/chemically induced , Marine Toxins/adverse effects , Okadaic Acid/adverse effects , Shellfish Poisoning/diagnosis , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Mice , Mice, Inbred ICR , Specific Pathogen-Free Organisms
4.
J Cancer Res Clin Oncol ; 144(12): 2339-2349, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30341686

ABSTRACT

PURPOSE: The okadaic acid class of tumor promoters, which are inhibitors of protein phosphatases 1 and 2A (PP1 and PP2A), induced tumor promotion in mouse skin, rat glandular stomach, and rat liver. Endogenous protein inhibitors of PP2A, SET and CIP2A, were up-regulated in various human cancers, so it is vital to review the essential mechanisms of tumor promotion by the okadaic acid class compounds, together with cancer progression by SET and CIP2A in humans. RESULTS AND DISCUSSION: The first part of this review introduces the okadaic acid class compounds and the mechanism of tumor promotion: (1) inhibition of PP1 and PP2A activities of the okadaic acid class compounds; (2) some topics of tumor promotion; (3) TNF-α gene expression as a central mediator in tumor promotion; (4) exposure to the okadaic acid class of tumor promoters in relation to human cancer. The second part emphasizes the overexpression of SET and CIP2A in cancer progression, and the anticancer activity of SET antagonists as follows: (5) isolation and characterization of SET; (6) isolation and characterization of CIP2A; (7) progression of leukemia with SET; (8) progression of breast cancer with SET and CIP2A; (9) progression of lung cancer with SET; (10) anti-carcinogenic effects of SET antagonists OP449 and FTY720; and also (11) TNF-α-inducing protein of Helicobacter pylori, which is a clinical example of the okadaic acid pathway. CONCLUSIONS: The overexpression of endogenous protein inhibitors of PP2A, SET and CIP2A, is tightly linked to the progression of various human cancers, as well as Alzheimer's disease.


Subject(s)
Autoantigens/metabolism , Histone Chaperones/metabolism , Membrane Proteins/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Okadaic Acid/adverse effects , Protein Phosphatase 2/metabolism , Transcription Factors/metabolism , Animals , Autoantigens/genetics , Biomarkers, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins , Disease Progression , Environmental Exposure/adverse effects , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/chemistry , Gene Expression Regulation, Neoplastic , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/genetics , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Okadaic Acid/chemistry , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Mol Carcinog ; 55(11): 1573-1583, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26377854

ABSTRACT

Lysophosphatidic acid (LPA) signaling via LPA receptors (LPA1 to LPA6 ) mediates a variety of cellular functions, including cell motility. In the present study, we investigated the effects of LPA receptors on cell motile activity during multi-stage hepatocarcinogenesis in rat liver epithelial WB-F344 cells treated with chemical liver carcinogens. Cells were treated with a initiator (N-nitrosodiethylamine (DEN)) and three promoters (phenobarbital (PB), okadaic acid (OA) and clofibrate) every 24 h for 2 days. Cell motile activity was elevated by DEN, correlating with Lpar3 expression. PB, OA, and clofibrate elevated Lpar1 expression and inhibited cell motile activity. To evaluate the effects of long-term treatment on cell motility, cells were treated with DEN and/or PB for at least 6 months. Lpar3 expression and cell motile activity were significantly elevated by the long-term DEN treatment with or without further PB treatment. In contrast, long-term PB treatment with or without further DEN elevated Lpar1 expression and inhibited cell motility. When the synthesis of extracellular LPA was blocked by a potent ATX inhibitor S32826 before cell motility assay, the cell motility induced by DEN and PB was markedly suppressed. These results suggest that activation of the different LPA receptors may regulate the biological functions of cells treated with chemical carcinogens. © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinogens/pharmacology , Diethylnitrosamine/adverse effects , Epithelial Cells/drug effects , Liver Neoplasms, Experimental/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Animals , Cell Line , Cell Movement/drug effects , Clofibrate/adverse effects , Clofibrate/pharmacology , Epithelial Cells/cytology , Gene Expression Regulation/drug effects , Liver Neoplasms, Experimental/chemically induced , Okadaic Acid/adverse effects , Okadaic Acid/pharmacology , Phenobarbital/adverse effects , Phenobarbital/pharmacology , Rats , Rats, Inbred F344
6.
Nutrition ; 30(3): 337-42, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24484683

ABSTRACT

OBJECTIVE: Green tea polyphenols (GTPs) are now being considered possible protective agents in neurodegenerative diseases such as Alzheimer's disease (AD). Previous studies suggested that GTPs could inhibit amyloid fibril formation and protect neurons from toxicity induced by ß-amyloid. However, whether GTPs can ameliorate learning and memory impairments and also reduce tau hyperphosphorylation induced by okadaic acid (OA) in rats remains unclear. The aim of this study was to determine if GTPs have neuroprotection against OA-induced neurotoxicity. METHODS: In this work, rats were pretreated with GTPs by intragastric administration for 4 wk. Then OA was microinjected into the right dorsal hippocampus. Morris water maze tests were used to test the ethologic changes in all groups, and tau protein hyperphosphorylation was detected both in vivo and in vitro. RESULTS: The ethologic test indicated that the staying time and swimming distance in the target quadrant were significantly decreased after OA treatment, whereas rats pretreated with GTPs stayed longer in the target quadrant. Methyl thiazolyl tetrazolium assay and lactate dehydrogenase leakage showed that GTPs greatly ameliorated primary hippocampal neurons damage induced by OA. Furthermore, reduced hyperphosphorylated tau protein was detected with GTPs pretreatment. CONCLUSION: Taken together, our results suggest that GTPs have neuroprotection against OA-induced neurotoxicity.


Subject(s)
Learning/drug effects , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Okadaic Acid/adverse effects , Polyphenols/pharmacology , Tea/chemistry , Alzheimer Disease , Animals , Antioxidants/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Maze Learning , Memory Disorders/chemically induced , Okadaic Acid/administration & dosage , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , tau Proteins/genetics , tau Proteins/metabolism
7.
Neurobiol Aging ; 35(2): 322-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24011540

ABSTRACT

Superficial layers I to III of the human cerebral cortex are more vulnerable toward Aß peptides than deep layers V to VI in aging. Three models of layers were used to investigate this pattern of frailty. First, primary neurons from E14 and E17 embryonic murine cortices, corresponding respectively to future deep and superficial layers, were treated either with Aß(1-42), okadaic acid, or kainic acid. Second, whole E14 and E17 embryonic cortices, and third, in vitro separated deep and superficial layers of young and old C57BL/6J mice, were treated identically. We observed that E14 and E17 neurons in culture were prone to death after the Aß and particularly the kainic acid treatment. This was also the case for the superficial layers of the aged cortex, but not for the embryonic, the young cortex, and the deep layers of the aged cortex. Thus, the aged superficial layers appeared to be preferentially vulnerable against Aß and kainic acid. This pattern of vulnerability corresponds to enhanced accumulation of senile plaques in the superficial cortical layers with aging and Alzheimer's disease.


Subject(s)
Aging/pathology , Cell Survival/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neurons/drug effects , Neurons/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/adverse effects , Animals , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Humans , Kainic Acid/adverse effects , Mice , Mice, Inbred C57BL , Okadaic Acid/adverse effects , Peptide Fragments/adverse effects , Plaque, Amyloid/metabolism
8.
Eur J Pharmacol ; 715(1-3): 381-94, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23685326

ABSTRACT

Okadaic acid (OKA) has been observed to cause memory impairment in human subjects having seafood contaminated with dinoflagellate (Helicondria okadai). OKA induces tau hyperphosphorylation and oxidative stress leading to memory impairment as our previous study has shown. Curcumin a natural antioxidant has demonstrated neuroprotection in various models of neurodegeneration. However, the effect of curcumin has not been explored in OKA induced memory impairment. Therefore, present study evaluated the effect of curcumin on OKA (100ng, intracerebrally) induced memory impairment in male Swiss albino mice as evaluated in Morris water maze (MWM) and passive avoidance tests (PAT). OKA administration resulted in memory impairment with a decreased cerebral blood flow (CBF) (measured by laser doppler flowmetry), ATP level and increased mitochondrial (Ca(2+))i, neuroinflammation (increased TNF-α, IL-1ß, COX-2 and GFAP), oxidative-nitrosative stress, increased Caspase-9 and cholinergic dysfunction (decreased AChE activity/expression and α7 nicotinic acetylcholine receptor expression) in cerebral cortex and hippocampus of mice brain. Oral administration of curcumin (50mg/kg) for 13 days significantly improved memory function in both MWM and PAT along with brain energy metabolism, CBF and cholinergic function. It decreased mitochondrial (Ca(2+))i, and ameliorated neuroinflammation and oxidative-nitrostative stress in different brain regions of OKA treated mice. Curcumin also inhibited astrocyte activation as evidenced by decreased GFAP expression. This neuroprotective effect of curcumin is due to its potent anti-oxidant action thus confirming previous studies. Therefore, use of curcumin should be encouraged in people consuming sea food (contaminated with dinoflagellates) to prevent cognitive impairment.


Subject(s)
Curcumin/pharmacology , Memory/drug effects , Memory/physiology , Neuroprotective Agents/pharmacology , Okadaic Acid/adverse effects , Acetylcholine , Acetylcholinesterase/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Atrophy/prevention & control , Avoidance Learning/drug effects , Avoidance Learning/physiology , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain/physiology , Calcium/metabolism , Energy Metabolism/drug effects , Glutathione/metabolism , Male , Malondialdehyde/metabolism , Maze Learning/drug effects , Maze Learning/physiology , Mice , Microcirculation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Motor Activity/drug effects , Neurons/cytology , Neurons/drug effects , Organ Size/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects
9.
J Expo Sci Environ Epidemiol ; 23(4): 435-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22760440

ABSTRACT

Phycotoxins, secondary phytoplankton metabolites, are considered as an important food safety issue because their accumulation by shellfish may render them unfit for human consumption. However, the likely intakes of phycotoxins via shellfish consumption are almost unknown because both contamination and consumption data are very scarce. Thus, two 1-year surveys were conducted (through the same population: recreational shellfish harvesters and from the same geographical area) to assess: shellfish consumption and contamination by major toxins (domoic acid (DA) group, okadaic acid (OA) group and spirolides (SPXs)). Recreational shellfish harvesters had been targeted as an at-risk subpopulation because they consume more shellfish than general population and because they eat not only commercial shellfish species controlled by official authorities but also their own harvests of shellfish species may be in non-controlled areas and more over shellfish species non-considered in the official control species. Then, these two kinds of data were combined with deterministic and probabilistic approaches for both acute and chronic exposures, on considering the impact of shellfish species and cooking on phycotoxin levels. For acute risk, monitoring programs seem to be adequate for DAs, whereas OAs could be a matter of concern for high consumers (their acute intakes were up to ninefold the acute reference dose (ARfD)). About chronic risk, OAs are a matter of concern. The daily OAs intakes were close to the ARfD, which is, by definition, greater than the tolerable daily intake. Moreover, SPX contamination is low but regular, no (sub)chronic SPX toxicity data exist; but in case of (sub)chronic toxicity, SPX exposure should be considered.


Subject(s)
Diet/adverse effects , Marine Toxins/adverse effects , Shellfish/adverse effects , Adult , Aged , Aged, 80 and over , Diet/statistics & numerical data , Food Contamination/statistics & numerical data , France/epidemiology , Humans , Kainic Acid/adverse effects , Kainic Acid/analogs & derivatives , Middle Aged , Models, Statistical , Okadaic Acid/adverse effects , Phytoplankton , Young Adult
10.
Hum Mol Genet ; 22(3): 466-82, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23118350

ABSTRACT

Failures in neurotrophic support and signalling play key roles in Alzheimer's disease (AD) pathogenesis. We previously demonstrated that downregulation of the neurotrophin effector Kinase D interacting substrate (Kidins220) by excitotoxicity and cerebral ischaemia contributed to neuronal death. This downregulation, triggered through overactivation of N-methyl-D-aspartate receptors (NMDARs), involved proteolysis of Kidins220 by calpain and transcriptional inhibition. As excitotoxicity is at the basis of AD aetiology, we hypothesized that Kidins220 might also be downregulated in this disease. Unexpectedly, Kidins220 is augmented in necropsies from AD patients where it accumulates with hyperphosphorylated tau. This increase correlates with enhanced Kidins220 resistance to calpain processing but no higher gene transcription. Using AD brain necropsies, glycogen synthase kinase 3-ß (GSK3ß)-transgenic mice and cell models of AD-related neurodegeneration, we show that GSK3ß phosphorylation decreases Kidins220 susceptibility to calpain proteolysis, while protein phosphatase 1 (PP1) action has the opposite effect. As altered activities of GSK3ß and phosphatases are involved in tau aggregation and constitute hallmarks in AD, a GSK3ß/PP1 imbalance may also contribute to Kidins220 decreased clearance, accumulation and hampered neurotrophin signalling from early stages of the disease pathogenesis. These results encourage searches for mutations in Kidins220 gene and their possible associations to dementias. Finally, our data support a model where the effects of excitotoxicity drastically differ when occurring in cerebral ischaemia versus progressively sustained toxicity along AD progression. The striking differences in Kidins220 stability resulting from chronic versus acute brain damage may also have important implications for the therapeutic intervention of neurodegenerative disorders.


Subject(s)
Alzheimer Disease/metabolism , Calpain/metabolism , Glycogen Synthase Kinase 3/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Phosphatase 1/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Calpain/genetics , Cell Death , Cells, Cultured , Disease Models, Animal , Down-Regulation , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/pathology , Okadaic Acid/adverse effects , Phosphorylation , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/genetics , Proteolysis , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , tau Proteins/genetics
11.
J Proteomics ; 75(7): 2038-52, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22270013

ABSTRACT

Okadaic acid (OA) is a principal diarrhetic shellfish poisoning toxin produced by marine dinoflagellates. This study compared protein profiles of mice small intestines at four time points (0, 3, 6 and 24 h) after a single oral administration of 750 µg/kg OA, and identified the differentially expressed proteins using 2-D DIGE and MALDI-TOF-TOF mass spectrometry. The results showed that the toxin content of the intestines reached its peak 3h after oral administration and then decreased rapidly. OA remarkably inhibited the intestinal PP activity but it recovered to the normal levels within 6 to 24 h. Electron microscope revealed the collapse of the villous architecture and the intestinal microvilli fell off at 3 h, but were repaired within 24h. Notable damage to the intestinal ultrastructure was observed after oral administration. Comparison of the small intestine protein profiles at four time points revealed that 58 proteins were remarkably altered in abundance, and these proteins were involved in macromolecular metabolism, cytoskeleton reorganization, signal transduction, molecular chaperoning and oxidative stress, suggesting that OA toxicity in mouse intestines was complex and diverse, and that multiple proteins other than PP were involved in the diarrhetic process. Villin 1 and hnRNP F might be the key triggers inducing diarrhea in the mouse small intestines.


Subject(s)
Diarrhea/metabolism , Enzyme Inhibitors/adverse effects , Gene Expression Regulation/drug effects , Intestine, Small/metabolism , Okadaic Acid/adverse effects , Proteome/metabolism , Shellfish Poisoning/metabolism , Animals , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred ICR , Okadaic Acid/pharmacology , Proteomics/methods , Time Factors
12.
Zhong Xi Yi Jie He Xue Bao ; 8(10): 955-60, 2010 Oct.
Article in Chinese | MEDLINE | ID: mdl-20939986

ABSTRACT

OBJECTIVE: To investigate the effects of gensenoside Rg1 on expressions of phosphorylated tau protein (P-tau), protein phosphatase 2A (PP2A) and tau protein in Alzheimer's disease-like tau phosphorylation rat brain slices, and to explore the mechanisms of gensenoside Rg1 in inhibiting tau phosphorylation. METHODS: Brains of 5-week-old Wistar rats were cut into slices which were 400 µm thick. These brain slices were randomly divided into blank control group, untreated group, low-dose ginsenoside Rg1 group, medium-dose ginsenoside Rg1 group and high-dose ginsenoside Rg1 group, with 10 slices in each group. All brain slices were cultured with artificial cerebrospinal fluid (ACSF). And brain slices in the ginsenoside R1 groups were administered with ginsenoside Rg1 (60, 120 and 240 µmol/L respectively) in ACSF for 2 h firstly. After 2-hour culture, okadaic acid (OA) was administered into ACSF of the untreated group and the ginsenoside Rg1 groups separately for 3 h to induce tau phosphorylation to prepare AD models. The concentration of OA in each group was 1 µmol/L. There was no any intervention for the brain slices in the blank control group. Expressions of P-tau, PP2A and tau proteins in the brain slices were determined by immunohistochemical method, and the results were analyzed by image acquisition and analysis system. RESULTS: Compared with the blank control group, the expression of P-tau protein was significantly increased (P<0.01) and the expressions of tau and PP2A proteins were decreased (P<0.01, P<0.05) in the untreated group. Compared with the untreated group, the expression of P-tau was significantly decreased (P<0.01) and the expressions of tau and PP2A proteins were increased (P<0.01, P<0.05) in the ginsenoside Rg1 groups, especially in the high-dose ginsenoside Rg1 group. CONCLUSION: Ginsenoside Rg1 can promote dephosphorylation of P-tau by increasing the expression of PP2A in Alzheimer's disease-like tau phosphorylation rat brain slices, so as to inhibit tau phosphorylation.


Subject(s)
Brain/metabolism , Ginsenosides/pharmacology , tau Proteins/metabolism , Animals , In Vitro Techniques , Male , Okadaic Acid/adverse effects , Phosphorylation/drug effects , Protein Phosphatase 2/metabolism , Rats , Rats, Wistar
13.
Pathobiology ; 75(3): 186-94, 2008.
Article in English | MEDLINE | ID: mdl-18550916

ABSTRACT

OBJECTIVE: We investigated whether neural stem cells (NSC) with transgenic expression of human nerve growth factor (hNGF) transplanted into the brain could offer a therapeutic option for the treatment of Alzheimer's disease (AD). METHODS: We infused okadaic acid into rat lateral ventricles to establish a chronic AD animal model. In addition, NSC were stably transduced with hNGF and enhanced green fluorescent protein (eGFP) genes (NSC-hNGF-eGFP) by using a recombination adeno-associated virus serotype 2 (rAAV2) vector. These genetically modified stem cells were grafted into the cerebral cortex of AD rats. RESULTS: AD model rats showed significant damage in learning and memory function, with the formation of senile plaques and neurofibrillary tangles in the cerebral cortex. The transferred hNGF gene conferred stable and high levels of protein expression in NSC in vitro. Moreover, the NSC-hNGF-eGFP, but not the NSC, survived, integrating into the host brain and enhancing cognitive performance after transplantation. CONCLUSION: The injection of okadaic acid into rat lateral ventricles constitutes a promising animal model for investigating selective aspects of AD. rAAV2-mediated hNGF delivery can render long-term and stable transduction of hNGF in NSC. NSC-hNGF-eGFP transplantation may offer a viable therapeutic approach for treatment of AD.


Subject(s)
Alzheimer Disease/psychology , Alzheimer Disease/therapy , Nerve Growth Factor/genetics , Stem Cell Transplantation , Alzheimer Disease/chemically induced , Animals , Dependovirus , Disease Models, Animal , Fetus , Genetic Vectors , Humans , Learning , Male , Nerve Growth Factor/biosynthesis , Neurons/metabolism , Okadaic Acid/adverse effects , Rats , Recombinant Proteins/genetics , Stem Cells/metabolism , Transduction, Genetic
14.
Eur J Pharmacol ; 451(3): 237-43, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12242084

ABSTRACT

We demonstrated that exposure of cells to 50 nM okadaic acid for 2 h induced a reduction in cellular glutathione transferase, glutathione reductase and catalase activity. Likewise, this acid prompted an increase in lipid peroxidation. Treatment of cells with 10(-5) M melatonin or 0.5 microg/ml vitamin C prevented the effects of okadaic acid. These results indicate that okadaic acid induces an oxidative stress imbalance, while melatonin and vitamin C prevent the oxidative stress induced by okadaic acid. Likewise, these data indicate the great importance of oxidative stress in both this experimental model and in the development and course of neurodegenerative disease, especially Alzheimer's disease. They show that melatonin is much more efficient than vitamin C in reducing the extent of oxidative stress. This phenomenon was demonstrated by the smaller dose of melatonin needed to obtain effects similar to those obtained with vitamin C on lipid peroxidation and by the protective effect of melatonin on antioxidant enzyme activity.


Subject(s)
Alzheimer Disease/drug therapy , Ascorbic Acid/pharmacology , Lipid Peroxidation/drug effects , Melatonin/pharmacology , Neuroblastoma/enzymology , Okadaic Acid/antagonists & inhibitors , Oxidative Stress/drug effects , Alzheimer Disease/chemically induced , Animals , Catalase/drug effects , Catalase/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Mice , Neuroblastoma/drug therapy , Okadaic Acid/adverse effects , Tumor Cells, Cultured
15.
Toxicol In Vitro ; 16(4): 357-63, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12110273

ABSTRACT

We have studied the death response induced by yessotoxin (YTX) in cultured HeLa cells, and have compared it to that triggered by okadaic acid (OA) in the same experimental system. Sub-nanomolar concentrations of YTX were found to induce HeLa cell death after a 48-96-h incubation. YTX caused loss of intact poly(ADP-ribose)-polymerase (PARP) in HeLa cells, and detection of the 85kDa fragment, which is indicative of proteolytic attack by caspases. Measurements of caspase activities using extracts prepared from YTX-treated cells and substrates of the caspase-3/7 and caspase-2 isoforms, showed that the relative proteolysis of caspase-3/7 substrate was about eight-fold higher than that of caspase-2, the levels of which were about twice those measured with extracts from control cells. These findings were matched by Western blot analyses of caspase-2, -3 and -7 in HeLa cell extracts, which showed that the levels of pro-caspase-2 were not greatly affected by YTX treatment, whereas pro-caspase-3 and -7 were activated in YTX-treated cells. Taken together, these data complement others previously obtained with OA, and support the notion that caspase isoforms involved in cell death induced by OA and YTX are cell- and toxin-specific.


Subject(s)
Caspases/pharmacology , Cell Death , Enzyme Inhibitors/adverse effects , Ethers, Cyclic/adverse effects , Mollusk Venoms/adverse effects , Okadaic Acid/adverse effects , Oxocins/adverse effects , Blotting, Western , Dose-Response Relationship, Drug , HeLa Cells , Humans , Isomerism
16.
Gut ; 41(5): 642-5, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9414971

ABSTRACT

BACKGROUND: Antisecretory factor (AF) is a recently identified regulatory protein which inhibits the intestinal fluid secretion induced by cholera toxin. AIMS: To test the effect of AF on: (a) inflammation and hypersecretion induced by toxin A from Clostridium difficile; and (b) morphological changes and hypersecretion induced by okadaic acid (the blue mussel toxin) in rat intestinal mucosa. METHODS: Morphological changes and fluid accumulation were observed in intestinal loops challenged with 1 microgram of toxin A or 3 micrograms of okadaic acid administered before or after injection of 0.1 microgram of recombinant AF (rAF). RESULTS: The cytotoxic and inflammatory reaction caused by toxin A was abolished after treatment with rAF given either intraveneously or intraluminally prior to the toxin or one hour after the toxin. The intestinal fluid response induced by toxin A and okadaic acid was reduced 55-80% by rAF. However, the characteristic increase in goblet cells at the tips of villi in the okadaic acid treated mucosa was not inhibited by rAF. CONCLUSION: Results suggest that AF might be involved in protection against inflammation and in counteracting dehydration caused by enterotoxins. Both effects are probably mediated via the enteric nervous system.


Subject(s)
Antidiarrheals/therapeutic use , Bacterial Toxins/adverse effects , Clostridioides difficile , Enterotoxins/adverse effects , Intestines/drug effects , Neuropeptides/therapeutic use , Okadaic Acid/adverse effects , Animals , Enzyme Inhibitors/adverse effects , Injections, Intravenous , Intestinal Secretions/drug effects , Intestines/pathology , Ligation , Male , Phosphoprotein Phosphatases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...