Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Anat Sci Educ ; 17(2): 307-318, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37885424

ABSTRACT

Among the basic medical sciences, anatomy and physiology (anatomy & physiology) is a fundamental subject for students majoring in nursing. Due to its diversity and difficulty, nursing students experience stress when studying it. Previous graduates generally presented lower achievements in anatomy & physiology than in other nursing-related subjects in the National Council Licensure Examination-Registered Nurse, indicating that anatomy & physiology education requires improvement. Accordingly, we examined the impact of innovative teaching on students' motivation and performance when learning anatomy & physiology through a quasi-experimental pre-/post-test design. For innovative teaching, we used the novel interactive audio human organ model, followed by team-based learning. The participants were 200 lower-grade students in the nursing department of a junior college in Taiwan, divided into two groups receiving innovative teaching (experimental group) or traditional teaching (control group). Questionnaire surveys were administered, and the collected data were statistically analyzed. The innovative teaching in anatomy & physiology improved learning motivation, especially in terms of affect, executive volition, and learning performance. The essential components of learning motivation, such as value, expectation, affect, and executive volition, were positively correlated with the reaction levels of learning performance. Regarding the improvement in academic performance, the experimental group performed significantly better than the control group. The use of innovative teaching in class enhances students' learning motivation and learning performance when studying anatomy & physiology. Interactive teaching aids enhance the enjoyment of learning anatomy & physiology while facilitating in-depth exploration of the human organs and systems.


Subject(s)
Anatomy , Physiology , Students, Nursing , Humans , Motivation , Anatomy/education , Learning , Curriculum , Teaching , Physiology/education
2.
Biomedicines ; 12(1)2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38255143

ABSTRACT

Patients with diabetes mellitus can experience hyperglycemia, which affects brain function and produces cognitive impairment or neurodegeneration. Neuroinflammation is an important cause of cognitive dysfunction. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are antihyperglycemic agents that reportedly possess anti-inflammatory properties and may produce beneficial cognitive effects. We hypothesized that SGLT2 inhibitors alleviate hyperglycemia-related inflammation in brain immune cells. Cultured BV-2 microglia were exposed to high glucose (HG) in the absence or presence of SGLT2 inhibitors including canagliflozin (Cana), dapagliflozin (Dapa), empagliflozin (Empa), and ertugliflozin (Ertu). Afterward, we evaluated the cytotoxic and inflammatory responses by specific biochemical assays. Treatments with non-toxic Cana or Dapa, but not Empa or Ertu, inhibited proliferation without cell death. Only Cana rescued BV-2 microglia from HG-induced cytotoxicity, including apoptosis or autophagic degradation. None of SGLT2 inhibitors affected the HG-stimulated induction of stress proteins HO-1 and HSP70. Also, compared to the other three SGLT2 inhibitors, Cana was better at inhibiting HG-induced oxidative/inflammatory stress, as evidenced by its ability to repress proinflammatory factors (e.g., oxygen free radicals, iNOS, NLRP3, IL-1ß, and TNF-α) other than COX-2. Cana's action to alleviate HG insults was mediated not by altering SGLT2 protein expression, but by reducing HG-stimulated signaling activities of NFκB, JNK, p38, and PI3K/Akt pathways. Particularly, Cana imitated the effects of NFκB inhibitor on HG-induced iNOS and COX-2. Of the four SGLT2 inhibitors, Cana provided BV-2 microglia with the best protection against HG-induced inflammatory toxicity. Thus, Cana may help to reduce innate neuroimmune damage caused by hyperglycemia.

3.
Cancers (Basel) ; 14(5)2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35267664

ABSTRACT

The formation of new blood vessels in solid tumors is regulated by various endothelial trophic factors. We identified that CLEC11A, an extracellular C-type lectin, was over-expressed in lung cancer cell lines harboring mutated EGFR. CLEC11A expression was also frequently elevated in lung adenocarcinoma (LAC) tissues with EGFR mutation. CLEC11A-expressing H1299 cells formed larger tumors in nude mice than did the control cells. The CLEC11A-expressing tumors contained more CD31-positive cells, suggesting that they had a higher angiogenic activity. CLEC11A per se did not induce blood vessel formation, but enhanced angiogenesis triggered by VEGF-A or basic FGF in vivo. Additionally, the expression of small hairpin RNA against CLEC11A (shCLEC11A) in HCC827 LAC cells suppressed their tumorigenic ability. Purified CLEC11A exhibited a chemotactic ability, which is dependent on its integrin-binding RGD and LDT motifs, toward endothelial cells. This chemotactic activity was not affected by the presence of a VEGFR inhibitor. Conditioned medium produced by HCC827-shCLEC11A cells had diminished chemotactic ability toward endothelial cells. CLEC11A treatments increased the levels of active integrin ß1 that were not associated with activation of focal adhesion kinases in endothelial cells. Our results indicated that CLEC11A was a factor of angiogenic potential and was involved in lung cancer tumorigenesis.

4.
Int J Mol Sci ; 22(20)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34681608

ABSTRACT

Bisphenol A (BPA) is an environmental contaminant widely suspected to be a neurological toxicant. Epidemiological studies have demonstrated close links between BPA exposure, pathogenetic brain degeneration, and altered neurobehaviors, considering BPA a risk factor for cognitive dysfunction. However, the mechanisms of BPA resulting in neurodegeneration remain unclear. Herein, cultured N2a neurons were subjected to BPA treatment, and neurotoxicity was assessed using neuronal viability and differentiation assays. Signaling cascades related to cellular self-degradation were also evaluated. BPA decreased cell viability and axon outgrowth (e.g., by down-regulating MAP2 and GAP43), thus confirming its role as a neurotoxicant. BPA induced neurotoxicity by down-regulating Bcl-2 and initiating apoptosis and autophagy flux inhibition (featured by nuclear translocation of apoptosis-inducing factor (AIF), light chain 3B (LC3B) aggregation, and p62 accumulation). Both heme oxygenase (HO)-1 and AMP-activated protein kinase (AMPK) up-regulated/activated by BPA mediated the molecular signalings involved in apoptosis and autophagy. HO-1 inhibition or AIF silencing effectively reduced BPA-induced neuronal death. Although BPA elicited intracellular oxygen free radical production, ROS scavenger NAC exerted no effect against BPA insults. These results suggest that BPA induces N2a neurotoxicity characterized by AIF-dependent apoptosis and p62-related autophagy defects via HO-1 up-regulation and AMPK activation, thereby resulting in neuronal degeneration.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Benzhydryl Compounds/pharmacology , Heme Oxygenase-1/metabolism , Phenols/pharmacology , Animals , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/genetics , Caspase 3/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Mice , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sequestosome-1 Protein/metabolism
5.
Sci Rep ; 9(1): 840, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30696869

ABSTRACT

Diabetes mellitus is associated with an increased risk of Alzheimer's dementia and cognitive decline. The cause of neurodegeneration in chronic diabetic patients remains unclear. Changes in brain microglial activity due to glycemic fluctuations may be an etiological factor. Here, we examined the impact of acute ambient glucose fluctuations on BV-2 microglial activity. Biochemical parameters were assayed and showed that the shift from normal glucose (NG; 5.5 mM) to high glucose (HG; 25 mM) promoted cell growth and induced oxidative/inflammatory stress and microglial activation, as evidenced by increased MTT reduction, elevated pro-inflammatory factor secretion (i.e., TNF-α and oxygen free radicals), and upregulated expression of stress/inflammatory proteins (i.e., HSP70, HO-1, iNOS, and COX-2). Also, LPS-induced inflammation was enlarged by an NG-to-HG shift. In contrast, the HG-to-NG shift trapped microglia in a state of metabolic stress, which led to apoptosis and autophagy, as evidenced by decreased Bcl-2 and increased cleaved caspase-3, TUNEL staining, and LC3B-II expression. These stress episodes were primarily mediated through MAPKs, PI3K/Akt, and NF-κB cascades. Our study demonstrates that acute glucose fluctuation forms the stress that alters microglial activity (e.g., inflammatory activation or self-degradation), representing a novel pathogenic mechanism for the continued deterioration of neurological function in diabetic patients.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus/pathology , Glucose/metabolism , Inflammation/metabolism , Microglia/metabolism , Animals , Caspase 3/metabolism , Cell Line , Cyclooxygenase 2/metabolism , Glucose Transporter Type 2/metabolism , Lipopolysaccharides/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/physiology
6.
J Steroid Biochem Mol Biol ; 185: 200-211, 2019 01.
Article in English | MEDLINE | ID: mdl-30194976

ABSTRACT

Bisphenol A (BPA), an endocrine-disrupting chemical, is capable of producing reproductive toxicity. BPA results in mitochondrial DNA (mtDNA) deletion and mitochondrial dysfunction; however, the effect of BPA on the mitochondria of ovarian granulosa cells is not clear. Further, 1,25-dihydroxyvitamin D3 (1,25D3) may play a role in reproduction, because its receptor, VDR, contributes to the inhibition of oxidative stress and predominantly exists in the nuclei of granulosa cells. Hence, the role of 1,25D3 in BPA-mediated effects on mitochondrial function was examined in this study. Primary rat granulosa cells treated with BPA, 1,25D3, or both were subjected to molecular/biochemical assays to measure cell survival, mtDNA content, mtDNA deletion, superoxide dismutase activity, levels of proteins related to mitochondrial biogenesis, and mitochondrial function. We found that cell viability was dose-dependently reduced and reactive oxygen species (ROS) levels were increased by BPA treatment. BPA administration elevated Mn-superoxide dismutase (MnSOD) expression but negatively regulated total SOD activity. 1,25D3 treatment alone increased 17ß-estradiol secretion, ATP production, and cellular oxygen consumption. In cells treated with both agents, 1,25D3 enhanced BPA-induced MnSOD protein upregulation and blocked the BPA-mediated decline in total SOD activity. Furthermore, 1,25D3 attenuated BPA-mediated mtDNA deletion but showed no effect on BPA-induced increases in mtDNA content. Although BPA had no influence on the levels of peroxisome proliferator-activated receptor-γ coactivator-1 α, nuclear respiratory factor-1, mitochondrial transcription factor A, or cytochrome c oxidase subunit IV, 1,25D3 plus BPA markedly increased mitochondrial biogenesis-related protein expression via the PI3K-Akt pathway. Moreover, BPA-mediated negative regulation of cytochrome c oxidase subunit I levels and 17ß-estradiol secretion was attenuated by 1,25D3 pre-treatment. Our results suggest that 1,25D3 attenuates BPA-induced decreases in 17ß-estradiol and that treatment with 1,25D3 plus BPA regulates granulosa cell mitochondria by elevating mitochondrial biogenesis-related protein levels.


Subject(s)
Benzhydryl Compounds/toxicity , Calcitriol/pharmacology , Endocrine Disruptors/toxicity , Estradiol/metabolism , Granulosa Cells/metabolism , Mitochondria/pathology , Phenols/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , DNA, Mitochondrial/genetics , Electron Transport Complex IV/metabolism , Female , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Calcitriol/metabolism , Sequence Deletion/drug effects , Sequence Deletion/genetics , Superoxide Dismutase/metabolism
7.
Int J Mol Sci ; 18(8)2017 Aug 11.
Article in English | MEDLINE | ID: mdl-28800073

ABSTRACT

There have been striking associations of cardiovascular diseases (e.g., atherosclerosis) and hypercholesterolemia with increased risk of neurodegeneration including Alzheimer's disease (AD). Low-density lipoprotein (LDL), a cardiovascular risk factor, plays a crucial role in AD pathogenesis; further, L5, a human plasma LDL fraction with high electronegativity, may be a factor contributing to AD-type dementia. Although L5 contributing to atherosclerosis progression has been studied, its role in inducing neurodegeneration remains unclear. Here, PC12 cell culture was used for treatments with human LDLs (L1, L5, or oxLDL), and subsequently cell viability and nerve growth factor (NGF)-induced neuronal differentiation were assessed. We identified L5 as a neurotoxic LDL, as demonstrated by decreased cell viability in a time- and concentration-dependent manner. Contrarily, L1 had no such effect. L5 caused cell damage by inducing ATM/H2AX-associated DNA breakage as well as by activating apoptosis via lectin-like oxidized LDL receptor-1 (LOX-1) signaling to p53 and ensuring cleavage of caspase-3. Additionally, sublethal L5 long-termly inhibited neurite outgrowth in NGF-treated PC12 cells, as evidenced by downregulation of early growth response factor-1 and neurofilament-M. This inhibitory effect was mediated via an interaction between L5 and LOX-1 to suppress NGF-induced activation of PI3k/Akt cascade, but not NGF receptor TrkA and downstream MAPK pathways. Together, our data suggest that L5 creates a neurotoxic stress via LOX-1 in PC12 cells, thereby leading to impairment of viability and NGF-induced differentiation. Atherogenic L5 likely contributes to neurodegenerative disorders.


Subject(s)
Lipoproteins, LDL/metabolism , Neuronal Outgrowth , Scavenger Receptors, Class E/metabolism , Animals , Apoptosis , Cell Survival , MAP Kinase Signaling System , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/metabolism , PC12 Cells , Rats
8.
Sci Rep ; 7(1): 5241, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701747

ABSTRACT

Expression of neuroendocrine-associated phosphatase (NEAP, also named as dual specificity phosphatase 26, [DUSP26]) is restricted to neuroendocrine tissues. We found that NEAP, but not its phosphatase-defective mutant, suppressed nerve growth factor (NGF) receptor TrkA and fibroblast growth factor receptor 1 (FGFR1) activation in PC12 cells upon NGF stimulation. Conversely, suppressing NEAP expression by RNA interference enhanced TrkA and FGFR1 phosphorylation. NEAP was capable of de-phosphorylating TrkA and FGFR1 directly in vitro. NEAP-orthologous gene existed in zebrafish. Morpholino (MO) suppression of NEAP in zebrafish resulted in hyper-phosphorylation of TrkA and FGFR1 as well as abnormal body postures and small eyes. Differentiation of retina in zebrafishes with NEAP MO treatment was severely defective, so were cranial motor neurons. Taken together, our data indicated that NEAP/DUSP26 have a critical role in regulating TrkA and FGFR1 signaling as well as proper development of retina and neuronal system in zebrafish.


Subject(s)
Dual-Specificity Phosphatases/physiology , Embryo, Nonmammalian/cytology , Mitogen-Activated Protein Kinase Phosphatases/physiology , Motor Neuron Disease/pathology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, trkA/antagonists & inhibitors , Retinal Diseases/pathology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Cell Differentiation , Dual-Specificity Phosphatases/genetics , Embryo, Nonmammalian/metabolism , Mitogen-Activated Protein Kinase Phosphatases/genetics , Morpholinos/pharmacology , Motor Neuron Disease/genetics , Motor Neuron Disease/metabolism , PC12 Cells , Phosphorylation , Rats , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Retinal Diseases/genetics , Retinal Diseases/metabolism , Signal Transduction , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
9.
J Neurochem ; 142(2): 231-245, 2017 07.
Article in English | MEDLINE | ID: mdl-28444734

ABSTRACT

Atherogenic risk factors, such as hypercholesterolemia, are associated with increased risk of neurodegeneration, especially Alzheimer's dementia. Human plasma electronegative low-density lipoprotein [LDL(-)], especially L5, may serve as an important contributing factor. L5 promoting an inflammatory action in atherosclerosis has been extensively studied. However, the role of L5 in inducing neuroinflammation remains unknown. Here, we examined the impact of L5 on immune activation and cell viability in cultured BV-2 microglia. BV-2 cells treated with lipopolysaccharide or human LDLs (L1, L5, or oxLDL) were subjected to molecular/biochemical assays for measuring microglial activation, levels of inflammatory factors, and cell survival. A transwell BV-2/N2a co-culture was used to assess N2a cell viability following BV-2 cell exposure to L5. We found that L5 enables the activation of microglia and elicits an inflammatory response, as evidenced by increased oxygen/nitrogen free radicals (nitric oxide, reactive oxygen species, and peroxides), elevated tumor necrosis factor-α levels, decreased basal interleukin-10 levels, and augmented production of pro-inflammatory proteins (inducible nitric oxide synthase and cyclooxygenase-2). L5 also triggered BV-2 cell death primarily via apoptosis. These effects were markedly disrupted by the application of signaling pathway inhibitors, thus demonstrating that L5 interacts with Toll-like receptor 4 to modulate multiple pathways, including MAPKs, PI3K/Akt, and NF-κB. Decreased N2a cell viability in a transwell co-culture was mainly ascribed to L5-induced BV-2 cell activation. Together, our data suggest that L5 creates a neuroinflammatory stress via microglial Toll-like receptor 4, thereby leading to the death of BV-2 microglia and coexistent N2a cells. Atherogenic L5 possibly contributes to neuroinflammation-related neurodegeneration.


Subject(s)
Cyclooxygenase 2/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Microglia/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Cell Survival/drug effects , Coculture Techniques , Humans , Inflammation/metabolism , Interleukin-10/metabolism , Microglia/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
10.
Nitric Oxide ; 54: 38-50, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26891889

ABSTRACT

Oxidative/nitrosative stress contributes to the etiology of the neurological disorders, including ischemic stroke and chronic neurodegeneration. Neurotoxic modifications mediated by reactive oxygen species (ROS) or reactive nitrogen species (RNS) are closely associated with the destruction of key macromolecules and inactivation of antioxidant enzymes, which compromises antioxidant defenses. Approaches to expel ROS/RNS and alleviate toxic oxidative/nitrosative stress in neurons have not completely been defined. Here, we aimed to evaluate the efficacy of various antioxidants that serve as the neuroprotectors under a toxic stress created by ROS plus nitric oxide (NO). Sublytic concentrations of hydrogen peroxide (H2O2) plus NO donor S-nitroso-N-acetyl-D, L-penicillamine (SNAP) enabled to induce a toxic oxidative/nitrosative stress through activating both p38 MAPK and p53 cascades, and cause DNA damage and protein tyrosine nitration in primary neuronal cultures. After comparing six antioxidants, including superoxide dimutase (SOD), catalase, 2,2,6,6-tetramethyl-1-piperidinoxyl (TEMPO), N-acetylcysteine, dimethylthiourea, and uric acid, TEMPO was the superior antioxidant that comprehensively and efficaciously decreased H2O2 plus SNAP-evoked activation of stress cascades of p38 MAPK and p53, production of NO, ROS, and peroxynitrite, double-strand breaks of DNA, and nitration of protein tyrosine residues. SOD increased the peroxynitrite formation and was unable to reduce the level of protein nitration. All antioxidants tested, except SOD, effectively reduced neuronal damage and DNA breakage caused by the toxic H2O2/SNAP combination. In conclusion, these results suggest that TEMPO ensures excellent ROS/RNS clearance and stress-signaling inhibition, thus effectively rescuing neurons from ROS/H2O2 plus NO/SNAP-induced insult. This study reveals a potential strategy for nitroxide antioxidants as a therapeutic agent against oxidative/nitrosative neurotoxicity.


Subject(s)
Antioxidants/pharmacology , Hydrogen Peroxide/pharmacology , Neurons/drug effects , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Catalase/pharmacology , Cyclic N-Oxides/pharmacology , L-Lactate Dehydrogenase/metabolism , Neurons/metabolism , Nitric Oxide Donors/pharmacology , Nitrites/metabolism , Peroxynitrous Acid/metabolism , Rats , Rats, Sprague-Dawley , S-Nitroso-N-Acetylpenicillamine/pharmacology , Superoxide Dismutase/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Uric Acid/pharmacology
11.
Reprod Biol Endocrinol ; 12: 90, 2014 Sep 20.
Article in English | MEDLINE | ID: mdl-25239217

ABSTRACT

BACKGROUND: Aromatase converts testosterone into 17beta-estradiol in granulosa cells, and the converted 17beta-estradiol contributes to follicular maturation. Additionally, excessive testosterone inhibits aromatase activity, which can lead to concerns regarding polycystic ovary syndrome (PCOS). Generally, 1,25-dihydroxyvitamin D3 (1,25D3) supplements help to improve the symptoms of PCOS patients who exhibit low blood levels of 1,25D3. Therefore, this study investigated the interaction effects of 1,25D3 and testosterone on estrogenesis and intercellular connections in rat granulosa cells. METHODS: Primary cultures of granulosa cells were treated with testosterone or testosterone plus 1,25D3, or pre-treated with a calcium channel blocker or calcium chelator. Cell lysates were subjected to western blot analysis to determine protein and phosphorylation levels, and 17beta-estradiol secretion was examined using a radioimmunoassay technique. Cell viability was evaluated by MTT reduction assay. Connexin 43 (Cx43) mRNA and protein expression levels were assessed by qRT-PCR, western blot, and immunocytochemistry. RESULTS: Testosterone treatment (0.1 and 1 microg/mL) increased aromatase expression and 17beta-estradiol secretion, and the addition of 1,25D3 attenuated testosterone (1 microg/mL)-induced aromatase expression but improved testosterone-induced 17beta-estradiol secretion. Furthermore, testosterone-induced aromatase phosphotyrosine levels increased at 10 min, 30 min and 1 h, whereas 1,25D3 increased the longevity of the testosterone effect to 6 h and 24 h. Within 18-24 h of treatment, 1,25D3 markedly enhanced testosterone-induced 17beta-estradiol secretion. Additionally, pre-treatment with a calcium channel blocker nifedipine or an intracellular calcium chelator BAPTA-AM reduced 1,25D3 and testosterone-induced 17beta-estradiol secretion. Groups that underwent testosterone treatment exhibited significantly increased estradiol receptor beta expression levels, which were not affected by 1,25D3. Neither testosterone nor 1,25D3 altered 1,25D3 receptor expression. Finally, at high doses of testosterone, Cx43 protein expression was decreased in granulosa cells, and this effect was reversed by co-treatment with 1,25D3. CONCLUSIONS: These data suggest that 1,25D3 potentially increases testosterone-induced 17beta-estradiol secretion by regulating aromatase phosphotyrosine levels, and calcium increase is involved in both 1,25D3 and testosterone-induced 17beta-estradiol secretion. 1,25D3 reverses the inhibitory effect of testosterone on Cx43 expression in granulosa cells.


Subject(s)
Calcitriol/metabolism , Connexin 43/metabolism , Estradiol/metabolism , Gene Expression Regulation, Developmental , Granulosa Cells/metabolism , Testosterone/metabolism , Up-Regulation , Animals , Aromatase/chemistry , Aromatase/metabolism , Calcium Channel Blockers/pharmacology , Calcium Chelating Agents/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Connexin 43/agonists , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Down-Regulation/drug effects , Estradiol/agonists , Estradiol/chemistry , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Granulosa Cells/cytology , Granulosa Cells/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats, Sprague-Dawley , Receptors, Estradiol/agonists , Receptors, Estradiol/antagonists & inhibitors , Receptors, Estradiol/metabolism , Testosterone/agonists , Testosterone/antagonists & inhibitors , Up-Regulation/drug effects
12.
Toxicol Appl Pharmacol ; 265(2): 241-52, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23022510

ABSTRACT

Methamphetamine (METH) is a drug of abuse which causes neurotoxicity and increased risk of developing neurodegenerative diseases. We previously found that METH induces heme oxygenase (HO)-1 expression in neurons and glial cells, and this offers partial protection against METH toxicity. In this study, we investigated the effects of l-ascorbate (vitamin C, Vit. C) on METH toxicity and HO-1 expression in neuronal/glial cocultures. Cell viability and damage were evaluated by 3-(4,5-dimethylthianol-2-yl)-2,5 diphenyl tetrazolium bromide (MTT) reduction and lactate dehydrogenase (LDH) release, respectively. Neuronal and glial localization of HO-1 were identified by double immunofluorescence staining. Reactive oxygen species (ROS) production was measured using the fluorochrome 2',7'-dichlorofluorescin diacetate. HO-1 mRNA and protein expression were examined by RT-qPCR and Western blotting, respectively. Results show that Vit. C induced HO-1 mRNA and protein expressions in time- and concentration-dependent manners. Inhibition of p38 mitogen-activated protein kinase (MAPK) but not extracellular signal-regulated kinase (ERK) significantly blocked induction of HO-1 by Vit. C. HO-1 mRNA and protein expressions were significantly elevated by a combination of Vit. C and METH, compared to either Vit. C or METH alone. Pretreatment with Vit. C enhanced METH-induced HO-1 expression and attenuated METH-induced ROS production and neurotoxicity. Pharmacological inhibition of HO activity abolished suppressive effects of Vit. C on METH-induced ROS production and attenuated neurotoxicity. We conclude that induction of HO-1 expression contributes to the attenuation of METH-induced ROS production and neurotoxicity by Vit. C. We suggest that HO-1 induction by Vit. C may serve as a strategy to alleviate METH neurotoxicity.


Subject(s)
Ascorbic Acid/pharmacology , Cerebral Cortex/drug effects , Heme Oxygenase-1/biosynthesis , Methamphetamine/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cell Survival/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Coculture Techniques , Drug Interactions , Enzyme Induction/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Immunohistochemistry , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , RNA/chemistry , RNA/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
13.
J Biol Chem ; 286(12): 10177-84, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21262974

ABSTRACT

Vaccinia H1-related phosphatase (VHR) is classified as a dual specificity phosphatase. Unlike typical dual specificity phosphatases, VHR lacks the MAPK-binding domain and shows poor activity against MAPKs. We found that EGF receptor (EGFR) was a direct substrate of VHR and that overexpression of VHR down-regulated EGFR phosphorylation, particularly at Tyr-992 residue. Expression of VHR inhibited the activation of phospholipase Cγ and protein kinase C, both downstream effectors of Tyr-992 phosphorylation of EGFR. Decreasing VHR expression by RNA interference caused higher EGFR phosphorylation at Tyr-992. In addition to EGFR, VHR also directly dephosphorylated ErbB2. Consistent with these results, suppression of VHR augmented the foci formation ability of H1299 non-small cell lung cancer (NSCLC) cells, whereas overexpression of VHR suppressed cell growth in both two- and three-dimensional cultures. Expression of VHR also suppressed tumor formation in a mouse xenograft model. Furthermore, VHR expression was significantly lower in NSCLC tissues in comparison to that in normal lung tissues. Collectively, this study shows that down-regulation of VHR expression enhances the signaling of ErbB receptors and may be involved in NSCLC pathogenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Down-Regulation , Dual Specificity Phosphatase 3/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/enzymology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Dual Specificity Phosphatase 3/genetics , ErbB Receptors/genetics , Female , Gene Expression Regulation, Enzymologic/genetics , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Signal Transduction/genetics , Transplantation, Heterologous
14.
Dev Neurobiol ; 70(8): 604-21, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20506231

ABSTRACT

The A2A adenosine receptor (A2AR) is a G-protein-coupled receptor. We previously reported that the C terminus of the A2AR binds to translin-associated protein X (TRAX) and modulates nerve growth factor (NGF)-evoked neurite outgrowth in PC12 cells. Herein, we show that neuritogenesis of primary hippocampal neurons requires p53 because blockage of p53 suppressed neurite outgrowth. The impaired neuritogenesis caused by p53 blockage was rescued by activation of the A2AR (designated the A2A rescue effect) in a TRAX-dependent manner. Importantly, suppression of a TRAX-interacting protein (kinesin heavy chain member 2A, KIF2A) inhibited the A2A rescue effect, whereas overexpression of KIF2A caused a rescue effect. Expression of a KIF2A fragment (KIF2A514), which disturbed the interaction between KIF2A and TRAX, blocked the rescue effect. Transient colocalization of TRAX and KIF2A was detected in the nucleus of PC12 cells upon NGF treatment. These data suggest that functional interaction between KIF2A and TRAX is critical for the A2A rescue effect. Moreover, p53 blockage during NGF treatment prevented the redistribution of KIF2A from the nucleus to the cytoplasmic region. Expression of a nuclear-retained KIF2A variant (NLS-KIF2A) did not rescue the impaired neurite outgrowth as did the wild-type KIF2A. Therefore, redistribution of KIF2A to the cytoplasmic fraction is a prerequisite for neurite outgrowth. Collectively, we demonstrate that KIF2A functions downstream of p53 to mediate neuritogenesis of primary hippocampal neurons and PC12 cells. Stimulation of the A2AR rescued neuritogenesis impaired by p53 blockage via an interaction between TRAX and KIF2A.


Subject(s)
DNA-Binding Proteins/metabolism , Kinesins/metabolism , Neurites/physiology , Neurogenesis , Receptor, Adenosine A2A/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Hippocampus/cytology , Hippocampus/physiology , Humans , Mice , Mice, Knockout , Nerve Growth Factor/metabolism , Neurons/cytology , Neurons/physiology , PC12 Cells , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
15.
J Neurochem ; 107(6): 1544-55, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19014381

ABSTRACT

Neuroendocrine-associated phosphatase (NEAP), an atypical dual specificity phosphatase is preferentially expressed in neuroendocrine cells. In this study we found that NEAP, but not NEAP-(C152S) mutant, evidently reduced epidermal growth factor (EGF) receptor (EGFR) downstream signaling, and impaired cell growth in response to EGF stimulation in PC12 cells. These phenomena were associated with NEAP-mediated down-regulation of EGFR mRNA and protein. NEAP had no significant effect on ErbB2/3 expression and phosphorylation levels in response to heregulin, indicating that the negative effect of NEAP on EGFR was selective. We showed that NEAP suppressed EGFR expression via decreasing the EGFR promoter activity and this was mediated through down-regulations of the Akt pathway and Wilms' tumor gene product (WT1). Consistent with these results, expression of WT1 reversed the suppressive effect of NEAP on EGFR promoter activity. Additionally, NEAP knockdown by RNA interference enhanced EGFR protein expression and nerve growth factor-induced differentiation, and an EGFR-specific inhibitor could reverse the later event. Taken together, our study indicated that NEAP modulates PC12 differentiation via suppression of EGFR expression and signaling.


Subject(s)
Cell Differentiation/physiology , Down-Regulation/physiology , ErbB Receptors/metabolism , Phosphoric Monoester Hydrolases/metabolism , Animals , Cell Differentiation/drug effects , Down-Regulation/drug effects , Mutation , Nerve Growth Factor/pharmacology , Neurites/drug effects , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/genetics , RNA, Small Interfering/pharmacology , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , WT1 Proteins/metabolism
16.
J Neurochem ; 98(1): 89-101, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16805799

ABSTRACT

The biochemical and biological properties of a novel neuroendocrine-associated phosphatase (NEAP) were characterized. NEAP had a sequence characteristic of a dual-specificity phosphatase (DSP), and was preferentially expressed in neuroendocrine cells/tissues as well as in skeletal muscle and heart. Expression of NEAP was up-regulated in nerve growth factor (NGF)-treated, differentiated PC12 cells. NEAP was cytosolic and did not apparently have effects against extracellular signal-regulated kinase, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase activated by various stimuli. Although NEAP and MAPK phosphatase (MPK)-1 showed similar phosphatase activity towards p-nitro phenylphosphate (pNPP), in contrast to MKP-1, NEAP did not dephosphorylate JNK and p38-MAPK in vitro. Overexpression of NEAP, but not the C152S mutant, in PC12 cells suppressed NGF-induced phosphorylation of the p85 subunit of phosphatidylinositol 3-kinase (PI3K) and Akt activation. Overexpression of NEAP also suppressed neurite outgrowth induced by NGF and sensitized PC12 cells to cisplatin-induced apoptosis. Suppression of NEAP by RNA interference enhanced NGF-induced neurite outgrowth and Akt activation. Our results indicated that, unlike other DSPs, down-regulation of conventional MAPKs was not the major function of NEAP. Furthermore, NEAP might be involved in neuronal differentiation via regulation of the PI3K/Akt signaling.


Subject(s)
Cell Differentiation/physiology , Neurosecretory Systems/metabolism , Phosphoric Monoester Hydrolases/physiology , Signal Transduction/physiology , Animals , Cell Differentiation/drug effects , Cell Fractionation/methods , Cell Line , Dose-Response Relationship, Drug , Dual-Specificity Phosphatases , Enzyme Activation/drug effects , Enzyme Activation/physiology , Gene Expression/drug effects , Humans , Immunoprecipitation/methods , Mitogen-Activated Protein Kinase Phosphatases , Mitogen-Activated Protein Kinases/metabolism , Mutation/physiology , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/physiology , Neurosecretory Systems/cytology , Phosphoric Monoester Hydrolases/chemistry , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment/methods , Signal Transduction/drug effects , Time Factors , Transfection/methods
17.
J Neurosci Res ; 72(4): 508-19, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12704812

ABSTRACT

Oxidative stress in the brain has been increasingly associated with the development of numerous human neurological diseases. Microglia, activated upon neuronal injury or inflammatory stimulation, are known to release superoxide anion (*O(2) (-)), hydrogen peroxide (H(2)O(2)), and nitric oxide (NO), thereby further contributing to oxidative neurotoxicity. The reaction of NO and *O(2) (-), forming the toxic peroxynitrite (ONOO(-)), has been proposed to play a pathogenic role in neuronal injury. However, the interactions between H(2)O(2) and NO during oxidative stress, which may promote or diminish cell death, is less clear. In this study, we explored oxidative neurotoxicity induced by H(2)O(2) plus NO in primary cultures of rat cerebral cortex neurons. As the mechanisms may involve reactions between H(2)O(2) and NO, we monitored the production of ONOO(-)and reactive oxygen species (ROS) throughout the experiments. Results indicated that the NO donor S-nitroso-N-acetyl-D, L-penicillamine (SNAP) and H(2)O(2) by themselves elicited neuronal death in a concentration- and time-dependent manner. Sublytic concentrations of H(2)O(2) plus SNAP were sufficient to induce neuronal apoptosis as determined by DNA laddering and fluorescent staining of apoptotic nuclei. Transient ONOO(-)increase was accompanied by rapid H(2)O(2) decay and NO production, whereas ROS slowly decreased following treatment. Furthermore, p38 mitogen-activated protein kinase (MAPK) activation and the cleavage of caspase-3 were observed. Conversely, inhibition of p38 MAPK and caspase-3 significantly reduced apoptotic death induced by H(2)O(2) plus SNAP. These data suggest that H(2)O(2) and NO act synergistically to induce neuronal death through apoptosis in which activation of p38 MAPK and caspase-3 is involved.


Subject(s)
Hydrogen Peroxide/metabolism , Neurons/drug effects , Neurons/pathology , Nitric Oxide/metabolism , Oxidative Stress/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Caspase 3 , Caspases/drug effects , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA Fragmentation , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/pharmacology , Extracellular Space/chemistry , Fetus , Hydrogen Peroxide/analysis , Hydrogen Peroxide/pharmacology , Immunohistochemistry , In Situ Nick-End Labeling , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Nitric Oxide/analysis , Nitric Oxide Donors/pharmacology , Oxidants/pharmacology , Peroxynitrous Acid/analysis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/analysis , S-Nitroso-N-Acetylpenicillamine/pharmacology , p38 Mitogen-Activated Protein Kinases
18.
Ann N Y Acad Sci ; 962: 318-31, 2002 May.
Article in English | MEDLINE | ID: mdl-12076984

ABSTRACT

Increasing evidence has suggested that inflammation in the brain is closely associated with the pathogenesis of several degenerative neurologic disorders, including Parkinson's disease, Alzheimer's diseases, multiple sclerosis, amyotrophic lateral sclerosis, and AIDS dementia. The hallmark of brain inflammation is the activation of glial cells, especially that of microglia that produce a variety of proinflammatory and neurotoxic factors, including cytokines, fatty acid metabolites, free radicals--such as nitric oxide (NO) and superoxide. Excessive production of NO, as a consequence of nitric oxide synthase induction in activated glia, has been attributed to participate in neurodegeneration. Using primary mixed neuron-glia cultures and glia-enriched cultures prepared from embryonic rodent brain tissues, we have systemically studied the relationship between the production of NO and neurodegeneration in response to stimulation by the inflammagen lipopolysaccharide. This review summarizes our recent findings on the kinetics of NO generation, the relative contribution of microglia and astrocytes to NO accumulation, the relationship between NO production and neurodegeneration, and points of intervention along the pathways associated with NO generation to achieve neuroprotection. We also describe our results relating to the effect of several opioid-related agents on microglial activation and neuroprotection. Among these agents, the opioid receptor antagonist naloxone, especially its non-opioid enantiomer (+)-naloxone, promises to be of potential therapeutic value for the treatment of inflammation-related diseases.


Subject(s)
Inflammation/physiopathology , Nerve Degeneration , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Humans , Lipopolysaccharides/metabolism , Narcotics/metabolism , Neuroglia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...