Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add filters








Year range
1.
Experimental Neurobiology ; : 270-278, 2019.
Article in English | WPRIM | ID: wpr-739539

ABSTRACT

Chronic immobilization stress (CIS) induces low levels of glutamate (Glu) and glutamine (Gln) and hypoactive glutamatergic signaling in the mouse prefrontal cortex (PFC), which is closely related to the Glu-Gln cycle. A Gln-supplemented diet ameliorates CIS-induced deleterious changes. Here, we investigated the effects of CIS and Gln supplementation on Glu-Gln cycle-related proteins to characterize the underlying mechanisms. Using the CIS-induced depression mouse model, we examined the expression of 11 proteins involved in the Glu-Gln cycle in the PFC. CIS decreased levels of glutamate transporter 1 (GLT1) and sodium-coupled neutral amino acid transporter (SNAT) 1, SANT2, SNAT3, and SNAT5. Gln supplementation did not affect the non-stressed group but significantly increased GLT1 and SNATs of the stressed group. By immunohistochemical analysis, we confirmed that SNAT1 and SNAT2 were decreased in neurons and GLT1, SNAT3, and SNAT5 were decreased in astrocytes in the medial PFC of the stressed group, but Gln-supplemented diet ameliorated these decrements. Collectively, these results suggest that CIS may cause depressive-like behaviors by decreasing Glu and Gln transportation in the PFC and that a Gln-supplemented diet could prevent the deleterious effects of CIS.


Subject(s)
Animals , Mice , Amino Acid Transport System X-AG , Amino Acid Transport Systems , Astrocytes , Depression , Depressive Disorder , Diet , Glutamic Acid , Glutamine , Immobilization , Neurons , Prefrontal Cortex , Transportation
2.
The Korean Journal of Physiology and Pharmacology ; : 335-344, 2019.
Article in English | WPRIM | ID: wpr-761802

ABSTRACT

Obesity causes inflammation and impairs thermogenic functions in brown adipose tissue (BAT). The adipokine lipocalin 2 (LCN2) has been implicated in inflammation and obesity. Herein, we investigated the protective effects of caloric restriction (CR) on LCN2-mediated inflammation and oxidative stress in the BAT of high-fat diet (HFD)-fed mice. Mice were fed a HFD for 20 weeks and then either continued on the HFD or subjected to CR for the next 12 weeks. CR led to the browning of the white fat-like phenotype in HFD-fed mice. Increased expressions of LCN2 and its receptor in the BAT of HFD-fed mice were significantly attenuated by CR. Additionally, HFD+CR-fed mice had fewer neutrophils and macrophages expressing LCN2 and iron-positive cells than HFD-fed mice. Further, oxidative stress and mitochondrial fission induced by a HFD were also significantly attenuated by CR. Our findings indicate that the protective effects of CR on inflammation and oxidative stress in the BAT of obese mice may be associated with regulation of LCN2.


Subject(s)
Animals , Mice , Adipokines , Adipose Tissue, Brown , Caloric Restriction , Diet, High-Fat , Inflammation , Lipocalins , Macrophages , Mice, Obese , Mitochondrial Dynamics , Neutrophils , Obesity , Oxidative Stress , Phenotype
3.
Anatomy & Cell Biology ; : 274-283, 2018.
Article in English | WPRIM | ID: wpr-718952

ABSTRACT

Hyper-O-GlcNAcylation is a general feature of cancer which contributes to various cancer phenotypes, including cell proliferation and cell growth. Quercetin, a naturally occurring dietary flavonoid, has been reported to reduce the proliferation and growth of cancer. Several reports of the anticancer effect of quercetin have been published, but there is no study regarding its effect on O-GlcNAcylation. The aim of this study was to investigate the anticancer effect of quercetin on HeLa cells and compare this with its effect on HaCaT cells. Cell viability and cell death were determined by MTT and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labelling assays. O-GlcNAcylation of AMP-activated protein kinase (AMPK) was examined by succinylated wheat germ agglutinin pulldown and immunoprecipitation. Immunofluorescence staining was used to detect the immunoreactivitiy of O-linked N-acetylglucosamine transferase (OGT) and sterol regulatory element binding protein 1 (SREBP-1). Quercetin decreased cell proliferation and induced cell death, but its effect on HaCaT cells was lower than that on HeLa cells. O-GlcNAcylation level was higher in HeLa cells than in HaCaT cells. Quercetin decreased the expression of global O-GlcNAcylation and increased AMPK activation by reducing the O-GlcNAcylation of AMPK. AMPK activation due to reduced O-GlcNAcylation of AMPK was confirmed by treatment with 6-diazo-5-oxo-L-norleucine. Our results also demonstrated that quercetin regulated SREBP-1 and its transcriptional targets. Furthermore, immunofluorescence staining showed that quercetin treatment decreased the immunoreactivities of OGT and SREBP-1 in HeLa cells. Our findings demonstrate that quercetin exhibited its anticancer effect by decreasing the O-GlcNAcylation of AMPK. Further studies are needed to explore how quercetin regulates O-GlcNAcylation in cancer.


Subject(s)
Humans , Adenosine , AMP-Activated Protein Kinases , Cell Death , Cell Proliferation , Cell Survival , Deoxyuridine , Diazooxonorleucine , Fluorescent Antibody Technique , HeLa Cells , Immunoprecipitation , Phenotype , Protein Kinases , Quercetin , Sterol Regulatory Element Binding Protein 1 , Transferases , Triticum , Uterine Cervical Neoplasms
4.
The Korean Journal of Physiology and Pharmacology ; : 301-309, 2018.
Article in English | WPRIM | ID: wpr-727588

ABSTRACT

Statins mediate vascular protection and reduce the prevalence of cardiovascular diseases. Recent work indicates that statins have anticonvulsive effects in the brain; however, little is known about the precise mechanism for its protective effect in kainic acid (KA)-induced seizures. Here, we investigated the protective effects of atorvastatin pretreatment on KA-induced neuroinflammation and hippocampal cell death. Mice were treated via intragastric administration of atorvastatin for 7 days, injected with KA, and then sacrificed after 24 h. We observed that atorvastatin pretreatment reduced KA-induced seizure activity, hippocampal cell death, and neuroinflammation. Atorvastatin pretreatment also inhibited KA-induced lipocalin-2 expression in the hippocampus and attenuated KA-induced hippocampal cyclooxygenase-2 expression and glial activation. Moreover, AKT phosphorylation in KA-treated hippocampus was inhibited by atorvastatin pretreatment. These findings suggest that atorvastatin pretreatment may protect hippocampal neurons during seizures by controlling lipocalin-2-associated neuroinflammation.


Subject(s)
Animals , Mice , Atorvastatin , Brain , Cardiovascular Diseases , Cell Death , Cyclooxygenase 2 , Hippocampus , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Kainic Acid , Neurons , Phosphorylation , Prevalence , Seizures
5.
The Korean Journal of Physiology and Pharmacology ; : 63-70, 2018.
Article in English | WPRIM | ID: wpr-727937

ABSTRACT

Cilostazol is a selective inhibitor of type 3 phosphodiesterase (PDE3) and has been widely used as an antiplatelet agent. Cilostazol mediates this activity through effects on the cyclic adenosine monophosphate (cAMP) signaling cascade. Recently, it has attracted attention as a neuroprotective agent. However, little is known about cilostazol's effect on excitotoxicity induced neuronal cell death. Therefore, this study evaluated the neuroprotective effect of cilostazol treatment against hippocampal neuronal damage in a mouse model of kainic acid (KA)-induced neuronal loss. Cilostazol pretreatment reduced KA-induced seizure scores and hippocampal neuron death. In addition, cilostazol pretreatment increased cAMP response element-binding protein (CREB) phosphorylation and decreased neuroinflammation. These observations suggest that cilostazol may have beneficial therapeutic effects on seizure activity and other neurological diseases associated with excitotoxicity.


Subject(s)
Animals , Mice , Adenosine Monophosphate , Cell Death , Cyclic AMP Response Element-Binding Protein , Hippocampus , Kainic Acid , Neurons , Neuroprotective Agents , Phosphorylation , Seizures , Therapeutic Uses
6.
The Korean Journal of Physiology and Pharmacology ; : 451-460, 2015.
Article in English | WPRIM | ID: wpr-727351

ABSTRACT

Sirtuin 1 (SIRT1) is a mammalian NAD+-dependent protein deacetylase that regulates cellular metabolism and inflammatory response. The organ-specific deletion of SIRT1 induces local inflammation and insulin resistance in dietary and genetic obesity. Macrophage-mediated inflammation contributes to insulin resistance and metabolic syndrome, however, the macrophage-specific SIRT1 function in the context of obesity is largely unknown. C57/BL6 wild type (WT) or myeloid-specific SIRT1 knockout (KO) mice were fed a high-fat diet (HFD) or normal diet (ND) for 12 weeks. Metabolic parameters and markers of hepatic steatosis and inflammation in liver were compared in WT and KO mice. SIRT1 deletion enhanced HFD-induced changes on body and liver weight gain, and increased glucose and insulin resistance. In liver, SIRT1 deletion increased the acetylation, and enhanced HFD-induced nuclear translocation of nuclear factor kappa B (NF-kappaB), hepatic inflammation and macrophage infiltration. HFD-fed KO mice showed severe hepatic steatosis by activating lipogenic pathway through sterol regulatory element-binding protein 1 (SREBP-1), and hepatic fibrogenesis, as indicated by induction of connective tissue growth factor (CTGF), alpha-smooth muscle actin (alpha-SMA), and collagen secretion. Myeloid-specific deletion of SIRT1 stimulates obesity-induced inflammation and increases the risk of hepatic fibrosis. Targeted induction of macrophage SIRT1 may be a good therapy for alleviating inflammation-associated metabolic syndrome.


Subject(s)
Animals , Mice , Acetylation , Actins , Collagen , Connective Tissue Growth Factor , Diet , Diet, High-Fat , Fibrosis , Glucose , Inflammation , Insulin Resistance , Liver , Macrophages , Metabolism , NF-kappa B , Obesity , Sirtuin 1 , Sterol Regulatory Element Binding Protein 1 , Weight Gain
7.
Anatomy & Cell Biology ; : 91-100, 2014.
Article in English | WPRIM | ID: wpr-137046

ABSTRACT

Excessive immune responses induced by ischemia-reperfusion injury (IRI) are known to lead to necrotic and apoptotic cell death, and calcineurin plays a major role in this process. Calcineurin dephosphorylates the nuclear factor of activated T-cells (NFAT), permitting its translocation into the nucleus. As a result, calcineurin promotes the release of pro-inflammatory cytokines, such as tumor necrosis factor-alpha. The overproduction of pro-inflammatory cytokines causes renal cell death. Calcineurin activity is regulated by calpain, a cysteine protease present in the nucleus. Calpain-mediated proteolysis increases the phosphatase activity of calcineurin, resulting in NFAT dephosphorylation. This process has been studied in cardiomyocytes but its role in renal IRI is unknown. Thus, we examined whether calpain regulates calcineurin in renal tubule nuclei. We established an in vivo renal IRI model in mice and identified the protective role of a calcineurin inhibitor, FK506, in this process. Calcineurin is expressed in the nucleus, where it is present in its calpain-cleaved form. FK506 reduced nuclear expression of calcineurin and prevented calcineurin-mediated NFAT activation. Our study shows clearly that FK506 reduces calpain-mediated calcineurin activity. Consequently, calcineurin could not maintain NFAT activation. FK506 reduced renal cell death by suppressing the transcription of pro-inflammatory cytokine genes. This study provides evidence that FK506 protects against inflammation in a renal IRI mouse model. We also provided a mechanism of calcineurin action in the nucleus. Therefore, FK506 could improve renal function by decreasing calcineurin activity in both the cytoplasm and the nucleus of renal tubule cells.


Subject(s)
Animals , Mice , Calcineurin , Calpain , Cell Death , Cysteine Proteases , Cytokines , Cytoplasm , Inflammation , Myocytes, Cardiac , Proteolysis , Reperfusion Injury , T-Lymphocytes , Tacrolimus , Tumor Necrosis Factor-alpha
8.
Anatomy & Cell Biology ; : 91-100, 2014.
Article in English | WPRIM | ID: wpr-137040

ABSTRACT

Excessive immune responses induced by ischemia-reperfusion injury (IRI) are known to lead to necrotic and apoptotic cell death, and calcineurin plays a major role in this process. Calcineurin dephosphorylates the nuclear factor of activated T-cells (NFAT), permitting its translocation into the nucleus. As a result, calcineurin promotes the release of pro-inflammatory cytokines, such as tumor necrosis factor-alpha. The overproduction of pro-inflammatory cytokines causes renal cell death. Calcineurin activity is regulated by calpain, a cysteine protease present in the nucleus. Calpain-mediated proteolysis increases the phosphatase activity of calcineurin, resulting in NFAT dephosphorylation. This process has been studied in cardiomyocytes but its role in renal IRI is unknown. Thus, we examined whether calpain regulates calcineurin in renal tubule nuclei. We established an in vivo renal IRI model in mice and identified the protective role of a calcineurin inhibitor, FK506, in this process. Calcineurin is expressed in the nucleus, where it is present in its calpain-cleaved form. FK506 reduced nuclear expression of calcineurin and prevented calcineurin-mediated NFAT activation. Our study shows clearly that FK506 reduces calpain-mediated calcineurin activity. Consequently, calcineurin could not maintain NFAT activation. FK506 reduced renal cell death by suppressing the transcription of pro-inflammatory cytokine genes. This study provides evidence that FK506 protects against inflammation in a renal IRI mouse model. We also provided a mechanism of calcineurin action in the nucleus. Therefore, FK506 could improve renal function by decreasing calcineurin activity in both the cytoplasm and the nucleus of renal tubule cells.


Subject(s)
Animals , Mice , Calcineurin , Calpain , Cell Death , Cysteine Proteases , Cytokines , Cytoplasm , Inflammation , Myocytes, Cardiac , Proteolysis , Reperfusion Injury , T-Lymphocytes , Tacrolimus , Tumor Necrosis Factor-alpha
9.
The Korean Journal of Physiology and Pharmacology ; : 333-339, 2014.
Article in English | WPRIM | ID: wpr-728460

ABSTRACT

Exendin-4 (Ex-4), a glucagon-like peptide-1 receptor (GLP-1R) agonist, has been known to reverse hepatic steatosis in ob/ob mice. Although many studies have evaluated molecular targets of Ex-4, its mechanism of action on hepatic steatosis and fibrosis has not fully been determined. In the liver, glucose transporter 4 (GLUT4) is mainly expressed in hepatocytes, endothelial cells and hepatic stellate cells (HSCs). In the present study, the effects of Ex-4 on GLUT4 expression were determined in the liver of ob/ob mice. Ob/ob mice were treated with Ex-4 for 10 weeks. Serum metabolic parameters, hepatic triglyceride levels, and liver tissues were evaluated for hepatic steatosis. The weights of the whole body and liver in ob/ob mice were reduced by long-term Ex-4 treatment. Serum metabolic parameters, hepatic steatosis, and hepatic fibrosis in ob/ob mice were reduced by Ex-4. Particularly, Ex-4 improved hepatic steatosis by enhancing GLUT4 via GLP-1R activation in ob/ob mice. Ex-4 treatment also inhibited hepatic fibrosis by decreasing expression of connective tissue growth factor in HSCs of ob/ob mice. Our data suggest that GLP-1 agonists exert a protective effect on hepatic steatosis and fibrosis in obesity and type 2 diabetes.


Subject(s)
Animals , Mice , Connective Tissue Growth Factor , Endothelial Cells , Fatty Liver , Fibrosis , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Glucose Transport Proteins, Facilitative , Hepatic Stellate Cells , Hepatocytes , Liver , Obesity , Triglycerides , Weights and Measures
10.
The Korean Journal of Physiology and Pharmacology ; : 267-274, 2013.
Article in English | WPRIM | ID: wpr-727720

ABSTRACT

A beneficial radioprotective agent has been used to treat the radiation-induced lung injury. This study was performed to investigate whether curcumin, which is known to have anti-inflammatory and antioxidant properties, could ameliorate radiation-induced pulmonary inflammation and fibrosis in irradiated lungs. Rats were given daily doses of intragastric curcumin (200 mg/kg) prior to a single irradiation and for 8 weeks after radiation. Histopathologic findings demonstrated that macrophage accumulation, interstitial edema, alveolar septal thickness, perivascular fibrosis, and collapse in radiation-treated lungs were inhibited by curcumin administration. Radiation-induced transforming growth factor-beta1 (TGF-beta1), connective tissue growth factor (CTGF) expression, and collagen accumulation were also inhibited by curcumin. Moreover, western blot analysis revealed that curcumin lowered radiation-induced increases of tumor necrosis factor-alpha (TNF-alpha), TNF receptor 1 (TNFR1), and cyclooxygenase-2 (COX-2). Curcumin also inhibited the nuclear translocation of nuclear factor-kappa B (NF-kappaB) p65 in radiation-treated lungs. These results indicate that long-term curcumin administration may reduce lung inflammation and fibrosis caused by radiation treatment.


Subject(s)
Animals , Rats , Blotting, Western , Collagen , Connective Tissue Growth Factor , Curcumin , Cyclooxygenase 2 , Edema , Fibrosis , Inflammation , Lung , Lung Injury , Macrophages , Pneumonia , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha
11.
The Korean Journal of Physiology and Pharmacology ; : 159-165, 2012.
Article in English | WPRIM | ID: wpr-728106

ABSTRACT

Squamous cell carcinoma (SCC) and adenocarcinoma (AC) are the major histological types of non-small cell lung carcinoma (NSCLC). Although both SCCs and ACs have been characterized histologically and clinically, the precise mechanisms underlying their migration and invasion are not yet known. Here, we address the involvement in NSCLC of the p21-associated kinase1 (Pak1)/LIM kinase1 (LIMK1)/cofilin pathway, which recently has been reported to play a critical role in tumor migration and invasion. The Pak1/LIMK1/cofilin pathway was evaluated in tumors from SCC (n=35) and AC (n=35) patients and in SCC- and AC-type cell lines by western blotting, immunohistochemistry, and in vitro migration and invasion assays. The levels of phosphorylated Pak1, LIMK1, and cofilin in lung tumor tissues from SCC patients were increased as compared to normal tissues. In addition, immunohistochemistry showed greater expression of phosphorylated cofilin in SCC tissues. Expression of phosphorylated Pak1 and LIMK1 proteins was also significantly higher in SCC-type cells than in AC-type cells. Moreover, migration and invasion assays revealed that a higher percentage of SCC type cells exhibited migration and invasion compared to AC type cells. Migration was also decreased in LIMK1 knockdown SK-MES-1 cells. These findings suggest that the activation of the Pak1/LIMK1/cofilin pathway could preferentially contribute to greater tumor migration and invasion in SCC, relative to that in AC.


Subject(s)
Humans , Adenocarcinoma , Blotting, Western , Carcinoma, Squamous Cell , Cell Line , Immunohistochemistry , Lung , Lung Neoplasms , Proteins
12.
Anatomy & Cell Biology ; : 194-203, 2011.
Article in English | WPRIM | ID: wpr-23479

ABSTRACT

AMP-activated protein kinase (AMPK), an enzyme involved in energy homeostasis, regulates inflammatory responses, but its precise mechanisms are not fully understood. Recent evidence has shown that resveratrol (RES), an AMPK activator, reduces prostaglandin E2 production in lipopolysaccharide (LPS)-treated microglia. Here, we examined the effect of RES on nuclear factor kappa B (NF-kappaB) dependent cyclooxygenase (COX)-2 activation in LPS-treated RWA 264.7 macrophages. We found that treatment with RES increased AMPK activation. AMPK and acetyl CoA carboxylase phosphorylation were attenuated in cells treated with LPS+RES, compared to cells treated with LPS alone. RES inhibited tumor necrosis factor (TNF)-alpha and TNF receptor 1 in LPS-treated cells. Finally, RES inhibited LPS-induced NF-kappaB translocation into the nucleus and COX-2 expression. Moreover, the effects of 5-aminoimidazole-4-carboxamide ribose and compound C were consistent with the effects of RES in LPS-treated cells. Taken together, these results suggest that the anti-inflammatory action of RES in RAW 264.7 macrophages is dependent on AMPK activation and is associated with inhibition of the LPS-stimulated NF-kappaB-dependent COX-2 signaling pathway.


Subject(s)
Acetyl-CoA Carboxylase , AMP-Activated Protein Kinases , Dinoprostone , Homeostasis , Macrophages , Microglia , NF-kappa B , Phosphorylation , Prostaglandin-Endoperoxide Synthases , Receptors, Tumor Necrosis Factor , Ribose , Stilbenes , Tumor Necrosis Factor-alpha
13.
Anatomy & Cell Biology ; : 150-156, 2010.
Article in English | WPRIM | ID: wpr-31958

ABSTRACT

Oxidative stress-induced cell death leads to phosphorylation of 14-3-3zeta at serine 58. 14-3-3zeta is detected at significant levels in cerebrospinal fluid after kainic acid (KA)-induced seizures. Here we examined temporal changes in 14-3-3zeta phosphorylation in the hippocampus and amygdala of mice after KA treatment. Mice were killed at 2, 6, 24, or 48 h after KA (30 mg/kg) injection. We observed an increase in TUNEL and Fluoro-Jade B (FJB)-stained neurons in the hippocampus and amygdala of KA-treated mice. Phospho (p)-14-3-3zeta and p-JNK expression was increased in the hippocampus 2 and 6 h after KA treatment, respectively. In immunohistochemical analysis, p-14-3-3zeta-positive cells were present in the CA3 region of the hippocampus and the central nucleus of amygdala (CeA) of KA-treated mice. Thus, phosphorylation of 14-3-3zeta at serine 58 may play an important role in KA-induced hippocampal and amygdaloid neuronal damage.


Subject(s)
Animals , Mice , Amygdala , Cell Death , Fluoresceins , Hippocampus , In Situ Nick-End Labeling , Kainic Acid , Neurons , Phosphorylation , Seizures , Serine
14.
Anatomy & Cell Biology ; : 194-200, 2010.
Article in English | WPRIM | ID: wpr-49865

ABSTRACT

It is well known that chronic ethanol treatment affects the synthesis of RNA and protein in the brain and the maintenance and function of nervous system. The changes in myelination-related genes are most prominent in human alcoholics. Previously, our cDNA microarray study showed altered Proteolipid protein (PLP), a major protein of central myelin. The present study aimed to gain more understanding of the expression of PLP after chronic ethanol treatment. Male Sprague-Dawley rats were daily treated with ethanol (15% in saline, 3 g/kg, i.p.) or saline for 14 days. Messenger RNAs from hippocampus of each group were subjected to cDNA expression array hybridization to determine the differential gene expressions. Among many ethanol responsive genes, PLP was negatively regulated by ethanol treatment, which is one of the most abundant proteins in the CNS and has an important role in the stabilization of myelin sheath. Using northern blot and immunohistochemical analysis, we showed the change in expression level of PLP mRNA and protein after ethanol treatment. PLP mRNA and protein were decreased in hippocampus of rat with chronic ethanol exposure, suggesting that ethanol may affect the stabilization of myelin sheath through the modulation of PLP expression and induce the pathophysiology of alcoholic brain.


Subject(s)
Animals , Humans , Male , Rats , Alcoholics , Blotting, Northern , Brain , Chimera , DNA, Complementary , Ethanol , Gene Expression , Hippocampus , Myelin Proteolipid Protein , Myelin Sheath , Nervous System , Oligonucleotide Array Sequence Analysis , Proteins , Rats, Sprague-Dawley , RNA , RNA, Messenger
15.
Korean Journal of Anatomy ; : 141-147, 2009.
Article in English | WPRIM | ID: wpr-647050

ABSTRACT

Adiponectin is an adipocyte-derived protein with anti-diabetic and anti-angiogenesis properties that improves both glucose metabolism and insulin resistance via the adenosine monophosphate-activated protein kinase (AMPK) cascade. Diabetic cognitive deficits are correlated with dysregulation of energy metabolism in the hippocampus. In the present study, we investigated the expression of adiponectin-mediated AMPK cascade proteins in the hippocampus of streptozotocin (STZ)-induced diabetic mice. Diabetes was induced by STZ (55 mg/kg) injection intraperitoneally. Twenty-four weeks after induction of diabetes, mice were sacrificed. Results showed that decreased serum adiponectin levels and increased expression of hippocampal adiponectin receptor 1 (AdipoR1) was expressed in diabetic mice. Phosphorylated AMPK, acetyl CoA carboxylase (ACC), and eNOS expression levels were increased in the hippocampus of diabetic mice. The immunoreactivity of glucose transporter 1 in the endothelium of hippocampal blood vessels was also increased. These results indicate that adiponectin-mediated AMPK cascade activation may play a role in catabolic process that is involved in diabetic neurodegeneration.


Subject(s)
Animals , Mice , Acetyl-CoA Carboxylase , Adenosine , Adiponectin , AMP-Activated Protein Kinases , Blood Vessels , Endothelium , Energy Metabolism , Glucose , Glucose Transport Proteins, Facilitative , Hippocampus , Insulin Resistance , Protein Kinases , Proteins , Receptors, Adiponectin , Streptozocin
16.
Korean Journal of Anatomy ; : 205-211, 2008.
Article in English | WPRIM | ID: wpr-647047

ABSTRACT

Reactive oxygen species (ROS) are important signaling molecules or mediators in many cellular responses, including the oxidative-burst defense response. Certain hormones are neuroprotective because they are modulators of neuronal activity or ROS scavengers. We have examined the effect of a hormone-free condition on ROS levels following glutamate-induced excitotoxicity in the mouse hippocampal HT22 cell line. We show that hormone starvation slightly elevates ROS and that continuous low concentrations of ROS induce expression of antioxidant enzymes, such as heme oxygenase-1 (HO-1). In addition, N-acetyl-L-cysteine (NAC) restores the expression of ERK1/2 protein in hormone-starved HT22 cells. These findings suggest that whereas high-dose ROS are cytotoxic and lead to tissue damage in the brain low-dose ROS may act in neuroprotective signaling.


Subject(s)
Animals , Mice , Acetylcysteine , Brain , Cell Line , Glutamic Acid , Heme Oxygenase-1 , Neurons , Reactive Oxygen Species , Starvation
17.
Korean Journal of Anatomy ; : 213-221, 2008.
Article in English | WPRIM | ID: wpr-647046

ABSTRACT

Seizure activity increases glucose utilization within the brain in response to neuronal injury. In this study, we investigated the expression of two brain glucose transporter (GLUT) proteins, GLUT1 and GLUT3, in the mouse hippocampus after kainic acid (KA) treatment. Forty-eight hours after KA (30 mg/kg) injection, mice were sacrificed and a histological evaluation of KA-treated hippocampus revealed cell death using cresyl violet staining and immunohistochemistry for caspase-3. In KA-treated hippocampus, reactive astrocytic changes were confirmed by increased immunoreactivity of glial fibrillary acidic protein (GFAP). Enhanced GLUT1-positive endothelial cells were present in the hippocampus after KA treatment. However, GLUT3-positive neurons were not localized to the KAtreated hippocampus. In particular, although GLUT-3 was not expressed in the hippocampus, pronounced GLUT3- positive cells were observed in the hypothalamic paraventricular nucleus (PVN), which controls energy metabolism. Thus, these results indicate that changes in endothelial GLUT1 and neuronal GLUT3 levels in response to neural injury may play important roles in neuroprotection against brain excitotoxicity.


Subject(s)
Animals , Mice , Benzoxazines , Brain , Caspase 3 , Cell Death , Endothelial Cells , Energy Metabolism , Glial Fibrillary Acidic Protein , Glucose , Glucose Transport Proteins, Facilitative , Hippocampus , Immunohistochemistry , Kainic Acid , Neurons , Paraventricular Hypothalamic Nucleus , Proteins , Seizures , Viola
18.
Korean Journal of Anatomy ; : 287-295, 2007.
Article in English | WPRIM | ID: wpr-652063

ABSTRACT

Vascular endothelial growth factor (VEGF) is closely involved in early retinal pathology of diabetes, including blood-retinal barrier breakdown, pericyte loss, neuro-retinal apoptosis, and cell proliferation. This study examines the involvement of VEGF in cell apoptosis and survival in the retina of animals with type 2 diabetes. We used retinas from 28-week-old Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a model of spontaneous type 2 diabetes, and Long-Evans Tokushima Otsuka (LETO) rats as controls. In parallel with evidence for pericyte loss, we found cell proliferation, apoptosis, and endothelial nitric oxide synthase (eNOS) (an indicator of endothelial cell proliferation/survival) and VEGF overexpression in the OLETF-retina, compared to control LETO. Furthermore, apoptotic signals were partly co-localized to only VEGF-positive cells in the OLETF-retina, but no apoptotic signals were found in VEGF- and eNOS-double-positive cells. These results suggest that upregulated VEGF is involved in apoptosis and eNOS-dependent cell survival in the retinas of type 2 diabetic rats.


Subject(s)
Animals , Rats , Apoptosis , Blood-Retinal Barrier , Cell Proliferation , Cell Survival , Endothelial Cells , Nitric Oxide Synthase Type III , Pathology , Pericytes , Rats, Inbred OLETF , Retina , Retinaldehyde , Vascular Endothelial Growth Factor A
19.
Korean Journal of Anatomy ; : 363-370, 2005.
Article in English | WPRIM | ID: wpr-647011

ABSTRACT

The principal aim of this study was to determine the effects of antipsychotics (haloperidol, sulpiride, and clozapine) on regulating dopamine (DA) D1 and D2 receptor mRNA levels in the rat caudate putamen (CPu), nucleus accumbens (NAc), and olfactory tubercle (OTu). Twenty male Sprague-Dawley rats (250 g) were treated with haloperidol (1mg/mL), sulpiride (40 mg/mL), clozapine (20 mg/mL), and the control group received only water. Drugs were administered orally for 4 weeks. Antipsychotic drugs had differential effects on DA D1 and D2 receptor gene expression. Haloperidol and sulpiride induced an increase of DA D1 and D2 receptor mRNA levels in the rat CPu, OTu, and NAc; haloperidol caused a greater increase than sulpiride. However, clozapine treatment had less effect on DA receptor mRNAs levels in the same area. Antipsychotic drugs differentially upregulated the expression of DA D1 and D2 receptor mRNAs in the rat brain. These changes may be related, at least in part, to changes of DA concentration following antipsychotics treatment.


Subject(s)
Animals , Humans , Male , Rats , Antipsychotic Agents , Brain , Clozapine , Dopamine , Gene Expression , Haloperidol , In Situ Hybridization , Nucleus Accumbens , Olfactory Pathways , Putamen , Rats, Sprague-Dawley , Receptors, Dopamine , RNA, Messenger , Sulpiride , Water
20.
Experimental & Molecular Medicine ; : 78-85, 2005.
Article in English | WPRIM | ID: wpr-90145

ABSTRACT

CD137, which is expressed on activated T cells, plays a critical role in inflammatory responses. However, the exact role that CD137 plays in monocytes is not fully known. Here we studied the expression and function of CD137 in human monocytic THP-1 cells, which we found constitutively expresses CD137 at the mRNA and protein level. Cross-linking of CD137 increased the secretion of IL-8 and TNF-alpha, promoted the expression of CD54 and CD11b, and increased adhesion to extracellular matrix (ECM) proteins. In particular CD137-induced adhesion of THP-1 cells was inhibited by an inhibitor of mitogen-activated protein kinase kinase (MEK), but not by a p38 kinase inhibitor. Taken together, these results show that the adhesion and cytokine production of THP-1 cells induced by CD137 occur via activation of MEK, which results in the activation of ERK-1/2 signaling pathways. Therefore, this study suggests that CD137 induces an activating and migrating signal during inflammatory processes.


Subject(s)
Humans , Antigens, CD/biosynthesis , CD11 Antigens/biosynthesis , Cell Adhesion , Cell Adhesion Molecules/biosynthesis , Cell Line , Cytokines/biosynthesis , Enzyme Activation , Extracellular Matrix Proteins/metabolism , Flow Cytometry , Immunity, Innate , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-8/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Monocytes/metabolism , Phosphorylation , Protein Binding , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL