Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Int Immunopharmacol ; 16(2): 232-42, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23623942

ABSTRACT

Advancements in rheumatoid-arthritis-(RA) therapies have shown considerable progresses in the comprehension of disease. However, the development of new potential agents with relative safety and efficacy continues and natural compounds have been considered as alternatives to identify new entities. Since previous in-vivo data and our in-vitro findings showed that torilin has a strong anti-inflammatory property, we further investigated its effect against collagen-induced-arthritis-(CIA) in mice. CIA-induced DBA/1J mice were treated with torilin or methotrexate (MTX) for 5-weeks. Arthritis severity was evaluated by arthritic score and joint histopathology. Draining lymph node (dLN), joint and peripheral-blood mononuclear-cell (PBMC) counts, and activation/localization of T-/B-lymphocytes, dendritic cells (DCs) and neutrophils were examined by FACS analysis. Serum anti-type-II-collagen-(CII) antibody levels and cultured-splenocyte and serum cytokines were also evaluated. Torilin markedly reduced CIA-induced arthritic score, histopathology and leukocyte counts. Besides, torilin suppressed CIA-activated T-cells including CD3+, CD3+/CD69+, CD8+, CD4+ and CD4+/CD25+ in dLNs or joints. It also modified CD19+ or CD20+/CD23+ (B-cells), MHCII+/CD11c+ (DCs) and Gr-1+/CD11b+ (neutrophil) subpopulations. It further depressed total anti-CII-IgG, anti-CII-IgG1 and anti-CII-IgG2a antibody productions. Moreover, while IFN-γ and IL-10 were not affected, torilin suppressed CIA-induced serum TNF-α, IL-1ß and IL-6 levels. Interestingly, torilin also blocked IFN-γ, IL-17 and IL-6 cytokines while it did not affect IL-10 but enhanced IL-4 in splenocytes. These results show that torilin attenuated arthritis severity, modified leukocyte activations in dLNs or joints, and restored serum and splenocyte cytokine imbalances. Torilin may have immunomodulatory and anti-inflammatory properties with the capacity to ameliorate the inflammatory response in CIA-mice.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/blood , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Collagen Type II/immunology , Cytokines/blood , Cytokines/immunology , Foot/pathology , Immunoglobulin G/blood , Joints/cytology , Leukocyte Count , Leukocytes/immunology , Lymph Nodes/cytology , Male , Mice , Mice, Inbred DBA , Sesquiterpenes, Guaiane/pharmacology , Sesquiterpenes, Guaiane/therapeutic use , Spleen/cytology
2.
J Nat Med ; 67(4): 807-13, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23463332

ABSTRACT

Mushrooms have a long history of dietary benefits in Asia due to their health-promoting effects. Phellinus baumii, a wild mushroom, has been reported to have anti-platelet, anti-inflammatory, anti-obesity and free radical scavenging activities. However, its anti-rheumatoid arthritis (RA) property remains poorly understood. Hence, we investigated the protective effect of Phellinus baumii ethyl acetate extract (PBEAE) against bovine collagen type II induced arthritis (CIA) in DBA/1 mice. PBEAE (50 and 150 mg/kg) reduced the CIA score and leukocyte count in draining lymph nodes (DLNs) and inflamed joints. PBEAE also attenuated the expressions of CD3⁺ (T cells), CD19⁺ (B cells), CD4⁺ (T-helper), CD8⁺ (T-cytotoxic), MHC class II/CD11c⁺ (antigen-presenting cells), double positives (B220⁺/CD23⁺ and CD3⁺/CD69⁺: early lymphocyte activation markers) and CD4⁺/CD25⁺ (activated T-helper) leukocyte subpopulations in DLNs. Likewise, CD3⁺ and Gr-1⁺CD11b⁺ (neutrophil) counts in inflamed joints were also decreased. Furthermore, PBEAE reduced the serum levels of anti-collagen type immunoglobulin G, tumor necrosis factor-α and interleukin (IL)-1ß and IL-6. Taken together, PBEAE impaired cellular recruitment to the inflamed joint and alleviated CIA, and thus could be considered as a potential agent against rheumatoid arthritis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Basidiomycota , Acetates/chemistry , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cattle , Cell Extracts/therapeutic use , Collagen Type II , Interleukin-1beta/blood , Interleukin-6/blood , Male , Mice , Mice, Inbred DBA , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/blood
3.
Am J Physiol Cell Physiol ; 298(3): C611-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20032508

ABSTRACT

Regulator of G protein signaling (RGS) family members, such as RGS2, interact with Galpha subunits of heterotrimeric G proteins, accelerating the rate of GTP hydrolysis and attenuating the intracellular signaling triggered by the G protein-coupled receptor-ligand interaction. They are also reported to regulate G protein-effector interactions and form multiprotein signaling complexes. Ischemic stress-induced changes in RGS2 expression have been described in astrocytes, and these changes are associated with intracellular signaling cascades, suggesting that RGS2 upregulation may be an important mechanism by which astrocytes may regulate RGS2 function in response to physiological stress. However, information on the functional roles of stress-induced modulation of RGS2 protein expression in astrocyte function is limited. We report the role of ischemic stress in RGS2 protein expression in rat C6 astrocytoma cells and primary mouse astrocytes. A marked increase in RGS2 occurred after ischemic stress induced by chemicals (sodium azide and 2-deoxyglucose) or oxygen-glucose deprivation (OGD, real ischemia). RGS2 mRNA expression was markedly enhanced by 1 h of exposure to chemical ischemia or 6 h of OGD followed by 2 or 6 h of recovery, respectively. This enhanced expression in primary astrocytes and C6 cells was restored to baseline levels after 12 h of recovery from chemically induced ischemic stress or 4-6 h of recovery from OGD. RGS2 protein was also significantly expressed at 12-24 h of recovery from ischemic insult. Ischemia-induced RGS2 upregulation was associated with enhanced apoptosis. It significantly increased annexin V-positive cells, cleaved caspase-3, and enhanced DNA ladder formation and cell cycle arrest. However, a small interfering RNA (siRNA)-mediated RGS2 knockdown reversed the apoptotic cell death associated with ischemia-induced RGS2 upregulation. Upregulated RGS2 was significantly inhibited by SB-203580, a p38 MAPK inhibitor. Rottlerin, a potent inhibitor of PKCdelta, completely abrogated the increased RGS2 expression. We also examine whether ischemia-induced RGS2-mediated apoptosis is affected by siRNA-targeted endogenous PKCdelta downregulation or its phosphorylation. Although RGS2 upregulation was not affected, siRNA transfection significantly suppressed endogenous PKCdelta mRNA and protein expressions. Ischemia-induced PKCdelta phosphorylation and caspase-3 cleavage were dose dependently inhibited by PKCdelta knockdown, and this endogenous PKCdelta suppression reversed ischemia-induced annexin V-positive cells. This study suggests that ischemic stress increases RGS2 expression and that this condition contributes to enhanced apoptosis in C6 cells and primary astrocytes. The signaling it follows may involve PKCdelta and p38 MAPK pathways.


Subject(s)
Apoptosis , Astrocytes/metabolism , Brain Ischemia/metabolism , RGS Proteins/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/pathology , Brain Ischemia/pathology , Caspase 3/metabolism , Cell Hypoxia , Cell Line, Tumor , Glucose/deficiency , Humans , Mice , Oxidative Stress , Phosphorylation , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Kinase Inhibitors/pharmacology , RGS Proteins/genetics , RNA Interference , RNA, Messenger/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Signal Transduction , Time Factors , Transfection , Up-Regulation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Eur J Pharmacol ; 627(1-3): 85-91, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-19913011

ABSTRACT

Platelets, though anucleated, possess several transcription factors, including NF-kappaB, that exert non-genomic functions regulating platelet activation. Since platelets have not only been recognized as central players of homeostasis, but also participated in pathological conditions such as thrombosis, atherosclerosis, and inflammation, we examined rat platelet NF-kappaB expression and evaluated the effects of anti-inflammatory drug BAY 11-7082, an inhibitor of NF-kappaB activation, in platelet physiology. Western blotting revealed that rat platelets express NF-kappaB. BAY 11-7082, dose dependently, inhibited collagen- or thrombin-induced-platelet aggregation. ATP release, TXB(2) formation, P-selectin expression, and intercellular Ca(2+) concentration activated by collagen were reduced in BAY 11-7082-treated platelets. BAY 11-7082 elevated intracellular levels of cAMP, but not cGMP, and its co-incubation with cAMP-activating agent (forskolin) or its hydrolyzing enzyme inhibitor (3-isobutyl-1-methylxanthine, IBMX), synergistically inhibited collagen-induced-platelet aggregation. In addition, vasodilator-stimulated-phosphoprotein (VASP) phosphorylation was enhanced in BAY 11-7082-treated platelets, which was partially inhibited by a protein kinase A (PKA) inhibitor, H-89. Moreover, while p38 mitogen-activated protein kinase (MAPK) was not affected, BAY 11-7082 attenuated c-Jun N-terminal kinase 1 (JNK1) and extracellular-signal-regulated protein kinase 2 (ERK2) phosphorylations. In conclusion, BAY 11-7082 inhibits platelet activation, granule secretion, and aggregation, and that this effect is mediated by inhibition of JNK1 and ERK2 phosphorylations, and partially by stimulation of cAMP-dependent PKA VASP phosphorylation. The ability of BAY 11-7082 to inhibit platelet function might be relevant in cases involving aberrant platelet activation where the drug is considered as anti-atherothrombosis, and anti-inflammatory therapy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Adhesion Molecules/metabolism , Cyclic AMP/metabolism , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Nitriles/pharmacology , Phosphoproteins/metabolism , Platelet Aggregation/drug effects , Sulfones/pharmacology , Adenosine Triphosphate/metabolism , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Collagen/pharmacology , Cyclic AMP/biosynthesis , Cyclic GMP/biosynthesis , Cyclic GMP/metabolism , Gene Expression Regulation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , NF-kappa B/metabolism , P-Selectin/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Thrombin/pharmacology , Thromboxane A2/biosynthesis
5.
Biol Pharm Bull ; 31(11): 2012-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18981565

ABSTRACT

Salmonellosis is a major bacterial zoonosis that causes a variety of disease syndromes, from self-limiting enteritis to fatal infection in animals and food-borne infection and typhoid fever in humans. Recently, the emergence of multidrug-resistant strains of Salmonella sp. has caused more serious problems in public health. The present study investigated the antibacterial effects of Houttuynia cordata water extract (HCWE) against murine salmonellosis. In RAW 264.7 cells, there was no detectable cytotoxic effect of HCWE at any concentration between 25 and 100 microg/ml after 8-h incubation. The antibacterial activity of HCWE was then examined in a Salmonella enterica serovar (Salmonella typhimurium), and was found to increase in a dose-dependent manner at concentrations from 25 to 100 microg/ml during 8-h incubation. HCWE also affected RAW 264.7 cells including morphologic change and bacterial uptake, but there was no significant difference in bacterial replication in RAW 264.7 cells. With HCWE alone, nitric oxide (NO) production by RAW 264.7 cells did not increase, but when RAW 264.7 cells were infected by S. typhimurium, with or without HCWE, NO production with HCWE was 2-fold higher than that without HCWE. Treatment with HCWE did not affect inducible NO synthase (iNOS) mRNA expression by RAW 264.7 cells, but when RAW 264.7 cells with HCWE were infected by S. typhimurium, iNOS mRNA expression was increased during 8-h incubation. Furthermore, HCWE showed virulence reduction effects in S. typhimurium-infected BALB/c mice. After a lethal dose of S. typhimurium, the mortality rate in the HCWE untreated group was 100% at 7 d, but the HCWE 25, 50, and 100 microg/ml groups survived until 11, 17, and 23 d, respectively. These data suggest that HCWE is stable and beneficial in the treatment of bacterial infection including intracellularly replicating pathogens and may solve antimicrobial misuse and overuse.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Houttuynia/chemistry , Macrophages/microbiology , Salmonella typhimurium/drug effects , Animals , Anti-Bacterial Agents/isolation & purification , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/therapeutic use , Female , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Phagocytosis/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Salmonella Infections, Animal/drug therapy , Salmonella typhimurium/pathogenicity , Virulence , Water
6.
J Pharm Pharmacol ; 60(11): 1531-6, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18957175

ABSTRACT

Ginsenoside Rg3, a single ginseng saponin, is known to be a major anti-platelet component of protopanaxadiol that is isolated from Korean red ginseng. In this study, we investigated whether dihydroginsenoside Rg3, a stable chemical derivative of ginsenoside Rg3, also demonstrated anti-platelet activity. Dihydroginsenoside Rg3 inhibited thrombin-induced platelet aggregation in a concentration-dependent manner with an IC50 (concentration producing 50% inhibition) of 18.8 +/- 0.4 microM. Ginsenoside Rg3 inhibited platelet aggregation which was induced by thrombin (0.1 U mL(-1)) with an IC50 of 40.2 +/- 0.9 microM. We next determined whether dihydroginsenoside Rg3 affected different types of ligand-induced platelet aggregation. We found that dihydroginsenoside Rg3 inhibited collagen-induced platelet aggregation with an IC50 of 20.0 +/- 0.9 microM. To elucidate the inhibitory mechanism of dihydroginsenoside Rg3 on aggregation, we analysed its downstream signalling pathway. It was interesting to note that dihydroginsenoside Rg3 elevated cyclic AMP production in resting platelets, but did not affect cyclic GMP production. In addition, we found that dihydroginsenoside Rg3 potently suppressed phosphorylation of extracellular signal-regulated kinase 2 (ERK2), which was stimulated by collagen (2.5 microg mL(-1)), but not of p38 mitogen-activated protein kinase. Taken together, our results indicate that dihydroginsenoside Rg3 potently inhibited platelet aggregation via the modulation of downstream signalling components such as cAMP and ERK2.


Subject(s)
Ginsenosides/pharmacology , Mitogen-Activated Protein Kinase 1/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen/metabolism , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Ginsenosides/administration & dosage , Ginsenosides/isolation & purification , Inhibitory Concentration 50 , Male , Mitogen-Activated Protein Kinase 1/metabolism , Panax/chemistry , Phosphorylation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/isolation & purification , Rats , Rats, Sprague-Dawley , Thrombin/metabolism
7.
Phytother Res ; 22(10): 1389-95, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18688813

ABSTRACT

Aloe species are traditionally prescribed for hypertension, burning, and rheumatoid arthritis. To elucidate the mechanism of the antihypertensive and anti-inflammatory activities of this herb, the ethanol fraction from A. saponaria Haw. was evaluated for antioxidative activity using xanthine-xanthine oxidase (XO) assay, 2,2-Diphenyl-lpicrylhydrazyl radical (DPPH) assay, lipopolysaccharide (LPS)-induced nitric oxide (NO) production in RAW 264.7 cell, and antinociceptive activity using a tail-flick assay and hind paw pressure assay in cisplatin-treated hyperalgesic rats. The ethanol fraction displayed potent antioxidative activities in XO assay. In addition, ethanol fractions showed potent scavenging effects in DPPH assay. We next examined whether ethanol fractions showed anti-inflammatory activities. Ethanol fractions significantly suppressed NO production from LPS-activated RAW264.7 cells. As expected, ethanol fractions dose-dependently inhibited the messenger RNA expression of inducible NO synthase (iNOS). Moreover, ethanol fractions potently suppressed the expression of cycloxygenase (COX)-2 and granulocyte-macrophage colony-stimulating factor (GM-CSF), which are stimulated by LPS in RAW264.7 cells. In addition, ethanol fractions significantly blocked cisplatin-induced hyperalgesia using tail-flick assay and hind paw pressure test in rats. Taken altogether, ethanol extracts of aloe may be useful as a functional food or as a drug against reactive oxygen species (ROS) mediated diseases.


Subject(s)
Aloe/chemistry , Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Plant Extracts/pharmacology , Animals , Biphenyl Compounds , Cell Line , Cisplatin/pharmacology , Cyclooxygenase 2/drug effects , Drug Evaluation, Preclinical , Ethanol/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Mice , Nitric Oxide Synthase Type II/antagonists & inhibitors , Picrates/chemistry , Rats , Reverse Transcriptase Polymerase Chain Reaction , Xanthine Oxidase/metabolism
8.
Eur J Pharmacol ; 584(2-3): 361-7, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18313047

ABSTRACT

This study was designed to investigate the effect of davallialactone, which was isolated from the mushroom Inonotus xeranticus, on platelet aggregation induced by collagen, thrombin and ADP. We found that davallialactone dose-dependently inhibited platelet aggregation that was stimulated either by collagen (2.5 microg/ml), a potent ligand of integrin alpha2beta1 and glycoprotein VI, or by thrombin (0.1U/ml), a potent agonist of the protease-activated receptors (PARs) PAR1 and PAR3. In addition, davallialactone inhibited platelet aggregation induced by ADP, an agonist of P2Y receptor. To understand the mechanism of anti-platelet activity, we determined whether davallialactone affected the downstream signaling in collagen-activated platelets. Using the fura-2/AM fluorometric assay, we found that davallialactone dose-dependently inhibited intracellular calcium concentration levels ([Ca2+]i). Moreover, davallialactone inhibited the phosphorylation of extracellular signal-regulated protein kinase (ERK)-2 and p38 mitogen-activated protein kinase (MAPK), in a dose-dependent manner. The tyrosine phosphorylation of 60 and 85kDa proteins, which were activated by collagen, were differentially inhibited by davallialactone. Taken together, these data suggest that davallialactone may have potential anti-platelet aggregation activity via suppression of intracellular downstream signaling pathways.


Subject(s)
Blood Platelets/drug effects , Calcium Signaling/drug effects , Lactones/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Diphosphate/metabolism , Animals , Blood Platelets/enzymology , Blood Platelets/metabolism , Collagen/metabolism , Dose-Response Relationship, Drug , Phosphorylation , Phosphotyrosine/metabolism , Rats , Thrombin/metabolism
9.
Pharmazie ; 62(6): 453-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17663194

ABSTRACT

Suaeda asparagoides Miq. (Chenopodiaceae: S. asparagoides) is a salt-marsh plant that has long been prescribed in traditional Oriental medicine for the treatment of hypertension and hepatitis. In order to elucidate the pharmacological mechanisms of the herb, we conducted an examination of the anti-oxidative and anti-inflammatory properties of solvent-extracts of S. asparagoides. All of the solvent fractions showed potent anti-oxidative effects, as assessed using a radical generation assay system (xanthine oxidase assay) and an electron-donating activity system (DPPH [2,2-diphenyl-l-picrylhydrazyl radical] assay), with IC50 values ranging from 9 to 42 microg/ml. In agreement with this pattern, the total phenolic contents were widely distributed in the various solvent fractions, and ranged from 36.5 to 50.3 mg/g of dry weight. All of the solvent fractions significantly suppressed NO production in RAW264.7 cells induced by lipopolysaccharide (LPS, 0.1 microg/ml) and of the fractions, only the chloroform (CHC) fraction completely blocked the expression of inducible NO synthase (iNOS). Additionally, the hexane (HEX) and CHC fractions suppressed the mRNA expression of granulocyte/macrophage colony-stimulating factor (GM-CSF) and monocyte chemoattractant protein 1 (MCP-1), respectively, in the LPS-stimulated RAW264.7 cells. Therefore, these results suggest that the pharmacological action of S. asparagoides is due to its potent anti-oxidative effects and anti-inflammatory effects, and that therefore it can be applied to other diseases caused by oxidative stress and inflammation, such as cardiovascular diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Chenopodiaceae/chemistry , Animals , Biphenyl Compounds , Cell Line , Cell Proliferation/drug effects , Chemokines/biosynthesis , Cytokines/biosynthesis , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/antagonists & inhibitors , Phenols/analysis , Picrates/chemistry , Plant Extracts/pharmacology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Solvents , Tetrazolium Salts , Thiazoles , Xanthine Oxidase/antagonists & inhibitors
10.
Biochem Biophys Res Commun ; 349(1): 408-15, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16934753

ABSTRACT

Regulators of G protein signaling (RGSs) are inducibly expressed in response to various stimuli and the up-regulation of RGSs leads to significant decreases in GPCR responsiveness. Isoproterenol, an adrenergic receptor agonist, stimulated RGS2 mRNA in C6 rat astrocytoma cells. The up-regulation of RGS2 mRNA was abrogated by genistein, a protein tyrosine kinase inhibitor (PTK), and by broad-spectrum protein kinase C (PKC) inhibitors (staurosporine and GF109203X). alpha-Adrenergic antagonist (prazocin), beta-adrenergic antagonist (prazocin), and pertussis toxin only partially blocked the RGS2 up-regulation, suggesting that the RGS2 up-regulation is concomitantly mediated by Galphai, Galphas, and Galphaq. It is interesting to note that SB203580, a potent p38 mitogen-activated protein kinase (MAPK) inhibitor, completely inhibited the isoproterenol-mediated RGS2 expression. In addition, isoproterenol also markedly stimulated RGS2 mRNA in rat primary astrocytes, which were sensitive to SB203580 and staurosporine. Therefore, our data suggest that adrenergic receptor-mediated signaling (induced by isoproterenol) may be involved in the regulation of RGS2 expression in astrocytes via activating PTK, PKC, and p38 MAPK.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation, Neoplastic , Isoproterenol/pharmacology , RGS Proteins/biosynthesis , Up-Regulation , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , MAP Kinase Signaling System , Protein Kinase C/antagonists & inhibitors , RGS Proteins/chemistry , RGS Proteins/physiology , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL