Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 17(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38675401

ABSTRACT

Exposure to hypoxia results in the development of pulmonary arterial hypertension (PAH). An increase in the intracellular Ca2+ concentration ([Ca2+]i) in pulmonary artery smooth muscle cells (PASMCs) is a major trigger for pulmonary vasoconstriction and proliferation. This study investigated the mechanism by which KMUP-1, a xanthine derivative with phosphodiesterase inhibitory activity, inhibits hypoxia-induced canonical transient receptor potential channel 1 (TRPC1) protein overexpression and regulates [Ca2+]i through store-operated calcium channels (SOCs). Ex vivo PASMCs were cultured from Sprague-Dawley rats in a modular incubator chamber under 1% O2/5% CO2 for 24 h to elucidate TRPC1 overexpression and observe the Ca2+ release and entry. KMUP-1 (1 µM) inhibited hypoxia-induced TRPC family protein encoded for SOC overexpression, particularly TRPC1. KMUP-1 inhibition of TRPC1 protein was restored by the protein kinase G (PKG) inhibitor KT5823 (1 µM) and the protein kinase A (PKA) inhibitor KT5720 (1 µM). KMUP-1 attenuated protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA, 1 µM)-upregulated TRPC1. We suggest that the effects of KMUP-1 on TRPC1 might involve activating the cyclic guanosine monophosphate (cGMP)/PKG and cyclic adenosine monophosphate (cAMP)/PKA pathways and inhibiting the PKC pathway. We also used Fura 2-acetoxymethyl ester (Fura 2-AM, 5 µM) to measure the stored calcium release from the sarcoplasmic reticulum (SR) and calcium entry through SOCs in hypoxic PASMCs under treatment with thapsigargin (1 µM) and nifedipine (5 µM). In hypoxic conditions, store-operated calcium entry (SOCE) activity was enhanced in PASMCs, and KMUP-1 diminished this activity. In conclusion, KMUP-1 inhibited the expression of TRPC1 protein and the activity of SOC-mediated Ca2+ entry upon SR Ca2+ depletion in hypoxic PASMCs.

2.
Int J Mol Sci ; 23(24)2022 Dec 14.
Article in English | MEDLINE | ID: mdl-36555516

ABSTRACT

Autophagy facilitates the degradation of organelles and cytoplasmic proteins in a lysosome-dependent manner. It also plays a crucial role in cell damage. Whether loganin affects autophagy in chronic constriction injury (CCI)-induced neuropathic pain remains unclear. We investigated the neuroprotective effect of loganin on the autophagic-lysosomal pathway in the rat CCI model. Sprague-Dawley rats were divided into sham, CCI, sham + loganin, and CCI + loganin. Loganin (5 mg/kg/day) was intraperitoneally injected once daily, and rats were sacrificed on day 7 after CCI. This study focused on the mechanism by which loganin modulates autophagic flux after CCI. CCI enhanced the autophagic marker LC3B-II in the ipsilateral spinal cord. The ubiquitin-binding protein p62 binds to LC3B-II and integrates into autophagosomes, which are degraded by autophagy. CCI caused the accumulation of p62, indicating the interruption of autophagosome turnover. Loganin significantly attenuated the expression of Beclin-1, LC3B-II, and p62. Double immunofluorescence staining was used to confirm that LC3B-II and p62 were reduced by loganin in the spinal microglia and astrocytes. Loganin also lessened the CCI-increased colocalization of both proteins. Enhanced lysosome-associated membrane protein 2 (LAMP2) and pro-cathepsin D (pro-CTSD) in CCI rats were also attenuated by loganin, suggesting that loganin improves impaired lysosomal function and autophagic flux. Loganin also attenuated the CCI-increased apoptosis protein Bax and cleaved caspase-3. Loganin prevents CCI-induced neuropathic pain, which could be attributed to the regulation of neuroinflammation, neuronal autophagy, and associated cell death. These data suggest autophagy could be a potential target for preventing neuropathic pain.


Subject(s)
Cardiac Glycosides , Neuralgia , Animals , Rats , Autophagy , Constriction , Hyperalgesia/etiology , Hyperalgesia/complications , Iridoid Glycosides , Neuralgia/drug therapy , Neuralgia/etiology , Neuralgia/metabolism , Rats, Sprague-Dawley
3.
Biomed Pharmacother ; 145: 112447, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34808553

ABSTRACT

Eugenosedin-A (Eu-A) has been shown to protect against hyperglycemia- and hyperlipidemia-induced metabolic syndrome. We investigated the relationship of KATP channel activities and insulin secretion by Eu-A in vitro in pancreatic ß-cells, and examined the effect of Eu-A on streptozotocin (STZ)/nicotinamide (NA)-induced type 2 diabetes mellitus (T2DM) in vivo. We isolated pancreatic islets from adult male Wistar rats (250-350 g) and identified pancreatic ß-cells by the cell size, capacitance and membrane potential. Perforated patch-clamp and inside-out recordings were used to monitor the membrane potential (current-clamp mode) and channel activity (voltage-clamp mode) of ß-cells. The membrane potential of ß-cells was raised by Eu-A and reversed by the KATP channel activator diazoxide. Eu-A inhibited the KATP channel activity measured at - 60 mV and increased the intracellular calcium concentration ([Ca2+]i), resulting in enhanced insulin secretion. Eu-A also reduced Kir6.2 protein on the cell membrane and scattered in the cytosol under normal glucose conditions (5.6 mM). In our animal study, rats were divided into normal and STZ/NA-induced T2DM groups. Normal rats fed with regular chow were divided into control and control+Eu-A (5 mg/kg/day, i.p.) groups. The STZ/NA-induced diabetic rats fed with a high-fat diet (HFD) were divided into three groups: T2DM, T2DM+Eu-A (5 mg/kg/day, i.p.), and T2DM+glibenclamide (0.5 mg/kg/day, i.p.; a KATP channel inhibitor). Both Eu-A and glibenclamide decreased the rats' blood glucose, prevented weight gain, and enhanced insulin secretion. We found that Eu-A blocked pancreatic ß-cell KATP channels, caused membrane potential depolarization, and stimulated Ca2+ influx, thus increasing insulin secretion. Furthermore, Eu-A decreased blood glucose and increased insulin levels in T2DM rats. These results suggested that Eu-A might have clinical benefits for the control of T2DM and its complications.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hyperglycemia/drug therapy , Piperazines/pharmacology , Animals , Blood Glucose/drug effects , Diet, High-Fat , Glyburide/pharmacology , Hyperglycemia/etiology , Hypoglycemic Agents/pharmacology , Insulin Secretion/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , KATP Channels/metabolism , Male , Obesity/complications , Rats , Rats, Wistar
4.
Cells ; 9(9)2020 08 23.
Article in English | MEDLINE | ID: mdl-32842536

ABSTRACT

Diabetic peripheral neuropathy (DPN) is caused by hyperglycemia, which induces oxidative stress and inflammatory responses that damage nerve tissue. Excessive generation of reactive oxygen species (ROS) and NOD-like receptor protein 3 (NLRP3) inflammasome activation trigger the inflammation and pyroptosis in diabetes. Schwann cell dysfunction further promotes DPN progression. Loganin has been shown to have antioxidant and anti-inflammatory neuroprotective activities. This study evaluated the neuroprotective effect of loganin on high-glucose (25 mM)-induced rat Schwann cell line RSC96 injury, a recognized in vitro cell model of DPN. RSC96 cells were pretreated with loganin (0.1, 1, 10, 25, 50 µM) before exposure to high glucose. Loganin's effects were examined by CCK-8 assay, ROS assay, cell death assay, immunofluorescence staining, quantitative RT-PCR and western blot. High-glucose-treated RSC96 cells sustained cell viability loss, ROS generation, NF-κB nuclear translocation, P2 × 7 purinergic receptor and TXNIP (thioredoxin-interacting protein) expression, NLRP3 inflammasome (NLRP3, ASC, caspase-1) activation, IL-1ß and IL-18 maturation and gasdermin D cleavage. Those effects were reduced by loganin pretreatment. In conclusion, we found that loganin's antioxidant effects prevent RSC96 Schwann cell pyroptosis by inhibiting ROS generation and suppressing NLRP3 inflammasome activation.


Subject(s)
Glucose/adverse effects , Inflammasomes/metabolism , Iridoids/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Schwann Cells/metabolism , Animals , Humans , Rats
5.
Sci Rep ; 8(1): 16457, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30405207

ABSTRACT

Valproate (VPA) is a well-known drug for treating epilepsy and mania, but its action in neuropathic pain is unclear. We used a chronic constriction injury (CCI) model to explore whether VPA prevents neuropathic pain-mediated inflammation and neuronal death. Rats were treated with or without VPA. CCI + VPA rats were intraperitoneally injected with VPA (300 mg/kg/day) from postoperative day (POD) 1 to 14. We measured paw withdrawal latency (PWL) and paw withdrawal threshold (PWT) 1 day before surgery and 1, 3, 7, 14 days after CCI and harvested the sciatic nerves (SN), spinal cord (SC) and dorsal root ganglia (DRG) on POD 3, 7, and 14. PWL and PWT were reduced in CCI rats, but increased in CCI + VPA rats on POD 7 and POD 14. VPA lowered CCI-induced inflammatory proteins (pNFκB, iNOS and COX-2), pro-apoptotic proteins (pAKT/AKT and pGSK-3ß/GSK-3ß), proinflammatory cytokines (TNF-α and IL-1ß) and nuclear pNFκB activation in the SN, DRG and SC in CCI rats. COX-2 and pGSK-3 proteins were decreased by VPA on immunofluorescence analysis. VPA attenuated CCI-induced thermal and mechanical pain behaviors in rats in correlation with anti-neuroinflammation action involving reduction of pNFκB/iNOS/COX-2 activation and inhibition of pAKT/pGSK-3ß-mediated neuronal death from injury to peripheral nerves.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Neuralgia/physiopathology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Valproic Acid/pharmacology , Animals , Biomarkers , Cell Death/drug effects , Cytokines/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Neuralgia/drug therapy , Neuralgia/etiology , Proto-Oncogene Proteins c-akt/metabolism , Rats
6.
Pharmacol Rep ; 70(4): 746-752, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29936361

ABSTRACT

BACKGROUND: Diabetes mellitus is a metabolic disorder characterized by chronic hyperglycemia accompanied by impaired vascular and endothelial function. Activation of ATP-sensitive potassium (KATP) channels can protect endothelial function against hypertension and hyperglycemia. KMUP-1, a xanthine derivative, has been demonstrated to modulate K+-channel activity in smooth muscles. This study investigated protective mechanisms of KMUP-1 in impaired mesenteric artery (MA) reactivity in streptozotocin (STZ)-induced diabetic rats. METHODS: Rats were divided into three groups: control, STZ (65 mg/kg, ip) and STZ + KMUP-1 (5 or 10 mg/kg/day, ip). MA reactivity was measured by dual wire myograph. MA smooth muscle cells (MASMCs) were enzymatically dissociated and the KATP currents recorded by a whole-cell patch-clamp technique. RESULTS: STZ decreased MA KATP currents in a time-course dependent manner and achieved steady inhibition at day 14. In the MASMCs of STZ-treated rats, KMUP-1 partially recovered the KATP currents, suggesting that vascular KATP channels were activated by KMUP-1. K+ (80 mM KCl)-induced MA contractions in STZ-treated rats were higher than those of control rats. KMUP-1 significantly attenuated STZ-stimulated MA contractions in response to high K+, suggesting that KMUP-1 may partly restore the vascular reactivity of MAs. In addition, STZ decreased the expression of endothelial nitric oxide synthase (eNOS) and this effect was reversed by KMUP-1, suggesting that KMUP-1 could improve STZ-induced vascular endothelial dysfunction. CONCLUSION: KMUP-1 prevents STZ impairment of MA reactivity, eNOS levels and KATP channels, and accordingly protects against vascular dysfunction in diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , KATP Channels/metabolism , Mesenteric Arteries/physiopathology , Piperidines/pharmacology , Vascular Diseases/complications , Vascular Diseases/prevention & control , Xanthines/pharmacology , Animals , Diabetes Mellitus, Experimental/metabolism , Dose-Response Relationship, Drug , Male , Membrane Potentials/physiology , Mesenteric Arteries/metabolism , Myocytes, Smooth Muscle/physiology , Nitric Oxide Synthase Type III/biosynthesis , Potassium/pharmacology , Rats , Time Factors , Vascular Diseases/metabolism , Vascular Diseases/physiopathology
7.
Kaohsiung J Med Sci ; 34(3): 142-149, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29475461

ABSTRACT

This study examined the effects of eugenosedin-A (Eu-A) in a streptozotocin (STZ)/nicotinamide-induced rat model of type II diabetes mellitus (T2DM). Six-week-old Sprague-Dawley rats were randomly divided into three groups: (1) RD group, normal rats fed a regular diet (RD), (2) DM group, T2DM rats fed a high-fat diet, and (3) Eu-A group, T2DM rats fed a high fat diet plus oral Eu-A (5 mg/kg/day). After 30 days, the DM group had higher body weight, higher blood glucose and lower insulin levels than the RD group. The DM group also had increased protein expression of glycogen synthase kinase (GSK) in liver and skeletal muscle and decreased protein expression of insulin receptor (IR), insulin receptor substrate-1 (IRS-1), IRS-2, AMP-activated protein kinase (AMPK), glucose transporter-4 (GLUT-4), glucokinase (GCK), and peroxisome proliferator-activated receptor γ (PPAR-γ). STZ/nicotinamide-induced T2DM increased the expression of mitogen-activated protein kinases (MAPKs: p38, ERK, JNK) and inflammatory p65 protein. In the Eu-A treated T2DM rats, however, blood glucose was attenuated and the insulin concentration stimulated. Changes in IR, IRS-1 and IRS-2 proteins as well as AMPK, GLUT-4, GCK, GSK, PPAR-γ, MAPKs, and inflammatory p65 proteins were ameliorated. These results suggested that Eu-A alleviates STZ/nicotinamide-induced hyperglycemia by improving insulin levels and glucose metabolism, and inhibiting the MAPKs- and p65-mediated inflammatory pathway.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Mitogen-Activated Protein Kinases/genetics , Piperazines/pharmacology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat/adverse effects , Gene Expression Regulation , Glucokinase/genetics , Glucokinase/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Glycogen Synthase Kinases/genetics , Glycogen Synthase Kinases/metabolism , Hyperglycemia/chemically induced , Hyperglycemia/genetics , Hyperglycemia/pathology , Insulin/blood , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Niacinamide , PPAR gamma/genetics , PPAR gamma/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , Streptozocin , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
8.
Kaohsiung J Med Sci ; 32(12): 607-613, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27914611

ABSTRACT

We have demonstrated that KMUP-1 (7-[2-[4-(2-chlorobenzene)piperazinyl]ethyl]-1,3-dimethylxanthine) blunts monocrotaline-induced pulmonary arterial hypertension by altering Ca2+ sensitivity, K+-channel function, endothelial nitric oxide synthase activity, and RhoA/Rho kinase (ROCK) expression. This study further investigated whether KMUP-1 impedes uridine 5'-triphosphate (UTP)-inhibited delayed rectifying K+ (KDR) current in rat pulmonary arteries involved the RhoA/ROCK signaling. Pulmonary artery smooth muscle cells (PASMCs) were enzymatically dissociated from rat pulmonary arteries. KMUP-1 (30µM) attenuated UTP (30µM)-mediated membrane depolarization and abolished UTP-enhanced cytosolic Ca2+ concentration. Whole-cell patch-clamp electrophysiology was used to monitor KDR currents. A voltage-dependent KDR current was isolated and shown to consist of a 4-aminopyridine (5mM)-sensitive component and an insensitive component. The 4-aminopyridine sensitive KDR current was suppressed by UTP (30µM). The ROCK inhibitor Y27632 (30µM) abolished the ability of UTP to inhibit the KDR current. Like Y27632, KMUP-1 (30µM) similarly abolished UTP-inhibited KDR currents. Superfused protein kinase A and protein kinase G inhibitors (KT5720, 300nM and KT5823, 300nM) did not affect UTP-inhibited KDR currents, but the currents were restored by adding KMUP-1 (30µM) to the superfusate. KMUP-1 reversal of KDR current inhibition by UTP predominantly involves the ROCK inhibition. The results indicate that the RhoA/ROCK signaling pathway plays a key role in eliciting PASMCs depolarization caused by UTP, which would result in pulmonary artery constriction. KMUP-1 blocks UTP-mediated PASMCs depolarization, suggesting that it would prevent abnormal pulmonary vasoconstriction.


Subject(s)
Delayed Rectifier Potassium Channels/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Piperidines/pharmacology , Pulmonary Artery/cytology , Uridine Triphosphate/pharmacology , Xanthines/pharmacology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , 4-Aminopyridine/pharmacology , Amides/pharmacology , Animals , Calcium/metabolism , Cytosol/metabolism , Down-Regulation/drug effects , Female , Ion Channel Gating/drug effects , Myocytes, Smooth Muscle/drug effects , Pyridines/pharmacology , Rats, Sprague-Dawley , Signal Transduction/drug effects
9.
Drug Dev Res ; 75(2): 97-106, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24648214

ABSTRACT

In this study the effects of low-dose aspirin (5 mg/kg) on adhesion molecule and chemokine expression in a hyperlipidemic rat model. Six-week-old Sprague-Dawley (SD) rats were assigned to two control groups receiving either a regular diet or high-fat diet (HFD) and a treatment group fed HFD with 5 mg/kg aspirin for a 10-week period. Compared with the regular diet control group, the HFD control group had higher body weight, lower levels of high-density lipoprotein, higher concentrations of insulin, triglyceride, total cholesterol, and low-density lipoprotein, but no differences in blood glucose and glycated hemoglobin. The prothrombin time (PT) and activated partial thromboplastin time (aPTT) were clearly shortened in the HFD group. That group also had increased expression of intercellular adhesion molecule-1 (ICAM-1), ICAM-2, ICAM-3, vascular cell adhesion molecule (VCAM), platelet endothelial cell adhesion molecule (PECAM) and P-selectin in platelets and vascular adhesion protein-1 in lymphocyte and in aorta increased expressions of ICAM-1, ICAM-2, ICAM-3, VCAM, PECAM, E-selectin, monocyte chemoattractant protein-1 (MCP-1) and CCR2. The HFD rats also had increased PKCα, IκB kinase α (IKKα), p65, mitogen-activated protein kinases (MAPKs) (p38, c-Jun N-terminal kinases 1, extracellular signal-regulated kinase 1/2), and their phosphorylated forms. Low-dose aspirin improved HFD-induced hyperinsulinemia and hyperlipidemia, recovered PT and aPTT, inhibited upregulation of adhesion molecules and chemokines and reduced expression of PKCα, IKKα, p65, and MAPKs. Low-dose aspirin ameliorates HFD-induced hyperlipidemia and hyperinsulinemia, and prevents HFD-induced expression of adhesion molecules and chemokine formation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/therapeutic use , Cell Adhesion Molecules/blood , Chemokines/blood , Diet, High-Fat/adverse effects , Hyperlipidemias/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aspirin/administration & dosage , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Blood Platelets/immunology , Blood Platelets/metabolism , Body Weight/drug effects , Cell Adhesion Molecules/biosynthesis , Chemokines/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Hyperlipidemias/blood , Hyperlipidemias/complications , Hyperlipidemias/immunology , Lipids/blood , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Rats, Sprague-Dawley , Weight Gain/drug effects
10.
Kaohsiung J Med Sci ; 27(12): 538-43, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22208536

ABSTRACT

This study investigated whether KMUP-1, a xanthine-based derivative, inhibits L-type Ca(2+) currents (I(Ca,L)) in rat basilar artery smooth muscle cells (RBASMCs). We used whole cell patch-clamp recording to monitor Ba(2+) currents (I(Ba)) through L-type Ca(2+) channels (LTCCs). Under voltage-clamp conditions, holding at -40 mV, KMUP-1 (1, 3, 10 µM) inhibited I(Ba) in a concentration-dependent manner and its IC(50) value was 2.27 ± 0.45 µM. A high concentration of KMUP-1 (10 µM) showed without modifying the I(Ba) current-voltage relationship. On the other hand, the protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA, 1 µM) increase I(Ba) was inhibited by KMUP-1. Pretreatment with the PKC inhibitor chelerythrine (5 µM) intensified KMUP-1-inhibited I(Ba). However, the Rho kinase inhibitor Y-27632 (30 µM) failed to affect the I(Ba) inhibition by KMUP-1. In light of these results, we suggest that KMUP-1 inhibition of LTCCs in concentration- and voltage-dependent manners in RBASMCs may be due, at least in part, to its modulation of the PKC pathway.


Subject(s)
Basilar Artery/cytology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Myocytes, Smooth Muscle/drug effects , Piperidines/pharmacology , Protein Kinase C/metabolism , Xanthines/pharmacology , Animals , Barium Compounds/pharmacology , Benzophenanthridines/pharmacology , Chlorides/pharmacology , Female , Membrane Potentials/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction , rho-Associated Kinases/metabolism
11.
J Pharm Pharmacol ; 61(4): 517-25, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19298700

ABSTRACT

OBJECTIVES: Eugenosedin-A is a serotonin (5-hydroxytryptamine; 5-HT) 5-HT(1B/2A) and alpha(1)/alpha(2)/beta(1)-adrenoceptor blocker with anti-oxidative, anti-inflammatory and free-radical scavenging activities. Previous reports demonstrated that 5-HT(2A) blockers could diminish hyperlipidaemia. This study therefore aimed to investigate the possible uses and mechanisms of eugenosedin-A and other agents in treating hyperlipidaemia. METHODS: C57BL/6J mice were randomly divided into seven groups, fed a regular diet or a high-fat diet alone or supplemented with one of five agents: eugenosedin-A, ketanserin, prazosin, propranolol or atorvastatin (5 mg/kg p.o.) for 8 weeks. KEY FINDINGS: Compared with the regular diet, the mice fed the high-fat diet had significantly higher body weight and glucose, insulin and lipid levels. Brain malondialdehyde concentration was increased and liver glutathione peroxidase activity decreased. Addition of eugenosedin-A to the high-fat diet resulted in less weight gain and reduced hyperglycaemia, hyperinsulinaemia and hyperlipidaemia. Lipid and glucose homeostasis were related to decreased hepatic lipogenesis mRNAs and proteins (sterol regulatory element binding protein 1a, fatty acid synthase, sterol-CoA desaturase) and restored adipose peroxisome proliferator-activated receptor gamma expression. Eugenosedin-A also enhanced low-density lipoprotein receptor mRNA expression. CONCLUSIONS: Eugenosedin-A may improve plasma lipid metabolism by increasing low-density lipoprotein receptor and peroxisome proliferator-activated receptor gamma expression and diminishing sterol regulatory element binding protein 1a, fatty acid synthase and sterol-CoA desaturase. Reduction of plasma glucose and lipid levels may, in turn, reduce insulin concentration, which would explain the marked improvement in obesity-related hyperglycaemia and hyperlipidaemia. Furthermore, eugenosedin-A affected malondialdehyde concentration and glutathione peroxidase activity, suggesting it may have anti-peroxidation effects in mice fed a high-fat diet.


Subject(s)
Dietary Fats/pharmacology , Hyperglycemia/prevention & control , Hyperlipidemias/prevention & control , Lipid Peroxidation/drug effects , Piperazines/pharmacology , Adipose Tissue/metabolism , Animals , Atorvastatin , Blood Glucose/drug effects , Body Weight/drug effects , Brain/metabolism , Female , Glutathione Peroxidase/metabolism , Heptanoic Acids/pharmacology , Insulin/blood , Ketanserin/pharmacology , Lipogenesis/genetics , Liver/metabolism , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , Prazosin/pharmacology , Propranolol/pharmacology , Pyrroles/pharmacology , Superoxide Dismutase/metabolism , Weight Gain/drug effects
12.
J Cardiovasc Pharmacol ; 51(2): 154-61, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18287883

ABSTRACT

Eugenosedin-B is able to block serotonin (5-HT) and alpha/beta receptors and to inhibit platelet aggregation. In Wistar rats, intravenous injections of eugenosedin-B (2.4, 7.2, 12 micromoL/kg) caused a dose-dependent decrease in blood pressure and heart rate. In contrast, intracisternal injection of eugenosedin-B (0.3, 0.03 micromoL) and an alpha2-antagonist yohimbine (0.03 micromoL) increased blood pressure and heart rate. Eugenosedin-B and yohimbine prevented hypotension induced by intracisternal injection of an alpha2-agonist clonidine (38 pmol). In in vitro experiments, eugenosedin-B (10, 10, 10 M) competitively antagonized norepinephrine-, clonidine-, and 5-HT (10 to 10 M)-induced vasocontractions in isolated rat aorta. It also competitively antagonized the isoproterenol (10 to 10 M)-induced positive inotropic effects in isolated rat atrium. These findings clearly suggest that eugenosedin-B possesses alpha1, alpha2, beta1, and 5-HT2A receptor blocking activities. In isolated rabbit ear artery sensitized with 16 mM K, eugenosedin-B antagonized 5-nonyloxytryptamine- and 5-HT-induced vasocontractions, indicating it also blocked 5-HT1B and 5-HT2A receptors. In radioligand-binding experiments, eugenosedin-B had significant binding affinities on alpha1, alpha2, beta1, 5-HT1B, and 5-HT2A receptors. In human platelets, eugenosedin-B inhibited epinephrine and 5-HT-induced aggregations. It also had competitive binding effects in human platelet with [H]yohimbine (alpha2), [H]ketanserin (5-HT2A). We conclude that hypotensive and vasorelaxant effects of eugenosedin-B can be attributed to its multiple actions on the blockade of 5-HT1B, 5-HT2A, alpha1/2 and beta1 receptors, and its ability to reduce platelet aggregation attributed to its blockade of alpha2 and 5-HT2A receptors.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Blood Platelets/drug effects , Piperazines/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Serotonin Antagonists/pharmacology , Styrenes/pharmacology , Vasodilator Agents/pharmacology , Adrenergic alpha-Antagonists/administration & dosage , Adrenergic beta-Antagonists/administration & dosage , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Binding, Competitive , Blood Platelets/metabolism , Blood Pressure/drug effects , Ear/blood supply , Humans , Hypotension/chemically induced , Hypotension/prevention & control , In Vitro Techniques , Injections, Intravenous , Injections, Intraventricular , Male , Myocardial Contraction/drug effects , Piperazines/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Rabbits , Radioligand Assay , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1B/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, alpha-2/physiology , Serotonin 5-HT1 Receptor Antagonists , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/administration & dosage , Styrenes/administration & dosage , Vasoconstriction/drug effects , Vasodilator Agents/administration & dosage
13.
J Pharm Pharmacol ; 59(6): 879-89, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17637181

ABSTRACT

In this study, we investigate the protective effects of eugenosedin-A on p38 mitogen-activated protein kinase (MAPK), inflammatory nitric oxide (NO) and cyclooxygenase-2 (COX-2) pathways in a rat model of endotoxin shock. Rats were pretreated with eugenosedin-A, trazodone, yohimbine (1 mg kg(-1), i.v.), aminoguanidine or ascorbic acid (15 mg kg(-1), i.v.) 30 min before endotoxin challenge. Endotoxaemia was induced by a single i.v. injection of lipopolysaccharide (LPS, 10 mg kg(-1)). In rats not treated with eugenosedin-A, LPS increased plasma concentrations of NO and prostaglandin E(2) (PGE(2)), and levels of p38 MAPK, inducible NO synthase (iNOS) and COX-2 proteins in the liver, lung, aorta and lymphocytes. In the pre-treated rats, eugenosedin-A not only inhibited the LPS-induced NO and PGE(2) levels but also attenuated the LPS-induced increase in p38 MAPK and iNOS levels in the liver, aorta and lymphocytes. Eugenosedin-A also reduced LPS-induced COX-2 proteins in the aorta and lymphocytes. Likewise, aminoguanidine, ascorbic acid, yohimbine and trazodone were also found to decrease NO and PGE(2) concentrations after endotoxin challenge. While aminoguanidine and ascorbic acid also attenuated the LPS-induced increase in p38 MAPK, iNOS and COX-2 proteins in the aorta and lymphocytes, trazodone and yohimbine inhibited only the increase in p38 MAPK, iNOS and COX-2 proteins in lymphocytes. Finally, eugenosedin-A (10(-10)-10(-8) M) significantly inhibited the biphasic response induced by hydrogen peroxide (10(-6)-3 x 10(-5) M) in rat denudated aorta. Taken together, the results of this study indicate that eugenosedin-A, as well as ascorbic acid, can attenuate free-radical-mediated aortic contraction and relaxation. It may therefore be able to reduce the damage caused by septic shock by inhibiting formation of p38 MAPK, iNOS, COX-2 and free radicals.


Subject(s)
Antioxidants/pharmacology , Cyclooxygenase 2/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Piperazines/pharmacology , p38 Mitogen-Activated Protein Kinases/biosynthesis , Adrenergic alpha-Antagonists/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Ascorbic Acid/pharmacology , Blotting, Western , Endotoxemia/chemically induced , Endotoxemia/metabolism , Guanidines/pharmacology , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Lipopolysaccharides/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Organ Specificity , Rats , Rats, Wistar , Serotonin Antagonists/pharmacology , Trazodone/pharmacology , Up-Regulation , Yohimbine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...