Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Phytomedicine ; 92: 153734, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34536822

RESUMEN

BACKGROUND: Neuropathic pain has been shown to be modulated by the activation of the chemokine C-X-C motif ligand 12 (CXCL12)/chemokine CXC receptor 4 (CXCR4) dependent nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome. Loganin, an iridoid glycoside, was proven to prevent neuropathic pain, but its underlying mechanisms related to NLRP3 activation are still unknown. PURPOSE: This study investigated the underlying mechanisms of loganin's effect on chronic constriction injury (CCI)-induced NLRP3 inflammasome activation in the spinal cord. METHODS: Sprague-Dawley rats were randomly divided into four groups: sham, CCI, sham + loganin, and CCI + loganin. Loganin (5 mg/kg/day) was administered intraperitoneally starting the day after surgery. Paw withdrawal threshold (PWT) and latency (PWL) were assessed before CCI and on days 1, 3, 7 and 14 after CCI. Spinal cords were collected for western blots and immunofluorescence studies. RESULTS: Loganin prevented CCI-attenuated PWT and PWL, suggesting improved mechanical allodynia and thermal hyperalgesia. The expression of CXCL12, CXCR4, thioredoxin-interacting protein (TXNIP), NLRP3 inflammasome (NLRP3, ASC, and caspase-1), IL-1ß, and IL-18 were enhanced on day 7 after CCI, and all were reduced after loganin treatment. Dual immunofluorescence also showed that increased CXCL12, CXCR4, and NLRP3 were colocalized with NeuN (neuronal marker), GFAP (astrocyte marker), and Iba1 (microglial marker) on day 7 in the ipsilateral spinal dorsal horn (SDH). These immunoreactivities were attenuated in loganin-treated rats. Moreover, loganin decreased the assembly of NLRP3/ASC inflammasome after CCI in the ipsilateral SDH. Loganin appears to attenuate CCI-induced neuropathic pain by suppressing CXCL12/CXCR4-mediated NLRP3 inflammasome. CONCLUSION: Our findings suggest that loganin might be a suitable candidate for managing CCI-provoked neuropathic pain.


Asunto(s)
Inflamasomas , Neuralgia , Animales , Proteínas de Ciclo Celular , Hiperalgesia/tratamiento farmacológico , Iridoides , Proteína con Dominio Pirina 3 de la Familia NLR , Neuralgia/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Receptores CXCR4 , Receptores de Quimiocina , Médula Espinal
2.
Int J Med Sci ; 17(16): 2594-2602, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33029102

RESUMEN

Pulmonary hypertension (PH) is a lethal and rapidly progressing disorder if left untreated, but there is still no definitive therapy. An imbalance between vasoconstriction and vasodilation has been proposed as the mechanism underlying PH. Among the vasomediators of the pulmonary circulation is the renin-angiotensin system (RAS), the involvement of which in the development of PH has been proposed. Within the RAS, angiotensin-converting enzyme 2 (ACE2), which converts angiotensin (Ang) II into Ang-(1-7), is an important regulator of blood pressure, and has been implicated in cardiovascular disease and PH. In this study, we investigated the effects of the ACE2 activator diminazene aceturate (DIZE) on the development of PH secondary to left ventricular dysfunction. A model of PH secondary to left ventricular dysfunction was established in 6-week-old Wistar rats by ascending aortic banding for 42 days. The hemodynamics and pulmonary expression of ACE, Ang II, ACE2, Ang-(1-7), and the Ang-(1-7) MAS receptor were investigated in the early treatment group, which was administered DIZE (15 mg/kg/day) from days 1 to 42, and in the late treatment group, administered DIZE (15 mg/kg/day) from days 29 to 42. Sham-operated rats served as controls. DIZE ameliorated mean pulmonary artery pressure, pulmonary arteriolar remodeling, and plasma brain natriuretic peptide levels, in addition to reversing the overexpression of ACE and up-regulation of both Ang-(1-7) and MAS, in the early and late treatment groups. DIZE has therapeutic potential for preventing the development of PH secondary to left ventricular dysfunction through ACEII activation and the positive feedback of ANG-(1-7) on the MAS receptor. A translational study in humans is needed to substantiate these findings.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Diminazeno/análogos & derivados , Activadores de Enzimas/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Disfunción Ventricular Izquierda/complicaciones , Angiotensina I/metabolismo , Angiotensina II/metabolismo , Animales , Diminazeno/farmacología , Diminazeno/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Activadores de Enzimas/uso terapéutico , Humanos , Hipertensión Pulmonar/etiología , Fragmentos de Péptidos/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Disfunción Ventricular Izquierda/tratamiento farmacológico
3.
J Tradit Complement Med ; 10(4): 378-388, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32695655

RESUMEN

BACKGROUND AND AIM: Centella asiatica, Justicia gendarussa and Imperata cylindrica decoction (CJID) is efficacious for hypertension. NADPH (nicotinamide adenine dinucleotide phosphate) oxidase (NOX)-induced reactive oxygen species (ROS) generation modulates nuclear factor kappa B (NF-κB) activation and thus mediates hypertension-induced vascular remodeling. This research aims to investigate the anti-remodeling effect of CJID through the mechanism of NOXs-ROS-NF-κB pathway in spontaneously hypertensive rats (SHRs). EXPERIMENTAL PROCEDURE: CJID was orally administered once a day for five weeks in SHRs and normotensive-WKY (Wistar Kyoto) rats. All rats were sacrificed at the end of study and different assays were performed to determine whether CJID ameliorates vascular remodeling in SHRs, such as histological examination; lactate dehydrogenase (LDH), nitric oxide (NO), malondialdehyde (MDA) and superoxide dismutase (SOD) assays; superoxide and hydrogen peroxide (H2O2) generation assays, immunohistochemistry and immunofluorescence assays. . Changes in levels of inducible nitric oxide synthase (iNOS), NF-κB-p65, NF-κB inhibitor alpha/IκBα (inhibitory kappa B- alpha), phosphorylation of IκBα (p-IκBα) and NOX1, NOX2, NOX4 in the thoracic aorta were determined. RESULTS: Vascular remodeling indicators, media thickness, collagen and elastic accumulation in the thoracic aorta, of SHRs-treated CJID were attenuated. Redox homeostasis, aortic superoxide and hydrogen peroxide generation were decreased in SHRs-treated group. Aortic iNOS, p-IκBα, NF-κB-p65 and NOX1, NOX2, NOX4 expressions were suppressed. CONCLUSIONS: CJI treatment diminishes oxidative stress response in the thoracic aorta of SHRs via regulation of NOXs-ROS-NF-κB signaling pathway. These findings indicate that CJI possess protective effect against hypertension-induced vascular remodeling in SHRs.

4.
Phytomedicine ; 67: 153166, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31955133

RESUMEN

BACKGROUND: Peripheral nerve injury can produce chronic and ultimately neuropathic pain. The chronic constriction injury (CCI) model has provided a deeper understanding of nociception and chronic pain. Loganin is a well-known herbal medicine with glucose-lowering action and neuroprotective activity. PURPOSE: This study investigated the molecular mechanisms by which loganin reduced CCI-induced neuropathic pain. METHODS: Sprague-Dawley rats were randomly divided into four groups: sham, sham+loganin, CCI and CCI+loganin. Loganin (1 or 5 mg/kg/day) was injected intraperitoneally once daily for 14 days, starting the day after CCI. For behavioral testing, mechanical and thermal responses were assessed before surgery and on d1, d3, d7 and d14 after surgery. Sciatic nerves (SNs) were collected to measure proinflammatory cytokines. Proximal and distal SNs were collected separately for Western blotting and immunofluorescence studies. RESULTS: Thermal hyperalgesia and mechanical allodynia were reduced in the loganin-treated group as compared to the CCI group. Loganin (5 mg/kg/day) prevented CCI from inducing proinflammatory cytokines (TNF-α, IL-1ß), inflammatory proteins (TNF-α, IL-1ß, pNFκB, pIκB/IκB, iNOS) and receptor (TNFR1, IL-1R), adaptor protein (TRAF2) of TNF-α, and Schwann cell demyelination and axonal damage. Loganin also blocked IκB phosphorylation (p-IκB). Double immunofluorescent staining further demonstrated that pNFκB/pIκB protein was reduced by loganin in Schwann cells on d7 after CCI. In the distal stumps of injured SN, Schwann cell demyelination was correlated with pain behaviors in CCI rats. CONCLUSION: Our findings indicate that loganin improves CCI-induced neuroinflammation and pain behavior by downregulating TNF-α/IL-1ß-dependent NF-κB activation.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Iridoides/farmacología , FN-kappa B/metabolismo , Neuralgia/tratamiento farmacológico , Células de Schwann/efectos de los fármacos , Animales , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/metabolismo , Dolor Crónico/patología , Constricción , Citocinas/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/patología , Interleucina-1beta/metabolismo , Masculino , Neuralgia/metabolismo , Neuralgia/patología , Ratas Sprague-Dawley , Células de Schwann/metabolismo , Células de Schwann/patología , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Sci Rep ; 8(1): 5358, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29599473

RESUMEN

Metabolic syndrome (MetS) and ovarian hormone deficiency could affect bladder storage dysfunction. Epigallocatechin-3-gallate (EGCG), a polyphenolic compound in green tea, has been shown to protect against ovarian hormone deficiency induced overactive bladder (OAB). The present study investigated oxidative stress induced by MetS and bilateral ovariectomy (OVX), and elucidated the mechanism underlying the protective effect of EGCG (10 umol/kg/day) on bladder overactivity. Rats were fed with high fat high sugar (HFHS) diet to induce MetS and received ovariectomy surgery to deprive ovarian hormone. By dieting with HFHS for 6 months, rats developed MetS and OAB. MetS + OVX deteriorated bladder storage dysfunction more profound than MetS alone. MetS and MetS + OVX rats showed over-expression of inflammatory and fibrosis markers (1.7~3.8-fold of control). EGCG pretreatment alleviated storage dysfunction, and protected the bladders from MetS and OVX - induced interstitial fibrosis changes. Moreover, OVX exacerbated MetS related bladder apoptosis (2.3~4.5-fold of control; 1.8~2.6-fold of Mets group), enhances oxidative stress markers (3.6~4.3-fold of control; 1.8~2.2-fold of Mets group) and mitochondrial enzyme complexes subunits (1.8~3.7-fold of control; 1.5~3.4-fold of Mets group). EGCG pretreatment alleviated bladder apoptosis, attenuated oxidative stress, and reduced the mitochondrial and endoplasmic reticulum apoptotic signals. In conclusions, HFHS feeding and ovarian hormone deficiency enhances the generation of oxidative stress mediated through mitochondrial pathway. EGCG reduced the generation of oxidative stress and lessened bladder overactivity.


Asunto(s)
Apoptosis/efectos de los fármacos , Catequina/análogos & derivados , Síndrome Metabólico/complicaciones , Mitocondrias/efectos de los fármacos , Insuficiencia Ovárica Primaria/complicaciones , Vejiga Urinaria Hiperactiva/tratamiento farmacológico , Vejiga Urinaria Hiperactiva/etiología , Animales , Biomarcadores/sangre , Catequina/farmacología , Catequina/uso terapéutico , Dieta de Carga de Carbohidratos/métodos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Mitocondrias/metabolismo , Ovariectomía/métodos , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Ratas , Ratas Sprague-Dawley ,
6.
Kaohsiung J Med Sci ; 32(2): 55-67, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26944323

RESUMEN

KMUP-3 (7-{2-[4-(4-nitrobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) displays cardioprotection and increases cardiac output, and is suggested to increase cardiac performance and improve myocardial infarction. To determine whether KMUP-3 improves outcomes in hypoperfused myocardium by inducing Ca(2+) sensitization to oppose protein kinase (PK)G-mediated Ca(2+) blockade, we measured left ventricular systolic blood pressure, maximal rates of pressure development, mean arterial pressure and heart rate in rats, and measured contractility and expression of PKs/RhoA/Rho kinase (ROCK)II in beating guinea pig left atria. Hemodynamic changes induced by KMUP-3 (0.5-3.0 mg/kg, intravenously) were inhibited by Y27632 [(R)-(+)-trans-4-1-aminoethyl)-N-(4-Pyridyl) cyclohexane carboxamide] and ketanserin (1 mg/kg, intravenously). In electrically stimulated left guinea pig atria, positive inotropy induced by KMUP-3 (0.1-100µM) was inhibited by the endothelial NO synthase (eNOS) inhibitors N-nitro-l-arginine methyl ester (L-NAME) and 7-nitroindazole, cyclic AMP antagonist SQ22536 [9-(terahydro-2-furanyl)-9H-purin-6-amine], soluble guanylyl cyclase (sGC) antagonist ODQ (1H-[1,2,4] oxadiazolo[4,3-a] quinoxalin-1-one), RhoA inhibitor C3 exoenzyme, ß-blocker propranolol, 5-hydroxytryptamine 2A antagonist ketanserin, ROCK inhibitor Y27632 and KMUP-1 (7-{2-[4-(2-chlorobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) at 10µM. Western blotting assays indicated that KMUP-3 (0.1-10µM) increased PKA, RhoA/ROCKII, and PKC translocation and CIP-17 (an endogenous 17-kDa inhibitory protein) activation. In spontaneous right atria, KMUP-3 induced negative chronotropy that was blunted by 7-nitroindazole and atropine. In neonatal myocytes, L-NAME inhibited KMUP-3-induced eNOS phosphorylation and RhoA/ROCK activation. In H9c2 cells, Y-27632 (50µM) and PKG antagonist KT5823 [2,3,9,10,11,12-hexahydro-10R- methoxy-2,9-dimethyl-1-oxo-9S,12R-epoxy-1H-diindolo(1,2,3-fg:3',2',1'-kl) pyrrolo(3,4-i)(1,6)benzodiazocine-10-carboxylic acid, methyl ester] (3µM) reversed KMUP-3 (1-100µM)-induced Ca(2+)-entry blockade. GPCR agonist activity of KMUP-3 appeared opposed to KMUP-1, and increased cardiac output via Ca(2+) sensitization, and displayed cardioprotection via cyclic GMP/PKG-mediated myocardial preconditioning in animal studies.


Asunto(s)
Gasto Cardíaco/efectos de los fármacos , Cardiotónicos/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Piperidinas/farmacología , Xantinas/farmacología , Animales , Presión Sanguínea , Señalización del Calcio , Carbazoles/farmacología , Línea Celular , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Cobayas , Atrios Cardíacos/fisiopatología , Masculino , Transporte de Proteínas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo , Presión Ventricular
7.
J Lipid Res ; 56(11): 2070-84, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26351364

RESUMEN

The phosphodiesterase inhibitor (PDEI)/eNOS enhancer KMUP-1, targeting G-protein coupled receptors (GPCRs), improves dyslipidemia. We compared its lipid-lowering effects with simvastatin and explored hormone-sensitive lipase (HSL) translocation in hepatic fat loss. KMUP-1 HCl (1, 2.5, and 5 mg/kg/day) and simvastatin (5 mg/kg/day) were administered in C57BL/6J male mice fed a high-fat diet (HFD) by gavage for 8 weeks. KMUP-1 inhibited HFD-induced plasma/liver TG, total cholesterol, and LDL; increased HDL/3-hydroxy-3-methylglutaryl-CoA reductase (HMGR)/Rho kinase II (ROCK II)/PPARγ/ABCA1; and decreased liver and body weight. KMUP-1 HCl in drinking water (2.5 mg/200 ml tap water) for 1-14 or 8-14 weeks decreased HFD-induced liver and body weight and scavenger receptor class B type I expression and increased protein kinase A (PKA)/PKG/LDLRs/HSL expression and immunoreactivity. In HepG2 cells incubated with serum or exogenous mevalonate, KMUP-1 (10(-7)∼10(-5) M) reversed HMGR expression by feedback regulation, colocalized expression of ABCA1/apolipoprotein A-I/LXRα/PPARγ, and reduced exogenous geranylgeranyl pyrophosphate/farnesyl pyrophosphate (FPP)-induced RhoA/ROCK II expression. A guanosine 3',5'-cyclic monophosphate (cGMP) antagonist reversed KMUP-1-induced ROCK II reduction, indicating cGMP/eNOS involvement. KMUP-1 inceased PKG and LDLRs surrounded by LDL and restored oxidized LDL-induced PKA expresion. Unlike simvastatin, KMUP-1 could not inhibit (14)C mevalonate formation. KMUP-1 could, but simvastatin could not, decrease ROCK II expression by exogenous FPP/CGPP. KMUP-1 improves HDL via PPARγ/LXRα/ABCA1/Apo-I expression and increases LDLRs/PKA/PKG/HSL expression and immunoreactivity, leading to TG hydrolysis to lower hepatic fat and body weight.


Asunto(s)
Hiperlipoproteinemias/tratamiento farmacológico , Hipolipemiantes/farmacología , Piperidinas/farmacología , Xantinas/farmacología , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Dieta Alta en Grasa/efectos adversos , Evaluación Preclínica de Medicamentos , Células Hep G2 , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Hiperlipoproteinemias/etiología , Hipolipemiantes/uso terapéutico , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/fisiología , Lipoproteínas HDL/sangre , Lipoproteínas LDL/metabolismo , Hígado/patología , Masculino , Ácido Mevalónico/metabolismo , Ratones Endogámicos C57BL , PPAR gamma/metabolismo , Piperidinas/uso terapéutico , Receptores de LDL/metabolismo , Receptores Depuradores de Clase B/metabolismo , Sistemas de Mensajero Secundario , Esterol Esterasa/metabolismo , Xantinas/uso terapéutico
8.
J Ethnopharmacol ; 138(2): 373-81, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21963569

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: We investigated the antiproliferative effects of baicalein, isolated from Scutellaria baicalensis (Huang-qin), on ET-1-mediated pulmonary artery smooth muscle cells (PASMCs) proliferation and the mechanisms underlying these effects. MATERIALS AND METHODS: Intrapulmonary artery smooth muscle cells were isolated and cultured from female Sprague-Dawley rats and used during passages 3-6. The proliferation of PASMCs was quantified by cell counting and XTT assay. The protein expression of TRPC1 and PKCα were determined by western blotting. The cell cycle pattern was assayed by flow cytometry. The intracellular calcium concentrations ([Ca(2+)](i)) were measured using the fluorescent indicator fura-2-AM and flow cytometry. RESULTS: Baicalein (0.3-3 µM) inhibited PASMCs proliferation, promoted cell cycle progression, enhanced [Ca(2+)](i) levels, increased capacitative Ca(2+) entry (CCE), upregulated the canonical transient receptor potential 1 (TRPC1) channel and membrane protein kinase Cα (PKCα) expression induced by ET-1 (0.1 µM). The PKC activator PMA (1 µM) reversed the inhibitory effects of baicalein on ET-1-induced upregulation of TRPC1 expression and S phase accumulation, while the PKC inhibitor chelerythrine (1 µM) potentiated baicalein-mediated G(2)/M phase arrest and TRPC1 channel inhibition. CONCLUSION: Our findings suggest that baicalein protects against ET-1-induced PASMCs proliferation via modulation of the PKC-mediated TRPC channel.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Endotelina-1/fisiología , Flavanonas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Scutellaria baicalensis/química , Canales Catiónicos TRPC/antagonistas & inhibidores , Animales , Calcio/metabolismo , Células Cultivadas , Femenino , Flavanonas/aislamiento & purificación , Citometría de Flujo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteína Quinasa C-alfa/antagonistas & inhibidores , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPC/metabolismo
9.
Phytomedicine ; 17(10): 760-70, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20171070

RESUMEN

Baicalin isolated from Scutellaria baicalensis is a traditional Chinese herbal medicine used for cardiovascular dysfunction. The ionic mechanism of the vasorelaxant effects of baicalin remains unclear. We investigated whether baicalin relaxes mesenteric arteries (MAs) via large-conductance Ca2+-activated K+ (BK(Ca)) channel activation and voltage-dependent Ca2+ channel (VDCC) inhibition. The contractility of MA was determined by dual wire myograph. BK(Ca) channels and VDCCs were measured using whole-cell recordings in single myocytes, enzymatically dispersed from rat MAs. Baicalin (10-100 microM) attenuated 80 mM KCl-contracted MA in a concentration-related manner. L-NAME (30 microM) and indomethacin (10 microM) little affected baicalin (100 microM)-induced vasorelaxations. Contractions induced by iberiotoxin (IbTX, 0.1 microM), Bay K8644 (0.1 microM) or PMA (10 microM) were abolished by baicalin 100 microM. In MA myocytes, baicalin (0.3-30 microM) enhanced BK(Ca) channel activity in a concentration-dependent manner. Increased BK(Ca) currents were abolished by IbTX (0.1 microM). Baicalin-mediated (30 microM) BK(Ca) current activation was significantly attenuated by an adenylate cyclase inhibitor (SQ 22536, 10 microM), a soluble guanylate cyclase inhibitor (ODQ, 10 microM), competitive antagonists of cAMP and cGMP (Rp-cAMP, 100 microM and Rp-cGMP, 100 microM), and cAMP- and cGMP-dependent protein kinase inhibitors (KT5720, 0.3 microM and KT5823, 0.3 microM). Perfusate with PMA (0.1 microM) abolished baicalin-enhanced BK(Ca) currents. Additionally, baicalin (0.3-30 microM) reduced the amplitude of VDCC currents in a concentration-dependent manner and abolished VDCC activator Bay K8644-enhanced (0.1 microM) currents. Baicalin produced MA relaxation by activating BK(Ca) and inhibiting VDCC channels by endothelium-independent mechanisms and by stimulating the cGMP/PKG and cAMP/PKA pathways.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Flavonoides/farmacología , Arterias Mesentéricas/efectos de los fármacos , Canales de Potasio Calcio-Activados/agonistas , Scutellaria baicalensis/química , Animales , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Femenino , Flavonoides/aislamiento & purificación , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
10.
J Pharm Pharmacol ; 61(4): 517-25, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19298700

RESUMEN

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.


Asunto(s)
Grasas de la Dieta/farmacología , Hiperglucemia/prevención & control , Hiperlipidemias/prevención & control , Peroxidación de Lípido/efectos de los fármacos , Piperazinas/farmacología , Tejido Adiposo/metabolismo , Animales , Atorvastatina , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Femenino , Glutatión Peroxidasa/metabolismo , Ácidos Heptanoicos/farmacología , Insulina/sangre , Ketanserina/farmacología , Lipogénesis/genética , Hígado/metabolismo , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , PPAR gamma/genética , PPAR gamma/metabolismo , Prazosina/farmacología , Propranolol/farmacología , Pirroles/farmacología , Superóxido Dismutasa/metabolismo , Aumento de Peso/efectos de los fármacos
11.
J Nat Prod ; 71(6): 933-40, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18489163

RESUMEN

In this study, we investigate the anticancer effect of isoobtusilactone A (IOA), a constituent isolated from the leaves of Cinnamomum kotoense, on human non-small cell lung cancer (NSCLC) A549 cells. IOA was found to induce the arrest of G2-M phase, induce apoptosis, increase sub-G1, and inhibit the growth of these cells. Further investigation revealed that IOA's blockade of the cell cycle was associated with increased levels of p21/WAF1, p27 (kip1), and p53. In addition, IOA triggered the mitochondrial apoptotic pathway, as indicated by an increase in Bax/Bcl-2 ratios, resulting in a loss of mitochondrial membrane potential, release of cytochrome c, activation of caspase-9 and caspase-3, and cleavage of PARP. We also found the generation of reactive oxygen species (ROS) to be a critical mediator in IOA-induced inhibition of A549 cell growth. In antioxidant and NO inhibitor studies, we found that by pretreating A549 cells with either N-acetylcystenine (NAC), catalase, mannitol, dexamethasone, trolox, or L-NAME we could significantly decrease IOA production of ROS. Moreover, using NAC to block ROS, we could significantly suppress IOA-induced antiproliferation, antimigration, and anti-invasion. Finally, we found that IOA inhibited the migration and invasion of A549 cell migration and invasion. Taken together, these results suggest that IOA has anticancer effects on A549 cells.


Asunto(s)
Alcanos/aislamiento & purificación , Alcanos/farmacología , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Cinnamomum/química , Lactonas/aislamiento & purificación , Lactonas/farmacología , Mitocondrias/efectos de los fármacos , Plantas Medicinales/química , Alcanos/química , Antineoplásicos Fitogénicos/química , Caspasa 3/análisis , Caspasa 3/metabolismo , Caspasa 9/análisis , Caspasa 9/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/análisis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocromos c/análisis , Citocromos c/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Lactonas/química , Hojas de la Planta/química , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/química , Taiwán , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/metabolismo
12.
J Cardiovasc Pharmacol ; 44(3): 363-71, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15475835

RESUMEN

Urgosedin is a newly synthesized compound especially with serotonergic and alpha-adrenergic blocking actions. In rat isolated thoracic aorta, urgosedin competitively antagonized norepinephrine-, clonidine-, and serotonin-induced vasocontractions in a concentration-dependent manner. In radioligand binding experiments, urgosedin had significant binding affinities on alpha1/alpha2, 5-HT1A, 5-HT1B and 5-HT2A receptors. Intravenous injection of lipopolysaccharide (LPS) produced a biphasic hypotension in normotensive rats. Although intravenous injection of urgosedin caused minor depressor actions in the normotensive Wistar rat, urgosedin significantly attenuated the secondary prolonged hypotension produced by LPS. The plasma levels of cytokines (IL-1beta, IL-6, TNF-alpha, and IFN-gamma) and hypoglycemia induced by LPS were also reduced by urgosedin. Moreover, the acute survival rates (350 minutes) of endotoxic shock increased from 0% (LPS group) to 100% in the groups pretreated with urgosedin. In RAW264.7 cells, urgosedin inhibited LPS-induced inducible nitric oxide synthase (iNOS) expression. In conclusion, our data suggest that urgosedin was a newly potent serotonergic and mild alpha-adrenergic blocking agent. Its prevention of LPS-induced hypotension and hypoglycemia might partially mediate through its inhibition activities on the iNOS expression and cytokines formation. Urgosedin might be an effective pharmacological agent against LPS-induced hypotension, hypoglycemia, and the formation of pro-inflammatory mediators.


Asunto(s)
Derivados del Benceno/farmacocinética , Citocinas/antagonistas & inhibidores , Hipoglucemia/prevención & control , Hipotensión/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/efectos adversos , Piperazinas/farmacocinética , Administración Oral , Antagonistas Adrenérgicos alfa/administración & dosificación , Antagonistas Adrenérgicos alfa/química , Antagonistas Adrenérgicos alfa/farmacocinética , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Derivados del Benceno/administración & dosificación , Derivados del Benceno/química , Glucemia , Línea Celular , Citocinas/efectos adversos , Citocinas/biosíntesis , Citocinas/clasificación , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Hipoglucemia/inducido químicamente , Hipoglucemia/fisiopatología , Hipotensión/inducido químicamente , Hipotensión/mortalidad , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/antagonistas & inhibidores , Ratones , Ratones Endogámicos ICR , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Norepinefrina/farmacología , Piperazinas/administración & dosificación , Piperazinas/química , Profármacos/efectos adversos , Ratas , Ratas Wistar , Serotonina/farmacología , Serotoninérgicos/administración & dosificación , Serotoninérgicos/química , Serotoninérgicos/farmacocinética , Pruebas de Toxicidad Aguda/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA