Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Zhonghua Xin Xue Guan Bing Za Zhi ; 48(11): 930-935, 2020 Nov 24.
Artigo em Chinês | MEDLINE | ID: mdl-33210864

RESUMO

Objective: To evaluate the changes of left ventricular function in patients with ST segment elevation myocardial infarction (STEMI) before PCI and within 24 hours after PCI by layer-specific strain, and to explore the value of this new assessment method for quantitative monitoring on the myocardial function in STEMI patients. Methods: A total of 40 patients with acute anterior wall myocardial infarction, who underwent PCI in Affiliated Hospital of Jiangsu University during July 2017 to July 2018, were included in this prospective cohort study. According to the symptom to balloon time (STB), the patients were divided into STB ≤6 hours group (26 cases) and STB 6-12 hours group (14 cases). Echocardiography was performed before, immediately, 3 hours and 24 hours after PCI. Echocardiographic indexes including endocardial myocardial longitudinal strain (LS-endo), 18-segment full-thickness myocardial longitudinal strain (LS) of left ventricle and left ventricular global longitudinal strain (GLS) were measured. The mean LS-endo and LS values of myocardial segments in infarcted area (IALS-endo, IALS) and the mean LS-endo and LS values of myocardial segments in non-infarcted area (NIALS-endo, NIALS) were calculated. Results: There were 34 males and 6 females in this cohort and age was (62±10) years. In STB≤6 hours group, the IALS-endo value ((13.7±4.9)% vs. (10.0±2.7)%, P<0.05) and NIALS-endo value ((17.0±2.9)% vs. (14.6±2.9)%, P<0.05) were significantly higher at 24 hours after PCI than those before PCI. In the group of STB 6-12 hours, IALS-endo decreased immediately after PCI ((6.7±3.3)% vs. (11.9±6.5)%, P<0.05), and there was a rising trend at 3 hours after PCI (P>0.05). At 24 hours after PCI, the index was higher than that immediately after PCI ((13.6±8.4)% vs. (6.7±3.3)%, P<0.05). The NIALS-endo value was significantly higher at 24 hours after PCI than that before PCI ((17.1±2.1)% vs. (14.5±3.2)%, P<0.05). In the STB 6-12 hours group, the decrease rate of IALS-endo immediately after PCI was higher than that in the STB ≤6 hours group (93% (13/14) vs. 35% (9/26), P<0.001). In STB ≤6 hours group, the NIALS value at 24 hours after PCI was higher than that before PCI (P<0.05), and there was no significant difference in IALS, NIALS and GLS at other time points (P>0.05). Conclusions: Layered LS is superior to full-thickness LS and GLS in evaluating left ventricular function in STEMI patients. LS measured by echocardiography can continuously and quantitatively evaluate the changes of left ventricular myocardial function in STEMI patients before and after PCI.


Assuntos
Intervenção Coronária Percutânea , Infarto do Miocárdio com Supradesnível do Segmento ST , Ecocardiografia , Feminino , Humanos , Masculino , Estudos Prospectivos , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico por imagem , Infarto do Miocárdio com Supradesnível do Segmento ST/cirurgia , Função Ventricular Esquerda
2.
J Appl Microbiol ; 2018 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-30129993

RESUMO

AIMS: The purpose of this work was to find an efficient enzyme to synthesize d-3,4-dihydroxyphenyllactic acid (d-DSS). METHODS AND RESULTS: Nineteen lactic acid bacteria strains were screened for production of d-DSS using 3,4-dihydroxyphenylpyruvic acid (DPA) as a substrate. Lactobacillus reuteri JN516 exhibited the highest d-DSS yield. A nonspecific coenzyme, d-lactate dehydrogenase (d-LDH82319), from L. reuteri JN516 with high DPA reducing activity was identified. This enzyme reduced DPA to form d-DSS with excellent optical purity (enantioselectivity >99%). Its molecular weight was 35 kDa based on SDS-PAGE migration. The Michaelis-Menten constant (Km ), turnover number (kcat ), and catalytic efficiency (kcat /Km ) of d-LDH82319 for DPA were 0·09 mmol l-1 , 2·17 s-1 and 24·07 (mmol l-1 )-1  s-1 , respectively, with NADH as the coenzyme. The (Km ), (kcat ) and (kcat /Km ) of d-LDH82319 for DPA were 0·10 mmol l-1 , 0·13 s-1 and 1·30 (mmol l-1 )-1  s-1 , respectively, with NADPH as the coenzyme. The optimum temperature and pH of d-LDH82319 were 25°C and pH 8 respectively. Additionally, d-LDH82319 had a broad substrate range for alpha-keto acids, among which the activity of reducing pyruvate was the strongest; therefore, it belongs to the group of d-lactate dehydrogenases. d-LDH82319 and glucose dehydrogenase (GDH) were coexpressed to produce d-DSS from DPA. CONCLUSIONS: d-LDH82319 from L. reuteri JN516 with high DPA reducing activity has the characteristics of a nonspecific coenzyme. SIGNIFICANCE AND IMPACT OF THE STUDY: d-LDH82319 is the first reported coenzyme nonspecific d-lactate dehydrogenase with DPA-reducing activity. The coexpression system provided an effective method to produce d-DSS.

3.
Br J Pharmacol ; 152(2): 207-15, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17603552

RESUMO

BACKGROUND AND PURPOSE: Angiogenesis is a crucial step in tumour growth and metastasis. Ginsenoside-Rb1 (Rb1), the major active constituent of ginseng, potently inhibits angiogenesis in vivo and in vitro. However, the underlying mechanism remains unknown. We hypothesized that the potent anti-angiogenic protein, pigment epithelium-derived factor (PEDF), is involved in regulating the anti-angiogenic effects of Rb1. EXPERIMENTAL APPROACHES: Rb1-induced PEDF was determined by real-time PCR and western blot analysis. The anti-angiogenic effects of Rb1 were demonstrated using endothelial cell tube formation assay. Competitive ligand-binding and reporter gene assays were employed to indicate the interaction between Rb1 and the oestrogen receptor (ER). KEY RESULTS: Rb1 significantly increased the transcription, protein expression and secretion of PEDF. Targeted inhibition of PEDF completely prevented Rb1-induced inhibition of endothelial tube formation, suggesting that the anti-angiogenic effect of Rb1 was PEDF specific. Interestingly, the activation of PEDF occurred via a genomic pathway of ERbeta. Competitive ligand-binding assays indicated that Rb1 is a specific agonist of ERbeta, but not ERalpha. Rb1 effectively recruited transcriptional activators and activated an oestrogen-responsive reporter gene. Furthermore, Rb1-mediated PEDF activation and the subsequent inhibition of tube formation were blocked by the ER antagonist ICI 182,780 or transfection of ERbeta siRNA, indicating ERbeta dependence. CONCLUSIONS AND IMPLICATIONS: Here we show for the first time that the Rb1 suppressed the formation of endothelial tube-like structures through modulation of PEDF via ERbeta. These findings demonstrate a novel mechanism of the action of this ginsenoside that may have value in anti-cancer and anti-angiogenesis therapy.


Assuntos
Inibidores da Angiogênese/farmacologia , Células Endoteliais/efeitos dos fármacos , Receptor beta de Estrogênio/agonistas , Proteínas do Olho/metabolismo , Ginsenosídeos/farmacologia , Fatores de Crescimento Neural/metabolismo , Serpinas/metabolismo , Linhagem Celular , Células Endoteliais/fisiologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor beta de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Fulvestranto , Humanos , RNA Mensageiro/metabolismo
4.
Curr Med Chem ; 14(12): 1371-80, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17504218

RESUMO

Panax ginseng C.A. Meyer, one of the most popular and valued herbs, has been used extensively in traditional Chinese medicine for thousands of years. More than thirty ginsenosides, the pharmacologically active ingredients in ginseng, have been identified with various sugar moieties attached at the C-3, C-6 and C-20 positions of the steroidal skeleton. We herein review the current literature on the pharmacological effects of ginsenosides on the modulation of angiogenesis, dysregulations of which contribute towards many pathological conditions. Regarding the adaptogenic property of ginseng, the effects of ginsenosides on central nervous system are also discussed. Recent researches have pointed to the steroid hormone receptors as the target molecules to elicit the diverse cellular and physiological activities of ginseng. We believe that understanding the interaction between ginsenosides and various steroid hormone receptors may provide clues to unravel the secret of ginseng.


Assuntos
Ginsenosídeos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Cognição/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Ginsenosídeos/uso terapêutico , Humanos , Modelos Moleculares , Doenças Neurodegenerativas/tratamento farmacológico
5.
Neuropharmacology ; 52(3): 827-35, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17123556

RESUMO

Ginsenoside-Rg1, the pharmacologically active component isolated from ginseng, demonstrated neuroprotective effects on primary cultured rat nigral neurons against rotenone toxicity. Rotenone, a common household pesticide known for its specific and irreversible mitochondria complex I inhibition, has been suggested to be the causal agent of Parkinson's disease (PD) by inducing degeneration of cells in the substantial nigra. The present study demonstrated that co-treatment of rotenone and Rg1 could reduce rotenone-induced cell death by 58% (SEM=+/-5.60; N=3). Rotenone-induced mitochondria membrane potential (MMP, DeltaPsim) depletion was restored and elevated by at least 38% (SEM=+/-2.15; N=3) by Rg1. In addition, Rg1 prevented cytochrome c release from the mitochrondrial membrane and increased the phosphorylation inhibition of the pro-apoptotic protein Bad through activation of the PI3K/Akt pathway. The protective effects of Rg1 was blocked by glucocorticoid receptor antagonist RU486, indicating that the action of Rg1 is mediated through glucocorticoid receptor (GR). In conclusion, Rg1 inhibits the mitochondrial apoptotic pathway and increases the survival chance of the primary cultured nigral neurons against rotenone toxicity. Thus, Rg1 and its related compounds may be developed as protective agents against neurodegenerative diseases induced by mitochondrial toxins.


Assuntos
Ginsenosídeos/farmacologia , Inseticidas/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Rotenona/toxicidade , Substância Negra/citologia , Análise de Variância , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , Membranas Mitocondriais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo
6.
Biochem Pharmacol ; 72(4): 437-45, 2006 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-16793023

RESUMO

Aberrant angiogenesis is an essential step for the progression of solid tumors. Thus anti-angiogenic therapy is one of the most promising approaches to control tumor growth. In this study, we examined the ability of 20(R)-ginsenoside Rg3 (Rg3), one of the active compounds present in ginseng root, to interfere with the various steps of angiogenesis. Rg3 was found to inhibit the proliferation of human umbilical vein endothelial cells (HUVEC) with an IC50 of 10 nM in Trypan blue exclusion assay. Rg3 (1-10(3) nM) also dose dependently suppressed the capillary tube formation of HUVEC on the Matrigel in the presence or absence of 20 ng/ml vascular endothelial growth factor (VEGF). The VEGF-induced chemoinvasion of HUVEC and ex vivo microvascular sprouting in rat aortic ring assay were both significantly attenuated by Rg3. In addition, Rg3 (150 and 600 nM) remarkably abolished the basic fibroblast growth factor (bFGF)-induced angiogenesis in an in vivo Matrigel plug assay. The Matrix metalloproteinases (MMPs), such as MMP-2 and MMP-9, which play an important role in the degradation of basement membrane in angiogenesis and tumor metastasis present in the culture supernatant of Rg3-treated aortic ring culture were found to decrease in their gelatinolytic activities. Taken together, these data underpin the anti-tumor property of Rg3 through its angiosuppressive activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Ginsenosídeos/farmacologia , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/química , Animais , Aorta/efeitos dos fármacos , Aorta/enzimologia , Aorta/crescimento & desenvolvimento , Capilares/efeitos dos fármacos , Capilares/crescimento & desenvolvimento , Proliferação de Células/efeitos dos fármacos , Colágeno , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ginsenosídeos/administração & dosagem , Ginsenosídeos/química , Humanos , Técnicas In Vitro , Injeções Subcutâneas , Laminina , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Proteoglicanas , Ratos , Fatores de Crescimento do Endotélio Vascular/farmacologia
7.
FEBS Lett ; 580(13): 3211-6, 2006 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-16696977

RESUMO

We here provide definitive evidence that ginsenoside-Rg1, the pharmacologically active component of ginseng, is a functional ligand of the glucocorticoid receptor (GR) as determined by fluorescence polarization assay. Rg1 increased the phosphorylation of GR, phosphatidylinositol-3 kinase (PI3K), Akt/PKB and endothelial nitric oxide synthase (eNOS) leading to increase nitric oxide (NO) production in human umbilical vein endothelial cell. Rg1-induced eNOS phosphorylation and NO production were significantly reduced by RU486, LY294,002, or SH-6. Also, knockdown of GR completely eliminated the Rg1-induced NO production. This study revealed that Rg1 can indeed serve as an agonist ligand for GR and the activated GR can induce rapid NO production from eNOS via the non-transcriptional PI3K/Akt pathway.


Assuntos
Ginsenosídeos/farmacologia , Óxido Nítrico Sintase Tipo III/efeitos dos fármacos , Óxido Nítrico/biossíntese , Receptores de Glucocorticoides/agonistas , Cromonas/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Inibidores Enzimáticos/farmacologia , Ginsenosídeos/metabolismo , Humanos , Mifepristona/farmacologia , Morfolinas/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais
8.
Angiogenesis ; 8(3): 205-16, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16328162

RESUMO

The major active constituents of ginseng are ginsenosides, and Rg(1) is a predominant compound of the total extract. Recent studies have demonstrated that Rg(1) can promote angiogenesis in vivo and in vitro. In this study, we used a DNA microarray technology to elucidate the mechanisms of action of Rg(1). We report that Rg(1) induces the proliferation of HUVECs, monitored using [(3)H]-thymidine incorporation and Trypan blue exclusion assays. Furthermore, Rg(1) (150-600 nM) also showed an enhanced tube forming inducing effect on the HUVEC. Rg(1) was also demonstrated to promote angiogenesis in an in vivo Matrigel plug assay, and increase endothelial sprouting in the ex vivo rat aorta ring assay. Differential gene expression profile of HUVEC following treatment with Rg(1) revealed the expression of genes related to cell adhesion, migration and cytoskeleton, including RhoA, RhoB, IQGAP1, CALM2, Vav2 and LAMA4. Our results suggest that Rg(1) can promote angiogenesis in multiple models, and this effect is partly due to the modulation of genes that are involved in the cytoskeletal dynamics, cell-cell adhesion and migration.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Ginsenosídeos/metabolismo , Neovascularização Fisiológica/fisiologia , Panax/química , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Colágeno , Proteínas do Citoesqueleto/metabolismo , Primers do DNA , Combinação de Medicamentos , Ginsenosídeos/farmacologia , Humanos , Laminina , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Extratos Vegetais/metabolismo , Extratos Vegetais/farmacologia , Proteoglicanas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina/metabolismo , Trítio
9.
Angiogenesis ; 8(1): 3-12, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16132613

RESUMO

Sinomenine is an alkaloid extracted from the Chinese medicinal plant, Sinomenium acutum, which has been utilized to treat rheumatoid arthritis (RA) in China for over 2000 years. Sinomenine has been shown to mediate a wide range of pharmacological actions which includes anti-inflammatory and anti-rheumatic effects. RA has been classified as a chronic immune-mediated disease that exhibits overlapping manifestation of inflammatory, abnormal cellular and hormonal immune responses with synovial hyperplasia. Since, angiogenesis is recognized to play a critical role in the development of RA and anti-angiogenic therapy has been proposed as a new therapeutic strategy for treatment of RA, we would like to see if sinomenine possesses anti-angiogenic property. In this study, sinomenine inhibited bFGF-induced proliferation of human umbilical vein endothelial cells (HUVEC) and arrested its cell cycle in G1 phase. Sinomenine disrupted tube formation of HUVEC on Matrigel and suppressed the chemotaxis of HUVEC. In addition, sinomenine reduced neovascularization in Matrigel plug assay as well as microvascular outgrowth in rat aorta ring sprouting assay. These results suggest that sinomenine inhibited bFGF-induced angiogenesis in vitro and in vivo. As the leukocytes-endothelial adhesive interactions also play an important role in inflammation, we found that sinomenine reduced the transmigration of granulocytic differentiated HL60 cells across IL-1beta activated HUVEC monolayer. Therefore, the inhibition of leukocytes migration across blood vessel walls and the anti-angiogenic effect of sinomenine may contribute towards its therapeutic mechanisms in alleviating the pathogenesis of RA.


Assuntos
Inibidores da Angiogênese/farmacologia , Células Endoteliais/efeitos dos fármacos , Morfinanos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Extratos Vegetais/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Artrite Reumatoide/tratamento farmacológico , Feminino , Fatores de Crescimento de Fibroblastos , Células HL-60 , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Morfinanos/uso terapêutico , Neovascularização Patológica/induzido quimicamente , Fitoterapia , Extratos Vegetais/uso terapêutico , Ratos , Ratos Sprague-Dawley , Veias Umbilicais/citologia
10.
Br J Pharmacol ; 132(3): 760-6, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11159729

RESUMO

1. Somatostatin and the stable octapeptide analogues, octreotide and angiopeptin, were examined for their ability to stimulate the release of tritium from [(3)H]-arachidonic acid pre-loaded CHO-K1 cells expressing human recombinant sst(2), sst(3) or sst(5) receptors. 2. Somatostatin stimulated tritium release (pEC(50)) through the sst(2) (7.8+/-0.1) and sst(5) (7.3+/-0.2), but not the sst(3) receptor. Octreotide behaved as a full (sst(2) receptor) or partial agonist (sst(5) receptor), whereas angiopeptin behaved as a weak partial agonist at both receptor types. 3. Maximum responses to somatostatin through both receptor types were significantly reduced by pertussis toxin, whereas pEC(50) estimates were unaffected. 4. Inhibition of MEK1 or Src, but not PKA, PI 3-kinases or tyrosine kinases, by reportedly selective inhibitors reduced sst(2)-mediated responses by somatostatin, but not angiopeptin. A selective inhibitor of PKC (Ro-31-8220) reduced both somatostatin and angiopeptin responses. 5. These data provide further evidence for partial agonist activity of synthetic peptides of somatostatin. Furthermore, the somatostatin receptor signalling mechanisms which mediate arachidonic acid mobilization appear to be multiple and complex.


Assuntos
Ácido Araquidônico/metabolismo , Fármacos Cardiovasculares/farmacologia , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Animais , Transporte Biológico/efeitos dos fármacos , Células CHO , Cricetinae , Octreotida/farmacologia , Oligopeptídeos/farmacologia , Peptídeos/síntese química , Peptídeos/química , Peptídeos/farmacologia , Peptídeos Cíclicos , Toxina Pertussis , Receptores de Somatostatina/genética , Transdução de Sinais/efeitos dos fármacos , Somatostatina/farmacologia , Trítio , Fatores de Virulência de Bordetella/farmacologia
11.
J Pathol ; 192(2): 234-42, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11004701

RESUMO

In atherosclerosis, leukocyte migration into the plaque is thought to occur across the endothelium of the arterial lumen. However, intraplaque microvessels have been noted. While the significance of, and stimuli for these are uncertain, it seems possible that they may represent a second portal of entry for leukocytes into the plaque. This study performed a basic characterization of intraplaque microvessels and tested the hypothesis that the novel angiogenic factor thymidine phosphorylase (TP) is expressed in atherosclerosis. Immunocytochemistry was performed on aortic and coronary plaques and morphometry on coronary plaques. In plaques from both sites, macrophages, foam cells, and giant cells were immunoreactive for the angiogenic factors TP and vascular endothelial growth factor. Venule-like intraplaque microvessels expressed endothelial leukocyte adhesion molecules HLA-DR and ICAM-1, in contrast to the endothelium overlying the plaque. In coronary plaques, there was a correlation between severity of stenosis and plaque microvascular density. These results are consistent with a role for plaque macrophage/foam cell TP in stimulating plaque angiogenesis. While attention has focused on dysfunction of the endothelium overlying the plaque, microvascular endothelium may also represent a portal of entry for leukocytes into established plaques.


Assuntos
Arteriosclerose/patologia , Neovascularização Patológica/enzimologia , Timidina Fosforilase/metabolismo , Aorta/metabolismo , Aorta/patologia , Arteriosclerose/metabolismo , Doença da Artéria Coronariana/metabolismo , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/metabolismo , Células Espumosas/metabolismo , Células Gigantes/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Macrófagos/metabolismo
12.
J Asian Nat Prod Res ; 2(3): 237-43, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11256699

RESUMO

Five new abietane derivatives which have a commonly rearranged abietane skeleton contained a 17(15-->16),18(4-->3)-diabeo-abietane framework, mandarones D-H, were isolated from the stem of Clerodendrum nmantarinorum Diels (Verbenaceae). The structures were characterized as (16S)-12,16-epoxy-11-hydroxy-17(15-->16), 18(4-->3)-diabeo-abieta-3,5,8,11,13-pentaene-7-one (mandarone D, 1), 12,16-epoxy-11,14-dihydroxy-17(15-->16),18(4-->3)-diabeo-abieta-3,5,8,11, 13,15-hexaene-7-one (mandarone E, 2), 12,16-epoxy-6,11,14-trihydroxy-17(15-->16),18(4-->3)-diabeo-abieta-3,5,8,11,13,15-hexaene-7-one (mandarone F, 3),12,16-epoxy-11,14-dihydroxy-6-methoxy-17(15-->16),18(4-->3)-diabeo-abieta-3,5,8,11,13,15-hexaene-2,7-dione (mandarone G, 4) and 12,16-epoxy-11,14-dihydroxy-17(15-->16),18(4-->3)-diabeo-abieta-3,5,8,11,13,15-hexaene-1,7-dione (mandarone H, 5) respectively, mainly based on the spectral analysis and by comparison with those of closely related compounds.


Assuntos
Diterpenos/química , Medicamentos de Ervas Chinesas , Lamiaceae/química , Cromatografia , Diterpenos/isolamento & purificação , Humanos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Estrutura Molecular , Caules de Planta/química
13.
Br J Pharmacol ; 124(2): 323-30, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9641549

RESUMO

1. Somatostatin (SRIF) exerts antiproliferative effects, and angiopeptin (an sst2/sst5 receptor-selective analogue) has recently been evaluated in clinical trials for the prophylaxis of restenosis following coronary angioplasty. Using an in vitro model of cell growth we have examined the effects of SRIF and angiopeptin on cell proliferation in CHO-K1 cells stably transfected with the human or rat recombinant sst2 or sst5 receptor and compared these with their effects on rat aortic vascular smooth muscle cells (VSMC) expressing endogenous somatostatin receptors. 2. In CHO-KI cells, expressing either human or rat recombinant sst2 or sst5 receptors, or in rat aortic VSMC, SRIF and angiopeptin (0.1-1000 nM) had no effect on basal re-growth of cells into a denuded area of a previously confluent monolayer. In contrast, basic fibroblast growth factor (bFGF, 10 ng ml(-1)) stimulated re-growth of these cells. 3. SRIF (0.1-1000 nM) caused a concentration-dependent inhibition of the bFGF-stimulated re-growth in CHO-K1 cells expressing human sst2 (h sst2) or sst5 (h sst5) receptors (pIC50=8.05+/-0.03 and 8.56+/-0.12, respectively). In contrast, angiopeptin (0.1-1000 nM) acted as a partial agonist at the h sst2 receptor (44.6+/-2.7% inhibition of the bFGF-stimulated re-growth at 100 nM; pIC50=8.69+/-0.25) but was devoid of any agonist activity at the h sst5 receptor. 4. In CHO-K1 cells stably expressing rat recombinant sst2 (r sst2) or sst5 (r sst5) receptors, SRIF (0.1-1000 nM) was able to inhibit the bFGF-stimulated re-growth (pIC50=7.98+/-24 and 8.50+/-0.12, respectively). Angiopeptin (0.1-1000 nM) caused a concentration-dependent inhibition of bFGF-stimulated re-growth at the r sst2 receptor (pIC50=8.08+/-0.24) but acted as a partial agonist at the r sst5 receptor (maximum response= 57.7+/-3.6% inhibition of bFGF-stimulated re-growth at 100 nM; pIC50=8.60+/-0.16). 5. Although angiopeptin was inactive as an agonist at the h sst5 receptor, 100 nM angiopeptin potently antagonized the SRIF-induced inhibition of proliferation in CHO h sst5 (estimated pKB= 10.4+/-0.3). 5-Hydroxytryptamine (0.1 nM-10 microM) also inhibited bFGF-stimulated re-growth (pIC50=8.36+/-0.11) and angiopeptin had no effect on this response (pKB<7). 6. SRIF (0.1-1000 nM) caused a concentration-dependent (pIC50=8.04+/-0.08) inhibition of bFGF-stimulated re-growth in VSMC, whereas angiopeptin displayed weak agonist activity, only inhibiting bFGF-stimulated re-growth at concentrations greater than 100 nM. Angiopeptin (100 nM) caused a rightward displacement of the concentration-effect curve to SRIF with an estimated pKB value of 7.70+/-0.12. 7. These findings suggest that the low intrinsic activity of angiopeptin at the h sst2 receptor, combined with its lack of agonist activity at the h sst5 receptor, may explain the poor clinical efficacy of angiopeptin in trials for coronary artery restenosis, which contrasts with encouraging data found in equivalent in vivo animal studies.


Assuntos
Antineoplásicos/farmacologia , Fármacos Cardiovasculares/farmacologia , Antagonistas de Hormônios/farmacologia , Oligopeptídeos/farmacologia , Receptores de Somatostatina/efeitos dos fármacos , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Animais , Aorta Torácica , Células CHO , Divisão Celular/efeitos dos fármacos , Cricetinae , Relação Dose-Resposta a Droga , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Peptídeos Cíclicos , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/genética , Transfecção
14.
Br J Pharmacol ; 125(8): 1630-3, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9886753

RESUMO

We have investigated the actions of somatostatin (SRIF) and angiopeptin on cell proliferation of CHO-K1 cells expressing the recently cloned rat sst2(b) receptor (CHOsst2(b)) and compared these to their effects in cells expressing the sst2(a) receptor (CHOsst2(a)). In contrast to the sst2(a) receptor, the sst2(b) receptor did not mediate inhibition of bFGF (10 ng ml(-1))-stimulated re-growth and cell proliferation. Rather, SRIF (0.1-1000 nM) and angiopeptin (0.1-1000 nM) stimulated basal re-growth and proliferation of CHOsst2(b) cells in a concentration-dependent manner (estimated pEC50 values of 7.8 and 7.9, respectively). The opposite effects of SRIF on cell proliferation mediated through the two sst2 receptor isoforms were both abolished by 18 h pre-treatment with pertussis toxin. The proliferative effect via the sst2(b) receptor was also abolished by the tyrosine kinase inhibitor, genistein. In conclusion, the present study shows that the rat sst2(a) and sst2(b) receptor splice variants mediate opposite effects on cell proliferation.


Assuntos
Receptores de Somatostatina/fisiologia , Animais , Células CHO/citologia , Células CHO/efeitos dos fármacos , Células CHO/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Cricetinae , Relação Dose-Resposta a Droga , Fator 2 de Crescimento de Fibroblastos/farmacologia , Antagonistas de Hormônios/farmacologia , Oligopeptídeos/farmacologia , Peptídeos Cíclicos , Toxina Pertussis , Ratos , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/genética , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Fatores de Virulência de Bordetella/farmacologia
15.
Angiogenesis ; 2(1): 67-80, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-14517377

RESUMO

The processes of wound repair were investigated using an in vitro model of mechanical injury on confluent cell monolayers of either human umbilical vein endothelial cells (HUVEC), aortic endothelial (RAEC) or smooth muscle cells (VSMC) of the rat. A mechanical wounder was used to produce 11 parallel (400 microm wide) lesions across the monolayer and the movement of cells into the denuded area was quantified using image analysis. The lesioned area recovered completely in 72h, with proliferation occurring after 24h for endothelial cells and 18h for VSMC, as detected by an increase in cell numbers. The cell migration inhibitor Taxol (1ng/ml) abolished the increase in repair of HUVEC monolayers in the first 24h of repair, while actinomycin D had no effect before 24h but thereafter abolished the further repair which was associated with increased cell numbers. Repair of endothelial cells was accelerated by basic fibroblast growth factor (bFGF), vascular endothelial growth factor or platelet-derived growth factor-BB (PDGF), and in VSMC both bFGF and PDGF increased repair. This simple in vitro model of mechanical injury allows a quantitative study of the repair processes of a previously confluent monolayer and thus is a representation of mechanical damage in vivo.

17.
Br J Pharmacol ; 120(7): 1302-11, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9105706

RESUMO

1. The vasoconstrictor peptide antiotensin II (AII) can stimulate angiogenesis, an important process in wound healing, tumour growth and chronic inflammation. To elucidate mechanisms underlying AII-enhanced angiogenesis, we have studied a subcutaneous sponge granuloma model in the rat by use of 133Xe clearance, morphometry and quantitative in vitro autoradiography. 2. When injected directly into the sponge, AII (1 nmol day-1) increased 133Xe clearance from, and fibrovascular growth in sponge granulomas, indicating enhanced angiogenesis 6 to 12 days after implantation. This AII-enhanced angiogenesis was inhibited by daily doses (100 nmol/sponge) of the specific but subtype non-selective AII receptor antagonist (Sar1, Ile8)AII, and by the selective non-peptide AT1 receptor antagonists losartan and DuP 532. In contrast, AII-enhanced neovascularization was not inhibited by the AT2 receptor antagonist PD123319, nor was it mimicked by the AT2 receptor agonist CGP42112A (each at 100 nmol/sponge day-1). 3. AI (1 nmol/sponge day-1), the angiotensin converting enzyme (ACE) inhibitors captopril (up to 100 micrograms/sponge day-1) and lisinopril (40 micrograms/sponge day-1), or AII receptor antagonists did not affect angiogenesis in the absence of exogenous AII. 4. [125I]-(Sar1, Ile8)AII binding sites with characteristics of AT1 receptors were localized to microvessels and to non-vascular cells within the sponge stroma from 4 days after implantation, and were at higher density than in skin throughout the study. 5. [125I]-(Sar1, Ile8)AII binding sites with characteristics of AT2 receptors were localized to non-vascular stromal cells, were of lower density and appeared later than did AT1 sites. 6. The ACE inhibitor [125I]-351A bound to sites with characteristics of ACE, 14 days after sponge implantation. [125I]-351A bound less densely to sponge stroma than to skin. 7. We propose that AII can stimulate angiogenesis, acting via AT1 receptors within the sponge granuloma. AT1 and AT2 receptors and ACE develop sequentially during microvascular maturation, and the role of the endogenous angiotensin system in angiogenesis will depend on the balanced local expression of its various components. Pharmacological modulation of this balance may provide novel therapeutic approaches in angiogenesis-dependent diseases.


Assuntos
Granuloma de Corpo Estranho/metabolismo , Neovascularização Patológica , Peptidil Dipeptidase A/metabolismo , Receptores de Angiotensina/metabolismo , 1-Sarcosina-8-Isoleucina Angiotensina II/metabolismo , Antagonistas de Receptores de Angiotensina , Animais , Sítios de Ligação , Granuloma de Corpo Estranho/enzimologia , Radioisótopos do Iodo , Poríferos , Ratos , Receptores de Angiotensina/agonistas
18.
Br J Pharmacol ; 122(4): 663-70, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9375962

RESUMO

1. The aim of the present study was to determine the effect of somatostatin (SRIF) on mitogen-induced regeneration of rat aortic vascular smooth muscle cells (VSMC) and for comparison Chinese hamster ovary (CHO)-K1 cells expressing human recombinant sst5 receptors (CHOsst5), following partial denudation of a confluent cell monolayer. Regeneration was assessed by measuring areas of recovery into the denuded area and by counting total cell numbers. 2. In VSMC, SRIF (0.1 nM - 1 microM) had no effect on the basal levels of regeneration but caused a concentration-dependent inhibition (pIC50 8.0-8.6) of the stimulated regeneration induced by submaximal concentrations of basic fibroblast growth factor (bFGF, 10 ng ml[-1]), platelet-derived growth factor-BB (PDGF, 5 ng ml[-1]) or endothelin-1 (ET-1, 100 nM). SRIF (pIC50 8.8) also inhibited bFGF-induced regeneration of CHOsst5 cells. 3. In VSMC, the inhibitory action of SRIF on the regeneration induced by bFGF (10 ng ml[-1]) was due to an anti-proliferative effect, rather than an effect on cell migration, as SRIF (0.1 nM - 1 microM) abolished bFGF-induced increases in total cell numbers. The bFGF-induced increase in cell numbers was also abolished by actinomycin D (0.1 microg ml[-1]). 4. The sst5 receptor-selective agonist, L-362,855 (pIC50 10.5), was about 100 times more potent than SRIF at inhibiting bFGF-induced regeneration of both VSMC and CHOsst5 cells whilst the sst2 receptor-selective agonist, BIM-23027 (pIC50 6.8), was approximately 20 times weaker than SRIF. 5. The sst5 receptor antagonist, BIM-23056 (100 nM), antagonized SRIF-induced inhibition of bFGF-induced regeneration in both VSMC and CHOsst5 cells (estimated pKB values 8.8 and 8.3, respectively). 6. SRIF-induced inhibition of bFGF-induced regeneration of VSMC and CHOsst5 cells was abolished by pretreating cells with pertussis toxin (100 ng ml[-1]) for 20 h. 7. These findings suggest that SRIF-induced inhibition of the proliferation of rat aortic VSMC is mediated via activation of receptors which are similar to human sst5 receptors. Furthermore this inhibitory effect is transduced via pertussis toxin-sensitive Gi/Go proteins.


Assuntos
Aorta/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Somatostatina/análogos & derivados , Animais , Aorta/citologia , Células CHO , Divisão Celular , Células Cultivadas , Cricetinae , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Técnicas In Vitro , Masculino , Músculo Liso Vascular/citologia , Oligopeptídeos/farmacologia , Toxina Pertussis , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/genética , Proteínas Recombinantes/genética , Somatostatina/farmacologia , Fatores de Virulência de Bordetella/farmacologia
19.
Histochem J ; 28(11): 759-69, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8968728

RESUMO

Angiogenesis is an essential component of wound healing and inflammation. In the rat subcutaneous sponge implantation model, angiogenesis can be enhanced by administration of the sensory neuropeptide, substance P. We have used quantitative in vitro receptor autoradiography and immunohistochemistry to investigate the development of endogenous neurovascular regulatory systems in the newly-formed granulation tissue of this model. The fraction of endothelial cells immunoreactive for proliferating cell nuclear antigen, endothelial fractional area, and 133Xe clearance were used as measures of endothelial proliferation, neovascularization, and blood flow, respectively. Endothelial proliferation occurred predominantly in tissues surrounding the sponge, and peaked before neovascularization of sponge stroma and the establishment of sponge blood flow. Substance P-containing sensory nerves and specific, high affinity substance P binding sites with characteristics of neurokinin receptors of the NK1 subclass, were localized to microvessels surrounding the sponge at all time points. Lower density substance P binding sites were localized to newly formed microvessels within the sponge stroma, progressively increasing in density from day 4 to day 14. Nerve fibres were observed in the stroma of only 2 of 6 sponges at day 14, and none at earlier time points. These data support the hypothesis that substance P-enhanced angiogenesis in this model results from a direct action on microvascular NK1 receptors. Neovascularization is a sequential process, with early endothelial proliferation followed by new vessel formation and increased blood flow, with maturation of endogenous neurovascular regulatory systems occurring late in this process in inflamed tissues.


Assuntos
Dermatite/patologia , Neovascularização Patológica , Receptores da Neurocinina-1/metabolismo , Animais , Sítios de Ligação , Granuloma/patologia , Microcirculação , Poríferos , Ratos , Pele/irrigação sanguínea , Pele/inervação , Substância P/metabolismo
20.
Br J Pharmacol ; 118(7): 1584-91, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8842418

RESUMO

1. Subcutaneous implantation of sterile polyether sponges elicited a reproducible neovascular response in rats, as determined by blood flow measurement with a 133Xe clearance technique and confirmed histologically. This model was used to monitor the levels of two cytokines during angiogenesis and to compare the activities of angiostatic steroids and anti-inflammatory steroids. 2. Initial experiments followed the neovascular development over a 20-day period. Daily local injection of hydrocortisone caused a dose-dependent (0.5, 5 and 50 micrograms per sponge) inhibition of the basal sponge-induced angiogenesis. However, daily systemic treatment of hydrocortisone (2, 10 and 50 mg kg-1, s.c.) was less effective at inhibiting angiogenesis, and this inhibition was not sustained by day 20 after sponge implantation. 3. To investigate the involvement of cytokines during the course of angiogenesis, we measured the endogenous levels of tumour necrosis factor-alpha (TNF-alpha) and interleukin 6 (IL-6) in sponge implants. Levels of IL-6 and TNF-alpha peaked at day 7 and day 11 after implantation, respectively. These cytokine levels subsided through the completion of angiogenesis by day 20. 4. Subsequent experiments were carried out over a 14-day period. Among the three angiostatic steroids tested, U-24067 (6 alpha-fluoro-17,21 - dihydroxy-16 alpha-methylpregna -4,9(11)-diene-3,20-dione-21-acetate) showed a dose-dependent inhibition (0.5, 5 and 50 micrograms per sponge per day) of sponge-induced angiogenesis. Tetrahydro-S was also effective at 5 micrograms doses, but medroxyprogesterone failed to affect the angiogenic response. None of these steroids caused atrophies of the spleen and thymus. 5. Daily local injection of dexamethasone (0.5 microgram per sponge) inhibited the basal sponge-induced angiogenesis almost completely. Although higher doses of dexamethasone (5 and 50 micrograms per sponge) did not produce further inhibition of angiogenesis, they caused severe spleen and thymus weight losses, indicative of immunosuppression. 6. At the daily dose of 5 micrograms per sponge, dexamethasone inhibited angiogenesis and produced a marked reduction in the levels of TNF-alpha and IL-6 at day 14. In contrast, hydrocortisone, U-24067 and tetrahydro-S did not influence the levels of TNF-alpha and IL-6. 7. We concluded that the anti-angiogenic activity of angiostatic steroids and anti-inflammatory steroids in the rat sponge model is independent of their ability to reduce the production of TNF-alpha and IL-6. The differential effects of angiostatic and anti-inflammatory steroids suggest that U-24067 and its derivatives may have therapeutic potential in the management of angiogenic diseases such as rheumatoid arthritis.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Inflamação/metabolismo , Inflamação/patologia , Neovascularização Patológica/fisiopatologia , Animais , Linhagem Celular , Hidrocortisona/farmacologia , Inflamação/induzido quimicamente , Interleucina-6/metabolismo , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo , Radioisótopos de Xenônio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA