Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biol Pharm Bull ; 41(4): 555-563, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29607928

RESUMO

Artemisinin was discovered in 1971 as a constituent of the wormwood genus plant (Artemisia annua). This plant has been used as an herbal medicine to treat malaria since ancient times. The compound artemisinin has a sesquiterpene lactone bearing a peroxide group that offers its biological activity. In addition to anti-malarial activity, artemisinin derivatives have been reported to exert antitumor activity in cancer cells, and have attracted attention as potential anti-cancer drugs. Mechanisms that might explain the antitumor activities of artemisinin derivatives reportedly induction of apoptosis, angiogenesis inhibitory effects, inhibition of hypoxia-inducible factor-1α (HIF-1α) activation, and direct DNA injury. Reactive oxygen species (ROS) generation is involved in many cases. However, little is known about the mechanism of ROS formation from artemisinin derivatives and what types of ROS are produced. Therefore, we investigated the iron-induced ROS formation mechanism by using artesunate, a water-soluble artemisinin derivative, which is thought to be the underlying mechanism involved in artesunate-mediated cell death. The ROS generated by the coexistence of iron(II), artesunate, and molecular oxygen was a hydroxyl radical or hydroxyl radical-like ROS. Artesunate can reduce iron(III) to iron(II), which enables generation of ROS irrespective of the iron valence. We found that reduction from iron(III) to iron(II) was activated in the acidic rather than the neutral region and was proportional to the hydrogen ion concentration.


Assuntos
Antineoplásicos/farmacologia , Artemisininas/farmacologia , Ferro/farmacologia , Oxigênio/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Antimaláricos/farmacologia , Antipirina/análogos & derivados , Antipirina/farmacologia , Artesunato , Sobrevivência Celular/efeitos dos fármacos , Edaravone , Sequestradores de Radicais Livres/farmacologia , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Oxirredução
2.
Pharmacology ; 90(5-6): 324-31, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23052181

RESUMO

BACKGROUND/AIMS: Tumor necrosis factor-α (TNF-α) is known to involve the progression of renal dysfunction through its cytotoxicity and proinflammatory effects such as the induction of intercellular adhesion molecule (ICAM)-1 expression in vascular endothelial cells (ECs). Olmesartan, one of the angiotensin II type 1 receptor blockers (ARBs), has been reported to show protective effects on injured ECs by some causal factors of renal disorder other than angiotensin II. However, the effects of olmesartan on TNF-α-induced glomerular EC damage have not been investigated. In the present study, we investigated the effects of RNH-6270, an active metabolite of olmesartan, on TNF-α-induced human glomerular EC (HGEC) damage to clarify the renoprotective mechanisms of ARBs. METHODS: Cultured HGECs were stimulated by TNF-α, and then cell viability and cytotoxicity were measured by MTT assay and lactate dehydrogenase release assay, respectively. TNF-α-induced oxidative stress was estimated by dihydroethidium assay and lucigenin chemiluminescence assay. ICAM-1 expression and the phosphorylations of mitogen-activated protein kinases were measured using Western blotting assay. RESULTS: RNH-6270 suppressed cell death and the increase in ICAM-1 expression induced by TNF-α via the inhibition of reactive oxygen species in HGECs. CONCLUSION: Our findings suggested that olmesartan might have protective effects against TNF-α-induced glomerular EC dysfunction.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Imidazóis/farmacologia , Tetrazóis/farmacologia , Fator de Necrose Tumoral alfa , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Glomérulos Renais/citologia , Espécies Reativas de Oxigênio/metabolismo
3.
Chem Pharm Bull (Tokyo) ; 59(2): 208-14, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21297301

RESUMO

Recently, increasing evidence suggests that the antihypertensive drug nifedipine acts as a protective agent for endothelial cells, and that the activity is unrelated to its calcium channel blocking. Nifedipine is unstable under light and reportedly decomposes to a stable nitrosonifedipine (NO-NIF). NO-NIF has no antihypertensive effect, and it has been recognized as a contaminant of nifedipine. The present study for the first time demonstrated that NO-NIF changed to a NO-NIF radical in a time-dependent manner when it interacted with human umbilical vein endothelial cells (HUVECs). The electron paramagnetic resonance (EPR) signal of NO-NIF radicals in HUVECs showed an asymmetric pattern suggesting that the radicals were located in the membrane. The NO-NIF radicals had radical scavenging activity for 1,1-diphenyl-2-picrylhydrazyl, whereas neither NO-NIF nor nifedipine did. In addition, the NO-NIF radical more effectively quenched lipid peroxides than NO-NIF or nifedipine. Furthermore, NO-NIF attenuated the superoxide-derived free radicals in HUVECs stimulated with LY83583, and suppressed iron-nitrilotriacetic acid (Fe-NTA)-induced cytotoxicity in rat pheochromocytoma (PC12) cells. Our findings suggest that NO-NIF is a candidate for a new class of antioxidative drugs that protect cells against oxidative stress.


Assuntos
Antioxidantes/química , Antioxidantes/metabolismo , Nifedipino/química , Nifedipino/metabolismo , Fotólise , Animais , Antioxidantes/farmacologia , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Nifedipino/farmacologia , Células PC12 , Fotólise/efeitos dos fármacos , Ratos
4.
Nephrol Dial Transplant ; 25(2): 364-72, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19812233

RESUMO

BACKGROUND: Clinical studies have shown that angiotensin II (Ang II) type 1 (AT1) receptor blockers (ARBs) are able to provide renoprotection independent of their blood pressure lowering effects. ARBs also are reported to suppress oxidative stress, inflammation and certain other cellular responses in a receptor-independent manner. We investigated the effects of an ARB, olmesartan, on the cell migration induced by platelet-derived growth factor (PDGF), a major mitogen involved in the pathogenesis of glomerulonephritis in rat mesangial cells (RMCs). METHODS: Cell migration was determined by a modified Boyden chamber assay. The intracellular signalling pathway was examined by western blotting. AT1 receptor expression was knocked down by small interfering RNAs. The intracellular reactive oxygen species (ROS) was measured by using a fluorescent probe. The O(2)(.-) scavenging activities were studied by the electron paramagnetic resonance-spin trapping method. RESULTS: PDGF-induced cell migration was inhibited by olmesartan in AT1 receptor knockdown RMCs. Olmesartan attenuated big mitogen-activated protein (MAP) kinase 1 (BMK1) and Src activation by PDGF in AT1 receptor knockdown RMCs. PDGF-induced BMK1 activation was suppressed by the Src family tyrosine kinase inhibitors, indicating that Src exists upstream of BMK1. The NADPH oxidase inhibitors inhibited not only PDGF-induced BMK1 and Src activation but also RMC migration. The elevation in ROS generation induced by PDGF was decreased by olmesartan. Olmesartan displayed neither directly ROS scavenging activity nor the inhibition of ROS-mediated intracellular signalling in RMCs. CONCLUSIONS: Olmesartan attenuates ROS generation by PDGF, leading to the subsequent inhibition of Src/ BMK1/migration in an AT1 receptor-independent manner in RMCs.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Movimento Celular/efeitos dos fármacos , Imidazóis/farmacologia , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/fisiologia , Tetrazóis/farmacologia , Animais , Movimento Celular/fisiologia , Células Cultivadas , Masculino , Fator de Crescimento Derivado de Plaquetas/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
5.
Naunyn Schmiedebergs Arch Pharmacol ; 372(4): 257-66, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16362428

RESUMO

Pramipexole, a novel non-ergot dopamine (DA) agonist, has been successfully applied to the treatment of Parkinson's disease (PD). Although the specific cause of PD remains unknown, recent studies have provided evidence that oxidative stress plays a role in the parthenogenesis of the disease. In the present study, we examined the effect of pramipexole on hydrogen peroxide (H2O2, 100 microM)-induced PC12 cell death, and the intracellular mechanism of this effect. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay revealed that pretreatment of PC12 cells with pramipexole (1-100 microM) resulted in significant protection against H2O2-induced cell death in a concentration-dependent manner. The protective effect of pramipexole was not affected by pretreatment with the DA receptor antagonists sulpiride, spiperone or domperidone, suggesting that the effect of pramipexole is not mediated by DA receptors. In PC12 cells, pramipexole inhibited H2O2-induced lactate dehydrogenase (LDH) leakage, as well as H2O2-induced cytochrome c release and caspase-3 activation with the resultant apoptosis. It was also observed in PC12 cells that H2O2 stimulated phosphorylation of mitogen-activated protein (MAP) kinases, i.e., extracellular signal-regulated kinase1/2 (ERK1/2), c-Jun NH2-terminal kinase (JNK) and p38 MAP kinase. Pramipexole inhibited H2O2-induced JNK and p38 MAP kinase, but not ERK1/2 phosphorylation. Furthermore, in these cells experiments with a fluorescent probe, 2-[6-(4'-amino)phenoxy-3H-xanthen-3-on-9-yl]benzoic acid, revealed that pramipexole, the JNK inhibitor SP600125 and the p38 MAP kinase inhibitor SB203580 inhibited the generation of H2O2-induced reactive oxygen species. Caspase inhibitors Z-DEVD-FMK and Z-IETD-FMK, as well as SP600125 and SB203580, inhibited H2O2-induced PC12 cell death to a similar extent as pramipexole. These results suggest that pramipexole exerts a protective effect against oxidative stress-induced PC12 cell death in part through an inhibition of JNK and p38 MAP kinase.


Assuntos
Antiparkinsonianos/farmacologia , Fármacos Neuroprotetores/farmacologia , Tiazóis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Benzotiazóis , Caspase 3 , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocromos c/metabolismo , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Peróxido de Hidrogênio/toxicidade , L-Lactato Desidrogenase/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Pramipexol , Ratos
6.
J Med Invest ; 50(3-4): 126-35, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-13678381

RESUMO

The recent treatment of hematological malignancies appears to be unsatisfactory in child and adult patients with acute myeloid leukemia and adult patients with acute lymphocytic leukemia. A major problem in the treatment of leukemia is caused by the development of drug resistance to chemotherapeutic agents, which is already present at diagnosis or after chemotherapy as a minimal residual disease, their resistance having originated from genetic or epigenetic mutations during prior growth of the leukemia clone. It was suggested that the mechanisms of drug resistance consist of drug resistance proteins, which work as a drug efflux pump. These are the permeability-related glycoprotein (P-Gp), the multidrug-resistance associated protein (MRP), the lung resistance protein (LRP), and other MDR proteins such as the transporter associated with antigen processing (TAP), anthracyclin resistance associated protein (ARA), MRP 2-7, and breast cancer resistance protein (BCRP). In addition, anti-apoptosis mechanisms, alterations of tumor suppressor genes, altered immunogenicity, drug resistance mechanisms for individual drugs, and clinical risk factors such as white blood cell count, age, and other factors have been reported to act in drug resistance singly or in combinations. Here we describe the update of research on the biology of MDR in the hematological malignancies and also discuss how to overcome MDR and adapt the updated treatment methods in the clinical medical field.


Assuntos
Resistência a Múltiplos Medicamentos , Genes MDR , Neoplasias Hematológicas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Adulto , Apoptose , Criança , Resistência a Múltiplos Medicamentos/genética , Genes Supressores de Tumor , Neoplasias Hematológicas/genética , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Mutação , Proteínas de Neoplasias , Fenótipo , Partículas de Ribonucleoproteínas em Forma de Abóbada
7.
J Med Invest ; 58(1-2): 118-26, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21372496

RESUMO

Recently, increasing evidence suggests that the antihypertensive drug nifedipine acts as a protective agent for endothelial cells, and that the activity is unrelated to its calcium channel blocking. Nitrosonifedipine (NO-NIF) is metabolically and photochemically produced from nifedipine, and NO-NIF has been recognized as a contaminant of nifedipine because it has no antihypertensive effect. Treatment of tumor necrosis factor-α (TNF-α) suppressed the cell viability and facilitated the expression of Inter-Cellular Adhesion Molecule 1(ICAM-1) in human glomerular endothelial cells (HGECs) though, pretreatment of NO-NIF significantly recovered the TNF-α-induced cell damage to the same extent as Trolox-C did, and suppressed the ICAM-1 expression in a concentration dependent manner. In addition, NO-NIF inhibited the cell toxicity induced by cumene hydroperoxide, which hampers the integrity of cell membrane through oxidative stress, as effective as Trolox-c. These data suggest that NO-NIF is a candidate for a new class of antioxidative drug that protect cells against oxidative stress in glomerular endothelial cells.


Assuntos
Glomérulos Renais/efeitos dos fármacos , Nifedipino/análogos & derivados , Compostos Nitrosos/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/toxicidade , Antioxidantes/farmacologia , Derivados de Benzeno/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Nifedipino/farmacologia , Estresse Oxidativo/efeitos dos fármacos
8.
J Pharmacol Sci ; 109(1): 14-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19151535

RESUMO

Chronic kidney disease (CKD) is becoming a major public health problem worldwide. It is important to protect endothelial function in CKD treatment because injury of the endothelium is a critical event for the generation and progression of CKD. Recently, clinical studies showed that nifedipine, an antihypertensive drug, acts as a protective agent of endothelial cells (ECs). Nifedipine is reported to partially decompose to a nitrosonifedipine that has high reactivity against lipid-derived radicals in vitro. However, it is still unclear whether nitrosonifedipine is a biologically active agent against endothelial injury. We observed that nitrosonifedipine was converted to radical form by reaction with cultured ECs. The cumene hydroperoxide mediated cytotoxity was reduced by nitrosonifedipine in cultured human glomerular ECs (HGECs). Also nitrosonifedipine suppressed the expression of TNF-alpha-induced intercellular cell adhesion molecule-1 in HGECs. Chronic administration of N(omega)-nitro-L-arginine methyl ester (L-NAME) caused systemic arterial hypertension, endothelial injury, and renal dysfunction. In L-NAME-induced hypertensive rats, nitrosonifedipine treatment improved not only the acetylcholine-induced vasodilation of the aortic rings, but also renal dysfunction such as increasing the levels of serum creatinine and urinary protein excretion. Our preliminary data suggest that nitrosonifedipine is a new and useful drug for the treatment of CKD involving ameliorating effects on EC disorder.


Assuntos
Descoberta de Drogas/métodos , Células Endoteliais/efeitos dos fármacos , Falência Renal Crônica/tratamento farmacológico , Animais , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Falência Renal Crônica/fisiopatologia , Falência Renal Crônica/prevenção & controle , Estrutura Molecular , Nifedipino/análogos & derivados , Nifedipino/química , Nifedipino/farmacologia , Compostos Nitrosos/química , Compostos Nitrosos/farmacologia , Vasodilatadores/química , Vasodilatadores/farmacologia
9.
J Pharmacol Sci ; 109(2): 257-64, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19202317

RESUMO

Many epidemiologic studies have reported that dietary flavonoids provide protection against cardiovascular disease. Quercetin, a member of the bioflavonoids family, has been proposed to have anti-inflammatory, anti-atherogenic, and anti-hypertensive properties leading to the beneficial effects against cardiovascular diseases. Recent studies demonstrated that orally administered quercetin appeared in plasma as glucuronide-conjugated forms in rats and humans. Therefore, we examined the effect of chemically synthesized quercetin glucuronide on platelet-derived growth factor (PDGF)-induced cell migration and kinase activation in cultured rat aortic smooth muscle cells (RASMCs). PDGF-induced RASMC migration was inhibited by quercetin 3-O-beta-D-glucuronide (Q3GA). Q3GA also attenuated PDGF-induced cell proliferation in RASMCs. PDGF activated extracellular-signal regulated kinase (ERK) 1/2, c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein (MAP) kinase, and Akt in RASMCs. PDGF-induced JNK and Akt activations were suppressed by Q3GA, whereas ERK1/2 and p38 MAP kinase activations were not affected. We also confirmed that PDGF-induced JNK and Akt activations were inhibited by antioxidants, N-acetylcysteine and diphenyleneiodonium chloride, in RASMCs. These findings suggest Q3GA would be an active metabolite of quercetin in plasma and may possess preventing effects for cardiovascular diseases relevant to vascular smooth muscle cell disorders.


Assuntos
Antioxidantes/farmacologia , Glucuronídeos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Quercetina/análogos & derivados , Quercetina/farmacologia , Animais , Antioxidantes/química , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Glucuronídeos/química , MAP Quinase Quinase 4/metabolismo , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Quercetina/química , Ratos , Ratos Sprague-Dawley
10.
Hypertens Res ; 32(3): 188-93, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19262481

RESUMO

Adiponectin, an adipocyte-derived hormone, has been proposed to show antiatherogenic properties through the inhibitory effects against various growth factors. Insulin-like growth factor-1 (IGF-1) is one of the potent mitogens, which has been considered to play important roles in both atherogenesis and plaque stabilization in accordance to the phase of atherosclerosis. The aim of this study is to elucidate the adiponectin effects on IGF-1-induced cell migration and its intracellular signaling pathways in vascular smooth muscle cells (VSMCs). In this study, we assessed cell migration and several kinase activities in cultured rat aortic smooth muscle cells (RASMCs). Adiponectin pretreatment suppressed IGF-1-induced cell migration and extracellular signal-regulated kinase (ERK)1/2 activation, which is one of the major mediators for IGF-1-induced cell migration. In RASMCs, adiponectin and 5-aminoimidazole-4-carboxamide riboside (AICAR), a 5'-AMP-activated protein kinase (AMPK) activator, stimulated AMPK activation. AMPK activation by AICAR inhibited IGF-1-induced ERK1/2 activation and cell migration in RASMCs. On the other hand, phosphorylation of Akt and Bad, proapoptotic molecules of the Bcl-2 family, which were increased by IGF-1 stimulation, was not diminished by the pretreatment with adiponectin. It was shown that adiponectin inhibited IGF-1-induced VSMC migration through suppression of ERK1/2 activation, which might be implicated in AMPK activation. Furthermore, adiponectin selectively inhibited ERK1/2 pathway, not Akt-Bad pathway, stimulated by IGF-1. From these findings, it was implied that adiponectin suppressed IGF-1-induced VSMC migration and its signaling selectivity.


Assuntos
Adiponectina/farmacologia , Movimento Celular/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Miócitos de Músculo Liso/metabolismo , Proteína Oncogênica v-akt/fisiologia , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Western Blotting , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ribonucleotídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína de Morte Celular Associada a bcl/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Transfusion ; 43(1): 65-71, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12519432

RESUMO

BACKGROUND: The expression of ABO antigens on the surface of RBCs is regulated by ABO gene-encoded ABO transferase activity after the formation of the H antigen. The molecular mechanisms that control the expression of the ABO blood group antigens along with erythroid differentiation are one of the most important subjects of study in transfusion science. STUDY DESIGN AND METHODS: FUT1(H), ABO, and beta 2-microglobulin mRNA were determined by semiquantitative RT-PCR from 27 hematopoietic cultured cell lines showing various differentiation stages and cell lineages and normal PBMNCs. The expression level of each cell was analyzed with computer software (Image, NIH) and was shown as the ratio of ABO mRNA or H mRNA to beta 2-microglobulin mRNA. The H antigen was also determined by immunocytochemical methods with flow cytometry in some cell lines. RESULTS: The highest expression of H mRNA was found in KOPM-28 and HEL cell lines and a lower expression was found in the mature cells. In contrast, it was observed that H antigen expression began at the level of HEL and PL-21 cells and increased with cell maturation. The highest expression of ABO mRNA was found in K-562 and KOPM-28 cell lines and it decreased along with cell maturation. CONCLUSION: Based on these results, it is concluded that the transcription of both H and ABO genes starts early in immature peripheral blood progenitor cells but gradually decreases during cellular maturation and also that the H antigen maintains a high level of expression thereafter. These results may reflect the active synthesis of H and ABO antigens in normal hematopoietic peripheral blood progenitor cells.


Assuntos
Sistema ABO de Grupos Sanguíneos/genética , Fucosiltransferases/genética , Células-Tronco Hematopoéticas/fisiologia , Diferenciação Celular , Humanos , RNA Mensageiro/análise , Células Tumorais Cultivadas , Galactosídeo 2-alfa-L-Fucosiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA