Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
País como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Toxicol Mech Methods ; 25(2): 113-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25406855

RESUMO

Deoxycholic acid (DOA) is one of the secondary bile acids used as a mild detergent for the isolation of membrane associated proteins. This study examined whether the secondary bile acid, DOA, altered Ca(2+) movement, cell viability and apoptosis in SCM1 human gastric cancer cells. The Ca(2+)-sensitive fluorescent dye fura-2 was used to measure [Ca(2+)]i. DOA-evoked [Ca(2+)]i rises concentration dependently. The response was reduced by removing extracellular Ca(2+). DOA-evoked Ca(2+) entry was inhibited by store-operated Ca(2+) channel inhibitors (nifedipine, econazole and SKF96365), the protein kinase C (PKC) activator phorbol 12-myristate 13 acetate (PMA) and the PKC inhibitor GF109203X. In Ca(2+)-free medium, treatment with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin (TG) abolished DOA-evoked [Ca(2+)]i rises. Conversely, treatment with DOA abolished TG-evoked [Ca(2+)]i rises. Inhibition of phospholipase C with U73122 abolished DOA-evoked [Ca(2+)]i rises. At 100-500 µM, DOA decreased cell viability, which was not changed by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM). DOA between 100 and 300 µM also induced apoptosis. Collectively, in SCM1 cells, DOA-induced [Ca(2+)]i rises by evoking phospholipase C-dependent Ca(2+) release from the endoplasmic reticulum and Ca(2+) entry via store-operated Ca(2+) channels. DOA also caused Ca(2+)-independent apoptosis.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ácido Desoxicólico/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Bloqueadores dos Canais de Cálcio/farmacologia , Quelantes de Cálcio/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Fura-2/análogos & derivados , Fura-2/metabolismo , Humanos , Microscopia de Fluorescência , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fatores de Tempo
2.
J Nat Prod ; 75(12): 2101-7, 2012 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-23163425

RESUMO

Diallyl sulfide (1), diallyl disulfide (2), and diallyl trisulfide (3), which are major organosulfur compounds of garlic (Allium sativum), are recognized as a group of potential chemopreventive compounds. In this study, the early signaling effects of 3 were examined on Madin-Darby canine kidney (MDCK) cells loaded with the Ca(2+)-sensitive dye fura-2. It was found that 3 caused an immediate and sustained increase of [Ca(2+)](i) in a concentration-dependent manner (EC(50) = 40 µM). Compound 3 also induced a [Ca(2+)](i) elevation when extracellular Ca(2+) was removed, but the magnitude was reduced by 45%. In Ca(2+)-free medium, the 3-induced [Ca(2+)](i) level was abolished by depleting stored Ca(2+) with 1 µM thapsigargin (an endoplasmic reticulum Ca(2+) pump inhibitor). Elevation of [Ca(2+)](i) caused by 3 in the Ca(2+)-containing medium was not affected by modulation of protein kinase C activity. The 3-induced Ca(2+) influx was inhibited by nifedipine and nicardipine (1 µM). U73122, an inhibitor of phospholipase C, abolished ATP (but not the 3-induced [Ca(2+)](i) level). These findings suggest that 3 induced a significant [Ca(2+)](i) elevation in MDCK renal tubular cells by stimulating both extracellular Ca(2+) influx and thapsigargin-sensitive intracellular Ca(2+) release via as yet unidentified mechanisms. Furthermore, the order of the allyl sulfide-induced [Ca(2+)](i) elevation and cell viability was 1 < 2 < 3. The differential effect of allyl sulfides on Ca(2+) signaling and cell death appears to correlate with the number of sulfur atoms in the structure of these allyl sulfides.


Assuntos
Compostos Alílicos/farmacologia , Cálcio/análise , Dissulfetos/farmacologia , Alho/química , Óleos Voláteis/química , Proteína Quinase C/metabolismo , Sulfetos/farmacologia , Trifosfato de Adenosina/metabolismo , Compostos Alílicos/química , Compostos Alílicos/isolamento & purificação , Animais , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dissulfetos/química , Dissulfetos/isolamento & purificação , Retículo Endoplasmático/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Células Madin Darby de Rim Canino , Estrutura Molecular , Nicardipino/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Relação Estrutura-Atividade , Sulfetos/química , Sulfetos/isolamento & purificação , Fosfolipases Tipo C/antagonistas & inibidores
3.
Arch Toxicol ; 86(2): 231-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21879349

RESUMO

Diallyl disulfide (DADS), one of the major organosulfur compounds of garlic, is recognized as a group of potential chemopreventive compounds. In this study, we examines the early signaling effects of DADS on human colorectal cancer cells SW480 loaded with Ca(2+)-sensitive dye fura-2. It was found that DADS caused an immediate and sustained rise of [Ca(2+)](i) in a concentration-dependent manner (EC(50) = 232 µM). DADS also induced a [Ca(2+)](i) elevation when extracellular Ca(2+) was removed, but the magnitude was reduced by 45%. Depletion of intracellular Ca(2+) stores with 2 µM carbonylcyanide m-chlorophenylhydrazone, a mitochondrial uncoupler, didn't affect DADS's effect. In Ca(2+)-free medium, the DADS-induced [Ca(2+)](i) rise was abolished by depleting stored Ca(2+) with 1 µM thapsigargin (an endoplasmic reticulum Ca(2+) pump inhibitor). DADS-caused [Ca(2+)](i) rise in Ca(2+)-containing medium was not affected by modulation of protein kinase C activity. The DADS-induced Ca(2+) influx was blocked by nicardipine (10 µM). U73122, an inhibitor of phospholipase C, abolished ATP (but not DADS)-induced [Ca(2+)](i) rise. These findings suggest that DADS induced a significant rise in [Ca(2+)](i) in SW480 colon cancer cells by stimulating both extracellular Ca(2+) influx and thapsigargin-sensitive intracellular Ca(2+) release via as yet unidentified mechanisms.


Assuntos
Compostos Alílicos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Dissulfetos/farmacologia , Anticarcinógenos/farmacologia , Antioxidantes/farmacologia , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/prevenção & controle , Alho/química , Variação Genética , Genótipo , Humanos
4.
J Nat Prod ; 73(8): 1370-4, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20669930

RESUMO

The effect of [6]-shogaol (1) on cytosolic free Ca(2+) concentrations ([Ca(2+)](i)) and viability has not been explored previously in oral epithelial cells. The present study has examined whether 1 alters [Ca(2+)](i) and viability in OC2 human oral cancer cells. Compound 1 at concentrations > or = 5 microM increased [Ca(2+)](i) in a concentration-dependent manner with a 50% effective concentration (EC(50)) value of 65 microM. The Ca(2+) signal was reduced substantially by removing extracellular Ca(2+). In a Ca(2+)-free medium, the 1-induced [Ca(2+)](i) elevation was mostly attenuated by depleting stored Ca(2+) with thapsigargin (an endoplasmic reticulum Ca(2+) pump inhibitor). The [Ca(2+)](i) signal was inhibited by La(3+) but not by L-type Ca(2+) channel blockers. The elevation of [Ca(2+)](i) caused by 1 in a Ca(2+)-containing medium was not affected by modulation of protein kinase C activity, but was inhibited by 82% with the phospholipase A2 inhibitor aristolochic acid I (20 microM). U73122, a selective inhibitor of phospholipase C, abolished 1-induced [Ca(2+)](i) release. At concentrations of 5-100 microM, 1 killed cells in a concentration-dependent manner. These findings suggest that [6]-shogaol induces a significant rise in [Ca(2+)](i) in oral cancer OC2 cells by causing stored Ca(2+) release from the thapsigargin-sensitive endoplasmic reticulum pool in an inositol 1,4,5-trisphosphate-dependent manner and by inducing Ca(2+) influx via a phospholipase A2- and La(3+)-sensitive pathway.


Assuntos
Canais de Cálcio Tipo L/efeitos dos fármacos , Cálcio/análise , Catecóis/farmacologia , Catecóis/química , Linhagem Celular Tumoral , Citosol/metabolismo , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Estrenos/farmacologia , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Estrutura Molecular , Neoplasias Bucais , Inibidores de Fosfolipase A2 , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Pirrolidinonas/farmacologia , Tapsigargina/farmacologia
5.
Molecules ; 14(3): 959-69, 2009 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-19255554

RESUMO

The effect of [10]-gingerol on cytosol free Ca(2+) concentration ([Ca(2+)](i)) and viability is large unknown. This study examines the early signaling effects of [10]-gingerol on human colorectal cancer cells. It was found that this compound caused a slow and sustained rise of [Ca(2+)](i) in a concentration-dependent manner. [10]-Gingerol also induced a [Ca(2+)](i) rise when extracellular Ca(2+) was removed, but the magnitude was reduced by 38%. In a Ca(2+)-free medium, the [10]-gingerol-induced [Ca(2+)](i) rise was partially abolished by depleting stored Ca(2+) with thapsigargin (an endoplasmic reticulum Ca(2+) pump inhibitor). The elevation of [10]-gingerol-caused [Ca(2+)](i) in a Ca(2+)-containing medium was not affected by modulation of protein kinase C activity. The [10]-gingerol-induced Ca(2+) influx was insensitive to L-type Ca(2+) channel blockers. At concentrations of 10-100 mM, [10]-gingerol killed cells in a concentration-dependent manner. These findings suggest that [10]-gingerol induces [Ca(2+)](i) rise by causing Ca(2+) release from the endoplasmic reticulum and Ca(2+) influx from non-L-type Ca(2+) channels in SW480 cancer cells.


Assuntos
Cálcio/metabolismo , Catecóis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Álcoois Graxos/farmacologia , Antineoplásicos , Canais de Cálcio Tipo L , Catecóis/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Retículo Endoplasmático/metabolismo , Álcoois Graxos/uso terapêutico , Humanos , Proteína Quinase C/metabolismo , Células Tumorais Cultivadas
6.
Front Pharmacol ; 9: 1285, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30473665

RESUMO

Background: Chemotherapy is the main treatment for triple-negative breast cancer (TNBC), which lack molecular markers for diagnosis and therapy. Cancer cells activate chemoresistant pathways and lead to therapeutic failure for patients with TNBC. Several kinases have been identified as chemoresistant genes. However, the involvement of kinases in the chemoresistance in TNBC cells is not fully understood. Methods: We employed a kinome siRNA library to screen whether targeting any kinases could increase the chemosensitivity of TNBC cell lines. The effects of kinase on cell viability in various breast cancer cells were validated with ATP level and colony formation. Protein expression and phosphorylation were determined by immunoblotting. The Cancer Genome Atlas (TCGA) dataset was collected to analyze the correlation of Src expression with prognosis of TNBC patients. Results: Primary screening and validation for the initial hits showed that Src kinase was a potential doxorubicin-resistant kinase in the TNBC cell lines MDA-MB-231 and Hs578T. Both siRNA against Src and the Src inhibitor dasatinib enhanced the cytotoxic effects of doxorubicin in TNBC cells. Moreover, phosphorylation of AKT and signal transducer and activator of transcription 3 (STAT3), downstream effectors of Src, were accordingly decreased in Src-silenced or -inhibited TNBC cells. Additionally, TCGA data analysis indicated that Src expression levels in tumor tissues were higher than those in tumor-adjacent normal tissues in patients with TNBC. High co-expression level of Src and STAT3 was also significantly correlated with poor prognosis in patients. Conclusion: Our results showed that Src-STAT3 axis might be involved in chemoresistance of TNBC cells.

7.
Cell Signal ; 17(7): 847-55, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15763427

RESUMO

Methylglyoxal (2-oxopropanal), a physiological glucose metabolite, is a highly reactive dicarbonyl compound that can induce stress in cells and cause apoptotic cell death. This study examines the early signaling effects of methylglyxal on renal cells. It was found that methylglyoxal caused a slow and sustained rise of intracellular Ca2+ concentration ([Ca2+]i) in a concentration-dependent manner (EC50=1.8 mM). Methylglyoxal also induced a [Ca2+]i rise when extracellular Ca2+ was removed, but the magnitude was reduced by 80%. Depletion of intracellular Ca2+ stores with thapsigargin (TG), an endoplasmic reticulum (ER) Ca2+ pump inhibitor, did not affect methylglyoxal's effect. In Ca2+-free medium, the methylglyoxal-induced [Ca2+]i rise was abolished by depleting stored Ca2+ with carbonylcyanide m-chlorophenylhydrazone (CCCP; a mitochondrial uncoupler). Methylglyoxal-caused [Ca2+]i rise in the Ca2+-containing medium was not affected by modulation of protein kinase C activity, presence of voltage-gated Ca2+ channel blockers, or preincubation with thiol-containing antioxidants. U73122, an inhibitor of phospholipase C, abolished ATP (but not methylglyoxal)-induced [Ca2+]i rise. Furthermore, the [Ca2+]i-elevating effect of methylglyoxal was cell type-dependent, because methylglyoxal failed to cause [Ca2+]i rises in CHO-K1, neutrophils, or platelets. Pretreatment with methylglyoxal for 0-24 h decreased cell viability in a concentration- and time-dependent manner. Meanwhile, methylglyoxal-induced cell death involved apoptotic and necrotic events, the former being the dominant. These findings suggest that methylglyoxal induced a significant rise in [Ca2+]i in Madin-Darby canine kidney (MDCK) renal tubular cells by stimulating both extracellular Ca2+ influx and CCCP-sensitive intracellular Ca2+ release via as yet unidentified mechanisms. The cell type-specific Ca2+ signaling may play an important role in the early process of cytotoxic action of methylglyoxal.


Assuntos
Cálcio/metabolismo , Túbulos Renais/citologia , Aldeído Pirúvico/metabolismo , Animais , Apoptose , Células CHO , Sinalização do Cálcio , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cricetulus , Cães , Relação Dose-Resposta a Droga , Corantes Fluorescentes , Fura-2 , Humanos , Líquido Intracelular/metabolismo , Túbulos Renais/metabolismo , Necrose , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Aldeído Pirúvico/toxicidade , Coelhos , Transdução de Sinais , Fatores de Tempo , Fosfolipases Tipo C/fisiologia
8.
Cell Signal ; 17(7): 837-45, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15763426

RESUMO

The in vitro effect of desipramine on renal tubular cell is unknown. In Madin-Darby canine kidney (MDCK) cells, the effect of desipramine on intracellular Ca2+ concentration ([Ca2+]i) was measured by using fura-2. Desipramine (>25 microM) caused a rapid and sustained rise of [Ca2+]i in a concentration-dependent manner (EC50=50 microM). Desipramine-induced [Ca2+]i rise was prevented by 40% by removal of extracellular Ca2+ but was not altered by L-type Ca2+ channel blockers. In Ca2+-free medium, thapsigargin, an inhibitor of the endoplasmic reticulum Ca2+-ATPase, caused a monophasic [Ca2+]i rise, after which desipramine failed to release more Ca2+; in addition, pretreatment with desipramine partly decreased thapsigargin-induced [Ca2+]i increase. U73122, an inhibitor of phospholipase C, did not change desipramine-induced [Ca2+]i rise. Incubation with 10-100 microM desipramine enhances or inhibits cell proliferation in a concentration- and time-dependent manner. The inhibitory effect of desipramine on proliferation was not extracellular Ca2+-dependent. Apoptosis appears to contribute to desipramine-induced cell death. Together, these findings suggest that desipramine increases baseline [Ca2+]i in renal tubular cells by evoking both extracellular Ca2+ influx and intracellular Ca2+ release, and can cause apoptosis.


Assuntos
Antidepressivos/farmacologia , Cálcio/metabolismo , Desipramina/farmacologia , Túbulos Renais/efeitos dos fármacos , Animais , Apoptose , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Sinalização do Cálcio , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cães , Relação Dose-Resposta a Droga , Líquido Extracelular/metabolismo , Corantes Fluorescentes , Fura-2 , Inositol 1,4,5-Trifosfato/fisiologia , Túbulos Renais/citologia , Fatores de Tempo , Fosfolipases Tipo C/fisiologia
9.
PLoS One ; 11(9): e0160586, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27632526

RESUMO

Low-power laser irradiation (LPLI) is a non-invasive and safe method for cancer treatment that alters a variety of physiological processes in the cells. Autophagy can play either a cytoprotective role or a detrimental role in cancer cells exposed to stress. The detailed mechanisms of autophagy and its role on cytotoxicity in oral cancer cells exposed to LPLI remain unclear. In this study, we showed that LPLI at 810 nm with energy density 60 J/cm2 increased the number of microtubule associated protein 1 light chain 3 (MAP1LC3) puncta and increased autophagic flux in oral cancer cells. Moreover, reactive oxygen species (ROS) production was induced, which increased RelA transcriptional activity and beclin 1 (BECN1) expression in oral cancer cells irradiated with LPLI. Furthermore, ROS scavenger or knockdown of RelA diminished LPLI-induced BECN1 expression and MAP1LC3-II conversion. In addition, pharmacological and genetic ablation of autophagy significantly enhanced the effects of LPLI-induced apoptosis in oral cancer cells. These results suggest that autophagy may be a resistant mechanism for LPLI-induced apoptosis in oral cancer cells.


Assuntos
Autofagia , Proteína Beclina-1/metabolismo , Neoplasias Bucais/patologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição RelA/metabolismo , Humanos , Terapia com Luz de Baixa Intensidade , Neoplasias Bucais/imunologia , NF-kappa B/metabolismo
10.
J Plant Physiol ; 162(2): 129-38, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15779823

RESUMO

Endoplasmic reticulum (ER)-enriched vesicles from etiolated hypocotyls of mung bean seedlings (Vigna radiata) were successfully isolated using Ficoll gradient and two-phase (polyethylene glycol-dextran) partition. The ER-enriched vesicles contained inorganic pyrophosphate (PPi) hydrolysis and its associated proton translocating activities. Antiserum prepared against vacuolar H+-pyrophosphatase (V-PPase, EC 3.6.1.1) did not inhibit this novel pyrophosphatase-dependent proton translocation, excluding the possible contamination of tonoplast vesicles in the ER-enriched membrane preparation. The optimal ratios of Mg2+/PPi (inorganic pyrophosphate) for enzymatic activity and PPi-dependent proton translocation of ER-enriched vesicles were higher than those of vacuolar membranes. The PPi-dependent proton translocation of ER-enriched vesicles absolutely required the presence of monovalent cations with preference for K+, but could be inhibited by a common PPase inhibitor, F-. Furthermore, ER H+-pyrophosphatase exhibited some similarities and differences to vacuolar H+-PPases in cofactor/substrate ratios, pH profile, and concentration dependence of F-, imidodiphosphate (a PPi analogue), and various chemical modifiers. These results suggest that ER-enriched vesicles contain a novel type of proton-translocating PPase distinct from that of tonoplast from higher plants.


Assuntos
Retículo Endoplasmático/enzimologia , Fabaceae/enzimologia , Pirofosfatase Inorgânica/metabolismo , Cátions Bivalentes/farmacologia , Cátions Monovalentes/farmacologia , Difosfonatos/farmacologia , Concentração de Íons de Hidrogênio , Hipocótilo/enzimologia , Pirofosfatase Inorgânica/antagonistas & inibidores
11.
Life Sci ; 132: 55-60, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25916802

RESUMO

AIMS: Vibrio vulnificusis an opportunistic pathogen that causes primary septicemia and wound infection with high mortality rate. This pathogen produces an RTX toxin (RtxA1) which can cause host cell rounding, cell death and interference with internalization by host phagocytes. However, the mechanism of RtxA1-induced phagocyte paralysis is not clear. MAIN METHODS: Using the murine macrophage cell line RAW264.7, we measured cytotoxicity and phagocytosis of V. vulnificusin normal and calcium-depleted media. To deplete extracellular and cytosolic Ca(2+), cells were exposed to the calcium chelators ethylene glycol tetraacetic acid (EGTA) and 1,2-bis-(o-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid, tetraacetoxymethyl esteris (BAPTA-AM), respectively. The cytotoxicity was examined by measuring the activity of lactate dehydrogenase (LDH) released from the damaged cells. The gentamicin protection assay was conducted to determine the number of internalized bacteria, while acridine orange staining was applied to visualize the intracellular bacteria. The fluorescent indicator fura-2-acetoxymethyl ester (fura 2-AM) was used to measure the Ca(2+)signal post-infection. KEY FINDINGS: We revealed that extracellular Ca(2+)was essential for phagocytes to internalize V. vulnificus. Meanwhile, cytosolic Ca(2+)flux in RAW264.7 cells induced by an RtxA1 isogenic mutant was repressed by the parent strain. Furthermore, depletion of extracellular Ca(2+)level by EGTA significantly reduced the cytotoxicity but did not affect the antiphagocytic activity of RtxA1 toxin. SIGNIFICANCE: Our results indicated that RtxA1 may interfere with cytosolic Ca(2+)flux of phagocyte to promote bacteria colonization.


Assuntos
Toxinas Bacterianas/toxicidade , Cálcio/metabolismo , Fagocitose/efeitos dos fármacos , Vibrio vulnificus/metabolismo , Laranja de Acridina , Animais , Toxinas Bacterianas/metabolismo , Western Blotting , Ácido Egtázico/análogos & derivados , Fura-2/análogos & derivados , L-Lactato Desidrogenase/metabolismo , Macrófagos , Camundongos , Vibrio vulnificus/genética
12.
Kaohsiung J Med Sci ; 31(1): 1-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25600914

RESUMO

This study explored the effect of deltamethrin, a pesticide, on free Ca(2+) concentration [Ca(2+)]i, viability, and apoptosis in Madin-Darby canine kidney (MDCK) canine renal tubular cells. Deltamethrin at concentrations between 10µM and 40µM evoked [Ca(2+)]i rises in a concentration-dependent manner. The Ca(2+) entry was inhibited by nifedipine, econazole, phorbol 12-myristate 13-acetate, and SKF96365. Treatment with the endoplasmic reticulum Ca(2+) pump inhibitor 2,5-di-tert-butylhydroquinone (BHQ) in a Ca(2+)-free medium abolished deltamethrin-induced [Ca(2+)]i rise. Treatment with deltamethrin also abolished BHQ-induced [Ca(2+)]i rise. Inhibition of phospholipase C (PLC) activity with U73122 abolished deltamethrin-evoked [Ca(2+)]i rise. Deltamethrin killed cells at 30-60µM in a concentration-dependent manner. The cytotoxic effect of deltamethrin was not reversed by prechelating cytosolic Ca(2+) with the acetoxymethyl ester of 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid. Annexin V/propidium iodide staining data suggest that 30-50µM deltamethrin induced apoptosis. Together, in MDCK renal tubular cells, deltamethrin induced [Ca(2+)]i rises that involved Ca(2+) entry through protein kinase C-mediated store-operated Ca(2+) channels, and PLC-dependent Ca(2+) release from the endoplasmic reticulum. Deltamethrin also induced Ca(2+)-independent cell death that might involve apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Túbulos Renais/citologia , Nitrilas/farmacologia , Piretrinas/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Cães , Túbulos Renais/metabolismo , Células Madin Darby de Rim Canino
13.
Life Sci ; 72(15): 1733-43, 2003 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-12559394

RESUMO

2,2'-dithiodipyridine (2,2'-DTDP), a reactive disulphide that mobilizes Ca(2+) in muscle, induced an increase in cytoplasmic free Ca(2+)concentrations ([Ca(2+)](i)) in MG63 human osteosarcoma cells loaded with the Ca(2+)-sensitive dye fura-2. 2,2'-DTDP acted in a concentration-independent manner with an EC(50) of 50 microM. The Ca(2+) signal comprised an initial spike and a prolonged increase. Removing extracellular Ca(2+) did not alter the Ca(2+) signal, suggesting that the Ca(2+) signal was due to store Ca(2+) release. In Ca(2+)-free medium, the 2,2'-DTDP-induced [Ca(2+)](i) increase was not changed by depleting store Ca(2+) with 50 microM bredfeldin A (a Golgi apparatus permeabilizer), 2 microM carbonylcyanide m-chlorophenylhydrazone (CCCP, a mitochondrial uncoupler), 1 microM thapsigargin (an endoplasmic reticulum Ca(2+)pump inhibitor) or 5 microM ryanodine. Conversely, 2,2'-DTDP pretreatment abolished CCCP and thapsigargin-induced [Ca(2+)](i) increases. 2,2'-DTDP-induced Ca(2+) signals in Ca(2+)-containing medium were not affected by modulation of protein kinase C activity or suppression of phospholipase C activity. However, 2,2'-DTDP-induced Ca(2+) release was inhibited by a thiol-selective reducing reagent, dithiothreitol (5-25 microM) in a concentration-dependent manner. Collectively, this study shows that 2,2'-DTDP induced [Ca(2+)](i) increases in human osteosarcoma cells via releasing store Ca(2+)from multiple stores in a manner independent of protein kinase C or phospholipase C activity. The 2,2'-DTDP-induced store Ca(2+) release appeared to be dependent on oxidation of membranes.


Assuntos
2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/farmacologia , Neoplasias Ósseas/metabolismo , Cálcio/metabolismo , Dissulfetos/farmacologia , Osteossarcoma/metabolismo , Compostos de Sulfidrila/metabolismo , Brefeldina A/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes , Fura-2 , Humanos , Oxirredução , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Substâncias Redutoras/farmacologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Tapsigargina/farmacologia , Células Tumorais Cultivadas , Fosfolipases Tipo C/metabolismo
14.
Life Sci ; 72(23): 2571-80, 2003 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-12672503

RESUMO

The effect of NPC-14686, a potential anti-inflammatory drug, on cytosolic free Ca(2+) levels ([Ca(2+)](i)) in HA22/VGH human hepatoma cells was explored by using fura-2 as a fluorescent Ca(2+) indicator. NPC-14686 at concentrations above 10 microM increased [Ca(2+)](i) in a concentration-dependent manner with an EC(50) value of 100 microM. The Ca(2+) signal was reduced by removing extracellular Ca(2+) or by 10 microM nifedipine and was not changed by verapamil or diltiazem. Pretreatment with 1 microM thapsigargin (an endoplasmic reticulum Ca(2+) pump inhibitor) to deplete the endoplasmic reticulum Ca(2+) abolished 200 microM NPC-14686-induced Ca(2+) release; and conversely pretreatment with NPC-14686 abolished thapsigargin-induced Ca(2+) release. The Ca(2+) release induced by 200 microM NPC-14686 was not changed by inhibiting phospholipase C with 2 microM U73122. Together, the results suggest that in human hepatoma cells, NPC-14686 induced a [Ca(2+)](i) increase by causing store Ca(2+) release from the endoplasmic reticulum in an phospholipase C-independent manner, and by inducing nifedipine-sensitive Ca(2+) influx.


Assuntos
Aminobutiratos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Hepatócitos/efeitos dos fármacos , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Estrenos/farmacologia , Corantes Fluorescentes , Fura-2/metabolismo , Hepatócitos/metabolismo , Humanos , Pirrolidinonas/farmacologia , Tapsigargina/farmacologia , Células Tumorais Cultivadas , Fosfolipases Tipo C/antagonistas & inibidores
15.
Environ Toxicol Pharmacol ; 14(1-2): 1-7, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21782656

RESUMO

In human osteosarcoma MG63 cells, effect of triethyltin, an environmental toxicant, on intracellular Ca(2+) concentration ([Ca(2+)](i)) was measured by using fura-2. Triethyltin (1-50 µM) caused a rapid and sustained plateau rise of [Ca(2+)](i) in a concentration-dependent manner (EC(50)=10 µM). Triethyltin-induced [Ca(2+)](i) rise was prevented by 50% by removal of extracellular Ca(2+) but was not altered by voltage-gated Ca(2+) channel blockers. In Ca(2+)-free medium, thapsigargin, an inhibitor of the endoplasmic reticulum (ER) Ca(2+)-ATPase, caused a monophasic [Ca(2+)](i) rise, after which the increasing effect of triethyltin on [Ca(2+)](i) was attenuated by 60%; also, pretreatment with triethyltin abolished thapsigargin-induced [Ca(2+)](i) increase. Depletion of mitochondrial Ca(2+) with carbonylcyanide m-chlorophenylhydrazone (CCCP; 2 µM) did not affect triethyltin-induced Ca(2+) release. U73122, an inhibitor of phoispholipase C, abolished ATP (but not triethyltin)-induced [Ca(2+)](i) rise. A low concentration (1 µM) of triethyltin failed to alter ATP and bradykinin-induced [Ca(2+)](i) rises. These findings suggest that triethyltin rapidly increases [Ca(2+)](i) in osteoblasts by stimulating both extracellular Ca(2+) influx and intracellular Ca(2+) release via as yet unidentified mechanism(s).

16.
Nat Prod Res ; 26(20): 1881-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21988123

RESUMO

Investigation of the leaves' extract of Cinnamomum tenuifolium (Lauraceae) led to the isolation of one novel benzodioxocinone, 2,3-dihydro-6,6-dimethylbenzo-[b][1,5]dioxocin-4(6 H)-one (1). The structure was determined through in-depth spectroscopic and mass-spectrometric analyses. The antioxidant potential was evaluated using the following in vitro method: scavenging of 1,1-diphenyl-2-picrylhydrazyl radical. We also detected the anti-proliferative effect of 1 on human oral cancer cells and its IC(50) is 107.7 µM.


Assuntos
Antioxidantes/química , Antioxidantes/farmacologia , Cinnamomum/química , Folhas de Planta/química , Compostos de Bifenilo/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Picratos/química
17.
Nat Prod Res ; 26(14): 1318-22, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22077382

RESUMO

A new phenylalkanoid, (E)-3-hydroxy-1-(4'-dihydroxy-3',5'-dimethoxy-phenyl)-dodecan-6-en-5-one (1) was isolated from the rhizomes of Chinese ginger (Zingiber officinale Roscoe (Zingiberaceae)). The structure of this new phenylalkanoid was elucidated by chemical and physical evidences.


Assuntos
Rizoma/química , Zingiber officinale/química , Estrutura Molecular
18.
Nat Prod Commun ; 6(11): 1671-2, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22224285

RESUMO

The cytotoxicity of hexahydrocurcumin and its effect on the cell cycle in human colorectal cancer cells SW480 has been studied for the first time. The compound, extracted from Zingiber officinale, was shown to be cytotoxic to colorectal cancer cells. Treatment of SW480 cells with hexahydrocurcumin (100 microM) resulted in a massive accumulation of the cells in the G1/G0 phase of the cell cycle. The cytotoxic effect of hexahydrocurcumin may prove useful in cancer prevention.


Assuntos
Antineoplásicos Fitogênicos/análise , Ciclo Celular/efeitos dos fármacos , Curcumina/análogos & derivados , Curcumina/farmacologia , Zingiber officinale/química , Linhagem Celular Tumoral , Curcumina/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos
19.
Nat Prod Commun ; 6(9): 1297-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21941902

RESUMO

A novel amide, cinnabutamine (1), along with five known amides, cinnaretamine (2), N-trans-caffeoyl-5-hydroxytyramine (3), N-trans-feruloyltyramine (4), N-trans-feruloyl-5-methoxytyramine (5) and N-cis-feruloyl-5-methoxytyramine (6), were isolated from the stems of Cinnamomum burmannii (Lauraceae). Their structures were characterized and identified by spectral analysis.


Assuntos
Amidas/química , Cinnamomum/química , Caules de Planta/química , Estrutura Molecular
20.
Food Chem Toxicol ; 47(9): 2344-50, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19555733

RESUMO

Diallyl sulfide (DAS), one of the major organosulfur compounds (OSCs) of garlic, is recognized as a group of potential chemoproventive compounds. In this study, we examines the early signaling effects of DAS on renal cells loaded with Ca(2+)-sensitive dye fura-2. It was found that DAS caused an immediate and sustained rise of [Ca(2+)](i) in a concentration-dependent manner (EC(50)=2.32 mM). DAS also induced a [Ca(2+)](i) elevation when extracellular Ca(2+) was removed, but the magnitude was reduced by 45%. Depletion of intracellular Ca(2+) stores with CCCP, a mitochondrial uncoupler, did not affect DAS's effect. In Ca(2+)-free medium, the DAS-induced [Ca(2+)](i) rise was abolished by depleting stored Ca(2+) with thapsigargin (an endoplasmic reticulum Ca(2+) pump inhibitor). DAS-caused [Ca(2+)](i) rise in Ca(2+)-containing medium was not affected by modulation of protein kinase C activity. The DAS-induced Ca(2+) influx was blocked by nicardipine. U73122, an inhibitor of phospholipase C, abolished ATP (but not DAS)-induced [Ca(2+)](i) rise. Additionally, pretreatment with DAS for 24 h decreased cell viability in a concentration-dependent manner. Furthermore, DAS-induced cell death involved apoptotic events. These findings suggest that diallyl sulfide induced a significant rise in [Ca(2+)](i) in MDCK renal tubular cells by stimulating both extracellular Ca(2+) influx and thapsigargin-sensitive intracellular Ca(2+) release via as yet unidentified mechanisms.


Assuntos
Compostos Alílicos/farmacologia , Antioxidantes/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Alho/química , Túbulos Renais/efeitos dos fármacos , Sulfetos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/fisiologia , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cães , Relação Dose-Resposta a Droga , Estrenos/farmacologia , Corantes Fluorescentes/metabolismo , Fura-2/metabolismo , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Nicardipino/farmacologia , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Pirrolidinonas/farmacologia , Tapsigargina/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo , Desacopladores/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa