Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chin J Physiol ; 60(1): 32-40, 2017 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-28052644

RESUMO

Thymol is a phenolic compound that affects physiology in different cell models. However, whether thymol affects Ca²âº homeostasis in prostate cancer cells is unknown. The action of this compound on cytosolic Ca²âº concentrations ([Ca²âº]i) and viability in PC3 human prostate cancer cells was explored. The results show that thymol at concentrations of 100-1500 µM caused [Ca²âº]i rises in a concentration-dependent manner. Removal of extracellular Ca²âº reduced thymol's effect by approximately 80%. Thymol-induced Ca²âº entry was confirmed by Mn²âº entry-induced quench of fura-2 fluorescence, and was inhibited by approximately 30% by Ca²âº entry modulators (nifedipine, econazole, SKF96365), and the protein kinase C (PKC) inhibitor GF109203X. In Ca²âº-free medium, treatment with the endoplasmic reticulum Ca²âº pump inhibitor thapsigargin abolished thymol-induced [Ca²âº]i rises. Treatment with thymol also abolished thapsigargin-induced [Ca²âº]i rises. Thymol-induced Ca²âº release from the endoplasmic reticulum was abolished by the phospholipase C (PLC) inhibitor U73122. Thymol at 100-900 µM decreased cell viability, which was not reversed by pretreatment with the Ca²âº chelator 1,2-bis(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM). Together, in PC3 cells, thymol induced [Ca²âº]i rises by inducing PLC-dependent Ca²âº release from the endoplasmic reticulum and Ca²âº entry via PKC-sensitive store-operated Ca²âº channels and other unknown channels. Thymol also induced Ca²âº-dissociated cell death.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antifúngicos/uso terapêutico , Sinalização do Cálcio/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Timol/uso terapêutico , Antifúngicos/farmacologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Homeostase/efeitos dos fármacos , Humanos , Masculino , Timol/farmacologia
2.
Chin J Physiol ; 58(5): 285-93, 2015 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-26387652

RESUMO

The effect of the anti-inflammatory compound NPC-14686 on intracellular Ca²âº concentration ([Ca²âº](i)) and viability in OC2 human oral cancer cells was investigated. The Ca²âº-sensitive fluorescent probe fura-2 was used to examine [Ca²âº](i). NPC-14686 induced [Ca²âº](i) rises in a concentration-dependent fashion. The effect was reduced approximately by 10% by removing extracellular Ca²âº. NPC-14686- elicited Ca²âº signal was decreased by nifedipine, econazole, SKF96365, and GF109203X. In Ca²âº-free medium, incubation with the endoplasmic reticulum Ca²âº pump inhibitor thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) abolished NPC-14686-induced [Ca²âº](i) rises. Conversely, pretreatment with NPC-14686 abolished thapsigargin or BHQ-induced [Ca²âº](i) rises. Inhibition of phospholipase C with U73122 abolished NPC-14686-induced [Ca²âº](i) rises. At 20-100 µM, NPC-14686 inhibited cell viability, which was not reversed by chelating cytosolic Ca²âº with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'- tetraacetic acid-acetoxymethyl ester (BAPTA/AM). NPC-14686 between 20 µM and 40 µM also induced apoptosis. Collectively, in OC2 cells, NPC-14686 induced [Ca²âº](i) rises by evoking phospholipase C-dependent Ca²âº release from the endoplasmic reticulum and Ca²âº entry via protein kinase C-regulated store-operated Ca²âº channels. NPC-14686 also caused Ca²âº-independent apoptosis.


Assuntos
Aminobutiratos/uso terapêutico , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Bucais/tratamento farmacológico , Aminobutiratos/farmacologia , Canais de Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Retículo Endoplasmático/metabolismo , Fura-2 , Homeostase , Humanos , Fosfolipases Tipo C/metabolismo
3.
Chin J Physiol ; 58(5): 302-11, 2015 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-26387654

RESUMO

Safrole is a carcinogen found in plants. The effect of safrole on cytosolic free Ca²âº concentrations ([Ca²âº](i)) and viability in SCM1 human gastric cancer cells was explored. The Ca²âº-sensitive fluorescent dye fura-2 was applied to measure [Ca²âº](i). Safrole at concentrations of 150-450 µM induced a [Ca²âº](i) rise in a concentration-dependent manner. The response was reduced by 60% by removing extracellular Ca²âº. Safrole-evoked Ca²âº entry was not altered by nifedipine, econazole, SKF96365, and protein kinase C activator or inhibitor. In Ca²âº-free medium, treatment with the endoplasmic reticulum Ca²âº pump inhibitor thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) abolished safrole-evoked [Ca²âº](i) rises. Conversely, treatment with safrole abolished thapsigargin or BHQ-evoked [Ca²âº](i) rises. Inhibition of phospholipase C (PLC) with U73122 abolished safrole-induced [Ca²âº](i) rises. At 250-550 µM, safrole decreased cell viability concentration-dependently, which was not reversed by chelating cytosolic Ca²âº with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/acetoxy methyl (BAPTA/AM). Annexin V/propidium iodide staining data suggest that safrole (350-550 µM) induced apoptosis concentration-dependently. These studies suggest that in SCM1 human gastric cancer cells, safrole induced [Ca²âº](i) rises by inducing PLC-dependent Ca²âº release from the endoplasmic reticulum and Ca²âº influx via non-store-operated Ca²âº entry pathways. Safrole-induced cell death may involve apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Safrol/efeitos adversos , Estômago/efeitos dos fármacos , Morte Celular , Linhagem Celular Tumoral , Fura-2 , Humanos , Fosfolipases Tipo C/metabolismo
4.
J Recept Signal Transduct Res ; 35(5): 429-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26096164

RESUMO

Protriptyline, a tricyclic anti-depressant, is used primarily to treat the combination of symptoms of anxiety and depression. However, the effect of protriptyline on prostate caner is unknown. This study examined whether the anti-depressant protriptyline altered Ca(2+) movement and cell viability in PC3 human prostate cancer cells. The Ca(2+)-sensitive fluorescent dye fura-2 was used to measure [Ca(2+)](i). Protriptyline evoked [Ca(2+)](i) rises concentration-dependently. The response was reduced by removing extracellular Ca(2+). Protriptyline-evoked Ca(2+) entry was inhibited by store-operated channel inhibitors (nifedipine, econazole and SKF96365), protein kinase C activator (phorbol 12-myristate 13 acetate, PMA) and protein kinase C inhibitor (GF109203X). Treatment with the endoplasmic reticulum Ca(2+) pump inhibitor 2,5-di-tert-butylhydr-oquinone (BHQ) in Ca(2+)-free medium inhibited 60% of protriptyline-evoked [Ca(2+)](i) rises. Conversely, treatment with protriptyline abolished BHQ-evoked [Ca(2+)](i) rises. Inhibition of phospholipase C with U73122 suppressed 50% of protriptyline-evoked [Ca(2+)](i) rises. At concentrations of 50-70 µM, protriptyline decreased cell viability in a concentration-dependent manner; which were not reversed by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM). Collectively, in PC3 cells, protriptyline evoked [Ca(2+)](i) rises by inducing phospholipase C-associated Ca(2+) release from the endoplasmic reticulum and other stores, and Ca(2+) influx via protein kinase C-sensitive store-operated Ca(2+) channels. Protriptyline caused cell death that was independent of [Ca(2+)](i) rises.


Assuntos
Apoptose/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Protriptilina/administração & dosagem , Transporte Biológico Ativo/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Masculino , Neoplasias da Próstata/tratamento farmacológico
5.
Chin J Physiol ; 58(3): 178-87, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26014123

RESUMO

The effect of the antidepressant doxepin on cytosolic Ca²âº concentrations ([Ca²âº](i)) and viability in PC3 human prostate cancer cells was explored. The Ca²âº-sensitive fluorescent dye fura-2 was applied to measure [Ca²âº](i). Doxepin at concentrations of 500-1000 µM induced a [Ca²âº](i) rise in a concentration-dependent manner. The response was reduced partly by removing Ca²âº. Doxepin-evoked Ca²âº entry was suppressed by Ca²âº entry blockers (nifedipine, econazole, SK&F96365), and protein kinase C (PKC) modulators. In the absence of extracellular Ca²âº, incubation with the endoplasmic reticulum Ca²âº pump inhibitor thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) partly inhibit doxepin-induced [Ca²âº](i) rise. Incubation with doxepin nearly inhibited thapsigargin or BHQ-induced [Ca²âº](i) rise. Inhibition of phospholipase C (PLC) with U73122 failed to alter doxepin-induced [Ca²âº](i) rise. At concentrations of 200-250 µM, doxepin killed cells in a concentration-dependent manner. This cytotoxic effect was not reversed by chelating cytosolic Ca²âº with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'- tetraacetic acid/acetoxy methyl (BAPTA/AM). Annexin V/PI staining data implied that doxepin (200 and 250 µM) did not induce apoptosis. Collectively, in PC3 cells, doxepin induced a [Ca²âº](i) rise by evoking PLC-independent Ca²âº release from stores including the endoplasmic reticulum and Ca²âº entry via PKC-sensitive store-operated Ca²âº channels. Doxepin caused cell death that was independent of [Ca²âº](i) rises.


Assuntos
Antidepressivos/farmacologia , Cálcio/metabolismo , Doxepina/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Masculino , Fosfolipases Tipo C/fisiologia
6.
Chin J Physiol ; 58(1): 1-8, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25687486

RESUMO

Methoxychlor, an organochlorine pesticide, is thought to be an endocrine disrupter that affects Ca²âº homeostasis and cell viability in different cell models. This study explored the action of methoxychlor on cytosolic free Ca²âº concentrations ([Ca²âº]i) and apoptosis in HA59T human hepatoma cells. Fura-2, a Ca²âº-sensitive fluorescent dye, was applied to measure [Ca²âº]i. Methoxychlor at concentrations of 0.1-1 µM caused a [Ca²âº]i rise in a concentration-dependent manner. Removal of external Ca²âº abolished methoxychlor's effect. Methoxychlor-induced Ca²âº influx was confirmed by Mn²âº-induced quench of fura-2 fluorescence. Methoxychlor-induced Ca²âº entry was inhibited by nifedipine, econazole, SK&F96365, and protein kinase C modulators. Methoxychlor killed cells at concentrations of 10-130 µM in a concentration-dependent fashion. Chelation of cytosolic Ca²âº with 1,2-bis(2-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid/AM (BAPTA/AM) did not prevent methoxychlor's cytotoxicity. Methoxychlor (10 and 50 µM) induced apoptosis concentration-dependently as determined by using Annexin V/propidium iodide staining. Together, in HA59T cells, methoxychlor induced a [Ca²âº]i rise by inducing Ca²âº entry via protein kinase C-sensitive Ca²âº-permeable channels, without causing Ca²âº release from stores. Methoxychlor also induced apoptosis that was independent of [Ca²âº]i rises.


Assuntos
Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Carcinoma Hepatocelular/metabolismo , Homeostase/efeitos dos fármacos , Inseticidas/farmacologia , Neoplasias Hepáticas/metabolismo , Metoxicloro/farmacologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Neoplasias Hepáticas/patologia
7.
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
8.
Chin J Physiol ; 57(3): 158-66, 2014 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-24826784

RESUMO

NPC-14686 has been shown to have anti-inflammatory effect in previous studies, but the mechanisms are unclear. The effect of NPC-14686 on cytosolic Ca²âº concentrations ([Ca²âº]i) and viability in MG63 human osteosarcoma cells was explored. The Ca²âº-sensitive fluorescent dye fura-2 was applied to measure [Ca²âº]i. NPC-14686 at concentrations of 100-500 µM induced a [Ca²âº]i rise in a concentration-dependent manner. The response was reduced by 80% by removing Ca²âº. NPC-14686 induced Mn²âº influx leading to quenching of fura-2 fluorescence. NPC-14686-evoked Ca²âº entry was suppressed by nifedipine, econazole, SK&F96365, and protein kinase C inhibitor. Inhibition of phospholipase C with U73122 abolished NPC-14686-induced [Ca²âº]i rise. At 20-50 µM, NPC-14686 decreased cell viability, which was not reversed by chelating cytosolic Ca²âº with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/acetoxy methyl (BAPTA/AM). Annexin V/propidium iodide staining data suggest that NPC-14686 (30-50 µM) induced apoptosis in a concentration-dependent manner. NPC-14686 also increased levels of reactive oxygen species. Together, in human osteosarcoma cells, NPC-14686 induced a [Ca²âº]i rise by inducing phospholipase C-dependent Ca²âº release from the endoplasmic reticulum and Ca²âº entry via protein kinase C-sensitive store-operated Ca²âº channels. NPC-14686 induced cell death that might involve apoptosis via mitochondrial pathways.


Assuntos
Aminobutiratos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/patologia , Sinalização do Cálcio/efeitos dos fármacos , Osteossarcoma/patologia , Antifúngicos/farmacologia , Apoptose/fisiologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Quelantes/farmacologia , Econazol/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Humanos , Imidazóis/farmacologia , Nifedipino/farmacologia
9.
Chin J Physiol ; 57(2): 90-8, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24694198

RESUMO

Thymol is a natural essential oil present in many plants and has many different effects in various cell types. However, the effect of thymol on the physiology of MDCK renal tubular cells is unknown. The action of the phytochemical thymol on cytosolic Ca²âº concentrations ([Ca²âº]i) and apoptosis in Madin-Darby canine kidney (MDCK) renal tubular cells was explored. Fura-2, a Ca²âº-sensitive fluorescent dye, was used to assess [Ca²âº]i. Thymol at concentrations of 200-500 µM caused a [Ca²âº]i rise in a concentration-dependent manner. Removal of extracellular Ca²âº partially reduced the effects of thymol. Thymol-induced Ca²âº entry was inhibited by nifedipine, econazole, SK&F96365 and protein kinase C modulators. In a Ca²âº-free medium, treatment with the endoplasmic reticulum Ca²âº pump inhibitor thapsigargin inhibited thymol-induced [Ca²âº]i increases. Treatment with thymol also inhibited thapsigargin-induced [Ca²âº]i rise. Thymol killed cells at concentrations of 300-500 µM in a concentrationdependent fashion. Chelating cytosolic Ca²âº with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/AM (BAPTA/AM) did not prevent thymol cytotoxicity. Thymol (400 and 500 µM) induced apoptosis detected by using Annexin V/propidium iodide staining. At 400 or 500 µM, thymol increased levels of reactive oxygen species. Together, in MDCK cells, thymol induced a [Ca²âº]i rise by inducing Ca²âº release from the endoplasmic reticulum and Ca²âº entry via protein kinase C-sensitive store-operated Ca²âº channels. Our data suggest that thymol-induced apoptosis might involve reactive oxygen species (ROS) production.


Assuntos
Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Homeostase/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Timol/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cães , Relação Dose-Resposta a Droga , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
10.
Acta Physiol Hung ; 99(1): 74-86, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22425810

RESUMO

The effect of 2,4,6-trimethyl-N-(meta-3-trifluoromethyl-phenyl)-benzenesulfonamide (m-3M3FBS), a presumed phospholipase C activator, on cytosolic free Ca2+ concentrations ([Ca2+]i) in OC2 human oral cancer cells is unclear. This study explored whether m-3M3FBS changed basal [Ca2+]i levels in suspended OC2 cells by using fura-2 as a Ca2+-sensitive fluorescent dye. M-3M3FBS at concentrations between 10-60 µM increased [Ca2+]i in a concentration-dependent manner. The Ca2+ signal was reduced partly by removing extracellular Ca2+. M-3M3FBS-induced Ca2+ influx was inhibited by the store-operated Ca2+ channel blockers nifedipine, econazole and SK&F96365, and by the phospholipase A2 inhibitor aristolochic acid. In Ca2+-free medium, 30 µM m-3M3FBS pretreatment inhibited the [Ca2+]i rise induced by the endoplasmic reticulum Ca2+ pump inhibitors thapsigargin and 2,5-di-tert-butylhydroquinone (BHQ). Conversely, pretreatment with thapsigargin, BHQ or cyclopiazonic acid partly reduced m-3M3FBS-induced [Ca2+]i rise. Inhibition of inositol 1,4,5-trisphosphate formation with U73122 did not alter m-3M3FBS-induced [Ca2+]i rise. At concentrations between 5 and 100 µM m-3M3FBS killed cells in a concentration-dependent manner. The cytotoxic effect of m-3M3FBS was not reversed by prechelating cytosolic Ca2+ with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). Propidium iodide staining data suggest that m-3M3FBS (20 or 50 µM) induced apoptosis in a Ca2+-independent manner. Collectively, in OC2 cells, m-3M3FBS induced [Ca2+]i rise by causing inositol 1,4,5-trisphosphate-independent Ca2+ release from the endoplasmic reticulum and Ca2+ influx via phospholipase A2-sensitive store-operated Ca2+ channels. M-3M3FBS also induced Ca2+-independent cell death and apoptosis.


Assuntos
Cálcio/metabolismo , Neoplasias Bucais , Sulfonamidas/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Estrenos/farmacologia , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Inibidores de Fosfodiesterase/farmacologia , Pirrolidinonas/farmacologia
11.
Basic Clin Pharmacol Toxicol ; 110(4): 314-21, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21995587

RESUMO

The effect of the natural product 3,3'-diindolylmethane (DIM) on cytosolic Ca(2+) concentrations ([Ca(2+)](i)) and viability in MG63 human osteosarcoma cells was explored. The Ca(2+)-sensitive fluorescent dye fura-2 was applied to measure [Ca(2+)](i). DIM at concentrations of 40-80 µM induced a [Ca(2+)](i) rise in a concentration-dependent manner. The response was reduced partly by removing Ca(2+). DIM-evoked Ca(2+) entry was suppressed by nifedipine, econazole, SK&F96365 and protein kinase C modulators. In the absence of extracellular Ca(2+), incubation with the endoplasmic reticulum Ca(2+) pump inhibitors thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) inhibited or abolished DIM-induced [Ca(2+)](i) rise. Incubation with DIM also inhibited thapsigargin or BHQ-induced [Ca(2+)](i) rise. Inhibition of phospholipase C with U73122 abolished DIM-induced [Ca(2+)](i) rise. At concentrations of 10-50 µM, DIM killed cells in a concentration-dependent manner. This cytotoxic effect was not altered by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). Annexin V/propidium iodide staining data implicate that DIM (20 and 40 µM) induced apoptosis in a concentration-dependent manner. In sum, in MG63 cells, DIM induced a [Ca(2+)](i) rise by evoking phospholipase C-dependent Ca(2+) release from the endoplasmic reticulum and Ca(2+) entry via protein kinase C-sensitive store-operated Ca(2+) channels. DIM caused cell death that may involve apoptosis.


Assuntos
Anticarcinógenos/farmacologia , Cálcio/metabolismo , Indóis/farmacologia , Osteossarcoma/tratamento farmacológico , Anticarcinógenos/administração & dosagem , Apoptose/efeitos dos fármacos , Canais de Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Corantes Fluorescentes/metabolismo , Fura-2/metabolismo , Homeostase , Humanos , Indóis/administração & dosagem , Osteossarcoma/patologia , Fosfolipases Tipo C/metabolismo
12.
Drug Chem Toxicol ; 34(4): 454-61, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21770746

RESUMO

The effect of the environmental contaminant, bisphenol A, on cytosolic free Ca(2+) concentrations ([Ca(2+)](i)) in Madin-Darby canine kidney (MDCK) cells is unclear. This study explored whether bisphenol A changed basal [Ca(2+)](i) levels in suspended MDCK cells by using fura-2 as a Ca(2+)-sensitive fluorescent dye. Bisphenol A, at concentrations between 50 and 300 µM, increased [Ca(2+)](i) in a concentration-dependent manner. The Ca(2+) signal was reduced, partly, by removing extracellular Ca(2+). Bisphenol A induced Mn(2+) influx, leading to quenching of fura-2 fluorescence, suggesting Ca(2+) influx. This Ca(2+) influx was inhibited by phospholipase A2 inhibitor aristolochic acid, store-operated Ca(2+) channel blockers nifedipine and SK&F96365, and protein kinase C inhibitor GF109203X. In Ca(2+)-free medium, pretreatment with the mitochondrial uncoupler, carbonylcyanide m-chlorophenylhydrazone (CCCP), and the endoplasmic reticulum Ca(2+) pump inhibitors, thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ), inhibited bisphenol A-induced Ca(2+) release. Conversely, pretreatment with bisphenol A abolished thapsigargin (or BHQ)- and CCCP-induced [Ca(2+)](i) rise. Inhibition of phospholipase C with U73122 abolished bisphenol-induced [Ca(2+)](i) rise. Bisphenol A caused a concentration-dependent decrease in cell viability via apoptosis in a Ca(2+)-independent manner. Collectively, in MDCK cells, bisphenol A induced [Ca(2+)](i) rises by causing phospholipase C-dependent Ca(2+) release from the endoplasmic reticulum and mitochondria and Ca(2+) influx via phospholipase A2-, protein kinase C-sensitive, store-operated Ca(2+) channels.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Disruptores Endócrinos/toxicidade , Túbulos Renais/efeitos dos fármacos , Fenóis/toxicidade , Animais , Compostos Benzidrílicos , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Interpretação Estatística de Dados , Diploide , Cães , Relação Dose-Resposta a Droga , Citometria de Fluxo , Túbulos Renais/citologia , Túbulos Renais/enzimologia , Túbulos Renais/metabolismo , Inibidores de Fosfolipase A2 , Fosfolipases Tipo C/antagonistas & inibidores
13.
Basic Clin Pharmacol Toxicol ; 109(2): 103-10, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21371263

RESUMO

The effect of sertraline, an antidepressant, on cytosolic-free Ca(2+) levels ([Ca(2+) ](i) ) in human cancer cells is unclear. This study examined whether sertraline altered basal [Ca(2+) ](i) levels in suspended PC3 human prostate cancer cells by using fura-2 as a Ca(2+) -sensitive fluorescent probe. At concentrations of 10-150 µM, sertraline induced a [Ca(2+) ](i) rise in a concentration-dependent fashion. The Ca(2+) signal was reduced partly by removing extracellular Ca(2+) indicating that Ca(2+) entry and release both contributed to the [Ca(2+) ](i) rise. Sertraline induced Mn(2+) influx, leading to quench of fura-2 fluorescence suggesting Ca(2+) influx. This Ca(2+) influx was inhibited by the suppression of store-operated Ca(2+) channels or by the modulation of protein kinase C activity. In Ca(2+) -free medium, pre-treatment with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin or 2,5-di-(t-butyl)-1,4-hydroquinone nearly abolished sertraline-induced Ca(2+) release. Conversely, pre-treatment with sertraline greatly reduced the inhibitor-induced [Ca(2+) ](i) rise, suggesting that sertraline released Ca(2+) from the endoplasmic reticulum. Inhibition of phospholipase C inhibited sertraline-induced [Ca(2+) ](i) rise. At 20-30 µM, sertraline killed cells in a concentration-dependent manner. The cytotoxic effect of sertraline was enhanced by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/AM. Annexin V-FITC data suggest that sertraline (20 and 30 µM) evoked apoptosis in a concentration-dependent manner. Together, in PC3 human prostate cancer cells, sertraline induced [Ca(2+) ](i) rises by causing phospholipase C-dependent Ca(2+) release from the endoplasmic reticulum and via multiple Ca(2+) influx pathways that involve store-operated Ca(2+) channels. Sertraline also induced apoptosis that was not triggered by [Ca(2+) ](i) rise.


Assuntos
Antidepressivos/farmacologia , Cálcio/metabolismo , Neoplasias da Próstata/metabolismo , Sertralina/farmacologia , Apoptose , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Manganês/metabolismo , Neoplasias da Próstata/patologia , Fosfolipases Tipo C/fisiologia
14.
J Recept Signal Transduct Res ; 31(3): 247-55, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21410406

RESUMO

Effect of the carcinogen thapsigargin on human prostate cancer cells is unclear. This study examined if thapsigargin altered basal [Ca²âº](i) levels in suspended PC3 human prostate cancer cells by using fura-2 as a Ca²âº-sensitive fluorescent probe. Thapsigargin at concentrations between 10 nM and 10 µM increased [Ca²âº](i) in a concentration-dependent fashion. The Ca²âº signal was reduced partly by removing extracellular Ca²âº indicating that Ca²âº entry and release both contributed to the [Ca²âº](i) rise. This Ca²âº influx was inhibited by suppression of phospholipase A2, but not by inhibition of store-operated Ca²âº channels or by modulation of protein kinase C activity. In Ca²âº-free medium, pretreatment with the endoplasmic reticulum Ca²âº pump inhibitor 2,5-di-(t-butyl)-1,4-hydroquinone (BHQ) nearly abolished thapsigargin-induced Ca²âº release. Conversely, pretreatment with thapsigargin greatly reduced BHQ-induced [Ca²âº](i) rise, suggesting that thapsigargin released Ca²âº from the endoplasmic reticulum. Inhibition of phospholipase C did not change thapsigargin-induced [Ca²âº](i) rise. At concentrations of 1-10 µM, thapsigargin induced cell death that was partly reversed by chelation of Ca²âº with BAPTA/AM. Annexin V/propidium iodide staining data suggest that apoptosis was partly responsible for thapsigargin-induced cell death. Together, in PC3 human prostate cancer cells, thapsigargin induced [Ca²âº](i) rises by causing phospholipase C-independent Ca²âº release from the endoplasmic reticulum and Ca²âº influx via phospholipase A2-sensitive Ca²âº channels. Thapsigargin also induced cell death via Ca²âº-dependent pathways and Ca²âº-independent apoptotic pathways.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Tapsigargina/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Aristolóquicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Estrenos/farmacologia , Fluorescência , Fura-2/metabolismo , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Masculino , Manganês/metabolismo , Neoplasias da Próstata/enzimologia , Pirrolidinonas/farmacologia , Fosfolipases Tipo C/metabolismo
15.
Hum Exp Toxicol ; 30(10): 1635-43, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21247994

RESUMO

Effect of sertraline, an antidepressant, on cytosolic free Ca(2+) levels ([Ca(2+)](i)) in human cancer cells is unclear. This study examined if sertraline altered basal [Ca(2+)](i) levels in suspended OC2 human oral cancer by using fura-2 as a Ca(2+)-sensitive fluorescent probe. At concentrations of 10-100 µM, sertraline induced a [Ca(2+)](i) rise in a concentration-dependent fashion. The Ca(2+) signal was reduced partly by removing extracellular Ca(2+) indicating that Ca(2+) entry and release both contributed to the [Ca(2+)](i) rise. Sertraline induced Mn(2+) influx, leading to quench of fura-2 fluorescence suggesting Ca(2+) influx. This Ca(2+) influx was inhibited by suppression of phospholipase A2, inhibition of store-operated Ca(2+) channels or by modulation of protein kinase C activity. In Ca(2+)-free medium, pretreatment with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin or 2,5-di-(t-butyl)-1,4-hydroquinone (BHQ) nearly abolished sertraline-induced Ca(2+) release. Conversely, pretreatment with sertraline greatly reduced the inhibitor-induced [Ca(2+)](i) rise, suggesting that sertraline released Ca(2+) from the endoplasmic reticulum. Inhibition of phospholipase C did not change sertraline-induced [Ca(2+)](i) rise. Together, in human oral cancer cells, sertraline induced [Ca(2+)](i) rises by causing phospholipase C-independent Ca(2+) release from the endoplasmic reticulum and Ca(2+) influx via store-operated Ca(2+) channels.


Assuntos
Antidepressivos/farmacologia , Cálcio/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sertralina/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Fluorescência , Fura-2/metabolismo , Humanos , Manganês/metabolismo
16.
Basic Clin Pharmacol Toxicol ; 108(5): 341-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21205218

RESUMO

The effect of the insecticide methoxychlor on the physiology of oral cells is unknown. This study aimed to explore the effect of methoxychlor on cytosolic Ca(2+) concentrations ([Ca(2+)](i)) in human oral cancer cells (OC2) by using the Ca(2+)-sensitive fluorescent dye fura-2. Methoxychlor at 5-20 µM increased [Ca(2+)](i) in a concentration-dependent manner. The signal was reduced by 70% by removing extracellular Ca(2+). Methoxychlor-induced Ca(2+) entry was not affected by nifedipine, econazole, SK&F96365 and protein kinase C modulators but was inhibited by the phospholipase A2 inhibitor aristolochic acid. In Ca(2+)-free medium, treatment with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) inhibited or abolished methoxychlor-induced [Ca(2+)](i) rise. Incubation with methoxychlor also inhibited thapsigargin- or BHQ-induced [Ca(2+)](i) rise. Inhibition of phospholipase C with U73122 did not alter methoxychlor-induced [Ca(2+)](i) rise. At 5-20 µM, methoxychlor killed cells in a concentration-dependent manner. The cytotoxic effect of methoxychlor was not reversed by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/AM (BAPTA/AM). Annexin V-FITC data suggest that methoxychlor (10 and 20 µM) evoked apoptosis in a concentration-dependent manner. Together, in human OC2, methoxychlor induced a [Ca(2+)](i) rise probably by inducing phospholipase C-independent Ca(2+) release from the endoplasmic reticulum and Ca(2+) entry via phospholipase A(2)-sensitive channels. Methoxychlor induced cell death that may involve apoptosis.


Assuntos
Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Inseticidas/farmacologia , Metoxicloro/farmacologia , Neoplasias Bucais/patologia , Análise de Variância , Apoptose , Sinalização do Cálcio/efeitos dos fármacos , Morte Celular , Linhagem Celular Tumoral , Citosol/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/patologia , Estrenos/metabolismo , Fura-2 , Humanos , Hidroquinonas/metabolismo , Nifedipino/metabolismo , Proteína Quinase C/metabolismo , Pirrolidinonas/metabolismo , Fosfolipases Tipo C/antagonistas & inibidores
17.
Chin J Physiol ; 52(3): 128-35, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19777798

RESUMO

The effect of Antrodia camphorata (AC) on human oral cancer cells has not been explored. This study examined the effect of AC on the viability, apoptosis, mitogen-activated protein kinases (MAPKs) phosphorylation and Ca2+ regulation of OC2 human oral cancer cells. AC at a concentration of 25 microM induced an increase in cell viability, but AC at concentrations > or = 50 microg/ml decreased viability in a concentration-dependent manner. AC at concentrations of 100-200 microg/ml induced apoptosis in a concentration-dependent manner as demonstrated by propidium iodide staining. AC (25 microg/ml) did not alter basal [Ca2+]i, but decreased the [Ca2+]i increases induced by ATP, bradykinin, histamine and thapsigargin. ATP, bradykinin, and histamine increased cell viability whereas thapsigargin decreased it. AC (25 microg/ml) pretreatment failed to alter ATP-induced increase in viability, potentiated bradykinin-induced increase in viability, decreased histamine-induced increase in viability and reversed thapsigargin-induced decrease in viability. Immunoblotting suggested that AC induced phosphorylation of ERK and JNK MAPKs, but not p38 MAPK. Collectively, for OC2 cells, AC exerted multiple effects on their viability and [Ca2+]i, induced their ERK and JNK MAPK phosphorylation, and probably evoked their apoptosis.


Assuntos
Antrodia , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Carcinoma de Células Escamosas/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neoplasias Bucais/patologia , Extratos Vegetais/farmacologia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , MAP Quinase Quinase 4/metabolismo , Neoplasias Bucais/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Chin J Physiol ; 51(2): 78-84, 2008 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-18666710

RESUMO

Antrodia camphorata (AC) has been used as a health supplement in Asia to control different cancers; however, the cellular mechanisms of its effects are unclear. The effect of AC on cultured human prostate cancer cells (PC3) has not been explored. This study examined the effect of AC on viability, apoptosis, mitogen-activated protein kinases (MAPKs) phosphorylation and Ca2+ handling in PC3 cells. AC at concentrations of 5-50 microg/ml did not affect cell viability, but at 100-200 microg/ml decreased viability and induced apoptosis in a concentration-dependent manner. AC at concentrations of 25-200 microg/ml did not alter basal [Ca2+]i, but at a concentration of 25 microg/ml decreased the [Ca2+]i increases induced by ATP, bradykinin, histamine and thapsigargin. ATP, bradykinin and histamine increased cell viability whereas thapsigargin decreased it. AC (25 microg/ml) pretreatment inhibited ATP-, bradykinin-, and histamine-induced enhancement on viability, but reversed thapsigargin-induced cytotoxicity. Immunoblotting showed that AC (200 microg/ml) did not induce the phosphorylation of ERK, JNK, and p38 MAPKs. Collectively, in PC3 cells, AC exerted multiple effects on viability and [Ca2+]i, caused apoptosis via pathways unrelated to [Ca2+]i signal and phosphorylation of ERK, JNK and p38 MAPKs.


Assuntos
Agaricales , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Polyporales/química , Neoplasias da Próstata/tratamento farmacológico , Trifosfato de Adenosina/antagonistas & inibidores , Antineoplásicos/farmacologia , Bradicinina/antagonistas & inibidores , Sobrevivência Celular/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos/farmacologia , Humanos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Tapsigargina/antagonistas & inibidores , Células Tumorais Cultivadas
19.
Clin Exp Pharmacol Physiol ; 34(7): 581-5, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17581212

RESUMO

1. Short-wave diathermy (SWD) is a form of radiofrequency radiation that is used therapeutically by physiotherapists. The cellular mechanisms of SWD are unclear. The present study was performed to explore the effect of different conditions of short-wave exposure on the proliferation of cultured human chondrocytes. 2. Cells exposed to short waves once per day for seven consecutive days exhibited a significant increase in proliferation by 42% compared with the control cells. In cells that were treated with short waves twice per day for seven consecutive days, or only once on Day 1 and then examined for proliferation on Day 7, cell proliferation was greater than the control cells by 40% and 30%, respectively. 3. Given the importance of mitogen-activated protein kinases (MAPK) in the proliferation of different cell types, efforts were extended to explore the role of three major types of MAPK; that is, extracellular signal-regulated kinase (ERK), c-Jun NH(2)-terminal protein kinase (JNK) and p38. 4. It was found that the level of phosphorylated ERK (phospho-ERK 1 and ERK 2) increased significantly within 5-120 min following consecutive exposure to short waves for 7 days. Exposure to short waves failed to alter the intensity of phosphorylated JNK and p38 within 0-240 min. 5. Cells were exposed to short waves once for seven consecutive days in the presence of 0, 10 micromol/L, 20 micromol/L or 50 micromol/L PD98059 (an ERK inhibitor). PD98059 totally inhibited short waves-induced enhancement of proliferation without altering normal control viability. In the presence of short waves and PD98059, the cell viability was lower than the normal control. Together, the data suggest that short waves could increase proliferation in human chondrocytes through activation of the ERK pathway, which is also involved in maintaining normal cell proliferation under physiological conditions.


Assuntos
Proliferação de Células/efeitos da radiação , Condrócitos/efeitos da radiação , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Ondas de Rádio , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Condrócitos/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Flavonoides/farmacologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Fatores de Tempo
20.
Life Sci ; 80(4): 364-9, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17054998

RESUMO

Melittin, a peptide from bee venom, is thought to be a phospholipase A(2) activator and Ca(2+) influx inducer that can evoke cell death in different cell types. However, the effect of melittin on cytosolic free Ca(2+) concentration ([Ca(2+)](i)) and viability has not been explored in human osteoblast-like cells. This study examined whether melittin altered [Ca(2+)](i) and killed cells in MG63 human osteosarcoma cells. [Ca(2+)](i) changes and cell viability were measured by using the fluorescent dyes fura-2 and WST-1, respectively. Melittin at concentrations above 0.075 microM increased [Ca(2+)](i) in a concentration-dependent manner. The Ca(2+) signal was abolished by removing extracellular Ca(2+). Melittin-induced Ca(2+) entry was confirmed by Mn(2+) quenching of fura-2 fluorescence at 360 nm excitation wavelength which was Ca(2+)-insensitive. The melittin-induced Ca(2+) influx was unchanged by modulation of protein kinase-C activity with phorbol 12-myristate 13-acetate (PMA) and GF 109203X, or inhibition of phospholipase A(2) with AACOCF(3) and aristolochic acid; but was substantially inhibited by blocking L-type Ca(2+) channels. At concentrations of 0.5 microM and 1 microM, melittin killed 33% and 45% of cells, respectively, via inducing apoptosis. Lower concentrations of melittin failed to kill cells. The cytotoxic effect of 1 microM melittin was completely reversed by pre-chelating cytosolic Ca(2+) with BAPTA. Taken together, these data showed that in MG63 cells, melittin induced a [Ca(2+)](i) increase by causing Ca(2+) entry through L-type Ca(2+) channels in a manner independent of protein kinase-C and phospholipase A(2) activity; and this [Ca(2+)](i) increase subsequently caused apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Canais de Cálcio Tipo L/efeitos dos fármacos , Cálcio/metabolismo , Meliteno/farmacologia , Osteoblastos/efeitos dos fármacos , Fosfolipases A/metabolismo , Canais de Cálcio Tipo L/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/metabolismo , Fura-2/metabolismo , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteossarcoma , Fosfolipases A/antagonistas & inibidores , Fosfolipases A2 , Acetato de Tetradecanoilforbol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...