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








Base de dados
Intervalo de ano de publicação
1.
Int J Oncol ; 47(5): 1711-24, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26398114

RESUMO

Controversial effects of thalidomide for solid malignancies have been reported. In the present study, we evaluate the effects of thalidomide for transitional cell carcinoma (TCC), the most common type of bladder cancer. Thalidomide precipitates were observed when its DMSO solution was added to the culture medium. No precipitation was found when thalidomide was dissolved in 45% γ-cyclodextrin, and this concentration of γ-cyclodextrin elicited slight cytotoxicity on TCC BFTC905 and primary human urothelial cells. Thalidomide-γ-cyclodextrin complex exerted a concentration-dependent cytotoxicity in TCC cells, but was relatively less cytotoxic (with IC50 of 200 µM) in BFTC905 cells than the other 3 TCC cell lines, possibly due to upregulation of Bcl-xL and HIF-1α mediated carbonic anhydrase IX, and promotion of quiescence. Gemcitabine-resistant BFTC905 cells were chosen for additional experiments. Thalidomide induced apoptosis through downregulation of survivin and securin. The secretion of VEGF and TNF-α was ameliorated by thalidomide, but they did not affect cell proliferation. Immune-modulating lenalidomide and pomalidomide did not elicit cytotoxicity. In addition, cereblon did not play a role in the thalidomide effect. Oxidative DNA damage was triggered by thalidomide, and anti-oxidants reversed the effect. Thalidomide also inhibited TNF-α induced invasion through inhibition of NF-κB, and downregulation of effectors, ICAM-1 and MMP-9. Thalidomide inhibited the growth of BFTC905 xenograft tumors in SCID mice via induction of DNA damage and suppression of angiogenesis. Higher average body weight, indicating less chachexia, was observed in thalidomide treated group. Sedative effect was observed within one-week of treatment. These pre-clinical results suggest therapeutic potential of thalidomide for gemcitabine-resistant bladder cancer.


Assuntos
Desoxicitidina/análogos & derivados , Neovascularização Patológica/tratamento farmacológico , Talidomida/administração & dosagem , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Dano ao DNA , Desoxicitidina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , NF-kappa B/biossíntese , Proteínas de Neoplasias/biossíntese , Neovascularização Patológica/patologia , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
2.
Oncol Rep ; 31(2): 771-80, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24297644

RESUMO

Survivin is overexpressed in transitional cell carcinoma (TCC), the most common type of bladder cancer. Previous reports demonstrated that knockdown of survivin by siRNA induced apoptosis of TCC cells. The present study evaluated the therapeutic effects of sepantronium bromide (YM155), a novel small molecule survivin inhibitor under clinical trials, on TCC cells in vitro. BFTC905, a grade III TCC cell line derived from a patient of blackfoot disease in Taiwan, was the most gemcitabine-resistant cell line when compared to BFTC909, TSGH8301 and T24 in cytotoxicity assay, resulting from upregulation of securin and bcl-2 after gemcitabine treatment. YM155 caused potent concentration­dependent cytotoxicity in 4 TCC cell lines (IC50s ≤20 nM), but exhibited no cytotoxicity in survivin-null primary human urothelial cells. For BFTC905 cells, addition of gemcitabine and/or cisplatin, the standard TCC chemotherapy regimen, to YM155 did not exert additive cytotoxic effects. Molecular analyses indicated that YM155 inhibited the proliferation of BFTC905 cells by increasing p27kip1, suppressing Ki-67, and inducing quiescence. In addition, YM155 elicited apoptosis manifested with DNA fragmentation through suppressing the expression of survivin, securin and bcl-2. Furthermore, YM155 induced autophagy in BFTC905 cells as autophagic inhibitor, 3-methyladenine, attenuated YM155-induced LC3B-II levels and reversed the cytotoxicity of YM155. mTOR inhibitors sirolimus and everolimus did not increase YM155-induced expression of LC3B-II nor augment YM155-induced cytotoxicity. These results indicate that YM155 exerts its lethal effect on BFTC905 cells via apoptotic and autophagic death pathways and suggest that YM155 may be a potential drug for the therapy of gemcitabine-resistant bladder cancer.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Carcinoma de Células de Transição/tratamento farmacológico , Imidazóis/farmacologia , Naftoquinonas/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Adenina/análogos & derivados , Adenina/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Fragmentação do DNA/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Everolimo , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Antígeno Ki-67/biossíntese , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Securina/biossíntese , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Survivina , Serina-Treonina Quinases TOR/antagonistas & inibidores , Urotélio/efeitos dos fármacos , Urotélio/patologia , Gencitabina
3.
Pediatr Neonatol ; 53(3): 199-204, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22770110

RESUMO

BACKGROUND: Pediatric glioblastoma is a malignant disease with an extremely poor clinical outcome. Patients usually suffer from resistance to radiation therapy, so targeted drug treatment may be a new possibility for glioblastoma therapy. Survivin is also overexpressed in glioblastoma. YM155, a novel small-molecule survivin inhibitor, has not been examined for its use in glioblastoma therapy. METHODS: The human glioblastoma cell line M059K, which expresses normal DNA-dependent protein kinase (DNA-PK) activity and is radiation-resistant, and M059J, which is deficient in DNA-PK activity and radiation-sensitive, were used in the study. Cell viability, DNA fragmentation, and the expression of survivin and securin following YM155 treatment were examined using MTT (methylthiazolyldiphenyl-tetrazolium) assay, ELISA assay, and Western blot analysis, respectively. RESULTS: YM155 caused a concentration-dependent cytotoxic effect, inhibiting the cell viability of both M059K and M059J cells by 70% after 48 hours of treatment with 50 nM YM155. The half-maximal inhibitory concentration (IC50) was around 30-35 nM for both cell lines. Apoptosis was determined to have occurred in both cell lines because immunoreactive signals from the DNA fragments in the cytoplasm were increased 24 hours after treatment with 30 nM YM155. The expression of survivin and securin in the M059K cells was greater than that measured in the M059J cells. Treatment with 30 nM YM155, for both 24 and 48 hours, significantly suppressed the expression of survivin and securin in both cell lines. CONCLUSION: The novel survivin inhibitor YM155 elicits potent cytotoxicity in glioblastoma cells in vitro via DNA-PK-independent mechanisms. YM155 could be used as a new therapeutic agent for the treatment of human glioblastomas.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Proteína Quinase Ativada por DNA/fisiologia , Glioblastoma/tratamento farmacológico , Imidazóis/farmacologia , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Naftoquinonas/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glioblastoma/enzimologia , Humanos , Proteínas Inibidoras de Apoptose/análise , Proteínas de Neoplasias/análise , Securina , Survivina
4.
Food Chem Toxicol ; 50(3-4): 1073-81, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22166790

RESUMO

Neuronal death induced by I6 displayed apoptotic characteristics but the precise mechanism has not been fully elucidated. In the present studies, I6 at 24 h after intraperitoneal administration significantly decreased the density of surviving neurons and increased caspase-3 activity in frontal cortex, suggesting that peripherally administered I6 may cross BBB to induce CNS toxicity. In rat embryonic primary cortical cells, I6-induced reduction of mitochondrial viability and neuronal apoptosis was inhibited by vitamin E. In addition, I6-induced reactive oxygen species (ROS) caused the disruption of mitochondria membrane potential (MMP), the release of cytochrome c, the activation of caspase-9 and caspase-3, and cleavage of poly(ADP-ribose) polymerase (PARP), resulting in activation of mitochondrial-mediated intrinsic death pathway. Pre-treatment with antioxidant vitamin E or N-acetylcysteine (NAC) completely abolished the I6-induced generation of ROS, loss of MMP, release of cytochrome c, activation of caspase-9 and caspase-3, and cleavage of PARP. Carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone (FCCP), a mitochondrial uncoupler, significantly reduced I6-induced neuronal death as well as caspase-3 activation and PARP cleavage. These results suggest that I6 induces neuronal death by promoting intracellular ROS production to cause a loss of MMP that result in release of cytochrome c and activation of mitochondria-mediated intrinsic death pathway.


Assuntos
Apoptose/efeitos dos fármacos , Guaiacol/análogos & derivados , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Barreira Hematoencefálica , Western Blotting , Caspase 3/metabolismo , Células Cultivadas , Ativação Enzimática , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/enzimologia , Guaiacol/farmacocinética , Guaiacol/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
J Biomed Sci ; 18: 69, 2011 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-21933448

RESUMO

BACKGROUND: Perinatal brain injury is the leading cause of subsequent neurological disability in both term and preterm baby. Glutamate excitotoxicity is one of the major factors involved in perinatal hypoxic-ischemic encephalopathy (HIE). Glutamate transporter GLT1, expressed mainly in mature astrocytes, is the major glutamate transporter in the brain. HIE induced excessive glutamate release which is not reuptaked by immature astrocytes may induce neuronal damage. Compounds, such as ceftriaxone, that enhance the expression of GLT1 may exert neuroprotective effect in HIE. METHODS: We used a neonatal rat model of HIE by unilateral ligation of carotid artery and subsequent exposure to 8% oxygen for 2 hrs on postnatal day 7 (P7) rats. Neonatal rats were administered three dosages of an antibiotic, ceftriaxone, 48 hrs prior to experimental HIE. Neurobehavioral tests of treated rats were assessed. Brain sections from P14 rats were examined with Nissl and immunohistochemical stain, and TUNEL assay. GLT1 protein expression was evaluated by Western blot and immunohistochemistry. RESULTS: Pre-treatment with 200 mg/kg ceftriaxone significantly reduced the brain injury scores and apoptotic cells in the hippocampus, restored myelination in the external capsule of P14 rats, and improved the hypoxia-ischemia induced learning and memory deficit of P23-24 rats. GLT1 expression was observed in the cortical neurons of ceftriaxone treated rats. CONCLUSION: These results suggest that pre-treatment of infants at risk for HIE with ceftriaxone may reduce subsequent brain injury.


Assuntos
Astrócitos/metabolismo , Ceftriaxona/farmacologia , Transportador 2 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Animais , Animais Recém-Nascidos , Western Blotting , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/prevenção & controle , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Testes Neuropsicológicos , Ratos
6.
J Gastroenterol ; 46(6): 822-33, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21437599

RESUMO

BACKGROUND: An overproduction of proinflammatory mediators in severe acute pancreatitis contributes to the systemic inflammatory response, which may lead to multiorgan damage and even death. Thus, inflammatory cytokines, e.g., tumor necrosis factor (TNF)-α and interleukin (IL)-1ß, may be novel targets for the treatment of acute pancreatitis. The aim of this study was to investigate the therapeutic effects of pomalidomide (or CC-4047), a thalidomide analog and immunomodulatory agent, in acute pancreatitis. METHODS: Acute pancreatitis was induced in C57BL/6 mice by intraperitoneal administration of cerulein (100 µg/kg/h × 8). Pomalidomide was administered (0.5 mg/kg orally) 1 h before the first or 1 h after the last cerulein administration. The severity of the acute pancreatitis was evaluated biochemically and morphologically. RESULTS: Pretreatment with pomalidomide significantly reduced the plasma levels of amylase and lipase; the histological injury; and the expression of TNF-α, IL-1ß, monocyte chemotactic protein-1 (MCP-1), and inducible nitric oxide synthase (iNOS) in cerulein-induced acute pancreatitis. Post-treatment with pomalidomide also decreased the cerulein-induced elevation of plasma amylase and lipase and decreased the pancreatic damage. CONCLUSIONS: Treatment with pomalidomide ameliorated the severity of cerulein-induced acute pancreatitis in mice. Our data suggest that pomalidomide may become a new therapeutic agent in future clinical trials for the treatment of acute pancreatitis.


Assuntos
Fatores Imunológicos/farmacologia , Pancreatite/tratamento farmacológico , Talidomida/análogos & derivados , Doença Aguda , Administração Oral , Amilases/sangue , Amilases/efeitos dos fármacos , Animais , Ceruletídeo/toxicidade , Modelos Animais de Doenças , Fatores Imunológicos/administração & dosagem , Lipase/sangue , Lipase/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pancreatite/fisiopatologia , Índice de Gravidade de Doença , Talidomida/administração & dosagem , Talidomida/farmacologia
7.
Int J Oncol ; 37(4): 943-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20811716

RESUMO

BDNF (brain-derived neurotrophic factor) and its receptor TrkB (tropomyosin receptor kinase B) play important roles in the progression of cancer, including transitional cell carcinoma (TCC) cells reported in our previous investigation. In this study, we used a specific TrkB antibody (Ab) to evaluate its effects on survival, proliferation and migration/invasion in three TCC cell lines (BFTC905, T24 and TSGH8301) in vitro. The TrkB Ab at 1 and 3 microg/ml, but not the TrkA or TrkC Abs, significantly elicited cytotoxicity in TCC cells. The TrkB Ab at 3 microg/ml also induced apoptosis of TCC cells, which may result from up-regulation of phospho-p38 plus down-regulation of survivin and securin expression. The TrkB Ab at 0.5 microg/ml, which did not show cytotoxicity, suppressed migration of TCC cells and invasion of BFTC905 cells, possibly mediated through increased E-cadherin, decreased BDNF-stimulated phospho-PLCgamma1 and reduced MMP-9 activity. These results indicate that TrkB blockade may be a new strategy for TCC therapy.


Assuntos
Anticorpos/farmacologia , Carcinoma de Células de Transição/patologia , Movimento Celular/efeitos dos fármacos , Receptor trkB/antagonistas & inibidores , Antígenos CD , Apoptose/efeitos dos fármacos , Caderinas/metabolismo , Carcinoma de Células de Transição/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Proteínas Inibidoras de Apoptose , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação , Receptor trkB/imunologia , Receptor trkB/metabolismo , Securina , Survivina , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Oncol Rep ; 24(5): 1265-70, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20878119

RESUMO

BDNF (brain-derived neurotrophic factor) and TrkB (tropomyosin receptor kinase B) are expressed in several tumor types. However, the existence of BDNF and TrkB in human bladder cancer, especially transitional cell carcinoma (TCC), has not been established. In this study, commercial TCC tissue arrays were used. Slides of paraffin-fixed human bladder tissues included all grades of TCC (13, 30 and 22 tissue samples in grade I, II and III, respectively), superficial and invasive TCC (31 and 34 tissue samples, respectively), paired malignancy-uninvolved urothelium (35 tissue samples) and normal urothelial tissues (12 tissue samples). The intensities of BDNF and TrkB immunostaining were graded as background, mild and strong (score as 0, 1 and 2, respectively). The results showed significantly overexpressed BDNF and TrkB in TCC samples compared to normal urothelium. According to grade assignment of TCC samples, BDNF in grade III and TrkB in grade I and III appeared to be overexpressed. BDNF and TrkB were overexpressed in superficial TCC samples according to staging classification. The score between the paired TCC and its uninvolved urothelium was not statistically different. In conclusion, the existence of overexpressed BDNF and TrkB in human TCC has been demonstrated in our study. A strategy involving BDNF/TrkB blockade may be a new hope for TCC target therapy.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Carcinoma de Células de Transição/metabolismo , Receptor trkB/biossíntese , Neoplasias da Bexiga Urinária/metabolismo , Carcinoma de Células de Transição/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Estadiamento de Neoplasias , Neoplasias da Bexiga Urinária/patologia
9.
Int J Oncol ; 36(6): 1469-76, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20428771

RESUMO

Pathologically, >90% of bladder cancer is transitional cell carcinoma (TCC). Previously, brain-derived neurotrophic factor (BDNF) but not tropomyosin-related kinase B (TrkB) was found in normal urothelium. TrkB activation by BDNF has been shown to promote the progression of several cancers, however, the existence and functional roles of both BDNF and TrkB in TCC have not been elucidated. In this study, three human TCC cell lines, BFTC905, TSGH8301, and T24 were used for the investigation. Both BDNF and TrkB but not TrkA or TrkC identified by RT-PCR and Western blotting were found in these cell lines. Immunostaining demonstrated the cytosolic expression of BDNF and TrkB, as well as membranous expression of TrkB in these cells. BDNF released from three cell lines was also detected in culture medium by ELISA. The proliferation of BFTC905 cells was enhanced by recombinant human BDNF (rhBDNF) in vitro, which was associated with increased phospho-TrkB and phospho-ERK levels. In contrast, TrkB-Fc chimeric protein served as BDNF scavenger eliciting cytotoxicity. Addition of rhBDNF in these cell lines cultured in poly-HEMA [Poly(2-hydroxyethyl methacrylate)] coated dishes for 48 h did not confer resistance to anoikis. Increased phospho-Akt expression was observed transiently within an hour after rhBDNF administration but disappeared 24 h later. Weekly injections of 100 ng rhBDNF into the cancer cell-loading site for 6 weeks promoted BFTC905 xenograft growth in SCID mice. Daily injection of 5 microg TrkB-Fc chimeric protein into the tumor 2 weeks after tumor cell implantation delayed tumor growth concomitant with phospho-TrkB suppression in xenografts. These results indicate that BDNF binding to TrkB receptor is a survival signal for TCC cells. Drugs that block BDNF or TrkB may provide a new and potential approach for TCC therapy.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Carcinoma de Células de Transição/metabolismo , Ativação Enzimática/fisiologia , Receptor trkB/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Animais , Anoikis/fisiologia , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Carcinoma de Células de Transição/genética , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/fisiologia , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Receptor trkB/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Biomed Sci ; 17: 4, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20074377

RESUMO

BACKGROUND: Caffeine, a nonselective adenosine A1 and A(2A) receptor antagonist, is the most widely used psychoactive substance in the world. Evidence demonstrates that caffeine and selective adenosine A(2A) antagonists interact with the neuronal systems involved in drug reinforcement, locomotor sensitization, and therapeutic effect in Parkinson's disease (PD). Evidence also indicates that low doses of caffeine and a selective adenosine A(2A) antagonist SCH58261 elicit locomotor stimulation whereas high doses of these drugs exert locomotor inhibition. Since these behavioral and therapeutic effects are mediated by the mesolimbic and nigrostriatal dopaminergic pathways which project to the striatum, we hypothesize that low doses of caffeine and SCH58261 may modulate the functions of dopaminergic neurons in the striatum. METHODS: In this study, we evaluated the neuroadaptations in the striatum by using reverse-phase high performance liquid chromatography (HPLC) to quantitate the concentrations of striatal dopamine and its metabolites, dihydroxylphenylacetic acid (DOPAC) and homovanilic acid (HVA), and using immunoblotting to measure the level of phosphorylation of tyrosine hydroxylase (TH) at Ser31, following chronic caffeine and SCH58261 sensitization in mice. Moreover, to validate further that the behavior sensitization of caffeine is through antagonism at the adenosine A(2A) receptor, we also evaluate whether chronic pretreatment with a selective adenosine A(2A) antagonist SCH58261 or a selective adenosine A1 antagonist DPCPX can sensitize the locomotor stimulating effects of caffeine. RESULTS: Chronic treatments with low dose caffeine (10 mg/kg) or SCH58261 (2 mg/kg) increased the concentrations of dopamine, DOPAC and HVA, concomitant with increased TH phosphorylation at Ser31 and consequently enhanced TH activity in the striatal tissues in both caffeine- and SCH58261-sensitized mice. In addition, chronic caffeine or SCH58261 administration induced locomotor sensitization, and locomotor cross-sensitization to caffeine was observed following chronic treatment of mice with SCH58261 but not with DPCPX. CONCLUSIONS: Our study demonstrated that low dosages of caffeine and a selective adenosine A(2A) antagonist SCH58261 elicited locomotor sensitization and cross-sensitization, which were associated with elevated dopamine concentration and TH phosphorylation at Ser31 in the striatum. Blockade of adenosine A(2A) receptor may play an important role in the striatal neuroadaptations observed in the caffeine-sensitized and SCH58261-sensitized mice.


Assuntos
Antagonistas do Receptor A2 de Adenosina , Cafeína/farmacologia , Dopamina/metabolismo , Neostriado/metabolismo , Fármacos Neuroprotetores/farmacologia , Pirimidinas/farmacologia , Triazóis/farmacologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Cafeína/administração & dosagem , Ácido Homovanílico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Pirimidinas/administração & dosagem , Serina/química , Triazóis/administração & dosagem , Tirosina 3-Mono-Oxigenase/metabolismo , Xantinas/metabolismo
11.
Psychopharmacology (Berl) ; 204(2): 313-25, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19169672

RESUMO

RATIONALE: Caffeine, an antagonist of adenosine A(1) and A(2A) receptor, is the most widely used psychoactive substance in the world. Evidence indicates that caffeine interacts with the neuronal systems involved in drug reinforcing. Although adenosine A(1) and/or A(2A) receptor have been found to play important roles in the locomotor stimulation and probably reinforcing effect of caffeine, the relative contribution of the A(1) and/or A(2A) receptors to the acute and chronic motor activation and reinforcing effects of caffeine has not been completely investigated. OBJECTIVE: The roles of adenosine A(1) and/or A(2A) receptor and the association of phospho-Thr75-dopamine- and cAMP-regulated phosphoprotein of molecular weight 32 kDa (DARPP-32) in the motor activation and reinforcing effects of caffeine, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), a selective A(1) antagonist, and 5-amino-7-(beta-phenylethyl)-2-(8-furyl) pyrazolol [4,3-e]-1,2,4-triazolol [1,5-c] pyrimidine (SCH58261), a selective A(2A) receptor antagonist were examined. METHODS: Locomotor stimulation and behavioral sensitization of caffeine, DPCPX, and SCH58261 were studied in C57BL/6 male mice following acute and chronic administration. Conditioned place preference (CPP) paradigm was used to evaluate the drug-seeking potential of these compounds. Furthermore, the expression of phospho-Thr75-DARPP-32 in striatal membrane from behaviorally sensitized mice was analyzed by Western blot. RESULTS: Caffeine and SCH58261 but not DPCPX induced CPP and locomotor sensitization in C57BL/6 mice. The locomotor sensitization after chronic treatment was associated with increased DARPP-32 phosphorylation at Thr75 in the striatum. CONCLUSION: Caffeine-induced reinforcing effect and behavioral sensitization are mediated by antagonism at adenosine A(2A) receptor. These effects are associated with phosphorylation of DARPP-32 at Thr75 in the striatum.


Assuntos
Antagonistas do Receptor A2 de Adenosina , Cafeína/farmacologia , Condicionamento Operante/efeitos dos fármacos , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Recompensa , Animais , Western Blotting , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Antagonistas de Receptores Purinérgicos P1 , Pirimidinas/farmacologia , Triazóis/farmacologia , Xantinas/farmacologia
12.
Apoptosis ; 13(7): 883-94, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18483861

RESUMO

The objective of this study was to evaluate the cardiac toxicity induced by carboplatin, a second generation platinum-containing anti-cancer drug, and to test whether pravastatin can reduce this cardio-toxicity. In the present study, infusion of carboplatin (100 mg/kg) to mice resulted in decreased survival rates and abnormal cardiac histology, concomitant with increased cardiac apoptosis. In addition, treatment of cultured rat cardiomyocytes with carboplatin (100 muM for 48 h) caused marked apoptosis and increased caspase-3, -9, and cytochrome C, but decreased BCL-XL protein expression, and this was inhibited by reactive oxygen species (ROS) scavenger n-acetylcysteine. Furthermore, pretreatment of cardiomyocytes with pravastatin (20 microM) before carboplatin exposure significantly attenuated apoptosis and decreased caspase-3, -9, cytochrome C activity. Lastly, mice pre-treated with pravastatin before carboplatin treatment showed improved survival rate and cardiac function, with reduced cardiomyocyte apoptosis via activating Akt and restoring normal mitochondrial HAX-1 in heart tissue. In summary, our results show that carboplatin can induce cardiotoxicity in vivo and in cultured cells via a mitochondrial pathway related to ROS production, whereas pravastatin administration can reduce such oxidative stress thus prevented cardiac apoptosis. Therefore, pravastatin can be used as a cytoprotective agent prior to carboplatin chemotherapy.


Assuntos
Antineoplásicos/antagonistas & inibidores , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Carboplatina/antagonistas & inibidores , Carboplatina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Pravastatina/farmacologia , Animais , Antineoplásicos/administração & dosagem , Apoptose/fisiologia , Carboplatina/administração & dosagem , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/prevenção & controle , Proteínas de Transporte/metabolismo , Caspases/metabolismo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/fisiopatologia , Marcação In Situ das Extremidades Cortadas , Peptídeos e Proteínas de Sinalização Intracelular , Leucopenia/induzido quimicamente , Leucopenia/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pravastatina/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
13.
Mol Pharmacol ; 72(5): 1238-45, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17671096

RESUMO

Peroxisome proliferator-activated receptor-alpha (PPAR-alpha) is a transcription factor and has been reported to inhibit cisplatin-mediated proximal tubule cell death. In addition, doxorubicin (Adriamycin)-induced nephrosis in rats is a commonly used experimental model for pharmacological studies of human chronic renal diseases. In this study, we investigated the protective effect of PPAR-alpha on doxorubicin-induced apoptosis and its detailed mechanism in NRK-52E cells and animal models. The mRNA level of PPAR-alpha was found to be reduced by doxorubicin treatment in NRK-52E cells. PPAR-alpha overexpression in NRK-52E cells significantly inhibited doxorubicin-induced apoptosis and the quantity of cleaved caspase-3. Endogenous prostacyclin (PGI(2)) augmentation, which has been reported to protect NRK-52E cells from doxorubicin-induced apoptosis, induced the translocation and activation of PPAR-alpha. The transformation of PPAR-alpha short interfering RNA was applied to silence the PPAR-alpha gene, which abolished the protective effect of PGI(2) augmentation in doxorubicin-treated cells. To confirm the protective role of PPAR-alpha in vivo, PPAR-alpha activator docosahexaenoic acid (DHA) was administered to doxorubicin-treated mice, and it has been shown to significantly reduce the doxorubicin-induced apoptotic cells in renal cortex. However, this protective effect of DHA did not exist in PPAR-alpha-deficient mice. In NRK-52E cells, the overexpression of PPAR-alpha elevated the activity of catalase and superoxide dismutase and inhibited doxorubicin-induced reactive oxygen species (ROS). PPAR-alpha overexpression also inhibited the doxorubicin-induced activity of nuclear factor-kappaB (NF-kappaB), which was associated with the interaction between PPAR-alpha and NF-kappaB p65 subunit as revealed in immunoprecipitation assays. Therefore, PPAR-alpha is capable of inhibiting doxorubicin-induced ROS and NF-kappaB activity and protecting NRK-52E cells from doxorubicin-induced apoptosis.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Citoproteção , Doxorrubicina/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , PPAR alfa/fisiologia , Adenoviridae/genética , Animais , Apoptose , Linhagem Celular , Ciclo-Oxigenase 1/genética , Sistema Enzimático do Citocromo P-450/genética , Humanos , Oxirredutases Intramoleculares/genética , Túbulos Renais Proximais/metabolismo , PPAR alfa/antagonistas & inibidores , PPAR alfa/genética , RNA Mensageiro/metabolismo , Ratos , Espécies Reativas de Oxigênio , Transfecção
14.
Toxicol Appl Pharmacol ; 222(1): 97-104, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17540426

RESUMO

Cancer cells express survivin that facilitates tumorigenesis. Celecoxib has been shown to reduce human colorectal cancers. However, the role and regulation of survivin by celecoxib in colorectal carcinoma cells remain unclear. Treatment with 40-80 muM celecoxib for 24 h induced cytotoxicity and proliferation inhibition via a concentration-dependent manner in RKO colorectal carcinoma cells. Celecoxib blocked the survivin protein expression and increased the phosphorylation of H2AX at serine-193 (gamma-H2AX). The survivin gene knockdown by transfection with a survivin siRNA revealed that the loss of survivin correlated with the expression of gamma-H2AX. Meanwhile, celecoxib increased caspase-3 activation and apoptosis. Celecoxib activated the phosphorylation of p38 mitogen-activated protein (MAP) kinase. The phosphorylated proteins of p38 MAP kinase and gamma-H2AX were observed in the apoptotic cells. SB203580, a specific p38 MAP kinase inhibitor, protected the survivin protein expression and decreased the levels of gamma-H2AX and apoptosis in the celecoxib-exposed cells. The blockade of survivin expression increased the celecoxib-induced cytotoxicity; conversely, overexpression of survivin by transfection with a survivin-expressing vector raised the cancer cell proliferation and resisted the celecoxib-induced cell death. Our results provide for the first time that p38 MAP kinase participates in the down-regulation of survivin and subsequently induces the activation of gamma-H2AX for mediating apoptosis following treatment with celecoxib in human colorectal cancer cells.


Assuntos
Neoplasias Colorretais/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Histonas/biossíntese , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas de Neoplasias/biossíntese , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Celecoxib , Contagem de Células , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Imunofluorescência , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Inibidoras de Apoptose , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias/genética , Fosforilação , RNA Interferente Pequeno/farmacologia , Survivina , Transfecção
15.
Sleep ; 29(3): 285-93, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16553013

RESUMO

STUDY OBJECTIVE: To clarify the cortical evoked responses in the primary somatosensory cortex of the rat under states of waking, slow-wave sleep (SWS), paradoxical sleep (PS), and spike-wave discharges (SWDs), which are associated with absence seizure. DESIGN: Somatosensory evoked potentials (SEPs) in response to single- and paired-pulse stimulations under waking, SWS, PS, and SWDs were compared. SEPs to a single-pulse stimulus with regard to cortical spikes of sleep spindles and SWDs were also evaluated. PARTICIPANTS: Twenty Long Evans rats. INTERVENTIONS: Single- and paired-pulse innocuous electrical stimulations were applied to the tail of rats with chronically implanted electrodes in the primary somatosensory cortex and neck muscle under waking, SWS, PS, and SWDs. MEASUREMENTS AND RESULTS: SEPs displayed distinct patterns under waking/PS and SWS/SWDs. The short-latency P1-N1 wave of the SEP was severely impeded during SWDs but not in other states. Reduction of the P1-N1 magnitude to the second stimulus of the paired-pulse stimulus for interstimulus intervals of < or = 300 milliseconds appeared in waking and PS states, but the decrease occurred only at particular interstimulus intervals under SWS. Interestingly, augmentation was found under SWDs. Moreover, cyclic augmentation of the P1-N1 magnitude was associated with spindle spikes, but cyclic reduction was observed with SWD spikes. CONCLUSION: Changes in SEPs are not only behavior dependent, but also phase locked onto ongoing brain activity. Distinct short-term plasticity of SEPs during sleep spindles or SWDs may merit further studies for seizure control and tactile information processing.


Assuntos
Epilepsia Tipo Ausência/fisiopatologia , Potenciais Somatossensoriais Evocados/fisiologia , Córtex Somatossensorial/fisiopatologia , Vigília/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Lateralidade Funcional/fisiologia , Polissonografia , Ratos , Ratos Long-Evans , Sono/fisiologia
16.
Biochem Pharmacol ; 70(5): 658-67, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16004971

RESUMO

The cyclooxygenase-2 (COX-2) protein is highly expressed in a variety of human cancers and has been reported to promote tumor growth. Non-steroidal anti-inflammatory drugs such as etodolac and celecoxib have been shown to inhibit COX-2 activity and may play a role in the chemoprevention of cancer. Oxaliplatin is a third-generation platinum compound that exhibits a different spectrum of activity compared with cisplatin. Other cisplatin-resistant tumors can still respond to oxaliplatin. However, the anticancer ability of the combination of COX-2 inhibitors and oxaliplatin is still unknown. In this study, we investigated the effects of combination of COX-2 inhibitors and oxaliplatin on the cell growth and survival in human colon cancer cells. Treatments with etodolac (0.3-0.5 mM) or celecoxib (20-80 microM) for 24 h concentration-dependently induced the cytotoxicity in the RKO colon carcinoma cells. Etodolac and celecoxib did not alter the COX-2 protein levels but inhibited its enzyme activity to reduce prostaglandin E2 production. Furthermore, the cell survival was concentration-dependently decreased following oxaliplatin (1-100 microM, 24 h) treatment. Combination of oxaliplatin and etodolac additively increased the death and growth inhibition of RKO cells. Survivin, an inhibitor protein of apoptosis, mediates anti-apoptosis and promotes cell division in cancer cells. Oxaliplatin or COX-2 inhibitors significantly decreased the levels of survivin proteins. Moreover, survivin proteins were markedly diminished following co-treatment with oxaliplatin and etodolac. Together, this is the first report that combination of COX-2 inhibitors and oxaliplatin can increase the reduction of survivin protein expression, growth inhibition, and death in human colon cancer cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Inibidores de Ciclo-Oxigenase/farmacologia , Compostos Organoplatínicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/química , Neoplasias do Colo/patologia , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Dinoprostona/biossíntese , Sinergismo Farmacológico , Humanos , Proteínas Inibidoras de Apoptose , Proteínas de Membrana , Proteínas Associadas aos Microtúbulos/análise , Proteínas de Neoplasias/análise , Oxaliplatina , Prostaglandina-Endoperóxido Sintases/análise , Survivina
17.
Carcinogenesis ; 26(1): 53-63, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15471901

RESUMO

Arsenic compounds, which are well-documented human carcinogens, are now used in cancer therapy. Knowledge of the mechanism by which arsenic exerts its toxicity may help in designing a more effective regimen for therapy. In this study, we showed that arsenite could induce prominent mitotic arrest in CGL-2 cells and demonstrated the presence of damaged DNA in arsenite-arrested mitotic cells. We then explored why these cells with arsenite-induced DNA damage were arrested at mitosis instead of G2 stage. When synchronized CGL-2 cells were treated with arsenite at stage G1, S or G2, all progressed into, and arrested at, the mitotic stage and contained damaged DNA, as demonstrated by the appearance of the DNA double-strand break marker, phosphorylated histone H2A.X (gamma-H2AX). Since X-irradiation induced G2 arrest in CGL-2 cells, these cells clearly have a functional G2 DNA damage checkpoint. However, treatment of X-irradiated CGL-2 cells with arsenite resulted in a decrease in G2 cells and an increase in mitotic cells, suggesting that arsenite may inhibit activation of the G2 DNA damage checkpoint and thus allow cells with damaged DNA to proceed from G2 into mitosis. Immunoblot analysis confirmed that arsenite treatment reduced the X-irradiation-induced phosphorylation of both ataxia-telangiectasia, mutated at serine 1981 and Cdc25C at serine 216, events which are crucial for G2 checkpoint activation and G2 arrest. Moreover, a higher frequency of apoptotic cells is observed in mitotic CGL-2 cells arrested by arsenite than those arrested by nocodazole or taxol. Our results show that the combined effects of arsenite in inducing DNA damages, inhibiting the activation of G2 checkpoint, and arresting cells with damaged DNA in the mitotic stage may subsequently enhance the induction of apoptosis in arsenite-arrested mitotic CGL-2 cells.


Assuntos
Arsenitos/toxicidade , Proteínas de Ciclo Celular/efeitos dos fármacos , Genes cdc/efeitos dos fármacos , Mitose/efeitos dos fármacos , Teratogênicos/toxicidade , Apoptose/fisiologia , Western Blotting , Proteínas de Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Imunofluorescência , Fase G2 , Genes cdc/efeitos da radiação , Humanos , Mitose/efeitos da radiação
18.
Br J Pharmacol ; 140(5): 955-63, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14517180

RESUMO

N-methyl-d-aspartate (NMDA) receptors have been demonstrated to be a pivotal target for ethanol action. The present study examined the actions of acute ethanol exposure on NMDA-induced responses and the acute tolerance to ethanol actions in rat sympathetic preganglionic neurons (SPNs) in vitro and in vivo. NMDA (50 microM) applied every 5 min induced reproducible membrane depolarizations of SPNs in neonatal spinal cord slice preparations. Ethanol (50 - 100 mM) applied by superfusion for 15 min caused a sustained decrease in NMDA-induced depolarizations in a dose-dependent and reversible manner. When the superfusion time of ethanol (100 mm) was increased to 50 min, NMDA-induced depolarizations were attenuated initially but a gradual recovery was seen in approximately 40% of SPNs tested. Repeated injections of NMDA (2 nM) intrathecally at 30 min interval caused reproducible increases in mean arterial pressure (MAP) in urethane-anesthetized rats. Intravenous injections of ethanol (0.16 or 0.32 g, 1 ml) inhibited NMDA-induced pressor effects in a blood concentration-dependent manner. The inhibition by ethanol of NMDA-induced pressor effects was reduced over time during continuous infusion of ethanol or on the second injection 3.5 h after prior injection of a higher dose of ethanol. Ethanol, at concentrations significantly inhibited NMDA-induced responses, had no significant effects on alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-induced responses. The study demonstrated the selective inhibition by ethanol of NMDA-induced responses and the development of acute tolerance to the inhibitory effects in SPNs both in vitro and in vivo. These effects may play important roles in the ethanol regulation of cardiovascular function.


Assuntos
Fibras Autônomas Pré-Ganglionares/efeitos dos fármacos , Etanol/farmacologia , N-Metilaspartato/antagonistas & inibidores , N-Metilaspartato/farmacologia , Animais , Fibras Autônomas Pré-Ganglionares/fisiologia , Relação Dose-Resposta a Droga , Tolerância a Medicamentos/fisiologia , Técnicas In Vitro , Masculino , Ratos , Ratos Sprague-Dawley
19.
J Neurosci Methods ; 118(1): 77-87, 2002 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-12191760

RESUMO

A low-noise flexible system for the simultaneous recording and analysis of several electrical signals (EEG, ECG, EMG, and diaphragm EMG) from the same rat was constructed for studying changes in physiological functions during the sleep-wake cycle. The hardware in the system includes a multichannel amplifier, a video camera, a timer code generator, and a PC. A miniature buffer headstage with high-input impedance connected to a 6-channel amplifier was developed. All electrical activities devoid of 60 Hz interference could be consistently recorded by our low-cost amplifier with no shielding treatment. The analytical software was established in the LabVIEW environment and consisted of three major frames: temporal, spectral, and nonlinear analyses. These analytical tools demonstrated several distinct utilities. For example, the sleep-wake states could be successfully distinguished by combining temporal and spectral analyses. An obvious theta rhythm during rapid-eye-movement sleep (REMS) was recorded from parietal to occipital cortical areas but not from the frontal area. In addition, two types of sleep apnea with/without cardiac arrhythmias were observed under REMS condition. Moreover, the evoked potentials of the primary somatosensory cortex elicited by innocuous electrical pulses were modulated by vigilant states, especially under a slow-wave sleep state. These results show that our system delivers high-quality signals and is suitable for sleep investigations. The system can be easily expanded by combining other recording devices, like a plethysmograph. This compact system can also be easily modified and applied to other related physiological or pharmacological studies.


Assuntos
Eletroencefalografia/métodos , Fases do Sono/fisiologia , Vigília/fisiologia , Animais , Eletroencefalografia/instrumentação , Eletromiografia/métodos , Desenho de Equipamento , Ratos , Ratos Long-Evans , Processamento de Sinais Assistido por Computador , Síndromes da Apneia do Sono/fisiopatologia , Sono REM/fisiologia , Software
20.
Life Sci ; 71(9): 1035-45, 2002 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-12088763

RESUMO

Lead exposure elicited an increase in blood pressure and was considered to be a cardiovascular risk factor. The involvements of sympathetic nervous system and circulating catecholamines have been implicated in lead-induced hypertension. This study examined the effects of PbCl(2) on sympathetic preganglionic neurons (SPNs) in vitro and in vivo. In vitro electrophysiological study showed that superfusion of a low concentration (5 microM) of PbCl(2), which had no effects on membrane potential and spontaneous discharge rate, enhanced excitatory postsynaptic potentials (EPSPs) in some of the SPNs examined but inhibited inhibitory postsynaptic potentials (IPSPs) in other SPNs tested. A higher concentration (50 microM) of PbCl(2) inhibited both EPSPs and IPSPs in all SPNs examined. In vivo study showed that intrathecal injection of PbCl(2) (10 and 100 nmol) via an implanted cannula to the T7-T9 segments of urethane-anesthetized rats increased both the heart rate and mean arterial pressure. The pressor and tachycardic responses of intrathecal PbCl(2) (100 nmol) were attenuated by pretreatment with intravenous administration of hexamethonium (10 mg/kg) or intrathecal AP-5 (DL-2-amino-5-phosphonovaleric acid, 100 nmol), but were not significantly antagonized by prior intrathecal administration of CNQX (6-cyano-7-nitroquinoxaline-2,3-dione, 100 nmol). Taken together, these results demonstrated that lead may exert a stimulatory effect on SPNs, which may result from the enhancement of EPSPs and inhibition of IPSPs by low concentrations of lead.


Assuntos
Gânglios/efeitos dos fármacos , Chumbo/farmacologia , Neurônios/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos , 2-Amino-5-fosfonovalerato/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Gânglios/citologia , Hexametônio/farmacologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sistema Nervoso Simpático/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA