Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Arch Toxicol ; 88(1): 89-96, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23877120

RESUMO

Darbepoetin (DPO), an erythropoietin (EPO) derivative, was licensed in 2002 to treat patients with solid tumors suffering from chemotherapy-dependent anemia, although various tumors express EPO to improve vascularization, thus favoring tumor growth and spreading. Therefore, we wanted to investigate direct effects of DPO on the liver tumor cell lines HepG2, SkHep1, Huh-7, AKN1, HCC-T and HCC-M, as well as on primary human hepatocytes (hHeps). DPO (0-40 ng/ml) did not affect viability of hHeps, HepG2, SkHep1, AKN1, HCC-T and HCC-M cells, as determined by Resazurin conversion. However, Huh-7 cells' viability dose-dependently decreased from 5 ng/ml DPO on. Lack of LDH release into culture medium and negative DNA laddering excluded apoptosis or necrosis as the cause for the reduced Resazurin conversion. In Huh-7 cells, DPO increased the expression of p53. Interestingly, Huh-7 cells showed the highest basal TGF-ß1 expression as compared to the other cell lines. Upon inhibition of TGF-ß1 signaling, DPO no longer reduced viability in Huh-7 cells. On the contrary, co-incubation with TGF-ß1 made the other cell lines responsive to DPO. Summarizing our data, we show that DPO reduces the growth of Huh-7 cells by up-regulation of the tumor-suppressor gene p53. This mechanism seems to be dependent on a strong TGF-ß expression and corresponding signaling in these cells, as other cell lines became responsive to DPO with TGF-ß1 supplementation. The knowledge of this mechanism offers great perspectives for the understanding and treatment of solid liver tumors.


Assuntos
Eritropoetina/análogos & derivados , Neoplasias Hepáticas/tratamento farmacológico , Fator de Crescimento Transformador beta/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Darbepoetina alfa , Relação Dose-Resposta a Droga , Eritropoetina/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos
2.
Oncol Rep ; 29(2): 646-52, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23229825

RESUMO

The response of standard-risk hepatoblastoma (HB) to neoadjuvant cisplatin (CDDP) chemotherapy is excellent; however, in high-risk HB, drug resistance remains a major challenge. Alternative therapeutic strategies may consider combining cytotoxic drugs with apoptosis sensitizers as this has shown additive effects in various types of malignancies. Analysis of published expression databases have revealed an anti-apoptosis state in HB samples. Herein, we evaluated the synergistic effects of ABT-737 as a modulator of apoptosis in combination with CDDP in HB. To this end, clonogenic assays were performed with HepT1 and HUH6 HB cells to evaluate the synergistic effects of CDDP and ABT-737. Combination treatment with CDDP and ABT-737 reduced the clonogenicity of HB cells more than 5-fold compared to treatment with CDDP alone. Furthermore, the HUH6 mixed-type HB cells showed higher sensitivity to CDDP and combination treatment compared to the HepT1 embryonal-type cells. Subcutaneous HUH6 tumors in NOD/LtSz-scid IL2Rγnull mice were treated with CDDP (1.25 and 3 mg/kg body weight, n=6), ABT-737 (100 mg/kg, n=5) and the combination of both agents (n=5). Combined treatment led to a significantly reduced tumor growth compared to CDDP treatment alone (p<0.02). When using higher doses of CDDP (3 mg/kg) alone or in combination with ABT-737, dose-dependent toxicity was observed in this mouse strain. In conclusion, our results demonstrated the enhancement of chemotherapy efficacy by using modulators of apoptosis together with cytotoxic agents. Additive effects of ABT-737 may allow reduction in CDDP dosages with maintenance of antitumor activity. Sensitizing HB to apoptosis may also render resistant HB susceptible to established chemotherapy regimens.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Compostos de Bifenilo/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Hepatoblastoma/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Nitrofenóis/farmacologia , Sulfonamidas/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Compostos de Bifenilo/administração & dosagem , Compostos de Bifenilo/efeitos adversos , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Perfilação da Expressão Gênica , Hepatoblastoma/genética , Hepatoblastoma/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Nitrofenóis/administração & dosagem , Nitrofenóis/efeitos adversos , Piperazinas/administração & dosagem , Piperazinas/efeitos adversos , Piperazinas/farmacologia , Sulfonamidas/administração & dosagem , Sulfonamidas/efeitos adversos , Redução de Peso/efeitos dos fármacos
3.
Pediatr Surg Int ; 29(2): 121-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23187893

RESUMO

PURPOSE: Duplex drugs are promising anticancer agents. After in vivo cleavage into active nucleoside analogues, they exert their anti-tumour activity with reduced toxicity and side effects. Here we evaluated the impact of two duplex drugs on the viability of hepatoblastoma (HB) cells lines and their toxicity against human fibroblasts. METHODS: The duplex drugs 2'-deoxy-5-fluorouridylyl-(3'-5')- 3'-C-ethynylcytidine (5-FdU(3'-5')ECyd) and 3'-C-ethynylcytidinylyl-(5'→1-O)-2-O-octadecyl-sn-glycerylyl-(3'-Ο→5')-2'-deoxy-5-fluorouridine (ECyd-lipid-5-FdU) were analysed in two HB cell lines (HUH6, HepT1) and fibroblasts by MTT assay. The treatment potential was compared to the single substances 2'-deoxy-5-fluorourindine (5-FdU), 3'-C-ethynylycytidine (ECyd) and an equimolar mixture of both. Cell cycle analyses were performed using flow cytometry after 7-AAD staining. RESULTS: Both duplex drugs achieve a potent cytotoxic effect at low µM concentrations, which was more pronounced than the mixture of ECyd + 5-FdU. Further, both substances exert toxicity on fibroblasts of tumour samples, with less toxicity in foreskin fibroblasts cultures. Cell cycle analyses revealed a shift towards apoptotic cells for both drugs in HB cells. CONCLUSION: 5-FdU(3'-5')ECyd and ECyd-lipid-5-FdU exert a highly potent anti-tumoural effect on HB cells and might therefore be a treatment option in HB. Pharmacological formulations of both duplex drugs have to be evaluated in vivo to reduce possible side effects.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Monofosfato de Citidina/análogos & derivados , Fluordesoxiuridilato/análogos & derivados , Hepatoblastoma/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Oligonucleotídeos/administração & dosagem , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Monofosfato de Citidina/administração & dosagem , Citometria de Fluxo/métodos , Fluordesoxiuridilato/administração & dosagem , Humanos , Células Tumorais Cultivadas
4.
Neonatology ; 101(4): 285-92, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22286302

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC) and focal intestinal perforation (FIP) are major causes of morbidity in infants with extremely low birth weight (ELBW). OBJECTIVE: To evaluate the surgical procedures applied, and the survival and long-term outcome of ELBW infants with NEC and FIP in a single-center study. METHODS: Inborn and outborn ELBW infants (<1000 g) with NEC and FIP were analyzed retrospectively from 2002 to 2007. Data collected include surgical procedures, survival as well as complications, length of partial parenteral nutrition and hospital stay. The short-term and long-term outcomes after 2-7 years were assessed and compared with a matched control group. RESULTS: Out of 280 ELBW infants, 28 underwent surgery, 19 because of FIP and 9 for NEC. Fourteen infants in the FIP group were treated with primary laparotomy and 5 with peritoneal drainage (PD). In the NEC group, only 1 infant was treated with PD. PD was used for unstable patients and was always followed by secondary laparotomy after stabilization. Five of 28 (18%) surgically treated ELBW infants and 4 (14%) matched controls died. The following complications occurred in the surgical group: complete (n = 1) or minor wound dehiscence (n = 4), stoma prolapse (n = 5), parastomal hernia (n = 2), stoma fistula (n = 1), and wound infection (n = 2). Dependency on parenteral nutrition was significantly shorter in infants with FIP, while there were no differences in time to stoma closure and length of hospital stay between those with FIP and those with NEC. Eleven of 23 (47.8%) surviving patients with FIP or NEC showed developmental delay, compared with 9 of 24 (37.5%) in the controls. CONCLUSIONS: The management of EBLW infants with NEC and FIP remains challenging. Our treatment approach was associated with low mortality. Developmental delay seems to be caused by extreme prematurity rather than NEC- or FIP-related bowel perforation.


Assuntos
Recém-Nascido de Peso Extremamente Baixo ao Nascer , Doenças do Prematuro/cirurgia , Perfuração Intestinal/congênito , Perfuração Intestinal/cirurgia , Algoritmos , Enterocolite Necrosante/complicações , Enterocolite Necrosante/diagnóstico , Enterocolite Necrosante/mortalidade , Enterocolite Necrosante/cirurgia , Feminino , Humanos , Lactente , Recém-Nascido , Doenças do Prematuro/diagnóstico , Doenças do Prematuro/mortalidade , Perfuração Intestinal/complicações , Perfuração Intestinal/mortalidade , Masculino , Projetos de Pesquisa , Estudos Retrospectivos
5.
Liver Int ; 32(4): 574-81, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22176637

RESUMO

BACKGROUND: Multidrug resistance is a major reason for poor treatment results in advanced hepatoblastoma (HB). Several alternative treatment options are currently under investigation to improve the prognosis of affected patients AIMS: This study aimed to analyse the impact of sorafenib on the viability of HB cells and xenotransplanted HB tumours. METHODS: Cell viability and apoptosis were evaluated in two HB cell lines (HUH6 and HepT1) after treatment with sorafenib using MTT and Caspase 3 activation assay. Extracellular signal-regulated kinase (ERK) phosphorylation was investigated using Western blot. In addition, sorafenib (30 mg/kg) was administered orally to NMRI mice bearing subcutaneous HUH6 derived tumours. Tumour progression and viability were monitored by tumour volume and α-fetoprotein (AFP) levels, and apoptosis was assessed using TUNEL assay. Tumour angiogenesis and mean vascular density (MVD) was determined using CD31 staining, ERK phosphorylation was detected using indirect immunofluorescence. RESULTS: Treatment with sorafenib led to decreased ERK phosphorylation, reduced cell viability and induction of apoptosis in HepT1 and HUH6 cells. In HB xenografts, sorafenib significantly reduced tumour growth compared with control (P < 0.05). AFP levels were lower in the sorafenib group (P = 0.07). Relative apoptotic areas detected using TUNEL assay were increased (P = 0.003). CD31 staining revealed inhibition of angiogenesis, and mean vascular density was lower in the sorafenib group (P = 0.02). ERK phosphorylation was reduced in tumours tissues after sorafenib treatment. CONCLUSION: Treatment with sorafenib led to a potent inhibition of cell viability, tumour progression and angiogenesis. Sorafenib might therefore also be a promising treatment option for high risk or recurrent HB.


Assuntos
Antineoplásicos/farmacologia , Benzenossulfonatos/farmacologia , Hepatoblastoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Animais , Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Benzenossulfonatos/uso terapêutico , Western Blotting , Caspase 3 , Linhagem Celular Tumoral , Sobrevivência Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Fatores de Transcrição Forkhead/genética , Marcação In Situ das Extremidades Cortadas , Camundongos , Neovascularização Patológica/patologia , Niacinamida/análogos & derivados , Compostos de Fenilureia , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Sorafenibe , Sais de Tetrazólio , Tiazóis , Transplante Heterólogo , Transplantes , alfa-Fetoproteínas/metabolismo
6.
BMC Cancer ; 11: 362, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21854558

RESUMO

BACKGROUND: The primary goal of current chemotherapy in hepatoblastoma (HB) is reduction of tumour volume and vitality to enable complete surgical resection and reduce risk of recurrence or metastatic disease. Drug resistance remains a major challenge for HB treatment. In some malignancies inhibition of anti-apoptotic pathways using small BH3 mimetic molecules like ABT-737 shows synergistic effects in combination with cystotoxic agents in vitro. Now we analysed toxicology and synergistic effects of this approach in HB cells and HB xenografts. METHODS: Viability was monitored in HB cells (HUH6 and HepT1) and fibroblasts treated with paclitaxel, ABT-737 and a combination of both in a MTT assay. HUH6 xenotransplants in NOD/LtSz-scid IL2Rγnull mice (NSG) were treated accordingly. Tumour volume and body weight were monitored. Xenografted tumours were analysed by histology and immunohistochemistry (Ki-67 and TUNEL assay). RESULTS: ABT-737 reduced viability in HUH6 and HepT1 cells cultures at concentrations above 1 µM and also enhanced the cytotoxic effect of paclitaxel when used in combination. Thereby paclitaxel could be reduced tenfold to achieve similar reduction of viability of tumour cells. In contrast no toxicity in fibroblasts was observed at the same regiments. Subcutaneous HB (HUH6) treated with paclitaxel (12 mg/kg body weight, n = 7) led to delayed tumour growth in the beginning of the experiment. However, tumour volume was similar to controls (n = 5) at day 25. Combination treatment with paclitaxel and ABT-737 (100 mg/kg, n = 8) revealed significantly 10 fold lower relative tumour volumes compared to control and paclitaxel groups. Paclitaxel dependent toxicity was observed in this mice strain. CONCLUSIONS: Our results demonstrate enhancement of chemotherapy by using modulators of apoptosis. Further analyses should include improved pharmacological formulations of paclitaxel and BH3 mimetics in order to reduce toxicological effects. Sensitising HB to apoptosis may also render resistant HB susceptible to established chemotherapy regimens.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Compostos de Bifenilo/farmacologia , Hepatoblastoma/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Nitrofenóis/farmacologia , Paclitaxel/farmacologia , Sulfonamidas/farmacologia , Análise de Variância , Animais , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/farmacologia , Compostos de Bifenilo/administração & dosagem , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Nitrofenóis/administração & dosagem , Paclitaxel/administração & dosagem , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Sulfonamidas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Int J Oncol ; 38(2): 447-54, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21132272

RESUMO

Multidrug resistance (MDR) is a major reason for poor treatment results in hepatoblastoma (HB). The objective of this study was to establish a drug resistance model for HB to analyse alternative treatment options in vitro. Both HB cell lines HUH6 and HepT1 were xenotransplanted in NMRI mice (nu/nu) and 2 cycles of cisplatin (CDDP) treatment were administered. Thereafter, xenotransplants were excised and viable tumour cells were re-cultured. 3D cultures of HUH6 and HepT1 cells were generated on a low binding culture surface. Cell viability in response to CDDP/DOXO (doxorubicin) and apoptosis was assessed by MTT-assay and caspase 3 activity, respectively. Efflux of doxorubicin was measured by flow cytometry. Cellular levels of ABC-transporters (MDR1, MRP1, cMOAT and BRCP) were determined by real time rt-PCR. Only HepT1 cells isolated from HB xenografts showed resistance to CDDP, but did not survive repeated passages. Culturing HUH6 and HepT1 cells as spheroids was successful and 3D cultures showed an IC50-drift to higher drug concentrations for CDDP and DOXO compared to 2D cultures. Treatment with CDDP and DOXO led to homogeneous apoptosis in spheroids. Increased doxorubicin efflux in HUH6 spheroids was not influenced by the P-glycoprotein inhibitor tariquidar. Expression levels of MDR1, MRP1, cMOAT and BRCP in 3D cultures were similar to those in 2D cultures and were higher in HepT1 than in HUH6 cells. In conclusion, a 3D cell culture model for multidrug resistance was established for hepatoblastoma. The underlying mechanism involves altered accessibility of the cells for drugs rather than up-regulation of ABC-transporters.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Modelos Animais de Doenças , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Hepatoblastoma/tratamento farmacológico , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/administração & dosagem , Doxorrubicina/administração & dosagem , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hepatoblastoma/metabolismo , Hepatoblastoma/patologia , Humanos , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Nus , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
8.
Pediatr Blood Cancer ; 55(6): 1089-95, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20680965

RESUMO

BACKGROUND: An increased expression of anti-apoptotic proteins is regularly found in malignant cells, contributing to their clonal expansion by conferring an improved survival ability. In Hepatoblastoma (HB) apoptosis regulation contributes to resistance and therapy failure, therefore we modulated apoptosis sensitivity of HB cells for an improved cytotoxic activity of commonly used drugs. PROCEDURE: Apoptosis-related proteins were quantified in HB cells (HuH6 and HepT1) using protein assays. Interaction of ABT-737, a small-molecule inhibitor of Bcl-2, Bcl-xL, and Bcl-W with cytotoxic drugs was monitored in a proliferation assay. Apoptosis induction was measured by caspase-3 activity. RESULTS: We found high levels of the anti-apoptotic protein Bcl-2 and Bcl-X as well as low levels of pro-apoptotic protein Bax and Bad in both HB cell lines. ABT-737 induced apoptosis in HuH6 and HepT1 cells at concentrations higher than 1 µM. ABT-737 also enhanced the cytotoxic effect of cisplatin (CDDP), doxorubicin (DOXO), etoposide and paclitaxel when used as combination therapy. HuH6 expressed slightly higher pro-apoptotic and lower anti-apoptotic protein levels than HepT1, which may explain the stronger enhancement of cytostatic drug effects in HuH6 cells when treated in combination with ABT-737. CONCLUSION: The observed anti-apoptotic phenotype in HB cell lines may contribute to resistance to cytotoxic drugs used in the standard treatment protocol of HB. These pre-clinical results suggest that apoptosis sensitizers with BH-3 mimicry, such as ABT-737, should be further evaluated in preclinical models of HB.


Assuntos
Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Resistencia a Medicamentos Antineoplásicos , Hepatoblastoma/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Nitrofenóis/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Sulfonamidas/farmacologia , Proteína bcl-X/antagonistas & inibidores , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Hepatoblastoma/metabolismo , Hepatoblastoma/patologia , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo
9.
J Cell Sci ; 118(Pt 2): 421-31, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15654019

RESUMO

Calcium is a key regulator of cardiac function and is modulated through the Ca2+-sensor protein S100A1. S100 proteins are considered to exert both intracellular and extracellular functions on their target cells. Here we report the impact of an increased intracellular S100A1 protein level on Ca2+-homeostasis in neonatal ventricular cardiomyocytes in vitro. Specifically, we compare the effects of exogenously added recombinant S100A1 to those resulting from the overexpression of a transduced S100A1 gene. Extracellularly added S100A1 enhanced the Ca2+-transient amplitude in neonatal ventricular cardiomyocytes (NVCMs) through a marked decrease in intracellular diastolic Ca2+-concentrations ([Ca2+]i). The decrease in [Ca2+]i was independent of sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) activity and was probably the result of an increased sarcolemmal Ca2+-extrusion through the sodium-calcium exchanger (NCX). At the same time the Ca2+-content of the sarcoplasmic reticulum (SR) decreased. These effects were dependent on the uptake of extracellularly added S100A1 protein and its subsequent routing to the endosomal compartment. Phospholipase C and protein kinase C, which are tightly associated with this subcellular compartment, were found to be activated by endocytosed S100A1. By contrast, adenoviral-mediated intracellular S100A1 overexpression enhanced the Ca2+-transient amplitude in NVCMs mainly through an increase in systolic [Ca2+]i. The increased Ca2+-load in the SR was based on an enhanced SERCA2a activity while NCX function was unaltered. Overexpressed S100A1 colocalized with SERCA2a and other Ca2+-regulatory proteins at the SR, whereas recombinant S100A1 protein that had been endocytosed did not colocalize with SR proteins. This study provides the first evidence that intracellular S100A1, depending on its subcellular location, modulates cardiac Ca2+-turnover via different Ca2+-regulatory proteins.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Cálcio/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/farmacologia , Regulação da Expressão Gênica , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas S100 , Retículo Sarcoplasmático/metabolismo
10.
Trends Biotechnol ; 22(10): 487-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15450736

RESUMO

Heart failure affects 23 million people worldwide and results from cardiac dysfunction characterized by decreased responsiveness to beta-adrenergic stimulation. A recent publication by W.J. Koch and colleagues highlights evidence for targeted beta-adrenergic receptor kinase (betaARK1) inhibition by gene transfer to improve contractile function and beta-adrenergic responsiveness in failing human myocardium. This proof-of-concept study has great importance for future heart failure therapy because it provides evidence for the therapeutic effectiveness of betaARK1 inhibition in failing human myocardium.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Inibidores Enzimáticos , Terapia Genética , Insuficiência Cardíaca/terapia , Miócitos Cardíacos/enzimologia , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Camundongos , Miocárdio/citologia , Miocárdio/enzimologia , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Transgenes , Quinases de Receptores Adrenérgicos beta
11.
J Biol Chem ; 278(48): 48404-12, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-12960148

RESUMO

S100A1 is a Ca2+-binding protein of the EF-hand type that belongs to the S100 protein family. It is specifically expressed in the myocardium at high levels and is considered to be an important regulator of cardiac contractility. Because the S100A1 protein is released into the extracellular space during ischemic myocardial injury, we examined the cardioprotective potential of the extracellular S100A1 protein on ventricular cardiomyocytes in vitro. In this report we show that extracellularly added S100A1 protein is endocytosed into the endosomal compartment of neonatal ventricular cardiomyocytes via a Ca2+-dependent clathrin-mediated process. S100A1 uptake protects neonatal ventricular cardiomyocytes from 2-deoxyglucose and oxidative stress-induced apoptosis in vitro. S100A1-mediated anti-apoptotic effects involve specific activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) pro-survival pathway, including activation of phospholipase C, protein kinase C, mitogen-activated protein kinase kinase 1, and ERK1/2. In contrast, neither transsarcolemmal Ca2+ influx via the L-type channel nor protein kinase A activity seems to take part in the S100A1-mediated signaling pathway. In conclusion, this study provides evidence for the S100A1 protein serving as a novel cardioprotective factor in vitro. These findings warrant speculation that injury-dependent release of the S100A1 protein from cardiomyocytes may serve as an intrinsic mechanism to promote survival of the myocardium in vivo.


Assuntos
Apoptose , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Animais Recém-Nascidos , Antimetabólitos/farmacologia , Western Blotting , Cálcio/metabolismo , Células Cultivadas , Clatrina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Desoxiglucose/farmacologia , Relação Dose-Resposta a Droga , Endocitose , Ativação Enzimática , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Sistema de Sinalização das MAP Quinases , Microscopia de Contraste de Fase , Proteína Quinase 3 Ativada por Mitógeno , Modelos Biológicos , Miocárdio/metabolismo , Estresse Oxidativo , Fosforilação , Ligação Proteica , Proteína Quinase C/metabolismo , Proteínas S100 , Transdução de Sinais , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Azul Tripano/farmacologia , Fosfolipases Tipo C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...