Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
ACS Omega ; 6(4): 2626-2637, 2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33553880

RESUMO

Epidemiological evidence has accentuated the repurposing of metformin hydrochloride for cancer treatment. However, the extreme hydrophilicity and poor permeability of metformin hydrochloride are responsible for its poor anticancer activity in vitro and in vivo. Here, we report the synthesis and characterization of several lipophilic metformin salts containing bulky anionic permeation enhancers such as caprate, laurate, oleate, cholate, and docusate as counterions. Of various counterions tested, only docusate was able to significantly improve the lipophilicity and lipid solubility of metformin. To evaluate the impact of the association of anionic permeation enhancers with metformin, we checked the in vitro anticancer activity of various lipophilic salts of metformin using drug-sensitive (MYCN-2) and drug-resistant (SK-N-Be2c) neuroblastoma cells as model cancer cells. Metformin hydrochloride showed a very low potency (IC50 ≈ >100 mM) against MYCN-2 and SK-N-Be2c cells. Anionic permeation enhancers showed a considerably higher activity (IC50 ≈ 125 µM to 1.6 mM) against MYCN-2 and SK-N-Be2c cells than metformin. The association of metformin with most of the bulky anionic agents negatively impacted the anticancer activity against MYCN-2 and SK-N-Be2c cells. However, metformin docusate showed 700- to 4300-fold improvement in anticancer potency compared to metformin hydrochloride and four- to five-fold higher in vitro anticancer activity compared to sodium docusate, indicating a synergistic association between metformin and docusate. A similar trend was observed when we tested the in vitro activity of metformin docusate, sodium docusate, and metformin hydrochloride against hepatocellular carcinoma (HepG2) and triple-negative breast cancer (MDA-MB-231) cells.

3.
Cell Calcium ; 82: 102052, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31306997

RESUMO

Neuroblastoma (NB) is the most common extra-cranial pediatric solid tumor in children. Despite NB's relative rarity, high-risk NB patients have a poor prognosis with survival rate less than 50%. This is even worse for patients with relapsed or refractory NB. Finding effective alternative treatment strategies is therefore a must. Calcium is an intracellular messenger that is unequivocally present in normal physiology mediating proliferation, growth, migration, cell division, angiogenesis and cell death, as well as pathophysiological processes such as those included in Weinberg's hallmarks of cancer. Within the past 20 years, the molecular identity of most calcium channels has been revealed, however for some of these channels the precise gating mechanism and their role in normal physiology is still elusive. Here we review the recent findings of components of calcium signaling that are deregulating in the malignant progression of NB. We further integrate critical calcium signaling pathways using patient-derived expression analysis. Revealing the roles of these calcium pathways in tumor development, progression, microenvironment and importantly - protection against antineoplastic drugs may hopefully lead to novel treatment strategies in the future.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Neuroblastoma/metabolismo , Animais , Transformação Celular Neoplásica , Criança , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Humanos , Neuroblastoma/patologia , Microambiente Tumoral
4.
Oncotarget ; 8(66): 109985-109999, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29299124

RESUMO

Neuroblastoma (NB) is the most common extra-cranial pediatric solid tumor. High-risk NB is difficult to treat due to the lack of response to current therapies and aggressive disease progression. Despite novel drugs, alternative treatments and multi-modal treatments, finding an effective treatment strategy for these patients continues to be a major challenge. The current study focuses on examining the effects of FTY-720 or fingolimod, a drug that is FDA-approved for refractory multiple sclerosis, in NB. The results showed that FTY-720 regulates multiple pathways that result in various effects on calcium signaling, ion channel activation and cell survival/death pathways. FTY-720 rapidly inhibits TRPM7 channel activity, and inhibited TRPM7 kinase activity, modulates calcium signaling, induces a loss of mitochondrial membrane potential and opening of the mitochondrial permeability transition pore, and ultimately leads to cell death. Interestingly, the data also showed that low concentrations of FTY-720 sensitized drug-resistant NB cells to antineoplastic drugs. These results suggest that FTY-720 may be an attractive alternative for the treatment of NB.

5.
FEBS Open Bio ; 4: 966-75, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25426416

RESUMO

Neuroblastoma is an extra-cranial solid cancer in children. MYCN gene amplification is a prognostic indicator of poor outcome in neuroblastoma. Recent studies have shown that the multiple steps involved in cell migration are dependent on the availability of intracellular calcium (Ca(2+)). Although significant advances have been made in understanding the role of Ca(2+) during migration, little has been achieved towards understanding its impact on the progression of diseases such as cancer. Interestingly, previous studies showed that cancer cell migration is regulated by TRPM7, a calcium-permeable ion channel. The objective of the current study was to elucidate the mechanism by which MycN promotes NB cell migration and the mechanism regulating TRPM7 expression. The results showed that MycN increased TRPM7 expression, induced TRPM7 channel activity, increased intracellular Ca(2+) signaling, and promoted cell migration in NB cells. The results also showed that inhibition or down-regulation of ornithine decarboxylase (ODC) inhibited TRPM7 expression, a process that was reversed by spermidine. Overall, this study provides evidence that MycN promotes TRPM7 expression and cell migration through a mechanism that involves ODC synthesis of polyamines.

6.
Int J Oncol ; 42(4): 1219-28, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23440295

RESUMO

Neuroblastoma (NB) is the most common extracranial pediatric tumor. NB patients over 18 months of age at the time of diagnosis are often in the later stages of the disease, present with widespread dissemination, and often possess MYCN tumor gene amplification. MYCN is a transcription factor that regulates the expression of a number of genes including ornithine decarboxylase (ODC), a rate-limiting enzyme in the biosynthesis of polyamines. Inhibiting ODC in NB cells produces many deleterious effects including G(1) cell cycle arrest, inhibition of cell proliferation, and decreased tumor growth, making ODC a promising target for drug interference. DFMO treatment leads to the accumulation of the cyclin-dependent kinase inhibitor p27(Kip1) protein and causes p27(Kip1)/Rb-coupled G(1) cell cycle arrest in MYCN-amplified NB tumor cells through a process that involves p27(Kip1) phosphorylation at residues Ser10 and Thr198. While p27(Kip1) is well known for its role as a cyclin-dependent kinase inhibitor, recent studies have revealed a novel function of p27(Kip1) as a regulator of cell migration and invasion. In the present study we found that p27(Kip1) regulates the migration and invasion in NB and that these events are dependent on the state of phosphorylation of p27(Kip1). DFMO treatments induced MYCN protein downregulation and phosphorylation of Akt/PKB (Ser473) and GSK3-ß (Ser9), and polyamine supplementation alleviated the DFMO-induced effects. Importantly, we provide strong evidence that p27(Kip1) mRNA correlates with clinical features and the survival probability of NB patients.


Assuntos
Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Eflornitina/farmacologia , Neuroblastoma/metabolismo , Linhagem Celular Tumoral , Pré-Escolar , Inibidor de Quinase Dependente de Ciclina p27/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Lactente , Estimativa de Kaplan-Meier , Proteína Proto-Oncogênica N-Myc , Invasividade Neoplásica , Neuroblastoma/mortalidade , Proteínas Nucleares , Proteínas Oncogênicas , Ornitina Descarboxilase/genética , Ornitina Descarboxilase/metabolismo , Transporte Proteico/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transdução de Sinais
7.
Cancer Sci ; 101(7): 1624-31, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20412121

RESUMO

Our previous studies revealed that the expression of the 19-kDa protein prenylated Rab acceptor 1 domain family, member 2 (PRAF2) is elevated in cancer tissues of the breast, colon, lung, and ovary, when compared to noncancerous tissues of paired samples. PRAF2 mRNA expression also correlated with several genetic and clinical features and is a candidate prognostic marker in the pediatric cancer neuroblastoma. The PRAF2-related proteins, PRAF1 and PRAF3, play multiple roles in cellular processes, including endo/exocytic vesicle trafficking and glutamate uptake. PRAF2 shares a high sequence homology with these family members, but its function remains unknown. In this study, we examined PRAF2 mRNA and protein expression in 20 different human cancer types using Affymetrix microarray and human tissue microarray (TMA) analyses, respectively. In addition, we investigated the subcellular distribution of PRAF2 by immunofluorescence microscopy and cell fractionation studies. PRAF2 mRNA and protein expression was elevated in several cancer tissues with highest levels in malignant glioma. At the molecular level, we detected native PRAF2 in small, vesicle-like structures throughout the cytoplasm as well as in and around cell nuclei of U-87 malignant glioma cells. We further found that monomeric and dimeric forms of PRAF2 are associated with different cell compartments, suggesting possible functional differences. Importantly, PRAF2 down-regulation by RNA interference significantly reduced the cell viability, migration, and invasiveness of U-87 cells. This study shows that PRAF2 expression is elevated in various tumors with exceptionally high expression in malignant gliomas, and PRAF2 therefore presents a candidate molecular target for therapeutic intervention.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Glioma/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Sequência de Aminoácidos , Astrocitoma/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteínas de Transporte/química , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular , Glioblastoma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Proteínas de Membrana/química , Oligodendroglioma/genética , Análise de Sequência com Séries de Oligonucleotídeos , Análise Serial de Proteínas , Prenilação de Proteína , RNA Mensageiro/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
8.
Biochem Pharmacol ; 80(2): 170-8, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20362557

RESUMO

Syrbactins belong to a new class of proteasome inhibitors which include syringolins and glidobactins. These small molecules are structurally distinct from other, well-established proteasome inhibitors, and bind the eukaryotic 20S proteasome by a novel mechanism. In this study, we examined the effects of syringolin A (SylA) and glidobactin A (GlbA) as well as two synthetic SylA-analogs (SylA-PEG and SylA-LIP) in human neuroblastoma (SK-N-SH), human multiple myeloma (MM1.S, MM1.RL, and U266), and human ovarian cancer (SKOV-3) cells. While all four syrbactins inhibited cell proliferation in a dose-dependent manner, GlbA was most potent in both dexamethasone-sensitive MM1.S cells (IC(50): 0.004microM) and dexamethasone-resistant MM1.RL cells (IC(50): 0.005microM). Syrbactins also inhibited the chymotrypsin-like proteasome activity in a dose-dependent fashion, and GlbA was most effective in SK-N-SH cells (IC(50): 0.015microM). The GlbA-promoted inhibition of proteasomal activity in SK-N-SH cells resulted in the accumulation of ubiquitinated proteins and tumor suppressor protein p53 and led to apoptotic cell death in a time-dependent manner. GlbA treatment also promoted the activation of Akt/PKB via phosphorylation at residue Ser(473) and induced autophagy as judged by the presence of the lipidated form of microtubule-associated protein 1 light chain 3 (LC3) and autophagosomes. Collectively, our data suggest that syrbactins belong to a new and effective proteasome inhibitor class which promotes cell death. Proteasome inhibition is a promising strategy for targeted anticancer therapy and syrbactins are a new class of inhibitors which provide a structural platform for the development of novel, proteasome inhibitor-based drug therapeutics.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Inibidores de Proteases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Pré-Escolar , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Lactente , Masculino , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Peptídeos Cíclicos/farmacologia
9.
Int J Cancer ; 126(9): 2012-24, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19960435

RESUMO

High polyamine (PA) levels and ornithine decarboxylase (ODC) overexpression are well-known phenomena in many aggressive cancer types. We analyzed the expression of ODC and ODC-activity regulating genes antizymes 1-3 (OAZ1-3) and antizyme inhibitors 1-2 (AZ-IN1-2) in human neuroblastoma (NB) tumors and correlated these with genetic and clinical features of NB. Since ODC is a known target gene of MYCN, the correlation between ODC and MYCN was of special interest. Data were obtained from Affymetrix micro-array analysis of 88 NB tumor samples. In addition, mRNA expression levels of ODC, OAZ2 and MYCN in a MYCN-inducible NB cell line were determined by quantitative real-time reverse-transcriptase polymerase chain reaction (RT-PCR). ODC mRNA expression in NB tumors was significantly predictive of decreased overall survival probability and correlated with several unfavorable clinical NB characteristics (all p < 0.005). Interestingly, high ODC mRNA expression also showed significant correlation with poor survival prognosis in Kaplan-Meier analyses stratified for patients without MYCN amplification, suggesting an additional role for ODC independent of MYCN. Conversely, high OAZ2 mRNA expression correlated with increased survival and with several favorable clinical NB characteristics (all p < 0.003). In addition, we provide first evidence of a role for MYCN-associated transcription factors MAD2 and MAD7 in ODC regulation. In NB cell cultures, ectopic overexpression of MYCN altered ODC but not OAZ2 mRNA levels. In conclusion, these data suggest that elevated ODC and low OAZ2 mRNA expression levels correlate with several unfavorable genetic and clinical features in NB, offering new insights into PA pathways and PA metabolism-targeting therapy in NB.


Assuntos
Amplificação de Genes , Genes myc , Neuroblastoma/genética , Ornitina Descarboxilase/genética , Proteínas/genética , Linhagem Celular Tumoral , Humanos , Neuroblastoma/enzimologia , Neuroblastoma/patologia , RNA Mensageiro/análise
10.
Mol Cancer Ther ; 8(7): 2067-75, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19584241

RESUMO

S-adenosylmethionine decarboxylase (AdoMetDC) is an essential enzyme of polyamine (PA) biosynthesis, and both AdoMetDC and PA levels are often up-regulated in cancer cells. The second-generation inhibitor SAM486A inhibits AdoMetDC enzyme activity and has been evaluated in phase II clinical cancer trials. However, little is known about the mechanism of action and potential use of this therapeutic drug in the treatment of the pediatric cancer neuroblastoma (NB). Here, we show that p53 wild-type NB cells are highly sensitive to SAM486A treatment. Most notably, SAM486A treatment resulted in the rapid accumulation of proapoptotic proteins p53 and Mdm2. Concomitant with the increase of proteins at endogenous levels, the in vivo phosphorylation of p53 at residues Ser(46)/Ser(392) and Mdm2 at residue Ser(166) was observed. Moreover, the antiapoptotic protein Akt/protein kinase B was down-regulated and also dephosphorylated at residue Ser(473) in a dose- and time-dependent manner and NB cells entered apoptotic cell death. The results presented in this study highlight the importance of PA homeostasis and provide a direct link between PA metabolism and apoptotic cell signaling pathways in p53 wild-type NB cells. PA inhibitors such as SAM486A may be effective alternative agents for the treatment of NBs with or without MYCN amplification.


Assuntos
Adenosilmetionina Descarboxilase/antagonistas & inibidores , Amidinas/farmacologia , Apoptose/efeitos dos fármacos , Indanos/farmacologia , Neuroblastoma/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/farmacologia , Western Blotting , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Fosforilação/efeitos dos fármacos , S-Adenosilmetionina/farmacologia
11.
Cancer Res ; 68(23): 9825-31, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19047162

RESUMO

Ornithine decarboxylase (ODC) is a key enzyme in mammalian polyamine biosynthesis that is up-regulated in various types of cancer. We previously showed that treating human neuroblastoma (NB) cells with the ODC inhibitor alpha-difluoromethylornithine (DFMO) depleted polyamine pools and induced G1 cell cycle arrest without causing apoptosis. However, the precise mechanism by which DFMO provokes these changes in NB cells remained unknown. Therefore, we further examined the effects of DFMO, alone and in combination with phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 or Akt/protein kinase B (PKB) inhibitor IV, on the regulation of cell survival and cell cycle-associated pathways in LAN-1 NB cells. In the present study, we found that the inhibition of ODC by DFMO promotes cell survival by inducing the phosphorylation of Akt/PKB at residue Ser473 and glycogen synthase kinase-3beta at Ser9. Intriguingly, DFMO also induced the phosphorylation of p27Kip1 at residues Ser10 (nuclear export) and Thr198 (protein stabilization) but not Thr187 (proteasomal degradation). The combined results from this study provide evidence for a direct cross-talk between ODC-dependent metabolic processes and well-established cell signaling pathways that are activated during NB tumorigenesis. The data suggest that inhibition of ODC by DFMO induces two opposing pathways in NB: one promoting cell survival by activating Akt/PKB via the PI3K/Akt pathway and one inducing p27Kip1/retinoblastoma-coupled G1 cell cycle arrest via a mechanism that regulates the phosphorylation and stabilization of p27Kip1. This study presents new information that may explain the moderate efficacy of DFMO monotherapy in clinical trials and reveals potential new targets for DFMO-based combination therapies for NB treatment.


Assuntos
Eflornitina/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neuroblastoma/tratamento farmacológico , Neuroblastoma/enzimologia , Proteína Oncogênica v-akt/metabolismo , Inibidores da Ornitina Descarboxilase , Poliaminas Biogênicas/antagonistas & inibidores , Poliaminas Biogênicas/metabolismo , Linhagem Celular Tumoral , Cromonas/farmacologia , Inibidor de Quinase Dependente de Ciclina p27 , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Morfolinas/farmacologia , Proteína Oncogênica v-akt/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
12.
Neurosci Lett ; 436(2): 171-6, 2008 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-18395978

RESUMO

PRA1 domain family, member 2 (PRAF2) is a novel 19-kDa protein with a prenylated Rab acceptor 1 (PRA1) motif and four transmembrane domains. Our previous studies revealed that PRAF2 is highly expressed in the brain and serves as a candidate prognostic marker in neuroblastoma (NB). PRAF2 is related to proteins PRAF1 (PRA1, prenylin, Yip3) and PRAF3 (GTRAP3-18, JWA, Arl6-IP5), both of which are enriched in the brain and implicated in cellular transport and endo/exocytic vesicle trafficking. However, the function for PRAF2 remains unknown. In this study, we analyzed the distribution and localization of PRAF2 in the mature human brain using two new antibodies specific for the protein. Analysis by immunohistochemistry revealed that in the human cerebellum, the PRAF2 protein was strongly expressed in Purkinje cells and, more moderately, in cells of the molecular and the granular layers. In the cerebral cortex, hippocampus, and lateral ventricles, PRAF2 protein was detected in neuronal cells, but not in non-neuronal cells. Intriguingly, immunoblot analysis revealed that PRAF2 is enriched in synaptic vesicles (SVs) prepared from rat brains. The expression of PRAF2 in specific regions of the brain including SVs suggest an important physiological function for this novel protein, possibly by participating in multiple aspects of SV maturation, transport, and signal transmission.


Assuntos
Encéfalo/metabolismo , Encéfalo/ultraestrutura , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Vesículas Sinápticas/metabolismo , Encéfalo/anatomia & histologia , Linhagem Celular Tumoral , Expressão Gênica/fisiologia , Humanos , Neuroblastoma
13.
Clin Cancer Res ; 13(21): 6312-9, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17975142

RESUMO

PURPOSE: Prenylated Rab acceptor 1 domain family, member 2 (PRAF2) is a novel 19-kDa protein that has recently been implicated in human cancer. In the present study, we analyzed for the first time PRAF2 mRNA expression in a large set of human tumors. The high expression in neuroblastic tumors prompted us to analyze PRAF2 expression correlations with genetic and clinical features of these tumors. In addition, we determined the localization of PRAF2 protein in neuroblastoma cells and studied its regulation in apoptosis. EXPERIMENTAL DESIGN: Affymetrix microarray analysis was done with a set of 41 different tumor types (1,426 samples) in the public domain, a set of three different neuroblastic tumor types (110 samples), and a panel of 25 neuroblastoma cell lines. The subcellular localization of endogenous PRAF2 in neuroblastoma cells was identified by immunofluorescence microscopy and apoptosis detected by Annexin V staining and poly(ADP-ribose) polymerase cleavage. RESULTS: PRAF2 mRNA was detected in 970 of 1,426 samples in the public data set. All 110 neuroblastic tumors expressed PRAF2 at higher levels than any other tumor examined. Importantly, PRAF2 expression levels significantly correlated with the following clinical features: patient age at diagnosis (P = 6.19 x 10(-5)), survival (P = 1.32 x 10(-3)), International Neuroblastoma Staging System stage (P = 2.86 x 10(-4)), and MYCN amplification (P = 3.74 x 10(-3)). PRAF2 localized in bright cytoplasmic punctae and protein levels increased in neuroblastoma cells that underwent cerulenin-induced apoptosis. CONCLUSIONS: Elevated PRAF2 expression levels correlated with unfavorable genetic and clinical features, suggesting PRAF2 as a candidate prognostic marker of neuroblastoma.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Cerulenina/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/metabolismo , Neuroblastoma/metabolismo , Linhagem Celular Tumoral , Pré-Escolar , Endossomos/metabolismo , Humanos , Lactente , Recém-Nascido , Microscopia de Fluorescência , Neuroblastoma/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Estrutura Terciária de Proteína , Distribuição Tecidual
14.
J Exp Zool A Comp Exp Biol ; 303(4): 319-22, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15776417

RESUMO

The aim of this study was to determine whether hypo-osmolarity, which activates taurine transport through the volume-sensitive organic osmolyte channel in skate (Raja erinacea) erythrocytes, also activates the organic osmolyte channel activity of skate AE1 (skAE1) expressed in oocytes. When Xenopus laevis oocytes expressing skAE1 were incubated in hypo-osmotic ND 96 (210 mOsm) media, taurine was transported at a significantly higher rate than when incubated in ND 96 (235 mOsm), which is iso-osmotic to Xenopus plasma. Therefore, hypo-osmotic stress is part of the activation mechanism of the organic osmolyte channel in skAE1 expressing oocytes.


Assuntos
Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Oócitos/metabolismo , Rajidae/metabolismo , Taurina/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia , Xenopus laevis/metabolismo , Animais , Proteína 1 de Troca de Ânion do Eritrócito/genética , Transporte Biológico , Western Blotting , Feminino , Soluções Hipotônicas , Rajidae/genética , Xenopus laevis/fisiologia
15.
J Membr Biol ; 208(3): 241-50, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16604471

RESUMO

When swollen, skate red blood cells increase permeability and allow efflux of a number of solutes, including taurine. Hypoosmosis-induced taurine permeability appears to involve the red cell anion exchanger. However, three isoforms have been cloned from these cells. Therefore, to determine the ability of the individual isoform skate anion exchanger 1 (skAE1) to mediate hypoosmosis-induced taurine permeability as well as associated regulatory events, skAE1 was expressed in Xenopus oocytes. This study focused on investigating the role of tyrosine kinases and lipid rafts in the regulation of the channel. The results showed that tyrosine kinase inhibitors and lipid raft-disrupting agents inhibited the volume-sensitive organic osmolyte channel while protein tyrosine phosphatase inhibitors activated the channel in oocytes expressing skAE1. To study the role of lipid rafts in the activation of the volume-sensitive organic osmolyte channel, the cellular localization of skAE1 was investigated. Also, the role of tyrosine kinases was investigated by examining the tyrosine phosphorylation state of skAE1. Hypoosmotic stress induced mobilization of skAE1 into light membranes and the cell surface as well as tyrosine phosphorylation of skAE1. These events are involved in the activation of the volume-sensitive organic osmolyte channel in Xenopus oocytes expressing skAE1.


Assuntos
Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Oócitos/citologia , Oócitos/fisiologia , Transdução de Sinais/fisiologia , Rajidae/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Proteína 1 de Troca de Ânion do Eritrócito/genética , Tamanho Celular , Células Cultivadas , Proteínas Recombinantes/metabolismo , Rajidae/genética , Xenopus laevis
16.
J Biol Chem ; 279(38): 39447-53, 2004 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-15258156

RESUMO

Upon hypotonic volume expansion, skate erythrocytes lose solutes via a pathway that requires participation of anion exchangers (AEs). Three skate AE isoforms (skAEs) are expressed, and at least skAE1 has been shown to mediate this effect when expressed in oocytes. Under isoosmotic conditions, only a small fraction of skAE1 is expressed on the external plasma membrane. Under these conditions, a portion of skAE1 may be found in non-ionic detergent-insoluble regions. However, the detergent-insoluble material is found intracellularly. Cellular volume expansion by hypoosmotic volume expansion but not volume expansion by isoosmotic medium by permeant solutes (ethylene glycol, diethyl urea, or ammonium chloride) stimulates the appearance of skAE1 in the external plasma membrane, and a significant portion of this is found in detergent-insoluble regions. Upon hypoosmotic volume expansion nearly half of the skAE1 is found as oligomers. SkAE1 in these detergent-insoluble fractions is highly tyrosine phosphorylated. These data suggest that volume expansion by hypoosmotic medium stimulates movement of skAE1 from an intracellular pool contained in detergent-insoluble lipid rafts to the plasma membrane. This skAE1 associates to form oligomers that could be involved in the solute efflux that occurs upon volume expansion.


Assuntos
Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Exocitose/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Ânions/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Soluções Hipotônicas/farmacologia , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Fosfotirosina/metabolismo , Rajidae , Taurina/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA