Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Phys Chem Chem Phys ; 23(7): 4344-4352, 2021 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-33588428

RESUMO

Redox-active bidentate halogen bond donors based on halopyridinium groups as halogen-bond donating units were synthesized and their structures were elucidated by X-ray diffraction analyses and DFT calculations. Via reversible twofold reduction, these dicationic species can be transformed to neutral compounds which should be much weaker Lewis acids. The corresponding electrochemical data were obtained, and CV as well as UV-vis and NMR techniques were also used to determine binding constants of these halogen bond donors to halides. While all titrations agree on the relative order of binding strengths (with chloride being bound strongest), there are marked deviations in the overall affinity constants which are discussed. In contrast to earlier azo-bridge analogues, the ethylene-linked variants presented herein do not oxidize halides, and thus the novel halogen bond donors could also be used as Lewis acidic organocatalysts in a halide abstraction benchmark reaction, yielding a performance similar to bis(haloimidazolium)-derived catalysts.

2.
Clin Transl Radiat Oncol ; 2: 29-35, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29657997

RESUMO

The revival of cancer immunotherapy has taken place with the clinical success of immune checkpoint inhibition. However, the spectrum of immunotherapeutic approaches is much broader encompassing T cell engaging strategies, tumour-specific vaccination, antibodies or immunocytokines. This review focuses on the immunological effects of irradiation and the evidence available on combination strategies with immunotherapy. The available data suggest great potential of combined treatments, yet also poses questions about dose, fractionation, timing and most promising multimodal strategies.

3.
Cell Death Dis ; 7: e2039, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26775694

RESUMO

Glioblastoma is a very aggressive form of brain tumor with limited therapeutic options. Usually, glioblastoma is treated with ionizing radiation (IR) and chemotherapy after surgical removal. However, radiotherapy is frequently unsuccessful, among others owing to resistance mechanisms the tumor cells have developed. Antiapoptotic B-cell leukemia (Bcl)-2 family members can contribute to radioresistance by interfering with apoptosis induction in response to IR. Bcl-2 and the closely related Bcl-xL and Mcl-1 are often overexpressed in glioblastoma cells. In contrast to Bcl-2 and Bcl-xL, Mcl-1 is a short-lived protein whose stability is closely regulated by ubiquitylation-dependent proteasomal degradation. Although ubiquitin ligases facilitate degradation, the deubiquitylating enzyme ubiquitin-specific protease 9x (USP9x) interferes with degradation by removing polyubiquitin chains from Mcl-1, thereby stabilizing this protein. Thus, an inability to downregulate Mcl-1 by enhanced USP9x activity might contribute to radioresistance. Here we analyzed the impact of USP9x on Mcl-1 levels and radiosensitivity in glioblastoma cells. Correlating Mcl-1 and USP9x expressions were significantly higher in human glioblastoma than in astrocytoma. Downregulation of Mcl-1 correlated with apoptosis induction in established glioblastoma cell lines. Although Mcl-1 knockdown by siRNA increased apoptosis induction after irradiation in all glioblastoma cell lines, USP9x knockdown significantly improved radiation-induced apoptosis in one of four cell lines and slightly increased apoptosis in another cell line. In the latter two cell lines, USP9x knockdown also increased radiation-induced clonogenic death. The massive downregulation of Mcl-1 and apoptosis induction in A172 cells transfected with USP9x siRNA shows that the deubiquitinase regulates cell survival by regulating Mcl-1 levels. In contrast, USP9x regulated radiosensitivity in Ln229 cells without affecting Mcl-1 levels. We conclude that USP9x can control survival and radiosensitivity in glioblastoma cells by Mcl-1-dependent and Mcl-1-independent mechanisms.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Feminino , Glioblastoma/patologia , Humanos , Masculino , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Tolerância a Radiação , Transfecção
4.
Strahlenther Onkol ; 189(3): 246-54, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23361139

RESUMO

BACKGROUND AND PURPOSE: Tumor hypoxia is a major problem in radiation therapy of solid tumors because of the radiosensitizing effect of oxygen. Nitroimidazole-containing compounds are oxygen mimetics accumulating in hypoxic tumor areas. However, the broad use of 2-nitroimidazoles as a hypoxic radiosensitizer is limited by their partially low efficacy and/or high neurotoxicity. MATERIALS AND METHODS: Here, we characterized the in vitro hypoxic cytotoxicity and hypoxic radiosensitizing efficacy of N,N,N-tris [2-(2-nitro-1H-imidazol-1-yl)ethyl]amine (PRC) in a hypoxia-sensitive lymphoma and a hypoxia-resistant glioblastoma cell line by colony formation assay and flow cytometry. RESULTS: PRC exerted high hypoxic cytotoxic and radiosensitizing action on both cell lines at almost absent toxicity under normoxic conditions. In particular, under hypoxia, but not normoxia, PRC targeted the mitochondria resulting in oxidative stress, G(2)/M cell cycle arrest, and triggering of the intrinsic apoptosis pathway. CONCLUSION: Our in vitro findings suggest that PRC might be a promising new 2-nitroimidazole for improving radiation therapy of hypoxic tumors in vivo.


Assuntos
Antineoplásicos/farmacologia , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Etilaminas/farmacologia , Nitroimidazóis/farmacologia , Radiossensibilizantes/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Técnicas In Vitro , Células Jurkat , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio , Ensaio Tumoral de Célula-Tronco
5.
Apoptosis ; 14(5): 711-20, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19343496

RESUMO

Ionising radiation, hypoxia, and the cyclooxygenase-2 inhibitor Celecoxib are known agonists of the intrinsic apoptosis pathway that involves mitochondrial damage upstream of caspase activation. Mitochondrial integrity is regulated by the pro-apoptotic Bcl-2 protein family members Bak and Bax. Upstream of the mitochondria, many kinases and phosphatases control the apoptotic response. However, the role of the non-receptor tyrosine kinase p56/Lck during apoptosis is controversial. The present investigation demonstrate the existence of two JCaM1.6 subclones, one expressing and one deficient for Bak. The lack of p56/Lck expression in JCaM1.6 cells per se did hardly affect apoptosis induced by ionising radiation, hypoxia, or Celecoxib. Only the additional loss of Bak expression, as observed in one JCaM1.6 subclone, rendered the cells resistant. siRNA-mediated downregulation of Bak and p56/Lck mimicked the observed effects in the subclones. Earlier experiments performed with the Bak-negative clone might have lead to the wrong assumption that lack of p56/Lck alone, and not the additonal loss of Bak, was responsible for reduced sensitivity towards stimuli of the intrinsic apoptosis pathway.


Assuntos
Apoptose , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/deficiência , Transdução de Sinais , Proteína Killer-Antagonista Homóloga a bcl-2/deficiência , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Celecoxib , Linhagem Celular Tumoral , Células Clonais , Inativação Gênica/efeitos dos fármacos , Inativação Gênica/efeitos da radiação , Humanos , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Pirazóis/farmacologia , Radiação Ionizante , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Sulfonamidas/farmacologia , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
6.
Acta Physiol (Oxf) ; 187(1-2): 191-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16734755

RESUMO

Osmotic shock triggers eryptosis, a suicidal death of erythrocytes characterized by cell shrinkage, cell membrane blebbing and phosphatidylserine exposure at the cell surface. Phosphatidylserine-exposing erythrocytes are recognized by macrophages, engulfed, degraded and thus cleared from circulating blood. Eryptosis following osmotic shock is mediated by two distinct signalling pathways. On the one hand, osmotic shock stimulates a cyclooxygenase leading to formation of prostaglandin E2 and subsequent activation of Ca2+-permeable cation channels. On the other hand, osmotic shock activates a phospholipase A2 leading to release of platelet activating factor, which in turn activates a sphingomyelinase and thus stimulates the formation of ceramide. The increased cytosolic Ca2+ concentrations on the one hand and ceramide on the other trigger phospholipid scrambling of the cell membrane with the subsequent shift of phosphatidylserine from the inner to the outer cell membrane leaflet. Ca2+ further activates Ca2+-sensitive K+ channels leading to cellular KCl loss and further cell shrinkage. The cation channels are inhibited by Cl- anions, erythropoietin and dopamine. The sphingomyelinase is inhibited by high concentrations of urea. Thus, the high Cl- and urea concentrations in renal medulla presumably prevent the triggering of eryptosis despite hyperosmolarity. The mechanisms involved in eryptosis may not only affect the survival of erythrocytes but may be similarly operative in nucleated cells exposed to osmotic shock.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/patologia , Canais Iônicos , Transdução de Sinais/fisiologia , Animais , Apoptose , Cálcio/metabolismo , Cátions , Membrana Celular/metabolismo , Tamanho Celular , Ceramidas/metabolismo , Citosol/metabolismo , Humanos , Pressão Osmótica , Fosfatidilserinas/metabolismo , Fator de Ativação de Plaquetas/metabolismo
7.
J Membr Biol ; 205(3): 147-57, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16362503

RESUMO

Cell proliferation and apoptosis are paralleled by altered regulation of ion channels that play an active part in the signaling of those fundamental cellular mechanisms. Cell proliferation must--at some time point--increase cell volume and apoptosis is typically paralleled by cell shrinkage. Cell volume changes require the participation of ion transport across the cell membrane, including appropriate activity of Cl- and K+ channels. Besides regulating cytosolic Cl- activity, osmolyte flux and, thus, cell volume, most Cl- channels allow HCO3- exit and cytosolic acidification, which inhibits cell proliferation and favors apoptosis. K+ exit through K+ channels may decrease intracellular K+ concentration, which in turn favors apoptotic cell death. K+ channel activity further maintains the cell membrane potential, a critical determinant of Ca2+ entry through Ca2+ channels. Cytosolic Ca2+ may trigger mechanisms required for cell proliferation and stimulate enzymes executing apoptosis. The switch between cell proliferation and apoptosis apparently depends on the magnitude and temporal organization of Ca2+ entry and on the functional state of the cell. Due to complex interaction with other signaling pathways, a given ion channel may play a dual role in both cell proliferation and apoptosis. Thus, specific ion channel blockers may abrogate both fundamental cellular mechanisms, depending on cell type, regulatory environment and condition of the cell. Clearly, considerable further experimental effort is required to fully understand the complex interplay between ion channels, cell proliferation and apoptosis.


Assuntos
Apoptose/fisiologia , Proliferação de Células , Canais Iônicos/fisiologia , Canais de Potássio/fisiologia , Animais , Canais de Cálcio/fisiologia , Tamanho Celular , Genes ras/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Canais de Ânion Dependentes de Voltagem/fisiologia , Receptor fas/fisiologia
8.
Cell Death Differ ; 12(5): 415-28, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15746942

RESUMO

Hyperosmotic shock, energy depletion, or removal of extracellular Cl(-) activates Ca(2+)-permeable cation channels in erythrocyte membranes. Subsequent Ca(2+) entry induces erythrocyte shrinkage and exposure of phosphatidylserine (PS) at the erythrocyte surface. PS-exposing cells are engulfed by macrophages. The present study explored the signalling involved. Hyperosmotic shock and Cl(-) removal triggered the release of prostaglandin E(2) (PGE(2)). In whole-cell recording, activation of the cation channels by Cl(-) removal was abolished by the cyclooxygenase inhibitor diclophenac. In FACS analysis, phospholipase-A(2) inhibitors quinacrine and palmitoyltrifluoromethyl-ketone, and cyclooxygenase inhibitors acetylsalicylic acid and diclophenac, blunted the increase of PS exposure following Cl(-) removal. PGE(2) (but not thromboxane) induced cation channel activation, increase in cytosolic Ca(2+) concentration, cell shrinkage, PS exposure, calpain activation, and ankyrin-R degradation. The latter was attenuated by calpain inhibitors-I/II, while PGE(2)-induced PS exposure was not. In conclusion, hyperosmotic shock or Cl(-) removal stimulates erythrocyte PS exposure through PGE(2) formation and subsequent activation of Ca(2+)-permeable cation channels.


Assuntos
Apoptose/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Prostaglandinas E/metabolismo , Anquirinas/metabolismo , Anexinas/metabolismo , Cálcio/metabolismo , Canais de Cálcio/efeitos dos fármacos , Calpaína/metabolismo , Tamanho Celular/efeitos dos fármacos , Cloretos/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Citosol/efeitos dos fármacos , Diclofenaco/farmacologia , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Humanos , Modelos Biológicos , Pressão Osmótica/efeitos dos fármacos , Técnicas de Patch-Clamp , Fosfatidilserinas/metabolismo , Fosfolipases A/metabolismo , Prostaglandinas E/farmacologia , Quinacrina/farmacologia , Solução Salina Hipertônica
9.
Cell Death Differ ; 11(2): 231-43, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14615798

RESUMO

Erythrocytes lack nuclei and mitochondria, the organelles important for apoptosis of nucleated cells. However, following increase of cytosolic Ca(2+) activity, erythrocytes undergo cell shrinkage, cell membrane blebbing and breakdown of phosphatidylserine asymmetry, all features typical for apoptosis in nucleated cells. The same events are observed following osmotic shock, an effect mediated in part by activation of Ca(2+)-permeable cation channels. However, erythrocyte death following osmotic shock is blunted but not prevented in the absence of extracellular Ca(2+) pointing to additional mechanisms. As shown in this study, osmotic shock (950 mOsm) triggers sphingomyelin breakdown and formation of ceramide. The stimulation of annexin binding following osmotic shock is mimicked by addition of ceramide or purified sphingomyelinase and significantly blunted by genetic (aSM-deficient mice) or pharmacologic (50 microM 3,4-dichloroisocoumarin) knockout of sphingomyelinase. The effect of ceramide is blunted but not abolished in the absence of Ca(2+). Conversely, osmotic shock-induced annexin binding is potentiated in the presence of sublethal concentrations of ceramide. In conclusion, ceramide and Ca(2+) entry through cation channels concert to trigger erythrocyte death during osmotic shock.


Assuntos
Ceramidas/metabolismo , Eritrócitos/citologia , Eritrócitos/metabolismo , Animais , Anexinas/metabolismo , Cálcio/metabolismo , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Ceramidas/biossíntese , Ceramidas/farmacologia , Cumarínicos/farmacologia , Eritrócitos/efeitos dos fármacos , Fumonisinas/farmacologia , Humanos , Ionomicina/farmacologia , Isocumarinas , Camundongos , Camundongos Knockout , Pressão Osmótica/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/metabolismo
10.
Naunyn Schmiedebergs Arch Pharmacol ; 367(4): 391-6, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12690431

RESUMO

Even though lacking mitochondria and nuclei erythrocytes do undergo apoptotic cell death which is characterized by breakdown of phosphatidylserine asymmetry (leading to annexin binding), membrane blebbing and cell shrinkage. Previously, we have shown that erythrocyte apoptosis is triggered by osmotic shrinkage at least in part through activation of cell volume-sensitive cation channels and subsequent Ca2+ entry. The channels could not only be activated by cell shrinkage but as well by replacement of Cl- with gluconate. Both, channel activity and annexin binding were sensitive to high concentrations of amiloride (1 mM). The present study has been performed to search for more effective blockers. To this end channel activity has been evaluated utilizing whole-cell patch-clamp and annexin binding determined by FACS analysis as an indicator of erythrocyte apoptosis. It is shown that either, increase of osmolarity or replacement of Cl- by gluconate triggers the activation of the cation channel which is inhibited by amiloride at 1 mM but not at 100 microM. Surprisingly, the cation channel was significantly more sensitive to the amiloride analogue ethylisopropylamiloride (EIPA, IC(50)=0.6+/-0.1 microM, n=5). Exposure of the cells to osmotic shock by addition of sucrose (850 mOsm) led to stimulation of annexin binding which was inhibited similarly by EIPA (IC(50)=0.2+/-0.2 microM, n=4). Moreover, annexin binding was inhibited by higher concentrations of HOE 642 (IC(50)=10+/-5 microM, n=5) and HOE 694 (IC(50)=12+/-6 microM, n=4). It is concluded that osmotic shock stimulates a cation channel which participates in the triggering of erythrocyte apoptosis. EIPA is an effective inhibitor of this cation channel and of channel mediated triggering of erythrocyte apoptosis.


Assuntos
Amilorida/análogos & derivados , Amilorida/farmacologia , Apoptose/efeitos dos fármacos , Eritrócitos/fisiologia , Canais Iônicos/antagonistas & inibidores , Amilorida/administração & dosagem , Anexinas/metabolismo , Apoptose/fisiologia , Cátions/metabolismo , Tamanho Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eritrócitos/citologia , Eritrócitos/metabolismo , Guanidinas/farmacologia , Humanos , Técnicas In Vitro , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Pressão Osmótica/efeitos dos fármacos , Técnicas de Patch-Clamp , Sulfonas/farmacologia
11.
Cell Death Differ ; 10(2): 249-56, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12700653

RESUMO

Erythrocytes are devoid of mitochondria and nuclei and were considered unable to undergo apoptosis. As shown recently, however, the Ca(2+)-ionophore ionomycin triggers breakdown of phosphatidylserine asymmetry (leading to annexin binding), membrane blebbing and shrinkage of erythrocytes, features typical for apoptosis in nucleated cells. In the present study, the effects of osmotic shrinkage and oxidative stress, well-known triggers of apoptosis in nucleated cells, were studied. Exposure to 850 mOsm for 24 h, to tert-butyl-hydroperoxide (1 mM) for 15 min, or to glucose-free medium for 48 h, all elicit erythrocyte shrinkage and annexin binding, both sequelae being blunted by removal of extracellular Ca(2+) and mimicked by ionomycin (1 microM). Osmotic shrinkage and oxidative stress activate Ca(2+)-permeable cation channels and increase cytosolic Ca(2+) concentration. The channels are inhibited by amiloride (1 mM), which further blunts annexin binding following osmotic shock, oxidative stress and glucose depletion. In conclusion, osmotic and oxidative stress open Ca(2+)-permeable cation channels in erythrocytes, thus increasing cytosolic Ca(2+) activity and triggering erythrocyte apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Cátions/metabolismo , Eritrócitos/fisiologia , Canais Iônicos/fisiologia , Estresse Oxidativo/fisiologia , Amilorida/farmacologia , Anexinas/metabolismo , Apoptose/fisiologia , Cálcio/farmacocinética , Contagem de Células , Tamanho Celular/efeitos dos fármacos , Citosol/química , Citosol/efeitos dos fármacos , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Glucose/metabolismo , Humanos , Canais Iônicos/antagonistas & inibidores , Ionomicina/farmacologia , Ionóforos/farmacologia , Pressão Osmótica/efeitos dos fármacos , Técnicas de Patch-Clamp , terc-Butil Hidroperóxido/farmacologia
12.
Pflugers Arch ; 445(1): 60-6, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12397388

RESUMO

The serum- and glucocorticoid-dependent kinase SGK1 was originally identified as a glucocorticoid-sensitive gene. Subsequently, the two homologous kinases SGK2 and SGK3 have been cloned, being products of distinct genes, which are differentially expressed and share 80% identity in amino acid sequence in their catalytic domains. While SGK1 has been shown to activate ion channels, including K(+) channels, the functions of SGK2 and SGK3 have not been examined. The present study was therefore performed to elucidate the effect of SGK1, SGK2, and SGK3 on electrical properties of renal epithelial cells. To this end human embryonic kidney (HEK293) cells were transfected with the kinases and ion-channel activity determined using the patch-clamp technique. In non-transfected cells and in cells transfected with the empty GFP construct a voltage-gated K(+) current was observed amounting to 303+/-19 pA ( n=13) and 299+/-29 pA ( n=23), respectively. Transfection with SGK1, SGK2 or SGK3 increased the voltage-gated K(+) current to 1056+/-152 pA ( n=17), 555+/-47 pA ( n=17), and 775+/-98 pA ( n=16), respectively. The K(+) current was fully blocked by 3 mM tetraethylammonium chloride and inhibited 45% by the Kv1 channel blocker margatoxin (10 nM). In dual electrode voltage-clamp experiments SGK isoforms up-regulated Kv1 voltage-gated K(+)channels expressed in Xenopus laevis oocytes. The present observations thus reveal a powerful stimulating effect of all three isoforms of SGK on K(+) channels. Those effects may participate in regulation of epithelial transport, cell proliferation, and neuromuscular excitability.


Assuntos
Proteínas Nucleares , Canais de Potássio/fisiologia , Proteínas Serina-Treonina Quinases/farmacologia , Animais , Linhagem Celular , Clonagem Molecular , Condutividade Elétrica , Humanos , Proteínas Imediatamente Precoces , Oócitos , Técnicas de Patch-Clamp , Canais de Potássio/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Isoformas de Proteínas/farmacologia , Regulação para Cima , Xenopus laevis
13.
Pflugers Arch ; 445(1): 74-9, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12397390

RESUMO

In a wide variety of cells, mitogenic factors release Ca(2+) from intracellular stores. The fall of the [Ca(2+)] within the lumen of the Ca(2+)-storing organelles triggers in many cells capacitative Ca(2+) entry (CCE). The present study was performed to elucidate the effect of insulin-like growth factor (IGF-1) on CCE in human embryonic kidney (HEK 293) cells. After depletion of Ca(2+) stores by thapsigargin, CCE was assessed by the increase in cytosolic free [Ca(2+)] (Fura-2 fluorescence imaging) when raising extracellular [Ca(2+)] from 0 to physiological concentrations. IGF-1 exposure (50 ng/ml) for 4 h in serum-free medium markedly enhanced CCE, while a 24-h exposure to IGF-1 depressed CCE profoundly. As some Ca(2+) channels are highly sensitive to the cell membrane potential, and as IGF-1 has been reported to enhance K(+) channel activity, the influence of K(+) channel blockers on the IGF-1-dependent stimulation of CCE was also tested. TEA, charybdotoxin and margatoxin decreased CCE. Similar to the total capacitative calcium entry, the fraction of CCE that was sensitive to K(+) channel blockers was increased after 4 h and decreased after 24 h exposure to IGF-1. Taken together, these data suggest that IGF-1 induces a transient increase followed by a decrease of CCE, and that these effects are at least partly dependent on IGF-1-induced K(+) channel activity.


Assuntos
Cálcio/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Rim/embriologia , Fenômenos Fisiológicos Sanguíneos , Linhagem Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Humanos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Fatores de Tempo
14.
Mol Genet Genomics ; 267(6): 757-71, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12207223

RESUMO

The phytopathogenic basidiomycete Ustilago maydis produces sexual teliospores only after infection of its host plant, maize. To investigate the process of spore formation, we have isolated Ssp1, a protein that is abundantly expressed in mature teliospores. The corresponding gene, ssp1, is expressed at low levels in haploid sporidia; however, transcriptional levels are drastically induced in mature teliospores. Transcriptional regulation of ssp1 involves positive and negative promoter elements, and is subject to control by the cAMP signaling cascade and histone deacetylase-mediated repression. Ssp1 shows similarity to linoleate diol synthase, a fatty acid dioxygenase from the fungus Gaeumannomyces graminis. In agreement with this presumed function, Ssp1 is localized on lipid bodies in germinating teliospores, suggesting a role in the mobilization of storage lipids.


Assuntos
Oxigenases/genética , Esporos Fúngicos/genética , Ustilago/genética , Sequência de Aminoácidos , Northern Blotting , AMP Cíclico , Expressão Gênica , Proteínas de Fluorescência Verde , Histona Desacetilases/metabolismo , Proteínas Luminescentes , Microscopia de Fluorescência , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão , Alinhamento de Sequência , Deleção de Sequência , Ustilago/enzimologia
15.
Pflugers Arch ; 443(4): 625-34, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11907830

RESUMO

Involvement of voltage-gated (Kv) potassium channels in IGF-1-induced proliferation of HEK293 cells was studied by patch-clamp, RT-PCR and FACS analysis. IGF-1 up-regulated outwardly rectifying whole-cell K+ current starting after 1 h of incubation and reaching a maximum after 4-6 h. The IGF-1-stimulated current was voltage-gated with an activation threshold of -30 mV to -40 mV, a half-maximal activation at +5.3+/-1.8 mV, and time constants for activation and inactivation of 4.5+/-0.4 ms and 43.5+/-5.6 ms ( n=10), respectively. The current was inhibited by TEA, margatoxin, agitoxin-2 and stichodactyla toxin. PCR amplification of different Kv subunits from HEK293 cDNA demonstrated the expression of Kv1.1, Kv1.2, Kv1.3, Kv1.4, Kv3.1 and Kv3.4 mRNA. Quantitative RT-PCR showed up-regulation of Kv1.1, 1.2 and 1.3 mRNA by IGF-1. The effect of IGF-1 on K+ current was blocked by inhibitors of phosphatidylinositol 3-kinase (PI3-kinase), wortmannin and LY294002, and mimicked by overexpression of human 3-phosphoinositide-dependent protein kinase-1 (hPDK1) or serum- and glucocorticoid-dependent kinase-1 (hSGK1), both sequential downstream targets of PI3-kinase. IGF-1-induced proliferation of HEK293 cells was inhibited by both K+ channel blockers and inhibitors of PI3-kinase. In conclusion, IGF-1 through PI3-kinase, PDK1 and SGK1 up-regulates Kv channels, an effect required for the proliferative action of the growth factor.


Assuntos
Fator de Crescimento Insulin-Like I/farmacologia , Proteínas Nucleares , Fosfatidilinositol 3-Quinases/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Androstadienos/farmacologia , Divisão Celular/fisiologia , Linhagem Celular , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Humanos , Proteínas Imediatamente Precoces , Rim/citologia , Canal de Potássio Kv1.3 , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Morfolinas/farmacologia , Neurotoxinas/farmacologia , Técnicas de Patch-Clamp , Inibidores de Fosfoinositídeo-3 Quinase , Potássio/metabolismo , Canais de Potássio/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/análise , Venenos de Escorpião , Transdução de Sinais/fisiologia , Wortmanina
16.
Dev Dyn ; 221(4): 464-9, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11500984

RESUMO

Abstract Expression of serum and glucocorticoid-dependent kinase 1 (SGK1) during development in mouse kidney (embryonic day [E] 14 to postnatal day [P] 1) was studied by in situ hybridization and immunofluorescence. In whole embryos, SGK1 mRNA was highly abundant in the developing metanephros, where SGK1 mRNA was expressed in the ureteric buds of the branching collecting duct system and in the mesenchymal blastema-derived comma- and s-shaped bodies. In E14 kidneys, SGK1 protein was below detection level, whereas at day E16, ureteric buds, s-shaped bodies and outgrowing loops of Henle expressed detectable amounts of SGK1 protein. SGK1 protein was also expressed in E16 primary tubules of the collecting duct system. In P1 kidneys, no or only faint SGK1 protein expression was apparent in comma- and s-shaped bodies, whereas SGK1 was continuously expressed by medullary collecting ducts. In conclusion, SGK1 is developmentally expressed in metanephrogenesis. High expression in developing collecting duct and in blastema-derived comma- and s-shaped bodies suggests a dual function of SGK1 in maturation of the reabsorbing collecting duct epithelium and in epithelial transition of the blastema cells.


Assuntos
Proteínas Nucleares , Proteínas Serina-Treonina Quinases/biossíntese , RNA Mensageiro/metabolismo , Animais , Linhagem Celular , Embrião de Mamíferos/metabolismo , Humanos , Proteínas Imediatamente Precoces , Immunoblotting , Hibridização In Situ , Rim/embriologia , Rim/metabolismo , Camundongos , Microscopia de Fluorescência , Fatores de Tempo
17.
Pflugers Arch ; 441(4): 551-8, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11212220

RESUMO

To identify the ion channels involved in erythrocyte volume regulation, whole-cell currents from human red blood cells (RBCs) were recorded in isotonic, hypotonic and hypertonic media. In isotonic NaCl bath solution, whole-cell currents rectified outwardly with reversal potentials (Vrev) between the equilibrium potential of Cl- (ECl) and that of nonselective cations (NSC), ENSC. Replacement of bath Cl by gluconate decreased outward conductance (G outward) by 43 +/- 6% (n = 5) and shifted Vrev with ECl indicating a high fractional Cl- conductance. Hypotonic cell swelling reversibly decreased G outward by 23 +/- 3% (n = 8) while hypertonic cell shrinkage reversibly increased G outward by 27 +/- 8% (n = 5). These shrinkage-activated and swelling-inactivated current fractions rectified outwardly with Vrev at ENSC suggesting that both fractions are generated by the same type of NSC channel. The shrinkage-activated deltaG outward decreased from 4.7 +/- 1.2 nS (n = 3) to 1.4 +/- 0.4 nS (n = 5) and 0.5 +/- 0.4 nS (n = 7) with the increase of pipette [Cl-] from 7 mM to 39 mM and 139 mM, respectively. Similarly, with this increase of pipette [Cl-], G outward under isotonic control conditions decreased from 8.2 +/- 1.4 nS (n = 5) to 7.4 +/- 1.0 nS (n = 20) and 4.1 +/- 0.7 nS (n = 17), due to the differing activity of these NSC channels. In conclusion, human RBCs express, besides a high fractional Cl- conductance, NSC channels that are regulated by cell volume and the cytosolic [Cl-].


Assuntos
Canais de Cloreto/fisiologia , Membrana Eritrocítica/fisiologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Tamanho Celular , Cloretos/metabolismo , Citosol/metabolismo , Condutividade Elétrica , Impedância Elétrica , Gluconatos/metabolismo , Gluconatos/farmacologia , Humanos , Soluções Hipotônicas , Potenciais da Membrana , Concentração Osmolar , Técnicas de Patch-Clamp , Solução Salina Hipertônica , Cloreto de Sódio , Soluções
18.
Artigo em Inglês | MEDLINE | ID: mdl-11913450

RESUMO

Ample pharmacological evidence points to a role of kinases in the regulation of cell volume. Given the limited selectivity of most inhibitors, however, the specific molecules involved have remained largely elusive. The search for cell volume regulated genes in liver HepG2 cells led to the discovery of the human serum- and glucocorticoid-dependent serine/threonine kinase hsgk1. Transcription and expression of hsgk1 is markedly and rapidly upregulated by osmotic and isotonic cell shrinkage. The effect of osmotic cell shrinkage on hsgk1 is mediated by p38 kinase. Further stimuli of hsgk1 transcription include glucocorticoids, aldosterone, TGF-beta1, serum, increase of intracellular Ca2+ and phorbolesters, whereas cAMP downregulates hsgk1 transcription. The hsgk1 protein is expressed in several epithelial tissues including human pancreas, intestine, kidney, and shark rectal gland. Co-expression of hsgk1 with the renal epithelial Na+-channel ENaC or the Na+/K+/2Cl(-)-cotransporter NKCC2 (BSC1) in Xenopus oocytes, accelerates insertion of the transport proteins into the cell membrane and thus, stimulates channel or transport activity. Thus, hsgk1 participates in the regulation of transport by steroids and secretagogues increasing intracellular Ca2+-activity. The stimulation of hsgk1 transcription by TGF-beta1 may further bear pathophysiological relevance.


Assuntos
Tamanho Celular , Epitélio/metabolismo , Proteínas Nucleares , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Transporte Biológico , Proteínas Imediatamente Precoces
19.
Am J Physiol Renal Physiol ; 279(1): F65-76, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10894788

RESUMO

The expression patterns of plasma membrane transporters that specify the epithelial cell type are acquired with ontogeny. To study this process during metanephrogenic mesenchyme-to-epithelium transition, branching ureteric buds with their adjacent mesenchymal blastema (mouse embryonic day E14) were dissected and explanted on a collagen matrix. In culture, induced mesenchymal cells condensed, aggregated, and converted to the comma- and S-shaped body. During in vitro condensation and aggregation, transcription factor Pax-2 protein was downregulated while the epithelial markers E-cadherin and beta-catenin proteins were upregulated. In addition, Wilms' tumor suppressor protein WT-1 was detectable upon condensation and downregulated in the S stage, where expression persisted in the long arm of the S. Patch-clamp, whole cell conductance (G, in nS/10 pF) of pre-epithelial condensed mesenchymal cells (n = 7) was compared with that of tubular proximal S-shaped-body epithelium (n = 6). Both stages expressed E-cadherin and WT-1 mRNA, as demonstrated by single-cell RT-PCR, testifying further to the epithelial as well as the nephrogenic commitment of the recorded cells. Mesenchymal cells exhibited whole cell currents (G = 6.7 +/- 1.3) with reversal potentials (V(rev), in mV) near equilibrium potential for Cl(-) (E(Cl)) (V(rev) = -40 +/- 7) suggestive of a high fractional Cl(-) conductance. Currents of the S-shaped-body cells (G = 4.0 +/- 1.1), in sharp contrast, had a V(rev) at E(K) (V(rev) = -82 +/- 6) indicating a high fractional K(+) conductance. Further, analysis of K(+)-selective whole cell tail currents and single-channel recording revealed a change in K(+) channel expression. Also, Kir6.1 K(+) channel mRNA and protein were downregulated between both stages, whereas K(v)LQT K(+) channel mRNA was abundant throughout. In conclusion, metanephrogenic mesenchyme-to-epithelium transition is accompanied by a profound reorganization of plasma membrane ion channel conductance.


Assuntos
Células Epiteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Canais Iônicos/metabolismo , Rim/embriologia , Rim/metabolismo , Mesoderma/citologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Transativadores , Animais , Caderinas/genética , Caderinas/metabolismo , Agregação Celular , Diferenciação Celular , Membrana Celular/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Condutividade Elétrica , Células Epiteliais/metabolismo , Imuno-Histoquímica , Canais Iônicos/genética , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Rim/citologia , Mesoderma/metabolismo , Camundongos , Fator de Transcrição PAX2 , Potássio/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Cloreto de Sódio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas WT1 , beta Catenina
20.
Pflugers Arch ; 440(2): 295-301, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10898530

RESUMO

The present study was performed to elucidate the effects of urea on vascular smooth muscle cells (SMC). Addition of urea (20, 50, 100 mM) to physiological salt solution blunted the vasoconstrictory effect of phenylephrine (by 17, 25 and 30%, respectively) and of an increased extracellular K+ concentration (by 7, 14 and 19%, respectively) without affecting the basal tone of rabbit arterial rings. According to Fura-2 fluorescence in cultured SMC (A7r5), urea had no effect on basal intracellular calcium activity ([Ca2+]i), but significantly blunted the increase of [Ca2+]i following an increase of extracellular K+. Whole-cell patch-clamp studies revealed that the Ca2+ current through voltage-sensitive Ca2+ channels is significantly inhibited in the presence of urea. As evident from calcein fluorescence, addition of urea leads to sustained cell shrinkage. The effects of urea on vascular tone, [Ca2+]i activity, voltage-gated Ca2+ channels and cell volume are mimicked by addition of raffinose or NaCl. However, the cell shrinkage induced by urea is sustained, whereas the addition of equiosmolar NaCl is only transient and followed by a regulatory cell volume increase. Moreover, hypertonic NaCl increases, whereas urea decreases, the transcription of cell-volume-regulated kinase hsgk. In conclusion, urea leads to sustained shrinkage of vascular smooth muscle cells, which is followed by inhibition of voltage-gated Ca2+ channels, a decrease of [Ca2+]i and thus blunts the vasoconstrictory action of phenylephrine and increased extracellular K+ concentration.


Assuntos
Cálcio/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Ureia/farmacologia , Vasoconstrição/efeitos dos fármacos , Animais , Canais de Cálcio/fisiologia , Células Cultivadas , Condutividade Elétrica , Técnicas In Vitro , Membranas Intracelulares/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Músculo Liso Vascular/citologia , Concentração Osmolar , Osmose , RNA Mensageiro/metabolismo , Coelhos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...