Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Eur Biophys J ; 45(7): 585-598, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27165704

RESUMO

K+ channels crosstalk with biochemical signaling cascades and regulate virtually all cellular processes by adjusting the intracellular K+ concentration, generating the membrane potential, mediating cell volume changes, contributing to Ca2+ signaling, and directly interacting within molecular complexes with membrane receptors and downstream effectors. Tumor cells exhibit aberrant expression and activity patterns of K+ channels. The upregulation of highly "oncogenic" K+ channels such as the Ca2+-activated IK channel may drive the neoplastic transformation, malignant progression, metastasis, or therapy resistance of tumor cells. In particular, ionizing radiation in doses used for fractionated radiotherapy in the clinic has been shown to activate K+ channels. Radiogenic K+ channel activity, in turn, contributes to the DNA damage response and promotes survival of the irradiated tumor cells. Tumor-specific overexpression of certain K+ channel types together with the fact that pharmacological K+ channel modulators are already in clinical use or well tolerated in clinical trials suggests that K+ channel targeting alone or in combination with radiotherapy might become a promising new strategy of anti-cancer therapy. The present article aims to review our current knowledge on K+ channel signaling in irradiated tumor cells. Moreover, it provides new data on molecular mechanisms of radiogenic K+ channel activation and downstream signaling events.


Assuntos
Neoplasias/patologia , Neoplasias/radioterapia , Canais de Potássio/metabolismo , Transdução de Sinais/efeitos da radiação , Animais , Humanos , Neoplasias/metabolismo
2.
Pflugers Arch ; 465(8): 1209-21, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23443853

RESUMO

Aberrant ion channel expression in the plasma membrane is characteristic for many tumor entities and has been attributed to neoplastic transformation, tumor progression, metastasis, and therapy resistance. The present study aimed to define the function of these "oncogenic" channels for radioresistance of leukemia cells. Chronic myeloid leukemia cells were irradiated (0-6 Gy X ray), ion channel expression and activity, Ca(2+)- and protein signaling, cell cycle progression, and cell survival were assessed by quantitative reverse transcriptase-polymerase chain reaction, patch-clamp recording, fura-2 Ca(2+)-imaging, immunoblotting, flow cytometry, and clonogenic survival assays, respectively. Ionizing radiation-induced G2/M arrest was preceded by activation of Kv3.4-like voltage-gated potassium channels. Channel activation in turn resulted in enhanced Ca(2+) entry and subsequent activation of Ca(2+)/calmodulin-dependent kinase-II, and inactivation of the phosphatase cdc25B and the cyclin-dependent kinase cdc2. Accordingly, channel inhibition by tetraethylammonium and blood-depressing substance-1 and substance-2 or downregulation by RNA interference led to release from radiation-induced G2/M arrest, increased apoptosis, and decreased clonogenic survival. Together, these findings indicate the functional significance of voltage-gated K(+) channels for the radioresistance of myeloid leukemia cells.


Assuntos
Ciclo Celular/genética , Sobrevivência Celular/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Canais de Potássio Shaw/genética , Canais de Potássio Shaw/metabolismo , Apoptose/genética , Proteína Quinase CDC2 , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Divisão Celular/genética , Linhagem Celular Tumoral , Células Cultivadas , Ciclina B/genética , Ciclina B/metabolismo , Quinases Ciclina-Dependentes , Fase G2/genética , Humanos , Células K562 , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Tolerância a Radiação/genética , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
3.
Ultrastruct Pathol ; 37(4): 241-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23758094

RESUMO

Ultraviolet radiation (UV) induces a series of morphological and ultrastructural alterations in human epidermis. Alterations observed in irradiated keratinocytes in morphological studies done before were cell retraction with loss of intercellular interactions, surface blebbing, and eventually cell death by apoptosis. The aim of this study was to investigate effect of UV-A, UV-B, and UV-C irradiation on the cytoskeleton of human keratinocytes. Keratinocytes were obtained by exfoliative scrubbing procedure from buccal mucosa of healthy individuals, and treated with UV-A, UV-B, and UV-C radiation. Afterward, treated cell were labeled with anti-alfa-tubulin and anti-human-cytokeratin and analyzed by light and confocal microscopy. The intensity of the cytokeratin labeling was found to be much higher in all irradiated cells than in control cells as observed by light microscope and measured with the Image J program. This measurement showed that with the decrease in the wavelengths of UV irradiation the intensity of the labeling of cells increases. Although the authors expected to find the collapse of microtubules toward the cell nucleus or their rearrangement in UV-treated cells, these alterations were not verified on cell smears labeled with anti-alfa tubulin observed by confocal microscope. When they used electron microscopy to examine in more detail the morphology of irradiated cells, they did not find apoptotic cells, but found features of autophagy in UV-treated keratinocytes. The authors assume that autophagy found as a result of UV radiation of human keratinocytes acts as a cytoprotective mechanism against UV-induced apoptosis.


Assuntos
Autofagia/efeitos da radiação , Citoesqueleto/efeitos da radiação , Queratinócitos/efeitos da radiação , Queratinócitos/ultraestrutura , Raios Ultravioleta/efeitos adversos , Adulto , Células Cultivadas , Feminino , Humanos , Imuno-Histoquímica , Masculino , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Adulto Jovem
4.
Cell Physiol Biochem ; 26(4-5): 597-608, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21063097

RESUMO

Genotoxic stress induces cell cycle arrest and DNA repair which may enable tumor cells to survive radiation therapy. Here, we defined the role of Ca(2+) signaling in the cell cycle control and survival of chronic myeloid leukemia (CML) cells subjected to ionizing radiation (IR). To this end, K562 erythroid leukemia cells were irradiated (0-10 Gy). Tumor survival was analyzed by clonogenic survival assay and cell cycle progression via flow cytometry. Plasma membrane cation conductance was assessed by patch-clamp whole-cell recording and the cytosolic free Ca(2+) concentration ([Ca(2+)](i)) was measured by fura-2 Ca(2+) imaging. Nuclear activity of Ca(2+)/calmodulin-dependent kinase II (CaMKII) was defined by Western blotting. In addition, the effect of IR (5 Gy) on the cation conductance of primary CML cells was determined. The results indicated that IR (10 Gy) induced a G(2)/M cell cycle arrest of K562 cells within 24 h post-irradiation (p.i.) and decreased the clonogenic survival to 0.5 % of that of the control cells. In K562 cells, G(2)/M cell cycle arrest was preceded by activation of TRPV5/6-like nonselective cation channels in the plasma membrane 1-5 h p.i., resulting in an elevated Ca(2+) entry as evident from fura-2 Ca(2+) imaging. Similarly, IR stimulated a Ca(2+)-permeable nonselective cation conductance in primary CML cells within 2-4 h p.i.. Ca(2+) entry, into K562 cells was paralleled by an IR-induced activation of nuclear CaMKII. The IR-stimulated accumulation in G(2) phase was delayed upon buffering [Ca(2+)](i) with the Ca(2+) chelator BAPTA-AM or inhibiting CaMKII with KN93 (1 nM). In addition, KN93 decreased the clonogenic survival of irradiated cells but not of control cells. In conclusion, the data suggest that IR-stimulated cation channel activation, Ca(2+) entry and CaMKII activity participate in control of cell cycle progression and survival of irradiated CML cells.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Tolerância a Radiação , Canais de Cátion TRPV/metabolismo , Canais de Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Divisão Celular , Quelantes/química , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/química , Ácido Egtázico/farmacologia , Fura-2/química , Fura-2/farmacologia , Fase G2 , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/radioterapia , Técnicas de Patch-Clamp , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Radiação Ionizante
5.
Front Pharmacol ; 11: 489, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32390841

RESUMO

Many tumor cells express highly elevated activities of voltage-gated K+ channels in the plasma membrane which are indispensable for tumor growth. To test for K+ channel function during DNA damage response, we subjected human chronic myeloid leukemia (CML) cells to sub-lethal doses of ionizing radiation (0-8 Gy, 6 MV photons) and determined K+ channel activity, K+ channel-dependent Ca2+ signaling, cell cycle progression, DNA repair, and clonogenic survival by whole-cell patch clamp recording, fura-2 Ca2+ imaging, Western blotting, flow cytometry, immunofluorescence microscopy, and pre-plating colony formation assay, respectively. As a result, the human erythroid CML cell line K562 and primary human CML cells functionally expressed hERG1. Irradiation stimulated in both cell types an increase in the activity of hERG1 K+ channels which became apparent 1-2 h post-irradiation. This increase in K+ channel activity was paralleled by an accumulation in S phase of cell cycle followed by a G2/M cell cycle arrest as analyzed between 8 and 72 h post-irradiation. Attenuating the K+ channel function by applying the hERG1 channel inhibitor E4031 modulated Ca2+ signaling, impaired inhibition of the mitosis promoting subunit cdc2, overrode cell cycle arrest, and decreased clonogenic survival of the irradiated cells but did not affect repair of DNA double strand breaks suggesting a critical role of the hERG1 K+ channels for the Ca2+ signaling and the cell cycle control during DNA damage response.

6.
Oxid Med Cell Longev ; 2016: 8026702, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26839633

RESUMO

Messenger RNA data of lymphohematopoietic cancer lines suggest a correlation between expression of the cation channel TRPM2 and the antiapoptotic protein Bcl-2. The latter is overexpressed in various tumor entities and mediates therapy resistance. Here, we analyzed the crosstalk between Bcl-2 and TRPM2 channels in T cell leukemia cells during oxidative stress as conferred by ionizing radiation (IR). To this end, the effects of TRPM2 inhibition or knock-down on plasma membrane currents, Ca(2+) signaling, mitochondrial superoxide anion formation, and cell cycle progression were compared between irradiated (0-10 Gy) Bcl-2-overexpressing and empty vector-transfected Jurkat cells. As a result, IR stimulated a TRPM2-mediated Ca(2+)-entry, which was higher in Bcl-2-overexpressing than in control cells and which contributed to IR-induced G2/M cell cycle arrest. TRPM2 inhibition induced a release from G2/M arrest resulting in cell death. Collectively, this data suggests a pivotal function of TRPM2 in the DNA damage response of T cell leukemia cells. Apoptosis-resistant Bcl-2-overexpressing cells even can afford higher TRPM2 activity without risking a hazardous Ca(2+)-overload-induced mitochondrial superoxide anion formation.


Assuntos
Pontos de Checagem do Ciclo Celular , Leucemia de Células T/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Canais de Cátion TRPM/metabolismo , Apoptose , Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Regulação Leucêmica da Expressão Gênica , Humanos , Células Jurkat , Mitocôndrias/metabolismo , Estresse Oxidativo , Técnicas de Patch-Clamp , Radiação Ionizante , Transdução de Sinais , Superóxidos/química
7.
Radiother Oncol ; 103(3): 373-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22516777

RESUMO

BACKGROUND AND PURPOSE: Solid tumor cells may adapt to an ischemic microenvironment by upregulation of sodium/glucose cotransport (SGLT) in the plasma membrane which supplies the tumor cell with glucose even at very low extracellular glucose concentration. Since SGLT activity has been shown to depend on the epithelial growth factor receptor (EGFR) and EGFR reportedly is activated by ionizing radiation, we tested for irradiation-induced SGLT activity. MATERIALS AND METHODS: A549 lung adenocarcinoma and FaDu head and neck squamous cancer cells were irradiated with 0 and 4 Gy X-ray and electrogenic SGLT transport activity was recorded by patch clamp current clamp in the presence and absence of extracellular glucose (5mM), the SGLT inhibitor phlorizin (500 µM), and the inhibitor of the EGFR tyrosine kinase activity erlotinib (1 µM). In addition, the effect of phlorizin and erlotinib on glucose uptake and clonogenic survival was tested in irradiated and control cells by tracer flux and colony formation assays, respectively. RESULTS: Irradiated A549 cells exhibited a significantly lower membrane potential 3h after irradiation than the control cells. Phlorizin, erlotinib or removal of extracellular glucose, hyperpolarized the irradiated A549 cells to a significantly higher extent than the control cells. Similarly, but less pronounced, glucose removal hyperpolarized irradiated FaDu cells. In addition, irradiated A549 cells exhibited a highly increased (3)H-glucose uptake which was sensitive to phlorizin. Finally, phlorizin radiosensitized the A549 and FaDu cells as evident from the colony formation assays. CONCLUSIONS: Taken together, these data suggest an irradiation-stimulated and EGFR-mediated increase in SGLT-generated glucose uptake which is required for the survival of the genotoxically stressed tumor cells.


Assuntos
Adenocarcinoma/fisiopatologia , Receptores ErbB/farmacologia , Neoplasias Pulmonares/fisiopatologia , Proteínas de Transporte de Sódio-Glucose/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/radioterapia , Adenocarcinoma de Pulmão , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/fisiopatologia , Linhagem Celular Tumoral/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cloridrato de Erlotinib , Glucose/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/fisiopatologia , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Florizina/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacologia , Doses de Radiação , Proteínas de Transporte de Sódio-Glucose/antagonistas & inibidores , Proteínas de Transporte de Sódio-Glucose/efeitos dos fármacos , Proteínas de Transporte de Sódio-Glucose/efeitos da radiação , Células Tumorais Cultivadas/efeitos da radiação
8.
Radiother Oncol ; 101(1): 122-6, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21704404

RESUMO

BACKGROUND AND PURPOSE: Glioblastoma cells express high levels of Ca(2+)-activated BK K(+) channels which have been proposed to be indispensable for glioblastoma proliferation and migration. Since migration of glioblastoma cells is reportedly stimulated by ionizing radiation (IR), we tested for an IR-induced increase in BK channel activity and its effect on cell migration. MATERIALS AND METHODS: T98G and U87MG cells were X-ray-irradiated with 0-2 Gy, BK channel activity was assessed by patch-clamp recording, migration by trans-well migration assay, and activation of the Ca(2+)/calmodulin-dependent kinase II (CaMKII) by immunoblotting. RESULTS: IR dose-dependently stimulated migration of glioblastoma cells which was sensitive to the BK channel inhibitor paxilline. Ca(2+)-permeabilization of T98G cells activated up to 350 BK channels per cells. Importantly, IR stimulated an increase in BK channel open probability but did not modify the total number of channels. Moreover, IR activated CaMKII in a paxilline-sensitive manner. Finally, inhibition of CaMKII by KN-93 abolished the IR-stimulated migration. CONCLUSIONS: We conclude that IR stimulates BK channel activity which results in activation of CaMKII leading to enhanced glioblastoma cell migration.


Assuntos
Neoplasias Encefálicas/radioterapia , Movimento Celular/efeitos da radiação , Glioblastoma/radioterapia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos da radiação , Canais de Potássio Cálcio-Ativados/efeitos da radiação , Benzilaminas/farmacologia , Neoplasias Encefálicas/patologia , Movimento Celular/efeitos dos fármacos , Glioblastoma/patologia , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio Cálcio-Ativados/metabolismo , Radiação Ionizante , Transdução de Sinais , Sulfonamidas/farmacologia , Células Tumorais Cultivadas
9.
Diabetes Care ; 30(8): 1964-7, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17519431

RESUMO

OBJECTIVE: The aim of the study was to assess the prevalence of cutaneous disorders and their relation to disease duration, metabolic control, and microvascular complications in children and adolescents with type 1 diabetes. RESEARCH DESIGN AND METHODS: The presence and frequency of skin manifestations were examined and compared in 212 unselected type 1 diabetic patients (aged 2-22 years, diabetes duration 1-15 years) and 196 healthy sex- and age-matched control subjects. Logistic regression was used to analyze the relation of cutaneous disorders with diabetes duration, glycemic control, and microvascular complications. RESULTS: One hundred forty-two (68%) type 1 diabetic patients had at least one cutaneous disorder vs. 52 (26.5%) control subjects (P < 0.01). Diabetes-associated skin lesions were found in 81 (38%) patients. Acquired ichthyosis, rubeosis faciei, diabetic hand, and necrobiosis lipoidica were seen in 22 vs. 3%, 7.1 vs. 0%, 2.3 vs. 0%, and 2.3 vs. 0% of type 1 diabetic and control subjects, respectively. The frequency of cutaneous reactions to insulin therapy was low (-2.7%). The prevalence of fungal infections in patients and control subjects was 4.7% and 1.5%, respectively. Keratosis pilaris affected 12% of our patients vs. 1.5% of control subjects. Diabetic hand was strongly (odds ratio 1.42 [95% CI 1.11-1.81]; P < 0.001), and rubeosis faciei weakly (1.22 [1.04-1.43]; P = 0.0087), associated with diabetes duration. Significant association was also found between acquired ichthyosis and keratosis pilaris (1.53 [1.09-1.79]; P < 0.001). CONCLUSIONS: Cutaneous manifestations are common in type 1 diabetic patients, and some of them, like acquired ichthyosis and keratosis pilaris, develop early in the course of the disease. Diabetic hand and rubeosis faciei are related to disease duration.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Dermatopatias/epidemiologia , Adolescente , Adulto , Idade de Início , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Prevalência , Valores de Referência , Análise de Regressão , Dermatopatias/classificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA