Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Biochim Biophys Acta ; 1848(10 Pt B): 2657-64, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25445673

RESUMO

Neoadjuvant, adjuvant or definitive fractionated radiation therapy are implemented in first line anti-cancer treatment regimens of many tumor entities. Ionizing radiation kills the tumor cells mainly by causing double strand breaks of their DNA through formation of intermediate radicals. Survival of the tumor cells depends on both, their capacity of oxidative defense and their efficacy of DNA repair. By damaging the targeted cells, ionizing radiation triggers a plethora of stress responses. Among those is the modulation of ion channels such as Ca2+-activated K+ channels or Ca2+-permeable nonselective cation channels belonging to the super-family of transient receptor potential channels. Radiogenic activation of these channels may contribute to radiogenic cell death as well as to DNA repair, glucose fueling, radiogenic hypermigration or lowering of the oxidative stress burden. The present review article introduces these channels and summarizes our current knowledge on the mechanisms underlying radiogenic ion channel modulation. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Assuntos
DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Radiação Ionizante , Canais de Potencial de Receptor Transitório/metabolismo , Morte Celular/efeitos da radiação , Dano ao DNA , Reparo do DNA , DNA de Neoplasias/química , DNA de Neoplasias/metabolismo , Humanos , Terapia Neoadjuvante , Neoplasias/genética , Neoplasias/patologia , Neoplasias/radioterapia , Canais de Potássio Cálcio-Ativados/genética , Tolerância a Radiação , Radioterapia Adjuvante , Transdução de Sinais , Canais de Potencial de Receptor Transitório/genética , Resultado do Tratamento
2.
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
3.
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
4.
Radiother Oncol ; 124(3): 462-467, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28711224

RESUMO

BACKGROUND: Radiotherapy (RT) is used to treat retinoblastoma (Rb), the most frequent ocular tumour in children. Besides eradicating the tumour, RT can cause severe side effects including secondary malignancies. This study aimed to define whether the radioprotector ortho-phospho-L-tyrosine (pTyr) prevents RT-induced side effects and affects local tumour control in a xenograft and a genetic orthotopic Rb mouse model. METHODS: B6;129-Rb1tm3Tyj/J (Rb+/-) and Y79-Rb cell-xenografted nude mice were fractionated external beam irradiated (15 fractions of 5Gy 6MV photons during 3weeks) with or without pTyr pre-treatment (100mg/kg BW, 16h prior to each irradiation). One, three, six and nine months after RT, tumour control and RT toxicity were evaluated using in vivo imaging and histology. We also analysed pTyr dependant post irradiation cell survival and p53 activity in vitro. RESULTS: In vitro pTyr pre-treatment showed no radioprotection on Y79 cells, but led to p53 stabilisation in unirradiated Y79 cells and to a facilitation of radiation-induced p21 up-regulation, confirming a modulation of p53 activity by pTyr. In both mouse models, secondary tumours were undetectable. In Rb+/- mice, pTyr significantly lowered RT-induced greying of the fur, retinal thickness reduction and photoreceptor loss. However, in the xenografted Rb model, pTyr considerably decreased RT-mediated tumour control, which was observed in 16 out of 22 control eyes but in none of the 24 pTyr treated eyes. CONCLUSIONS: In Rb+/- mice pTyr significantly prevents RT-induced greying of the fur as well as retinal degeneration. However, since non-irradiated control mice were not used in our study, a formal possibility exists that the effect shown in the retina of Rb+/- mice may be due to ageing of the animals and/or actions of pTyr alone. Unfortunately, as tested in a xenograft model, pTyr treatment reduced the control of Rb tumours.


Assuntos
Fracionamento da Dose de Radiação , Fosfotirosina/farmacologia , Protetores contra Radiação/farmacologia , Neoplasias da Retina/radioterapia , Retinoblastoma/radioterapia , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Camundongos , Retinoblastoma/patologia , Proteína Supressora de Tumor p53/fisiologia
5.
Oncotarget ; 8(56): 95896-95913, 2017 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-29221175

RESUMO

TRPM8 is a Ca2+-permeable nonselective cation channel belonging to the melastatin sub-group of the transient receptor potential (TRP) family. TRPM8 is aberrantly overexpressed in a variety of tumor entities including glioblastoma multiforme where it reportedly contributes to tumor invasion. The present study aimed to disclose further functions of TRPM8 in glioma biology in particular upon cell injury by ionizing radiation. To this end, TCGA data base was queried to expose the TRPM8 mRNA abundance in human glioblastoma specimens and immunoblotting was performed to analyze the TRPM8 protein abundance in primary cultures of human glioblastoma. Moreover, human glioblastoma cell lines were irradiated with 6 MV photons and TRPM8 channels were targeted pharmacologically or by RNA interference. TRPM8 abundance, Ca2+ signaling and resulting K+ channel activity, chemotaxis, cell migration, clonogenic survival, DNA repair, apoptotic cell death, and cell cycle control were determined by qRT-PCR, fura-2 Ca2+ imaging, patch-clamp recording, transfilter migration assay, wound healing assay, colony formation assay, immunohistology, flow cytometry, and immunoblotting. As a result, human glioblastoma upregulates TRPM8 channels to variable extent. TRPM8 inhibition or knockdown slowed down cell migration and chemotaxis, attenuated DNA repair and clonogenic survival, triggered apoptotic cell death, impaired cell cycle and radiosensitized glioblastoma cells. Mechanistically, ionizing radiation activated and upregulated TRPM8-mediated Ca2+ signaling that interfered with cell cycle control probably via CaMKII, cdc25C and cdc2. Combined, our data suggest that TRPM8 channels contribute to spreading, survival and radioresistance of human glioblastoma and, therefore, might represent a promising target in future anti-glioblastoma therapy.

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.
Integr Biol (Camb) ; 8(10): 1067-1078, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27713970

RESUMO

Although much is known about chemotaxis- induced by gradients of soluble chemical cues - the molecular mechanisms involved in haptotaxis (migration induced by substrate-bound protein gradients) are largely unknown. We used micropatterning to produce discontinuous gradients consisting of µm-sized fibronectin-dots arranged at constant lateral but continuously decreasing axial spacing. Parameters like gradient slope, protein concentration and size or shape of the fibronectin dots were modified to determine optimal conditions for directional cell migration in gradient patterns. We demonstrate that fibroblasts predominantly migrate uphill towards a higher fibronectin density in gradients with a dot size of 2 × 2 µm, a 2% and 6% slope, and a low fibronectin concentration of 1 µg ml-1. Increasing dot size to 3.5 × 3.5 µm resulted in stationary cells, whereas rectangular dots (2 × 3 µm) orientated perpendicular to the gradient axis preferentially induce lateral migration. During haptotaxis, the Golgi apparatus reorients to a posterior position between the nucleus and the trailing edge. Using pharmacological inhibitors, we demonstrate that actomyosin contractility and microtubule dynamics are a prerequisite for gradient recognition indicating that asymmetric intracellular forces are necessary to read the axis of adhesive gradients. In the haptotaxis signalling cascade, RhoA and Cdc42, and the atypical protein kinase C zeta (aPKCζ), but not Rac, are located upstream of actomyosin contractility.


Assuntos
Actomiosina/fisiologia , Polaridade Celular , Quimiotaxia/fisiologia , Fibroblastos/fisiologia , Fibronectinas/metabolismo , Mecanotransdução Celular/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Animais , Células Cultivadas , Embrião de Galinha , Matriz Extracelular/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Fibroblastos/citologia , Contração Muscular/fisiologia , Estresse Mecânico
8.
Sci Rep ; 5: 13450, 2015 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-26304588

RESUMO

Tumor cells can adapt to a hostile environment with reduced oxygen supply. The present study aimed to identify mechanisms that confer hypoxia resistance. Partially hypoxia/reoxygenation (H/R)-resistant proximal tubular (PT) cells were selected by exposing PT cultures to repetitive cycles of H/R. Thereafter, H/R-induced changes in mRNA and protein expression, inner mitochondrial membrane potential (ΔΨ(m)), formation of superoxide, and cell death were compared between H/R-adapted and control PT cultures. As a result, H/R-adapted PT cells exhibited lower H/R-induced hyperpolarization of ΔΨ(m) and produced less superoxide than the control cultures. Consequently, H/R triggered ΔΨ(m) break-down and DNA degradation in a lower percentage of H/R-adapted than control PT cells. Moreover, H/R induced upregulation of mitochondrial uncoupling protein-3 (UCP-3) in H/R-adapted PT but not in control cultures. In addition, ionizing radiation killed a lower percentage of H/R-adapted as compared to control cells suggestive of an H/R-radiation cross-resistance developed by the selection procedure. Knockdown of UCP-3 decreased H/R- and radioresitance of the H/R-adapted cells. Finally, UCP-3 protein abundance of PT-derived clear cell renal cell carcinoma and normal renal tissue was compared in human specimens indicating upregulation of UCP-3 during tumor development. Combined, our data suggest functional significance of UCP-3 for H/R resistance.


Assuntos
Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Canais Iônicos/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Proteínas Mitocondriais/metabolismo , Oxigênio/metabolismo , Idoso , Hipóxia Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Masculino , Estresse Oxidativo , Células Tumorais Cultivadas , Proteína Desacopladora 3 , Regulação para Cima
9.
Front Physiol ; 4: 212, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23966948

RESUMO

The standard treatment of many tumor entities comprises fractionated radiation therapy which applies ionizing radiation to the tumor-bearing target volume. Ionizing radiation causes double-strand breaks in the DNA backbone that result in cell death if the number of DNA double-strand breaks exceeds the DNA repair capacity of the tumor cell. Ionizing radiation reportedly does not only act on the DNA in the nucleus but also on the plasma membrane. In particular, ionizing radiation-induced modifications of ion channels and transporters have been reported. Importantly, these altered transports seem to contribute to the survival of the irradiated tumor cells. The present review article summarizes our current knowledge on the underlying mechanisms and introduces strategies to radiosensitize tumor cells by targeting plasma membrane ion transports.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA