Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35163464

RESUMEN

Novel compounds significantly interfering with the mitochondrial energy production may have therapeutic value in triple-negative breast cancer (TNBC). This criterion is clearly fulfilled by desethylamiodarone (DEA), which is a major metabolite of amiodarone, a widely used antiarrhythmic drug, since the DEA previously demonstrated anti-neoplastic, anti-metastasizing, and direct mitochondrial effects in B16F10 melanoma cells. Additionally, the more than fifty years of clinical experience with amiodarone should answer most of the safety concerns about DEA. Accordingly, in the present study, we investigated DEA's potential in TNBC by using a TN and a hormone receptor positive (HR+) BC cell line. DEA reduced the viability, colony formation, and invasive growth of the 4T1 cell line and led to a higher extent of the MCF-7 cell line. It lowered mitochondrial transmembrane potential and induced mitochondrial fragmentation. On the other hand, DEA failed to significantly affect various parameters of the cellular energy metabolism as determined by a Seahorse live cell respirometer. Cyclooxygenase 2 (COX-2), which was upregulated by DEA in the TNBC cell line only, accounted for most of 4T1's DEA resistance, which was counteracted by the selective COX-2 inhibitor celecoxib. All these data indicate that DEA may have potentiality in the therapy of TNBC.


Asunto(s)
Amiodarona/análogos & derivados , Antineoplásicos/farmacología , Celecoxib/farmacología , Ciclooxigenasa 2/metabolismo , Mitocondrias/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Amiodarona/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Metabolismo Energético/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos
2.
Biochem Pharmacol ; 162: 98-108, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30296409

RESUMEN

PURPOSE: The cytoprotective effect of poly(ADP-ribose) polymerase 1 (PARP1) inhibition is well documented in various cell types subjected to oxidative stress. Previously, we have demonstrated that PARP1 inhibition activates Akt, and showed that this response plays a critical role in the maintenance of mitochondrial integrity and in cell survival. However, it has not yet been defined how nuclear PARP1 signals to cytoplasmic Akt. METHODS: WRL 68, HeLa and MCF7 cells were grown in culture. Oxidative stress was induced with hydrogen peroxide. PARP was inhibited with the PARP inhibitor PJ34. ATM, mTOR- and NEMO were silenced using specific siRNAs. Cell viability assays were based on the MTT assay. PARP-ATM pulldown experiments were conducted; each protein was visualized by Western blotting. Immunoprecipitation of ATM, phospho-ATM and NEMO was performed from cytoplasmic and mitochondrial cell fractions and proteins were detected by Western blotting. In some experiments, a continually active Akt construct was introduced. Nuclear to cytoplasmic and mitochondrial translocation of phospho-Akt was visualized by confocal microscopy. RESULTS: Here we present evidence for a PARP1 mediated, PARylation-dependent interaction between ATM and NEMO, which is responsible for the cytoplasmic transport of phosphorylated (thus, activated) ATM kinase. In turn, the cytoplasmic p-ATM and NEMO forms complex with mTOR and Akt, yielding the phospho-ATM-NEMO-Akt-mTOR signalosome, which is responsible for the PARP-inhibition induced Akt activation. The phospho-ATM-NEMO-Akt-mTOR signalosome localizes to the mitochondria and is essential for the PARP-inhibition-mediated cytoprotective effects in oxidatively stressed cells. When the formation of the signalosome is prevented, the cytoprotective effects diminish, but cells can be rescued by constantly active Akt1, further confirming the critical role of Akt activation in cytoprotection. CONCLUSIONS: Taken together, the data presented in the current paper are consistent with the hypothesis that PARP inhibition suppresses the PARylation of ATM, which, in turn, forms an ATM-NEMO complex, which exits the nucleus, and combines in the cytosol with mTOR and Act, resulting in Act phosphorylation (i.e. activation), which, in turn, produces the cytoprotective action via the induction of Akt-mediated survival pathways. This mechanism can be important in the protective effect of PARP inhibitor in various diseases associated with oxidative stress. Moreover, disruption of the formation or action of the phospho-ATM-NEMO-Akt-mTOR signalosome may offer potential future experimental therapeutic checkpoints.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Quinasa I-kappa B/metabolismo , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citoprotección/fisiología , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Células MCF-7 , Mitocondrias/efectos de los fármacos , Fenantrenos/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
3.
Biochem Pharmacol ; 150: 86-96, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29378182

RESUMEN

Mitochondria fragmentation destabilizes mitochondrial membranes, promotes oxidative stress and facilitates cell death, thereby contributing to the development and the progression of several mitochondria-related diseases. Accordingly, compounds that reverse mitochondrial fragmentation could have therapeutic potential in treating such diseases. BGP-15, a hydroxylamine derivative, prevents insulin resistance in humans and protects against several oxidative stress-related diseases in animal models. Here we show that BGP-15 promotes mitochondrial fusion by activating optic atrophy 1 (OPA1), a GTPase dynamin protein that assist fusion of the inner mitochondrial membranes. Suppression of Mfn1, Mfn2 or OPA1 prevents BGP-15-induced mitochondrial fusion. BGP-15 activates Akt, S6K, mTOR, ERK1/2 and AS160, and reduces JNK phosphorylation which can contribute to its protective effects. Furthermore, BGP-15 protects lung structure, activates mitochondrial fusion, and stabilizes cristae membranes in vivo determined by electron microscopy in a model of pulmonary arterial hypertension. These data provide the first evidence that a drug promoting mitochondrial fusion in in vitro and in vivo systems can reduce or prevent the progression of mitochondria-related disorders.


Asunto(s)
Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/metabolismo , Dinámicas Mitocondriales/fisiología , Oximas/uso terapéutico , Piperidinas/uso terapéutico , Células A549 , Animales , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Células HeLa , Humanos , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/patología , Dinámicas Mitocondriales/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Oximas/farmacología , Piperidinas/farmacología , Ratas , Ratas Wistar , Resultado del Tratamiento
4.
PLoS One ; 12(12): e0189470, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29220397

RESUMEN

Bladder cancer (BC) is a common malignancy of the urinary tract that has a higher frequency in men than in women. Cytostatic resistance and metastasis formation are significant risk factors in BC therapy; therefore, there is great interest in overcoming drug resistance and in initiating research for novel chemotherapeutic approaches. Here, we suggest that desethylamiodarone (DEA)-a metabolite of amiodarone-may have cytostatic potential. DEA activates the collapse of mitochondrial membrane potential (detected by JC-1 fluorescence), and induces cell death in T24 human transitional-cell bladder carcinoma cell line at physiologically achievable concentrations. DEA induces cell cycle arrest in the G0/G1 phase, which may contribute to the inhibition of cell proliferation, and shifts the Bax/Bcl-2 ratio to initiate apoptosis, induce AIF nuclear translocation, and activate PARP-1 cleavage and caspase-3 activation. The major cytoprotective kinases-ERK and Akt-are inhibited by DEA, which may contribute to its cell death-inducing effects. DEA also inhibits the expression of B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1) and reduces colony formation of T24 bladder carcinoma cells, indicating its possible inhibitory effect on metastatic potential. These data show that DEA is a novel anti-cancer candidate of multiple cell death-inducing effects and metastatic potential. Our findings recommend further evaluation of its effects in clinical studies.


Asunto(s)
Amiodarona/análogos & derivados , Apoptosis/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/patología , Amiodarona/farmacología , Línea Celular Tumoral , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos
5.
Free Radic Biol Med ; 108: 770-784, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28457938

RESUMEN

Oxidative stress induces DNA breaks and PARP-1 activation which initiates mitochondrial reactive oxygen species (ROS) production and cell death through pathways not yet identified. Here, we show the mechanism by which PARP-1 influences these processes via PARylation of activating transcription factor-4 (ATF4) responsible for MAP kinase phosphatase-1 (MKP-1) expression and thereby regulates MAP kinases. PARP inhibitor, or silencing, of PARP induced MKP-1 expression by ATF4-dependent way, and inactivated JNK and p38 MAP kinases. Additionally, it induced ATF4 expression and binding to cAMP-response element (CRE) leading to MKP-1 expression and the inactivation of MAP kinases. In contrast, PARP-1 activation induced the PARylation of ATF4 and reduced its binding to CRE sequence in vitro. CHIP-qPCR analysis showed that PARP inhibitor increased the ATF4 occupancy at the initiation site of MKP-1. In oxidative stress, PARP inhibition reduced ROS-induced cell death, suppressed mitochondrial ROS production and protected mitochondrial membrane potential on an ATF4 and MKP-1 dependent way. Basically identical results were obtained in WRL-68, A-549 and T24/83 human cell lines indicating that the aforementioned mechanism can be universal. Here, we provide the first description of PARP-1-ATF4-MKP-1-JNK/p38 MAPK retrograde pathway, which is responsible for the regulation of mitochondrial integrity, ROS production and cell death in oxidative stress, and may represent a new mechanism of PARP in cancer therapy since cancer stem cells development is JNK-dependent.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Carcinogénesis/metabolismo , Fosfatasa 1 de Especificidad Dual/metabolismo , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción Activador 4/genética , Carcinogénesis/genética , Muerte Celular , Línea Celular Tumoral , AMP Cíclico/metabolismo , Daño del ADN , Fosfatasa 1 de Especificidad Dual/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Humanos , Mitocondrias/patología , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasa-1/genética , ARN Interferente Pequeño/genética , Elementos de Respuesta/genética , Transducción de Señal
6.
PLoS One ; 12(1): e0169372, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28046125

RESUMEN

Reactive oxygen species (ROS) play a critical role in the progression of mitochondria-related diseases. A novel insulin sensitizer drug candidate, BGP-15, has been shown to have protective effects in several oxidative stress-related diseases in animal and human studies. In this study, we investigated whether the protective effects of BGP-15 are predominantly via preserving mitochondrial integrity and reducing mitochondrial ROS production. BGP-15 was found to accumulate in the mitochondria, protect against ROS-induced mitochondrial depolarization and attenuate ROS-induced mitochondrial ROS production in a cell culture model, and also reduced ROS production predominantly at the complex I-III system in isolated mitochondria. At physiologically relevant concentrations, BGP-15 protected against hydrogen peroxide-induced cell death by reducing both apoptosis and necrosis. Additionally, it attenuated bacterial lipopolysaccharide (LPS)-induced collapse of mitochondrial membrane potential and ROS production in LPS-sensitive U-251 glioma cells, suggesting that BGP-15 may have a protective role in inflammatory diseases. However, BGP-15 did not have any antioxidant effects as shown by in vitro chemical and cell culture systems. These data suggest that BGP-15 could be a novel mitochondrial drug candidate for the prevention of ROS-related and inflammatory disease progression.


Asunto(s)
Citoprotección/efectos de los fármacos , Lipopolisacáridos/farmacología , Mitocondrias/química , Estrés Oxidativo/efectos de los fármacos , Oximas/farmacología , Piperidinas/farmacología , Animales , Bencimidazoles/metabolismo , Carbocianinas/metabolismo , Muerte Celular/efectos de los fármacos , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oxidación-Reducción/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo
7.
J Cancer ; 6(5): 470-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25874011

RESUMEN

Previously, we found that the unconventional small human heat-shock protein HSPB11 inhibits cell death by HSP90 mediated cholesterol-rich membrane microdomain dependent activation of phosphatidylinositol-3 kinase/protein kinase B pathway and by stabilising the mitochondrial membrane systems. Also, progressive cytoplasmic expression of HSPB11 correlated with brain tumor malignancy. In the present study we investigated how cytoplasmic abundance of HSPB11 augments tumor malignancy. We up- and downregulated the cytoplasmic level of HSPB11 before paclitaxel exposure in NIH3T3 and HeLa cells, which normally express low and high levels, respectively, of the HSPB11 protein. We examined the paclitaxel-mediated induction of cell death, mitochondrial fission, HSPB11 mitochondrial translocation and inhibitory phosphorylation of dynamin-like protein-1 (DLP1). We found that increasing cytoplasmic abundance of HSPB11 in NIH3T3 cells protected against paclitaxel-induced apoptosis, while suppressing HSPB11 in HeLa cells sensitised the cells toward paclitaxel. Also, paclitaxel enhanced mitochondrial translocation of HSPB11 in wild type HeLa but not in NIH3T3 cells. More importantly, increased cytoplasmic level of HSPB11 in NIH3T3 cells enhanced the inhibitory phosphorylation of DLP1 and attenuated paclitaxel-induced mitochondrial fission. All these results suggest that increased cytoplasmic abundance of HSPB11 augments inhibitory phosphorylation of DLP1 thereby reduces mitochondrial fission that eventually leads to decreased apoptosis. This novel mechanism may explain the resistance to apoptosis and increased malignancy of HSPB11-overexpressing tumours. The clinical significance of this mechanism has already been highlighted by the finding that the kinase inhibitor tyrphostin A9 induces cancer cell death by DLP1-mediated mitochondrial fragmentation.

8.
Int J Radiat Biol ; 90(12): 1152-61, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24937370

RESUMEN

UNLABELLED: Abstract Purpose: Sensitizing cancer cells to irradiation is a major challenge in clinical oncology. We aimed to define the signal transduction pathways involved in poly(ADP-ribose) polymerase (PARP) inhibitor-induced radiosensitization in various mammalian cancer lines. MATERIALS AND METHODS: Clonogenic survival assays and Western blot examinations were performed following telecobalt irradiation of cancer cells in the presence or absence of various combinations of PARP- and selective mitogen-activated protein kinase (MAPK) inhibitors. RESULTS: HO3089 resulted in significant cytotoxicity when combined with irradiation. In human U251 glioblastoma and A549 lung cancer cell lines, Erk1/2 and JNK/SAPK were found to mediate this effect of HO3089 since inhibitors of these kinases ameliorated it. In murine 4T1 breast cancer cell line, p38 MAPK rather than Erk1/2 or JNK/SAPK was identified as the main mediator of HO3089's radiosensitizing effect. Besides the aforementioned changes in kinase signaling, we detected increased p53, unchanged Bax and decreased Bcl-2 expression in the A549 cell line. CONCLUSIONS: HO3089 sensitizes cancer cells to photon irradiation via proapoptotic processes where p53 plays a crucial role. Activation of MAPK pathways is regarded the consequence of irradiation-induced DNA damage, thus their inhibition can counteract the radiosenzitizing effect of the PARP inhibitor.


Asunto(s)
Bencimidazoles/farmacología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Inhibidores de Proteínas Quinasas/farmacología , Tolerancia a Radiación/efectos de los fármacos , Animales , Línea Celular Tumoral , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
9.
Nutr Cancer ; 65(7): 1059-66, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24032376

RESUMEN

The goal of the present study was to compare the efficacy of treatment with irradiation (IR), temozolomide, and quercetin, alone, or in combinations, on 2 glioblastoma cell lines, DBTRG-05 and U-251. Cell viability assay, flow cytometry analysis, colony formation assay, and Western blot analysis were used to compare the effects of treatment on the 2 cell lines. The greatest reduction in cell viability and colony formation was observed when cells were treated with a combination of the agents including quercetin. The treatment of cells with the combination of IR and quercetin was equal to the efficiency of the combination of IR and temozolomide in decreasing cell viability as well as colony formation. Quercetin alone, or in combination with IR, increased the cleavage of caspase-3 and PARP-1 showing an activated apoptosis and significantly reduced the level of phospho-Akt. Moreover, these treatments increased the levels of phospho-ERK, phospho-JNK, phospho-p38, and phospho-RAF1. Our data indicate that the supplementation of standard therapy with quercetin increases efficacy of treatment of experimental glioblastoma through synergism in the induction of apoptosis via the cleavage of caspase-3 and PARP-1 and by the suppression of the actitivation of Akt pathway.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quercetina/farmacología , Transducción de Señal , Apoptosis/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimioradioterapia , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Humanos , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Temozolomida
10.
Int J Oncol ; 39(4): 1025-32, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21701777

RESUMEN

Growth hormone releasing hormone (GHRH) antagonists have been developed for the treatment of various cancers. We investigated the effects of a novel GHRH antagonist, MIA-602, on nine breast cancer cell lines, differing in their expression for estrogen-, progesterone- and HER-2 receptors. We detected the presence of pituitary-type GHRH receptors (pGHRH-R) on 6 of the 9 breast cancer cell lines. The main splice variant of pGHRH-R, SV1, was found on all 9 cell lines. MTT assay showed that following treatment with MIA-602, cell viability decreased significantly in all 9 cell lines. The reduction in cell viability was greater in cells positive for both pGHRH-R and SV1, than in cells positive for only SV1, but the difference was not significant. Using Western blotting, we demonstrated that the levels of phospho-Akt, -GSK3ß and -ERK1/2 decreased significantly following exposure to MIA-602 and the level of phospho-p38 increased after treatment. The reduction of the phosphorylated anti-apoptotic proteins was significantly greater in cells where both pGHRH-R and SV1 were present, than where only SV1 was expressed. In conclusion, our study shows that MIA-602 is effective against a wide range of breast cancer cells in vitro, independently of their receptor positivity, suggesting the potential use of GHRH antagonists also in the treatment of triple-negative breast cancer. The effect of MIA-602 was mediated nearly as well in tumors that expressed only the SV1 receptor compared to those in which both SV1 and pGHRH-R were present, although a difference could be detected at the level of cell signaling.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Hormona Liberadora de Hormona del Crecimiento/antagonistas & inhibidores , Sermorelina/análogos & derivados , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Receptor ErbB-2/biosíntesis , Receptores de Estrógenos/biosíntesis , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Receptores de Progesterona/biosíntesis , Sermorelina/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Eur J Cell Biol ; 89(11): 853-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20708296

RESUMEN

Tail-interacting protein (TIP47, also named PP17) has been implicated in lipid droplet metabolism and in the development of late endosomes, to date however, no data about its possible role in regulating cell death processes has been available. Here, we provide evidence for the role of TIP47 in the regulation of mitochondrial membrane stability and cell death. Overexpression of TIP47 protected NIH3T3 cells from taxol-induced cell death, while suppression of TIP47 by siRNA facilitated cell death. TIP47, but not its truncated form, t-TIP47, decreased taxol-induced cell death as determined by propidium iodide and fluorescent Annexin V staining. Recombinant TIP47, but not t-TIP47, partially prevented taxol-induced depolarization of mitochondria in vitro. Overexpression of TIP47, but not its truncated form, prevented the taxol-induced nuclear and cytoplasmic translocation of AIF and Endonuclease G, as well as the taxol-induced depolarization of mitochondria in NIH3T3 cells. Furthermore, overexpression of TIP47 facilitated Bcl-2 expression and suppressed Bax expression in taxol-treated cells. These data show that besides its previously known functions, TIP47 is involved in the regulation of mitochondria-related cell death by directly stabilizing the mitochondrial membrane system and by favorably affecting the expression of Bcl-2 homologues. Since TIP47 is overexpressed in certain tumors, it is possible that TIP47 contributes to the development of cytostatic resistance.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Paclitaxel/farmacología , Proteínas Gestacionales/metabolismo , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Perilipina-3 , Proteínas Gestacionales/biosíntesis , Proteínas Gestacionales/genética , ARN Interferente Pequeño/genética , Conejos , Transducción de Señal , Transfección , Proteínas de Transporte Vesicular
12.
J Biol Chem ; 285(3): 2140-51, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19901022

RESUMEN

We identified a sequence homologous to the Bcl-2 homology 3 (BH3) domain of Bcl-2 proteins in SOUL. Tissues expressed the protein to different extents. It was predominantly located in the cytoplasm, although a fraction of SOUL was associated with the mitochondria that increased upon oxidative stress. Recombinant SOUL protein facilitated mitochondrial permeability transition and collapse of mitochondrial membrane potential (MMP) and facilitated the release of proapoptotic mitochondrial intermembrane proteins (PMIP) at low calcium and phosphate concentrations in a cyclosporine A-dependent manner in vitro in isolated mitochondria. Suppression of endogenous SOUL by diced small interfering RNA in HeLa cells increased their viability in oxidative stress. Overexpression of SOUL in NIH3T3 cells promoted hydrogen peroxide-induced cell death and stimulated the release of PMIP but did not enhance caspase-3 activation. Despite the release of PMIP, SOUL facilitated predominantly necrotic cell death, as revealed by annexin V and propidium iodide staining. This necrotic death could be the result of SOUL-facilitated collapse of MMP demonstrated by JC-1 fluorescence. Deletion of the putative BH3 domain sequence prevented all of these effects of SOUL. Suppression of cyclophilin D prevented these effects too, indicating that SOUL facilitated mitochondrial permeability transition in vivo. Overexpression of Bcl-2 and Bcl-x(L), which can counteract the mitochondria-permeabilizing effect of BH3 domain proteins, also prevented SOUL-facilitated collapse of MMP and cell death. These data indicate that SOUL can be a novel member of the BH3 domain-only proteins that cannot induce cell death alone but can facilitate both outer and inner mitochondrial membrane permeabilization and predominantly necrotic cell death in oxidative stress.


Asunto(s)
Permeabilidad de la Membrana Celular , Hemoproteínas/química , Hemoproteínas/metabolismo , Membranas Mitocondriales/metabolismo , Estrés Oxidativo , Proteínas Gestacionales/química , Proteínas Gestacionales/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/química , Homología de Secuencia de Aminoácido , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bovinos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular , Peptidil-Prolil Isomerasa F , Ciclofilinas/farmacología , Regulación de la Expresión Génica , Células HeLa , Proteínas de Unión al Hemo , Hemoproteínas/deficiencia , Hemoproteínas/genética , Humanos , Peróxido de Hidrógeno/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Membranas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Células 3T3 NIH , Estrés Oxidativo/efectos de los fármacos , Proteínas Gestacionales/deficiencia , Proteínas Gestacionales/genética , Estructura Terciaria de Proteína , ARN Interferente Pequeño/genética , Ratas , Eliminación de Secuencia
13.
Neurotox Res ; 16(1): 68-76, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19526300

RESUMEN

Poly(ADP-ribose) polymerase (PARP) activation is considered as a major regulator of cell death in various pathophysiological conditions, however, no direct information is available about its role in chronic hypoperfusion-induced neuronal death. Here, we provide evidence for the protective effect of PARP inhibition on degenerative retinal damage induced by bilateral common carotid artery occlusion (BCCAO), an adequate chronic hypoperfusion murine model. We found that BCCAO in adult male Wistar rats led to severe degeneration of all retinal layers that was attenuated by a carboxaminobenzimidazol-derivative PARP inhibitor (HO3089) administered unilaterally into the vitreous body immediately following carotid occlusion and then 4 times in a 2-week-period. Normal morphological structure of the retina was preserved and the thickness of the retinal layers was increased in HO3089-treated eyes compared to the BCCAO eyes. For Western blot studies, HO3089 was administered immediately after BCCAO and retinas were removed 4 h later. According to Western blot analysis utilizing phosphorylation-specific primary antibodies, besides activating poly-ADP-ribose (PAR) synthesis, BCCAO induced phosphorylation of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). HO3089 inhibited PAR synthesis, and decreased the phosphorylation of these proapoptotic MAPKs. In addition, HO3089 treatment induced phosphorylation, that is activation, of the protective Akt/glycogen synthase kinase (GSK)-3beta and extracellular signal-regulated kinase (ERK1/2) signaling pathways. These data indicate that PARP activation has a major role in mediating chronic hypoperfusion-induced neuronal death, and inhibition of the enzyme prevents the pathological changes both in the morphology and the kinase signaling cascades involved. These results identify PARP inhibition as a possible molecular target in the clinical management of chronic hypoperfusion-induced neurodegenerative diseases including ocular ischemic syndrome.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Degeneración Retiniana/etiología , Degeneración Retiniana/prevención & control , Transducción de Señal/efectos de los fármacos , Animales , Bencimidazoles/toxicidad , Enfermedades de las Arterias Carótidas/complicaciones , MAP Quinasa Quinasa 4/metabolismo , Masculino , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Anticancer Res ; 29(2): 717-24, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19331227

RESUMEN

UNLABELLED: The aim of this study was to find a possible clinical use of the tail-interacting protein of 47 kDa (TIP47) and further document its expression in smear cytology, different cervical dysplasias, invasive cervical cancer and metastasis. PATIENTS AND METHODS: A new polyclonal anti-TIP47 antibody was developed and used on smears and histological cervix sections of sixty women with different cytological pathologies. Serum TIP47 level of patients with cervical intraepithelial neoplasia (CIN) or carcinoma in stage IIb, IIIa, and IIIb was monitored during treatment. RESULTS: TIP47 was expressed weakly in the dysplasias, stronger in invasive tumors and in lymph node metastasis. In patients with cervical carcinoma, the serum TIP47 level was found to be elevated; it decreased after therapy and elevated again in relapse. CONCLUSION: According to our results, TIP47 could be a good clinical marker for the early detection in blood of the recurrence of cervical carcinoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Gestacionales/metabolismo , Displasia del Cuello del Útero/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Animales , Biomarcadores de Tumor/sangre , Western Blotting , Línea Celular Tumoral , Citoplasma/metabolismo , Proteínas de Unión al ADN/sangre , Femenino , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/sangre , Ratones , Células 3T3 NIH , Perilipina-3 , Proteínas Gestacionales/sangre , Displasia del Cuello del Útero/sangre , Displasia del Cuello del Útero/patología , Neoplasias del Cuello Uterino/sangre , Neoplasias del Cuello Uterino/patología , Frotis Vaginal , Proteínas de Transporte Vesicular
15.
Eur J Cell Biol ; 86(3): 161-71, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17275951

RESUMEN

NCBI database analysis indicated that the human C1orf41 protein (small heat shock-like protein-Hsp16.2) has sequence similarity with small heat shock proteins (sHsps). Since sHsps have chaperone function, and so prevent aggregation of denatured proteins, we determined whether Hsp16.2 could prevent the heat-induced aggregation of denatured proteins. Under our experimental conditions, recombinant Hsp16.2 prevented aggregation of aldolase and glyceraldehyde-3-phosphate dehydrogenase, and protected Escherichia coli cells from heat stress indicating its chaperone function. Hsp16.2 also formed oligomeric complexes in aqueous solution. Hsp16.2 was found to be expressed at different levels in cell lines and tissues, and was mainly localized to the nucleus and the cytosol, but to a smaller extent, it could be also found in mitochondria. Hsp16.2 could be modified covalently by poly(ADP ribosylation) and acetylation. Hsp16.2 over-expression prevented etoposide-induced cell death as well as the release of mitochondrial cytochrome c and caspase activation. These data suggest that Hsp16.2 can prevent the destabilization of mitochondrial membrane systems and could represent a suitable target for modulating cell death pathways.


Asunto(s)
Apoptosis , Proteínas de Choque Térmico Pequeñas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Etopósido/farmacología , Expresión Génica/efectos de los fármacos , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Células HeLa , Proteínas de Choque Térmico Pequeñas/química , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estructura Cuaternaria de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Alineación de Secuencia , Homología de Secuencia , Temperatura
16.
Apoptosis ; 12(1): 97-112, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17136496

RESUMEN

AlphaB-crystallin homology, heat stress induction and chaperone activity suggested that a previously encloned gene product is a novel small heat shock protein (Hsp16.2). Suppression of Hsp16.2 by siRNA sensitized cells to hydrogen peroxide or taxol induced cell-death. Over-expressing of Hsp16.2 protected cells against stress stimuli by inhibiting cytochrome c release from the mitochondria, nuclear translocation of AIF and endonuclease G, and caspase 3 activation. Recombinant Hsp16.2 protected mitochondrial membrane potential against calcium induced collapse in vitro indicating that Hsp16.2 stabilizes mitochondrial membrane systems. Hsp16.2 formed self-aggregates and bound to Hsp90. Inhibition of Hsp90 by geldanamycin diminished the cytoprotective effect of Hsp16.2 indicating that this effect was Hsp90-mediated. Hsp16.2 over-expression increased lipid rafts formation as demonstrated by increased cell surface labeling with fluorescent cholera toxin B, and increased Akt phosphorylation. The inhibition of PI-3-kinase-Akt pathway by LY-294002 or wortmannin significantly decreased the protective effect of the Hsp16.2. These data indicate that the over-expression of Hsp16.2 inhibits cell death via the stabilization of mitochondrial membrane system, activation of Hsp90, stabilization of lipid rafts and by the activation of PI-3-kinase-Akt cytoprotective pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/farmacología , Proteínas de Choque Térmico/fisiología , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Células HeLa , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/genética , Humanos , Técnicas In Vitro , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Homología de Secuencia de Aminoácido , Cadena B de alfa-Cristalina/genética
17.
FEBS Lett ; 580(27): 6447-54, 2006 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-17098234

RESUMEN

We found that heme-binding protein 2/SOUL sensitised NIH3T3 cells to cell death induced by A23187 and etoposide, but it did not affect reactive oxygen species formation. In the presence of sub-threshold calcium, recombinant SOUL provoked mitochondrial permeability transition (mPT) in vitro that was inhibited by cyclosporine A (CsA). This effect was verified in vivo by monitoring the dissipation of mitochondrial membrane potential. Flow cytometry analysis showed that SOUL promoted necrotic death in A23187 and etoposide treated cells, which effect was prevented by CsA. These data suggest that besides its heme-binding properties SOUL promotes necrotic cell death by inducing mPT.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/farmacología , Hemoproteínas/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Gestacionales/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Calcimicina/farmacología , Proteínas Portadoras/genética , Ciclosporina/farmacología , Etopósido/farmacología , Proteínas de Unión al Hemo , Hemoproteínas/genética , Humanos , Inmunosupresores/farmacología , Ionóforos/farmacología , Ratones , Células 3T3 NIH , Necrosis/metabolismo , Permeabilidad/efectos de los fármacos , Proteínas Gestacionales/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
18.
Virus Res ; 113(2): 73-80, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15936106

RESUMEN

Application of improved molecular techniques in the detection and characterization of rotavirus strains has led to the recent description of several new combinations, specificities, and genetic variants of the outer capsid genes, VP7 and VP4. In spite of the enormous diversity of mammalian rotavirus strains, the few lapine rotaviruses characterized to date, appear to carry a narrow range of such antigen combinations; only P[14], G3 and, based on a more recent study, P[22], G3 rotaviruses have proved to be epidemiologically important in rabbits. In the present study, we characterized a lapine group A rotavirus with a super-short electropherotype detected in an outbreak of fatal enteritis in a Hungarian commercial rabbitry. Based on sequence and phylogenetic analysis of the VP7, VP4, and NSP4 genes, our lapine strain is a P[22], G3 rotavirus that carries the NSP4 genotype shared by most lapine rotaviruses. Although the P[22] VP4 specificity has been newly identified, the relatively high sequence variation between our strain and those identified in Italy (89.1-90.4% nucleotide identity; region VP8*) implies that these strains diversified far before they were described for the first time, strongly suggesting that this genotype may have circulated in rabbitries or in nature without prior detection. We conclude that genotype P[22] lapine rotaviruses show a wider geographical dispersal than previously thought, although understanding their true epidemiological significance needs further investigation.


Asunto(s)
Enteritis/veterinaria , Infecciones por Rotavirus/veterinaria , Rotavirus/genética , Crianza de Animales Domésticos , Animales , Proteínas de la Cápside/genética , Brotes de Enfermedades , Enteritis/epidemiología , Genes Virales , Glicoproteínas/genética , Hungría/epidemiología , Filogenia , Conejos , Infecciones por Rotavirus/epidemiología , Homología de Secuencia de Ácido Nucleico , Toxinas Biológicas/genética , Proteínas no Estructurales Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA