Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Lipid Res ; 53(6): 1134-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22454477

RESUMEN

Normally, cell proliferation and death are carefully balanced in higher eukaryotes, but one of the most important regulatory mechanisms, apoptosis, is upset in many malignancies, including hepatocellular-derived ones. Therefore, reinforcing cell death often is mandatory in anticancer therapy. We previously reported that a combination of tumor necrosis factor-α (TNF) and cycloheximide (CHX) efficiently kill HTC cells, a rat hepatoma line, in an apoptosis-like mode. Death is actively mediated by the lysosomal compartment, although lysosomal ceramide was previously shown not to be directly implicated in this process. In the present study, we show that TNF/CHX increase lysosomal ceramide that is subsequently converted into sphingosine. Although ceramide accumulation does not significantly alter the acidic compartment, the sphingosine therein generated causes lysosomal membrane permeabilization (LMP) followed by relocation of lysosomal cathepsins to the cytoplasm. TNF/CHX-induced LMP is effectively abrogated by siRNAs targeting acid sphingomyelinase or acid ceramidase, which prevent both LMP and death induced by TNF/CHX. Taken together, our results demonstrate that lysosomal accumulation of ceramide is not detrimental per se, whereas its degradation product sphingosine, which has the capacity to induce LMP, appears responsible for the observed apoptotic-like death.


Asunto(s)
Carcinoma Hepatocelular/patología , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Lisosomas/metabolismo , Esfingosina/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Ceramidasas/deficiencia , Ceramidasas/genética , Ceramidas/biosíntesis , Ceramidas/metabolismo , Cicloheximida/farmacología , Silenciador del Gen , Permeabilidad/efectos de los fármacos , ARN Interferente Pequeño/genética , Ratas , Receptores del Factor de Necrosis Tumoral/metabolismo , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Esfingosina/biosíntesis , Esfingosina/metabolismo
2.
Biochim Biophys Acta ; 1793(7): 1182-90, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19328214

RESUMEN

We previously showed that, in the rat hepatoma cell line HTC, TNF brings about a non-caspase-dependent, apoptosis-like process requiring NADPH oxidase activity, an iron-mediated pro-oxidant status, and a functional acidic vacuolar compartment. This process may thus involve mechanisms such as autophagy or relocation of lysosomal enzymes, perhaps secondary to the formation of ceramide by acidic sphingomyelinase. Here we investigated whether ceramide formation contributes to the apoptogenic process. HTC cells were found to be sensitive to exogenous ceramide and significantly protected against TNF by desipramine, an inhibitor of lysosomal acid sphingomyelinase. However, Bcl-2 transfection and Bcl-x(L) upregulation by dexamethasone significantly diminished the apoptogenic effect of ceramide but not that of TNF, suggesting that ceramide is not directly involved in TNF toxicity. Moreover, Bcl-x(L) silencing precluded dexamethasone-induced protection against ceramide and, by itself, induced massive death, demonstrating the strict dependence of HTC cells on Bcl-x(L) for survival also under standard culture conditions.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Ceramidas/toxicidad , Neoplasias Hepáticas Experimentales/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Dexametasona/farmacología , Glucocorticoides/farmacología , Neoplasias Hepáticas Experimentales/patología , Lisosomas , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Ratas , Células Tumorales Cultivadas , Proteína bcl-X/antagonistas & inhibidores , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
3.
Free Radic Biol Med ; 141: 205-219, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31207288

RESUMEN

The transcriptional regulator YAP plays an important role in cancer progression and is negatively controlled by the Hippo pathway. YAP is frequently overexpressed in human cancers, including bladder cancer. Interestingly, YAP expression and activity can be inhibited by pro-oxidant conditions; moreover, YAP itself can also affect the cellular redox status through multiple mechanisms. 4-Hydroxynonenal (HNE), the most intensively studied end product of lipid peroxidation, is a pro-oxidant agent able to deplete GSH and has an anti-tumoral effect by affecting multiple signal pathways, including the down-regulation of oncogene expressions. These observations prompted us to investigate the effect of HNE on YAP expression and activity. We demonstrated that HNE inhibited YAP expression and its target genes in bladder cancer cells through a redox-dependent mechanism. Moreover, the YAP down-regulation was accompanied by an inhibition of proliferation, migration, invasion, and angiogenesis, as well as by an accumulation of cells in the G2/M phase of cell cycle and by an induction of apoptosis. We also established the YAP role in inhibiting cell viability and inducing apoptosis in HNE-treated cells by using an expression vector for YAP. Furthermore, we identified a post-translational mechanism for the HNE-induced YAP expression inhibition, involving an increase of YAP phosphorylation and ubiquitination, leading to proteasomal degradation. Our data established that HNE can post-translationally down-regulate YAP through a redox-dependent mechanism and that this modulation can contribute to determining the specific anti-cancer effects of HNE.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aldehídos/farmacología , Regulación Neoplásica de la Expresión Génica , Procesamiento Proteico-Postraduccional , Factores de Transcripción/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Invasividad Neoplásica , Neovascularización Patológica , Oncogenes , Oxidación-Reducción , Fosfoproteínas/metabolismo , Transducción de Señal , Neoplasias de la Vejiga Urinaria/genética , Proteínas Señalizadoras YAP
4.
Free Radic Biol Med ; 115: 447-457, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29248722

RESUMEN

Redox adaptation plays an important role in cancer cells drug resistance. The antioxidant response is principally mediated by the transcription factor Nrf2, that induces the transcriptional activation of several genes involved in GSH synthesis, chemoresistance, and cytoprotection. YAP is emerging as a key mediator of chemoresistance in a variety of cancers, but its role in controlling the antioxidant status of the cells is yet elusive. Here, we show that impairing YAP protein expression reduced GSH content and Nrf2 protein and mRNA expression in bladder cancer cells. Moreover, in YAP knocked down cells the expression of FOXM1, a transcription factor involved in Nrf2 transcription, was down-regulated and the silencing of FOXM1 reduced Nrf2 expression. On the other hand, the silencing of Nrf2, as well as the depletion of GSH by BSO treatment, inhibited YAP expression, suggesting that cross-talk exists between YAP and Nrf2 proteins. Importantly, we found that silencing either YAP or Nrf2 enhanced sensitivity of bladder cancer cells to cytotoxic agents and reduced their migration. Furthermore, the inhibition of both YAP and Nrf2 expressions significantly increased cytotoxic drug sensitivity and synergistically reduced the migration of chemoresistant bladder cancer cells. These findings provide a rationale for targeting these transcriptional regulators in patients with chemoresistant bladder cancer, expressing high YAP and bearing a proficient antioxidant system.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/farmacología , Cisplatino/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fosfoproteínas/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/genética , Antioxidantes/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 2 Relacionado con NF-E2/genética , Oxidación-Reducción , Estrés Oxidativo , Fosfoproteínas/genética , ARN Interferente Pequeño/genética , Receptor Cross-Talk , Factores de Transcripción , Neoplasias de la Vejiga Urinaria/genética , Proteínas Señalizadoras YAP
5.
Free Radic Biol Med ; 97: 24-37, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27184956

RESUMEN

Several reports indicate that chemo-resistant cancer cells become highly adapted to intrinsic oxidative stress by up-regulating their antioxidant systems, which causes an increase of intracellular GSH content. Doxorubicin is one of the most widely used drugs for tumor treatment, able to kill cancer cells through several mechanisms. However, doxorubicin use is limited by its toxicity and cancer resistance. Therefore, new therapeutic strategies able to reduce doses and to overcome chemo-resistance are needed. A new class of glutathione-responsive cyclodextrin nanosponges (GSH-NS), is able to release anticancer drugs preferentially in cells having high GSH content. Doxorubicin-loaded GSH-NS, in the cancer cells with high GSH content, inhibited clonogenic growth, cell viability, topoisomerase II activity and induced DNA damage with higher effectiveness than free drug. Moreover, GSH-NS reduced the development of human tumor in xenograft models more than free drug. These characteristics indicate that GSH-NS can be a suitable drug delivery carrier for future applications in cancer therapy.


Asunto(s)
Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/genética , Neoplasias/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/química , Antioxidantes/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Glutatión/química , Glutatión/metabolismo , Humanos , Ratones , Nanoestructuras/administración & dosificación , Nanoestructuras/química , Neoplasias/metabolismo , Neoplasias/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Autophagy ; 11(12): 2184-98, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26566051

RESUMEN

Lysosomal membrane permeabilization (LMP) induced by oxidative stress has recently emerged as a prominent mechanism behind TNF cytotoxicity. This pathway relies on diffusion of hydrogen peroxide into lysosomes containing redox-active iron, accumulated by breakdown of iron-containing proteins and subcellular organelles. Upon oxidative lysosomal damage, LMP allows relocation to the cytoplasm of low mass iron and acidic hydrolases that contribute to DNA and mitochondrial damage, resulting in death by apoptosis or necrosis. Here we investigate the role of lysosomes and free iron in death of HTC cells, a rat hepatoma line, exposed to TNF following metallothionein (MT) upregulation. Iron-binding MT does not normally occur in HTC cells in significant amounts. Intracellular iron chelation attenuates TNF and cycloheximide (CHX)-induced LMP and cell death, demonstrating the critical role of this transition metal in mediating cytokine lethality. MT upregulation, combined with starvation-activated MT autophagy almost completely suppresses TNF and CHX toxicity, while impairment of both autophagy and MT upregulation by silencing of Atg7, and Mt1a and/or Mt2a, respectively, abrogates protection. Interestingly, MT upregulation by itself has little effect, while stimulated autophagy alone depresses cytokine toxicity to some degree. These results provide evidence that intralysosomal iron-catalyzed redox reactions play a key role in TNF and CHX-induced LMP and toxicity. The finding that chelation of intralysosomal iron achieved by autophagic delivery of MT, and to some degree probably of other iron-binding proteins as well, into the lysosomal compartment is highly protective provides a putative mechanism to explain autophagy-related suppression of death by TNF and CHX.


Asunto(s)
Autofagia/efectos de los fármacos , Hierro/metabolismo , Lisosomas/efectos de los fármacos , Metalotioneína/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Apoptosis/efectos de los fármacos , Autofagia/fisiología , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Neoplasias Hepáticas/metabolismo , Lisosomas/metabolismo , Necrosis/metabolismo , Ratas
7.
Antioxid Redox Signal ; 22(18): 1681-702, 2015 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-25365742

RESUMEN

SIGNIFICANCE: Oxidative stress provokes the peroxidation of polyunsaturated fatty acids in cellular membranes, leading to the formation of aldheydes that, due to their high chemical reactivity, are considered to act as second messengers of oxidative stress. Among the aldehydes formed during lipid peroxidation (LPO), 4-hydroxy-2-nonenal (HNE) is produced at a high level and easily reacts with both low-molecular-weight compounds and macromolecules, such as proteins and DNA. In particular, HNE-protein adducts have been extensively investigated in diseases characterized by the pathogenic contribution of oxidative stress, such as cancer, neurodegenerative, chronic inflammatory, and autoimmune diseases. RECENT ADVANCES: In this review, we describe and discuss recent insights regarding the role played by covalent adducts of HNE with proteins in the development and evolution of those among the earlier mentioned disease conditions in which the functional consequences of their formation have been characterized. CRITICAL ISSUES: Results obtained in recent years have shown that the generation of HNE-protein adducts can play important pathogenic roles in several diseases. However, in some cases, the generation of HNE-protein adducts can represent a contrast to the progression of disease or can promote adaptive cell responses, demonstrating that HNE is not only a toxic product of LPO but also a regulatory molecule that is involved in several biochemical pathways. FUTURE DIRECTIONS: In the next few years, the refinement of proteomical techniques, allowing the individuation of novel cellular targets of HNE, will lead to a better understanding the role of HNE in human diseases.


Asunto(s)
Aldehídos/metabolismo , Enfermedades Autoinmunes/metabolismo , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas/metabolismo , Aldehídos/química , Animales , Humanos , Inflamación/metabolismo , Peroxidación de Lípido , Redes y Vías Metabólicas , Estrés Oxidativo , Proteínas/química
8.
J Biomed Nanotechnol ; 11(12): 2169-85, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26510311

RESUMEN

4-hydroxynonenal (HNE), a lipid peroxidation product, is a promising anti-neoplastic drug due to its remarkable anti-cancer activities. However, this possibility has not been explored, because the delivery of HNE is very challenging as a result of its low solubility and its poor stability. This study intentionally designed a new type of lipid nanocapsules specifically for HNE delivery. They consist of a medium chain triglyceride liquid oil core surrounded by a polymer shell. A ß-cyclodextrin-poly(4-acryloylmorpholine) conjugate was selected as the shell component. HNE-loaded nanocapsules were about 350 nm in size with a negative surface charge. They were stable for two years when stored in suspensions at 4 degrees C. In vitro experiments showed that HNE was released from the nanocapsules at a considerable rate. Nanocapsule uptake into cells was evaluated using a fluorescent formulation that revealed rapid internalisation. Cytotoxicity studies demonstrated the safety of the formulation. Enhanced anti-tumoral activity against various cell lines, depending on increased HNE stability, was obtained by using HNE-loaded nanocapsules. In particular, we have demonstrated an increase in anti-proliferative, pro-apoptotic and differentiative activity in several tumour cell lines from different tissues. Moreover, we evaluated the effects of these new nanocapsules on a three-dimensional human reconstructed model of skin melanoma. Interestingly, the encouraging results obtained with topical administration on the epidermal surface could open new perspectives in melanoma treatments.


Asunto(s)
Aldehídos/química , Aldehídos/farmacología , Portadores de Fármacos/química , Lípidos/química , Melanoma/patología , Nanocápsulas/química , Acrilamidas/química , Transporte Biológico , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclodextrinas/química , Estabilidad de Medicamentos , Humanos , Morfolinas/química
9.
PLoS One ; 7(6): e40149, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761953

RESUMEN

PPARs are nuclear receptors activated by ligands. Activation of PPARγ leads to a reduction of adhesion and motility in some cancer models. PPARγ transcriptional activity can be negatively regulated by JNK-mediated phosphorylation. We postulated that the use of agents able to inhibit JNK activity could increase the effectiveness of PPARγ ligands. We analysed the effects of rosiglitazone (PPARγ ligand) and AS601245 (a selective JNK inhibitor) alone or in association on adhesion and migration of CaCo-2, HT29, and SW480 human colon cancer cells and investigated, through microarray analysis, the genes involved in these processes. Cell adhesion and migration was strongly inhibited by rosiglitazone and AS601245. Combined treatment with the two compounds resulted in a greater reduction of the adhesion and migration capacity. Affymetrix analysis in CaCo-2 cells revealed that some genes which were highly modulated by the combined treatment could be involved in these biological responses. Rosiglitazone, AS601245 and combined treatment down-regulated the expression of fibrinogen chains in all three cell lines. Moreover, rosiglitazone, alone or in association with AS601245, caused a decrease in the fibrinogen release. ARHGEF7/ß-PIX gene was highly down-regulated by combined treatment, and western blot analysis revealed that ß-PIX protein is down-modulated in CaCo-2, HT29 and SW480 cells, also. Transfection of cells with ß-PIX gene completely abrogated the inhibitory effect on cell migration, determined by rosiglitazone, AS601245 and combined treatment. Results demonstrated that ß-PIX protein is involved in the inhibition of cell migration and sustaining the positive interaction between PPARγ ligands and anti-inflammatory agents in humans.


Asunto(s)
Acetonitrilos/farmacología , Benzotiazoles/farmacología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/patología , Expresión Génica/efectos de los fármacos , Tiazolidinedionas/farmacología , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/genética , Fibrinógeno/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Rosiglitazona
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA