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











Base de datos
Intervalo de año de publicación
1.
Radiother Oncol ; 200: 110503, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39186982

RESUMEN

BACKGROUND AND PURPOSE: Radiotherapy (RT) is an integral treatment part for patients with head and neck squamous cell carcinoma (HNSCC), but radioresistance remains a major issue. Here, we use MitoTam, a mitochondrially targeted analogue of tamoxifen, which we aim to stimulate ferroptotic cell death with, and sensitize radioresistant cells to RT. MATERIALS AND METHODS: We assessed viability, reactive oxygen species (ROS) production, disruption of mitochondrial membrane potential, and lipid peroxidation in radiosensitive (UT-SCC-40) and radioresistant (UT-SCC-5) HNSCC cells following MitoTam treatment. To assess ferroptosis specificity, we used the ferroptosis inhibitor ferrostatin-1 (fer-1). Also, total antioxidant capacity and sensitivity to tert-butyl hydroperoxide were evaluated to assess ROS-responses. 53BP1 staining was used to assess radiosensitivity after MitoTam treatment. RESULTS: Our data revealed increased ROS, cell death, disruption of mitochondrial membrane potential, and lipid peroxidation following MitoTam treatment in both cell lines. Adverse effects of MitoTam on cell death, membrane potential and lipid peroxidation were prevented by fer-1, indicating induction of ferroptosis. Radioresistant HNSCC cells were less sensitive to the effects of MitoTam due to intrinsic higher antioxidant capacity. MitoTam treatment prior to RT led to superadditive residual DNA damage expressed by 53BP1 foci compared to RT or MitoTam alone. CONCLUSION: MitoTam induced ferroptosis in HNSCC cells, which could be used to overcome the elevated antioxidant capacity of radioresistant cells and sensitize such cells to RT. Treatment with MitoTam followed by RT could therefore present a promising effective therapy of radioresistant cancers. STATEMENT OF SIGNIFICANCE: Radiotherapy is applied in the treatment of a majority of cancer patients. Radioresistance due to elevated antioxidant levels can be overcome by promoting ferroptotic cell death combining ROS-inducing drug MitoTam with radiotherapy.


Asunto(s)
Ferroptosis , Neoplasias de Cabeza y Cuello , Peroxidación de Lípido , Tolerancia a Radiación , Especies Reactivas de Oxígeno , Humanos , Ferroptosis/efectos de los fármacos , Neoplasias de Cabeza y Cuello/radioterapia , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Tolerancia a Radiación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Línea Celular Tumoral , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Tamoxifeno/farmacología
2.
Oncogene ; 37(7): 963-970, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29059169

RESUMEN

The anti-diabetic biguanide metformin may exert health-promoting effects via metabolic regulation of the epigenome. Here we show that metformin promotes global DNA methylation in non-cancerous, cancer-prone and metastatic cancer cells by decreasing S-adenosylhomocysteine (SAH), a strong feedback inhibitor of S-adenosylmethionine (SAM)-dependent DNA methyltransferases, while promoting the accumulation of SAM, the universal methyl donor for cellular methylation. Using metformin and a mitochondria/complex I (mCI)-targeted analog of metformin (norMitoMet) in experimental pairs of wild-type and AMP-activated protein kinase (AMPK)-, serine hydroxymethyltransferase 2 (SHMT2)- and mCI-null cells, we provide evidence that metformin increases the SAM:SAH ratio-related methylation capacity by targeting the coupling between serine mitochondrial one-carbon flux and CI activity. By increasing the contribution of one-carbon units to the SAM from folate stores while decreasing SAH in response to AMPK-sensed energetic crisis, metformin can operate as a metabolo-epigenetic regulator capable of reprogramming one of the key conduits linking cellular metabolism to the DNA methylation machinery.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carbono/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Metilación de ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano , Metformina/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Biomarcadores de Tumor , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Complejo I de Transporte de Electrón/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Hipoglucemiantes/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Células Tumorales Cultivadas
3.
BMC Cancer ; 16: 309, 2016 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-27175805

RESUMEN

BACKGROUND: Pancreatic cancer is recognized as one of the most fatal tumors due to its aggressiveness and resistance to therapy. Statins were previously shown to inhibit the proliferation of cancer cells via various signaling pathways. In healthy tissues, statins activate the heme oxygenase pathway, nevertheless the role of heme oxygenase in pancreatic cancer is still controversial. The aim of this study was to evaluate, whether anti-proliferative effects of statins in pancreatic cancer cells are mediated via the heme oxygenase pathway. METHODS: In vitro effects of various statins and hemin, a heme oxygenase inducer, on cell proliferation were evaluated in PA-TU-8902, MiaPaCa-2 and BxPC-3 human pancreatic cancer cell lines. The effect of statins on heme oxygenase activity was assessed and heme oxygenase-silenced cells were used for pancreatic cancer cell proliferation studies. Cell death rate and reactive oxygen species production were measured in PA-TU-8902 cells, followed by evaluation of the effect of cerivastatin on GFP-K-Ras trafficking and expression of markers of invasiveness, osteopontin (SPP1) and SOX2. RESULTS: While simvastatin and cerivastatin displayed major anti-proliferative properties in all cell lines tested, pravastatin did not affect the cell growth at all. Strong anti-proliferative effect was observed also for hemin. Co-treatment of cerivastatin and hemin increased anti-proliferative potential of these agents, via increased production of reactive oxygen species and cell death compared to individual treatment. Heme oxygenase silencing did not prevent pancreatic cancer cells from the tumor-suppressive effect of cerivastatin or hemin. Cerivastatin, but not pravastatin, protected Ras protein from trafficking to the cell membrane and significantly reduced expressions of SPP1 (p < 0.05) and SOX2 (p < 0.01). CONCLUSIONS: Anti-proliferative effects of statins and hemin on human pancreatic cancer cell lines do not seem to be related to the heme oxygenase pathway. While hemin triggers reactive oxygen species-induced cell death, cerivastatin targets Ras protein trafficking and affects markers of invasiveness.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Humanos , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas , Proteínas ras/genética , Proteínas ras/metabolismo
4.
PLoS One ; 11(4): e0154544, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27123847

RESUMEN

Parkinson's disease is a complex age-related neurodegenerative disorder. Approximately 90% of Parkinson's disease cases are idiopathic, of unknown origin. The aetiology of Parkinson's disease is not fully understood but increasing evidence implies a failure in fundamental cellular processes including mitochondrial dysfunction and increased oxidative stress. To dissect the cellular events underlying idiopathic Parkinson's disease, we use primary cell lines established from the olfactory mucosa of Parkinson's disease patients. Previous metabolic and transcriptomic analyses identified deficiencies in stress response pathways in patient-derived cell lines. The aim of this study was to investigate whether these deficiencies manifested as increased susceptibility, as measured by cell viability, to a range of extrinsic stressors. We identified that patient-derived cells are more sensitive to mitochondrial complex I inhibition and hydrogen peroxide induced oxidative stress, than controls. Exposure to low levels (50 nM) of rotenone led to increased apoptosis in patient-derived cells. We identified an endogenous deficit in mitochondrial complex I in patient-derived cells, but this did not directly correlate with rotenone-sensitivity. We further characterized the sensitivity to rotenone and identified that it was partly associated with heat shock protein 27 levels. Finally, transcriptomic analysis following rotenone exposure revealed that patient-derived cells express a diminished response to rotenone-induced stress compared with cells from healthy controls. Our cellular model of idiopathic Parkinson's disease displays a clear susceptibility phenotype to mitochondrial stress. The determination of molecular mechanisms underpinning this susceptibility may lead to the identification of biomarkers for either disease onset or progression.


Asunto(s)
Apoptosis/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Proteínas de Choque Térmico HSP27/metabolismo , Mitocondrias/metabolismo , Mucosa Olfatoria/citología , Enfermedad de Parkinson/patología , Rotenona/farmacología , Supervivencia Celular , Células Cultivadas , Humanos , Peróxido de Hidrógeno/toxicidad , Mucosa Olfatoria/metabolismo , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/etiología
5.
Cell Death Dis ; 6: e1749, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25950479

RESUMEN

Respiratory complex II (CII, succinate dehydrogenase, SDH) inhibition can induce cell death, but the mechanistic details need clarification. To elucidate the role of reactive oxygen species (ROS) formation upon the ubiquinone-binding (Qp) site blockade, we substituted CII subunit C (SDHC) residues lining the Qp site by site-directed mutagenesis. Cell lines carrying these mutations were characterized on the bases of CII activity and exposed to Qp site inhibitors MitoVES, thenoyltrifluoroacetone (TTFA) and Atpenin A5. We found that I56F and S68A SDHC variants, which support succinate-mediated respiration and maintain low intracellular succinate, were less efficiently inhibited by MitoVES than the wild-type (WT) variant. Importantly, associated ROS generation and cell death induction was also impaired, and cell death in the WT cells was malonate and catalase sensitive. In contrast, the S68A variant was much more susceptible to TTFA inhibition than the I56F variant or the WT CII, which was again reflected by enhanced ROS formation and increased malonate- and catalase-sensitive cell death induction. The R72C variant that accumulates intracellular succinate due to compromised CII activity was resistant to MitoVES and TTFA treatment and did not increase ROS, even though TTFA efficiently generated ROS at low succinate in mitochondria isolated from R72C cells. Similarly, the high-affinity Qp site inhibitor Atpenin A5 rapidly increased intracellular succinate in WT cells but did not induce ROS or cell death, unlike MitoVES and TTFA that upregulated succinate only moderately. These results demonstrate that cell death initiation upon CII inhibition depends on ROS and that the extent of cell death correlates with the potency of inhibition at the Qp site unless intracellular succinate is high. In addition, this validates the Qp site of CII as a target for cell death induction with relevance to cancer therapy.


Asunto(s)
Complejo II de Transporte de Electrones/fisiología , Ubiquinona/genética , Ubiquinona/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Muerte Celular/fisiología , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Humanos , Mitocondrias/metabolismo , Mitocondrias/fisiología , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Ubiquinona/química
6.
Curr Med Chem ; 22(5): 552-68, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25245377

RESUMEN

Tumours exhibit higher basal levels of reactive oxygen species (ROS) and altered redox environment compared to normal cells. Excessive level of ROS can be toxic to these cells, thus they become more vulnerable to damage by further ROS insults induced by pharmacological agents. However, the upregulation of antioxidant capacity in adaptation to intrinsic oxidative stress in cancer cells can confer drug resistance. Therefore, abrogation of such drug-resistant mechanisms by redox modulation could have significant therapeutic implications. Many redox-modulating agents have been developed. The redox-active system epitomised by ascorbate-driven quinone redox cycling, and the group of redox-silent vitamin E analogues represented by α-tocopheryl succinate have been shown to induce selective cancer cell death in different types of cancer. These compounds synergistically act by destabilising organelles like mitochondria, unleashing their apoptogenic potential, which results in efficient death of malignant cells and suppression of tumour growth. Consistent with this notion, clinical trials that aim to examine the therapeutic performance of novel redox-modulating drugs in cancer patients are currently under way.


Asunto(s)
Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Apoptosis , Sinergismo Farmacológico , Humanos , Oxidación-Reducción
7.
Br J Cancer ; 102(8): 1224-34, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20332775

RESUMEN

BACKGROUND: A strategy to reduce the secondary effects of anti-cancer agents is to potentiate the therapeutic effect by their combination. A combination of vitamin K3 (VK3) and ascorbic acid (AA) exhibited an anti-cancer synergistic effect, associated with extracellular production of H(2)O(2) that promoted cell death. METHODS: The redox-silent vitamin E analogue alpha-tocopheryl succinate (alpha-TOS) was used in combination with VK3 and AA to evaluate their effect on prostate cancer cells. RESULTS: Prostate cancer cells were sensitive to alpha-TOS and VK3 treatment, but resistant to AA upto 3.2 mM. When combined, a synergistic effect was found for VK3-AA, whereas alpha-TOS-VK3 and alpha-TOS-AA combination showed an antagonist and additive effect, respectively. However, sub-lethal doses of AA-VK3 combination combined with a sub-toxic dose of alpha-TOS showed to induce efficient cell death that resembles autoschizis. Associated with this cell demise, lipid peroxidation, DNA damage, cytoskeleton alteration, lysosomal-mitochondrial perturbation, and release of cytochrome c without caspase activation were observed. Inhibition of lysosomal proteases did not attenuate cell death induced by the combined agents. Furthermore, cell deaths by apoptosis and autoschizis were detected. CONCLUSION: These finding support the emerging idea that synergistic combinations of some agents can overcome toxicity and other side-effects associated with high doses of single drugs creating the opportunity for therapeutically relevant selectivity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Muerte Celular/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Vitamina K 3/farmacología , alfa-Tocoferol/farmacología , Ácido Ascórbico/administración & dosificación , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Fibroblastos/efectos de los fármacos , Humanos , Lisosomas/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
8.
Oncogene ; 27(31): 4324-35, 2008 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-18372923

RESUMEN

Alpha-tocopheryl succinate (alpha-TOS) is a selective inducer of apoptosis in cancer cells, which involves the accumulation of reactive oxygen species (ROS). The molecular target of alpha-TOS has not been identified. Here, we show that alpha-TOS inhibits succinate dehydrogenase (SDH) activity of complex II (CII) by interacting with the proximal and distal ubiquinone (UbQ)-binding site (Q(P) and Q(D), respectively). This is based on biochemical analyses and molecular modelling, revealing similar or stronger interaction energy of alpha-TOS compared to that of UbQ for the Q(P) and Q(D) sites, respectively. CybL-mutant cells with dysfunctional CII failed to accumulate ROS and underwent apoptosis in the presence of alpha-TOS. Similar resistance was observed when CybL was knocked down with siRNA. Reconstitution of functional CII rendered CybL-mutant cells susceptible to alpha-TOS. We propose that alpha-TOS displaces UbQ in CII causing electrons generated by SDH to recombine with molecular oxygen to yield ROS. Our data highlight CII, a known tumour suppressor, as a novel target for cancer therapy.


Asunto(s)
Apoptosis , Sitios de Unión , Complejo II de Transporte de Electrones/metabolismo , Regulación de la Expresión Génica , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno , Ubiquinona/química , Vitamina E/análogos & derivados , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Ratones , Modelos Moleculares , Conformación Proteica , Tocoferoles , Vitamina E/farmacología
9.
Br J Cancer ; 90(8): 1644-53, 2004 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-15083198

RESUMEN

Malignant mesothelioma (MM) is a fatal type of neoplasia with poor therapeutic prognosis, largely due to resistance to apoptosis. We investigated the apoptotic effect of alpha-tocopheryl succinate (alpha-TOS), a strong proapoptotic agent, in combination with the immunological apoptogen TNF-related apoptosis-inducing ligand (TRAIL) on both MM and nonmalignant mesothelial cells, since MM cells show low susceptibility to the clinically intriguing TRAIL. All MM cell lines tested were sensitive to alpha-TOS-induced apoptosis, and exerted high sensitivity to TRAIL in the presence of subapoptotic doses of the vitamin E analogue. Neither TRAIL or alpha-TOS alone or in combination caused apoptosis in nonmalignant mesothelial cells. Isobologram analysis of the cytotoxicity assays revealed a synergistic interaction between the two agents in MM cells and their antagonistic effect in nonmalignant mesothelial cells. TRAIL-induced apoptosis and its augmentation by alpha-TOS were inhibited by the caspase-8 inhibitor Z-IETD-FMK and the pan-caspase inhibitor Z-VAD-FMK. Activation of caspase-8 was required to induce apoptosis, which was amplified by alpha-TOS via cytochrome c release following Bid cleavage, with ensuing activation of caspase-9. Enhancement of TRAIL-induced apoptosis in MM cells by alpha-TOS was also associated with upregulation of the TRAIL cognate death receptors DR4 and DR5. Our results show that alpha-TOS and TRAIL act in synergism to kill MM cells via mitochondrial pathway, and are nontoxic to nonmalignant mesothelial cells. These findings are indicative of a novel strategy for treatment of thus far fatal MM.


Asunto(s)
Apoptosis/efectos de los fármacos , Glicoproteínas de Membrana/farmacología , Mesotelioma/patología , Factor de Necrosis Tumoral alfa/farmacología , Vitamina E/análogos & derivados , Vitamina E/farmacología , Proteínas Reguladoras de la Apoptosis , Interacciones Farmacológicas , Humanos , Ligandos , Glicoproteínas de Membrana/farmacocinética , Mitocondrias/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF , Tocoferoles , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacocinética , Vitamina E/farmacocinética , Receptor fas
10.
Biochem Pharmacol ; 67(5): 841-54, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15104237

RESUMEN

Peroxisome proliferator-activated receptor-gamma (PPARgamma) has been suggested to upregulate CD36. Since free oxidized polyunsaturated fatty acids are PPARgamma ligands, we studied the effects of LDL modified by the simultaneous action of sPLA2 and 15-lipoxygenase (15LO) on CD36 expression and PPARgamma activation in monocytic cells. Exposure of MM6 cells, which do not express CD36 or other scavenger receptors, to such enzymatically modified LDL (enzLDL) resulted in upregulation of CD36 surface protein and mRNA expression. Similar effects were observed with free 13-hydroperoxyoctadecadienoic acid but not its esterified counterpart. Less pronounced effects were observed with LDL modified by 15LO alone. Upregulation of CD36 was inversely correlated to the state of cell differentiation, as showed by lower response to enzLDL of the scavenger receptor-expressing MM6-sr and THP1 cells. Importantly, LDL modified by sPLA2 and 15LO did not efficiently induce upregulation CD36 in PPARgamma-deficient macrophage-differentiated embryonic stem cells confirming a role of PPARgamma in CD36 expression in cells stimulated with enzLDL. Our data show that LDL modified with physiologically relevant enzymes stimulates CD36 expression in non-differentiated monocytes and that this process involves PPARgamma activation. These effects of enzLDL can be considered pro-atherogenic in the context of early atherosclerosis.


Asunto(s)
Antígenos CD36/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lipoproteínas LDL/farmacología , Monocitos/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Araquidonato 15-Lipooxigenasa/metabolismo , Antígenos CD36/genética , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Humanos , Ácidos Linoleicos/metabolismo , Monocitos/metabolismo , Oxidación-Reducción , Fosfolipasas A/metabolismo , Receptores de LDL/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
Br J Cancer ; 89(10): 1822-6, 2003 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-14612885

RESUMEN

Great hope has been given to micronutrients as anticancer agents, since they present natural compounds with beneficial effects for normal cells and tissues. One of these is vitamin E (VE), an antioxidant and an essential component of biological membranes and circulating lipoproteins. In spite of a number of epidemiological and intervention studies, little or no correlation between VE intake and incidence of cancer has been found. Recent reports have identified a redox-silent analogue of VE, alpha-tocopheryl succinate (alpha-TOS), as a potent anticancer agent with a unique structure and pharmacokinetics in vivo. alpha-TOS is highly selective for malignant cells, inducing them into apoptotic death largely via the mitochondrial route. The molecule of alpha-TOS may be modified so that analogues with higher activity are generated. Finally, alpha-TOS and similar agents are metabolised to VE, thereby yielding a compound with a secondary beneficial activity. Thus, alpha-TOS epitomises a group of novel compounds that hold substantial promise as future anticancer drugs. The reasons for this optimistic notion are discussed in the following paragraphs.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Vitamina E/análogos & derivados , Vitamina E/farmacología , Antineoplásicos/farmacocinética , Antioxidantes/farmacocinética , Apoptosis , Humanos , Neoplasias/tratamiento farmacológico , Tocoferoles , Vitamina E/farmacocinética
12.
Br J Cancer ; 88(12): 1948-55, 2003 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-12799642

RESUMEN

Recent results show that alpha-tocopheryl succinate (alpha-TOS) is a proapoptotic agent with antineoplastic activity. As modifications of the vitamin E (VE) molecule may affect its apoptogenic activity, we tested a number of newly synthesised VE analogues using malignant cell lines. Analogues of alpha-TOS with lower number of methyl substitutions on the aromatic ring were less active than alpha-TOS. Replacement of the succinyl group with a maleyl group greatly enhanced the activity, while it was lower for the glutaryl esters. Methylation of the free succinyl carboxyl group on alpha-TOS and delta-TOS completely prevented the apoptogenic activity of the parent compounds. Both Trolox and its succinylated derivative were inactive. alpha-tocotrienol (alpha-T3 H) failed to induce apoptosis, while gamma-T3 H was apoptogenic, and more so when succinylated. Shortening the aliphatic side chain of gamma-T3 by one isoprenyl unit increased its activity. Neither phytyl nor oleyl succinate caused apoptosis. These findings show that modifications of different functional moieties of the VE molecule can enhance apoptogenic activity. It is hoped that these observations will lead to the synthesis of analogues with even higher apoptogenic and, consequently, antineoplastic efficacy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Vitamina E/análogos & derivados , Humanos , Relación Estructura-Actividad , Células Tumorales Cultivadas , Vitamina E/síntesis química
13.
Br J Cancer ; 88(1): 153-8, 2003 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-12556975

RESUMEN

Activation of nuclear factor-kappaB (NF-kappaB) can interfere with induction of apoptosis triggered by the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL; Apo2L). Therefore, agents that suppress NF-kappaB activation may sensitise cells to TRAIL-dependent apoptosis. Exposure of Jurkat cells to TRAIL resulted in massive and saturable apoptosis induction, following an initial lag time. This lag was abolished by pretreatment of the cells with subapoptotic doses of alpha-tocopheryl succinate (alpha-TOS) or the proteasome inhibitor MG132. Exposure of the cells to TRAIL led to a rapid, transient activation of NF-kappaB, a process that was suppressed by cell pretreatment with alpha-TOS or MG132. Activation of NF-kappaB by TNF-alpha prior to TRAIL exposure increased resistance of the cells to TRAIL-mediated apoptosis. We conclude that alpha-TOS sensitises cells to TRAIL killing, at least in some cases, through inhibition of NF-kappaB activation. This further supports the possibility that this semisynthetic analogue of vitamin E is a potential adjuvant in cancer treatment, such as in the case of TRAIL-mediated inhibition of cancer.


Asunto(s)
Apoptosis , Glicoproteínas de Membrana/farmacología , FN-kappa B/metabolismo , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/farmacología , Vitamina E/análogos & derivados , Vitamina E/farmacología , Proteínas Reguladoras de la Apoptosis , Línea Celular , Interacciones Farmacológicas , Humanos , Células Jurkat , Linfoma , FN-kappa B/antagonistas & inhibidores , Ligando Inductor de Apoptosis Relacionado con TNF , Tocoferoles
14.
Biochem Soc Trans ; 30(4): 755-7, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12196187

RESUMEN

Pyridoxal isonicotinoyl hydrazone (PIH) analogues are effective iron chelators in vivo and in vitro, and may be of value for the treatment of secondary iron overload. The sensitivity of Jurkat cells to Fe-chelator complexes was enhanced several-fold by the depletion of the antioxidant glutathione, indicating the role of oxidative stress in their toxicity. K562 cells loaded with eicosapentaenoic acid, a fatty acid particularly susceptible to oxidation, were also more sensitive to the toxic effects of the Fe complexes, and toxicity was proportional to lipid peroxidation. Thus Fe-chelator complexes cause oxidative stress, which may be a major component of their toxicity. As was the case for their Fe complexes, the toxicity of PIH analogues was enhanced by glutathione depletion of Jurkat cells and eicosapentaenoic acid-loading of K562 cells. Thus the toxicity of the chelators themselves is also enhanced by compromised cellular redox status. In addition, the toxicity of the chelators was diminished by culturing Jurkat cells under hypoxic conditions, which may limit the production of the reactive oxygen species that initiate oxidative stress. A significant part of the toxicity of the chelators may be due to intracellular formation of Fe-chelator complexes, which oxidatively destroy the cell.


Asunto(s)
Quelantes/toxicidad , Isoniazida/análogos & derivados , Isoniazida/toxicidad , Piridoxal/análogos & derivados , Piridoxal/toxicidad , Ácido Ascórbico , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Humanos , Quelantes del Hierro/toxicidad , Células Jurkat , Células K562 , Cinética , Estructura Molecular , Oxidación-Reducción , Estrés Oxidativo , Relación Estructura-Actividad
15.
Apoptosis ; 7(2): 179-87, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11865203

RESUMEN

The incidence of cancer and atherosclerosis, two most common causes of death in developed countries, has been stagnating or, even, increasing. Drugs effective against such conditions are needed and, in this regard, the potential anti-atherosclerotic activity of vitamin E analogs has been studied extensively. Surprisingly, recent results indicate that these agents may also exert anti-neoplastic effects. Here we review the evidence that particular analogs of vitamin E may act as both antiatherogenic and anti-cancer agents, and discuss the possible molecular bases for these actions.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/terapia , Vitamina E/análogos & derivados , Animales , Anticolesterolemiantes/farmacología , Antioxidantes/farmacología , Proteínas Reguladoras de la Apoptosis , Glicoproteínas de Membrana/metabolismo , Ratones , Modelos Biológicos , Ligando Inductor de Apoptosis Relacionado con TNF , Tocoferoles , Factor de Necrosis Tumoral alfa/metabolismo , Vitamina E/metabolismo
16.
Redox Rep ; 6(3): 143-51, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11523588

RESUMEN

Recent evidence suggests that vitamin E and its analogues, which have been used for many years as antioxidants, may not only protect cells from free radical damage but also induce apoptotic cell death in various cell types. While alpha-tocopherol (alpha-TOH) is mainly known as an anti-apoptotic agent, its redox-silent analogues either have no influence on cell survival (alpha-tocopheryl acetate, alpha-TOA), or induce apoptosis (alpha-tocopheryl succinate, alpha-TOS). Although precise mechanisms of apoptosis induction by alpha-TOS remain to be elucidated, there is evidence that this process involves both the antiproliferative and membrane destabilising activities of the agent. Alpha-TOS has been shown to induce apoptosis in malignant cell lines but not, in general, in normal cells, and to inhibit tumorigenesis in vivo. These features suggest that this semi-synthetic analogue of vitamin E could be a promising antineoplastic agent.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Vitamina E/análogos & derivados , Vitamina E/farmacología , Adenocarcinoma/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Antioxidantes/uso terapéutico , Ciclo Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Ensayos de Selección de Medicamentos Antitumorales , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Radicales Libres , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Membranas Intracelulares/efectos de los fármacos , Lisosomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Estructura Molecular , Células Madre Neoplásicas/efectos de los fármacos , Oxidación-Reducción , Proteína Quinasa C/fisiología , Especies Reactivas de Oxígeno , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Tocoferoles , Células Tumorales Cultivadas/efectos de los fármacos , Vitamina E/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
17.
FEBS Lett ; 503(1): 46-50, 2001 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-11513852

RESUMEN

Generation of free radicals is often associated with the induction and progression of apoptosis. Therefore, antioxidants can prove anti-apoptotic, and can help to elucidate specific apoptotic pathways. Here we studied whether coenzyme Q, present in membranes in reduced (ubiquinol) or oxidised (ubiquinone) forms, can affect apoptosis induced by various stimuli. Exposure of Jurkat cells to alpha-tocopheryl succinate (alpha-TOS), hydrogen peroxide, anti-Fas IgM or TRAIL led to induction of apoptosis. Cell death due to the chemical agents was suppressed in cells enriched with the reduced form of coenzyme Q. However, coenzyme Q did not block cell death induced by the immunological agents. Ubiquinol-10 inhibited reactive oxygen species (ROS) generation in cells exposed to alpha-TOS, and a mitochondrially targeted coenzyme Q analogue also blocked apoptosis triggered by alpha-TOS or hydrogen peroxide. Therefore, it is plausible that ubiquinol-10 protects cells from chemically-induced apoptosis by acting as an antioxidant in mitochondria. Our results also indicate that generation of free radicals may not be a critical step in induction of apoptosis by immunological agents.


Asunto(s)
Antioxidantes/metabolismo , Apoptosis/fisiología , Mitocondrias/metabolismo , Ubiquinona/fisiología , Vitamina E/análogos & derivados , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Western Blotting , Humanos , Peróxido de Hidrógeno/farmacología , Inmunoglobulina M/farmacología , Células Jurkat , Glicoproteínas de Membrana/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF , Tocoferoles , Factor de Necrosis Tumoral alfa/farmacología , Vitamina E/farmacología
19.
Atherosclerosis ; 157(2): 257-83, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11472726

RESUMEN

Atherosclerosis is a disease involving both oxidative modifications and disbalance of the immune system. Vitamin E, an endogenous redox-active component of circulating lipoproteins and (sub)cellular membranes whose levels can be manipulated by supplementation, has been shown to play a role in the initiation and progression of the disease. Recent data reveal that the activities of vitamin E go beyond its redox function. Moreover, it has been shown that vitamin E can exacerbate certain processes associated with atherogenesis. In this essay we review the role of biology of atherosclerosis, and suggest that these two facets decide the clinical manifestation and outcome of the disease.


Asunto(s)
Arteriosclerosis/etiología , Vitamina E/fisiología , Animales , Progresión de la Enfermedad , Humanos , Sistema Inmunológico/fisiología , Oxidación-Reducción , Transducción de Señal/fisiología
20.
Biochemistry ; 40(15): 4686-92, 2001 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-11294636

RESUMEN

Suppression of NF kappa B activation has been involved in the elimination of survival programs during endothelial cell (EC) apoptosis. We used alpha-tocopheryl succinate (alpha-TOS) to trigger apoptosome formation and the subsequent activation of executioner caspases. The level of bcl-2 was reduced by alpha-TOS, and its downregulation potentiated and its overexpression suppressed pro-apoptotic effects of alpha-TOS, indicating a mitochondrial role in alpha-TOS-induced apoptosis in EC. alpha-TOS treatment was associated with induction of TUNEL-positive apoptosis in EC with a high but not with a low proliferation index. The use of the pan-caspase inhibitor z-VAD.fmk suggested the involvement of caspases in cleavage of p65, and in inhibition of nuclear translocation of p65 and NF kappa B-dependent transactivation of a gene construct encoding the green fluorescence protein elicited by TNF alpha in contact-arrested EC. The suppression by alpha-TOS of inflammatory EC responses induced by TNF alpha such as VCAM-1 mRNA and surface protein expression and shear-resistant arrest of monocytic cells were also reversed by z-VAD.fmk. NF kappa B-dependent transactivation was preserved in alpha-TOS-treated EC stably transfected with a caspase-noncleavable p65 mutant but not with its truncated form, thus establishing a direct link between alpha-TOS-induced effects and p65 cleavage. Our data infer a pathway by which caspase activation in EC inhibits NF kappa B-dependent inflammatory activation and monocyte recruitment, and provide evidence for a relationship between pro-apoptotic and anti-inflammatory pathways.


Asunto(s)
Caspasas/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/patología , FN-kappa B/metabolismo , Vitamina E/análogos & derivados , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasa 6 , Caspasas/fisiología , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Regulación hacia Abajo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Hidrólisis/efectos de los fármacos , Inflamación/enzimología , Inflamación/metabolismo , Monocitos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tocoferoles , Factor de Transcripción ReIA , Activación Transcripcional/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética , Vitamina E/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA