Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Hematol Oncol ; 42(3): e3281, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38775115

RESUMEN

The FLT3-ITD mutation represents the most frequent genetic alteration in newly diagnosed acute myeloid leukemia (AML) patient and is associated with poor prognosis. Mutation result in the retention of a constitutively active form of this receptor in the endoplasmic reticulum (ER) and the subsequent modification of its downstream effectors. Here, we assessed the impact of such retention on ER homeostasis and found that mutant cells present lower levels of ER stress due to the overexpression of ERO1α, one of the main proteins of the protein folding machinery at the ER. Overexpression of ERO1α resulted essential for ITD mutant cells survival and chemoresistance and also played a crucial role in shaping the type of glucose metabolism in AML cells, being the mitochondrial pathway the predominant one in those with a higher ER stress (non-mutated cells) and the glycolytic pathway the predominant one in those with lower ER stress (mutated cells). Our data indicate that FLT3 mutational status dictates the route for glucose metabolism in an ERO1α depending on manner and this provides a survival advantage to tumors carrying these ITD mutations.


Asunto(s)
Estrés del Retículo Endoplásmico , Retículo Endoplásmico , Leucemia Mieloide Aguda , Tirosina Quinasa 3 Similar a fms , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Retículo Endoplásmico/metabolismo , Mutación , Línea Celular Tumoral , Glicoproteínas de Membrana , Oxidorreductasas
2.
J Cell Physiol ; 236(1): 27-40, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32725819

RESUMEN

Several oncogenic pathways plus local microenvironmental conditions, such as hypoxia, converge on the regulation of cancer cells metabolism. The major metabolic alteration consists of a shift from oxidative phosphorylation as the major glucose consumer to aerobic glycolysis, although most of cancer cells utilize both pathways to a greater or lesser extent. Aerobic glycolysis, together with the directly related metabolic pathways such as the tricarboxylic acid cycle, the pentose phosphate pathway, or gluconeogenesis are currently considered as therapeutic targets in cancer research. Melatonin has been reported to present numerous antitumor effects, which result in a reduced cell growth. This is achieved with both low and high concentrations with no relevant side effects. Indeed, high concentrations of this indolamine reduce proliferation of cancer types resistant to low concentrations and induce cell death in some types of tumors. Previous work suggest that regulation of glucose metabolism and other related pathways play an important role in the antitumoral effects of high concentration of melatonin. In the present review, we analyze recent work on the regulation by such concentrations of this indolamine on aerobic glycolysis, gluconeogenesis, the tricarboxylic acid cycle and the pentose phosphate pathways of cancer cells.


Asunto(s)
Glucosa/metabolismo , Melatonina/administración & dosificación , Neoplasias/metabolismo , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Proliferación Celular/efectos de los fármacos , Gluconeogénesis/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos
3.
Oncol Rep ; 44(1): 293-302, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32319665

RESUMEN

The FMS­like tyrosine kinase 3 internal tandem duplication (FLT3­ITD) mutation represents the most frequent genetic alteration in acute myeloid leukemia (AML) and is associated with poor prognosis. The mutation promotes cancer cell survival and proliferation, and shifts their glucose metabolism towards aerobic glycolysis, a frequent alteration in cancer. In the present study, the impact of melatonin on the viability of AML cell lines with (MV­4­11 and MOLM­13) or without the FLT3­ITD mutation (OCI­AML3 and U­937) was evaluated. Melatonin induces cell death in AML cells carrying the FLT3­ITD mutation, but only inhibits the proliferation of AML cells without this mutation. Consistently, melatonin decreases tumor growth and increases animal survival in a xenograft model of FLT3­ITD AML. Toxicity is related to a decrease in glucose uptake, lactate dehydrogenase activity, lactate production and hypoxia­inducible factor­1α activation. Melatonin also regulates the expression of glucose metabolism­related genes, impairing the balance between anaplerosis and cataplerosis, through the upregulation of the expression of phosphoenolpyruvate carboxykinase 2 (PCK2). Collectively, the present findings highlight the regulation of glucose metabolism, currently considered a possible therapeutic target in cancer, as a key event in melatonin­induced cytotoxicity, suggesting its potential as a therapeutic tool for the treatment of patients with AML, particularly those carrying the FLT3­ITD mutation that results in low basal expression levels of PCK2.


Asunto(s)
Glucosa/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Melatonina/administración & dosificación , Mutación , Tirosina Quinasa 3 Similar a fms/genética , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Melatonina/farmacología , Ratones , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cancer Ther ; 17(3): 614-624, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29339551

RESUMEN

Internal tandem duplication (ITD) or tyrosine kinase domain mutations of FLT3 is the most frequent genetic alteration in acute myelogenous leukemia (AML) and are associated with poor disease outcome. Despite considerable efforts to develop single-target FLT3 drugs, so far, the most promising clinical response has been achieved using the multikinase inhibitor midostaurin. Here, we explore the activity of the indolocarbazole EC-70124, from the same chemical space as midostaurin, in preclinical models of AML, focusing on those bearing FLT3-ITD mutations. EC-70124 potently inhibits wild-type and mutant FLT3, and also other important kinases such as PIM kinases. EC-70124 inhibits proliferation of AML cell lines, inducing cell-cycle arrest and apoptosis. EC-70124 is orally bioavailable and displays higher metabolic stability and lower human protein plasma binding compared with midostaurin. Both in vitro and in vivo pharmacodynamic analyses demonstrate inhibition of FLT3-STAT5, Akt-mTOR-S6, and PIM-BAD pathways. Oral administration of EC-70124 in FLT3-ITD xenograft models demonstrates high efficacy, reaching complete tumor regression. Ex vivo, EC-70124 impaired cell viability in leukemic blasts, especially from FLT3-ITD patients. Our results demonstrate the ability of EC-70124 to reduce proliferation and induce cell death in AML cell lines, patient-derived leukemic blast and xenograft animal models, reaching best results in FLT3 mutants that carry other molecular pathways' alterations. Thus, its unique inhibition profile warrants EC-70124 as a promising agent for AML treatment based on its ability to interfere the complex oncogenic events activated in AML at several levels. Mol Cancer Ther; 17(3); 614-24. ©2018 AACR.


Asunto(s)
Carbazoles/farmacología , Indoles/farmacología , Leucemia Mieloide/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Enfermedad Aguda , Animales , Disponibilidad Biológica , Células CACO-2 , Carbazoles/farmacocinética , Carbazoles/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Femenino , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células HL-60 , Humanos , Indoles/farmacocinética , Indoles/uso terapéutico , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Ratones SCID , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-pim-1/genética , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Células THP-1 , Tirosina Quinasa 3 Similar a fms/metabolismo
5.
Mol Cell Endocrinol ; 434: 238-49, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27402602

RESUMEN

Melatonin (N-acetyl-5-methoxytryptamine) is a highly pleiotropic hormone with antioxidant, antiproliferative, oncolytic and neuroprotective properties. Here, we present evidence that the N-acetyl side chain plays a key role in melatonin's antiproliferative effect in HT22 and sw-1353 cells, but it does so at the expense of antioxidant and neuroprotective properties. Removal of the N-acetyl group enhances the antioxidant and neuroprotective properties of the indole, but it can lead to toxic methamphetamine-like effects in several cell lines. Inhibition of NFkB mimicked melatonin's antiproliferative and antioxidant effects, but not neuroprotection. Our results strongly suggest that neuroprotective and antiproliferative effects of melatonin rely on different parts of the molecule and are likely mediated by different mechanisms. We also predict that melatonin metabolism by target cells could determine whether melatonin inhibits cell proliferation, prevents toxicity or induces cell death (e.g. apoptosis or autophagy). These observations could have important implications for the rational use of melatonin in personalized medicine.


Asunto(s)
Antioxidantes/farmacología , Ácido Glutámico/toxicidad , Hipocampo/efectos de los fármacos , Melatonina/farmacología , Fármacos Neuroprotectores/farmacología , 5-Metoxitriptamina , Animales , Autofagia , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Hipocampo/citología , Humanos , Ratones
6.
PLoS One ; 10(8): e0135420, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26252771

RESUMEN

Melatonin kills or inhibits the proliferation of different cancer cell types, and this is associated with an increase or a decrease in reactive oxygen species, respectively. Intracellular oxidants originate mainly from oxidative metabolism, and cancer cells frequently show alterations in this metabolic pathway, such as the Warburg effect (aerobic glycolysis). Thus, we hypothesized that melatonin could also regulate differentially oxidative metabolism in cells where it is cytotoxic (Ewing sarcoma cells) and in cells where it inhibits proliferation (chondrosarcoma cells). Ewing sarcoma cells but not chondrosarcoma cells showed a metabolic profile consistent with aerobic glycolysis, i.e. increased glucose uptake, LDH activity, lactate production and HIF-1α activation. Melatonin reversed Ewing sarcoma metabolic profile and this effect was associated with its cytotoxicity. The differential regulation of metabolism by melatonin could explain why the hormone is harmless for a wide spectrum of normal and only a few tumoral cells, while it kills specific tumor cell types.


Asunto(s)
Glucólisis , Melatonina/farmacología , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Aerobiosis , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular , Condrosarcoma/metabolismo , Citometría de Flujo , Glucosa/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico/metabolismo , Melatonina/química , Potenciales de la Membrana , Fosfatidilinositol 3-Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
J Pineal Res ; 57(3): 308-16, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25163989

RESUMEN

Glioblastoma-initiating cells (GICs) represent a stem cell-like subpopulation within malignant glioblastomas responsible for tumor development, progression, therapeutic resistance, and tumor relapse. Thus, eradication of this subpopulation is essential to achieve stable, long-lasting remission. We have previously reported that melatonin decreases cell proliferation of glioblastoma cells both in vitro and in vivo and synergistically increases effectiveness of drugs in glioblastoma cells and also in GICs. In this study, we evaluated the effect of the indolamine alone in GICs and found that melatonin treatment reduces GICs proliferation and induces a decrease in self-renewal and clonogenic ability accompanied by a reduction in the expression of stem cell markers. Moreover, our results also indicate that melatonin treatment, by modulating stem cell properties, induces cell death with ultrastructural features of autophagy. Thus, data reported here reinforce the therapeutic potential of melatonin as a treatment of malignant glioblastoma both by inhibiting tumor bulk proliferation or killing GICs, and simultaneously enhancing the effect of chemotherapy.


Asunto(s)
Autofagia/fisiología , Neoplasias Encefálicas/patología , Glioma/patología , Melatonina/fisiología , Secuencia de Bases , Citometría de Flujo , Humanos , Melatonina/farmacología , Microscopía Electrónica , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Int J Mol Sci ; 14(4): 6597-613, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23528889

RESUMEN

It is well established that melatonin exerts antitumoral effects in many cancer types, mostly decreasing cell proliferation at low concentrations. On the other hand, induction of apoptosis by melatonin has been described in the last few years in some particular cancer types. The cytotoxic effect occurs after its administration at high concentrations, and the molecular pathways involved have been only partially determined. Moreover, a synergistic effect has been found in several cancer types when it is administered in combination with chemotherapeutic agents. In the present review, we will summarize published work on the pro-apoptotic effect of melatonin in cancer cells and the reported mechanisms involved in such action. We will also construct a hypothesis on how different cell signaling pathways may relate each other on account for such effect.


Asunto(s)
Apoptosis/efectos de los fármacos , Melatonina/farmacología , Neoplasias/patología , Animales , Relación Dosis-Respuesta a Droga , Humanos , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/ultraestructura
9.
J Neurosci Res ; 90(9): 1850-60, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22513717

RESUMEN

Parkinson's disease has been widely related to both apoptosis and oxidative stress. Many publications relate the loss of mitochondrial potential to an apoptosis-mediated cell death in different in vivo and in vitro models of this pathology. The present study used the dopaminegic specific neurotoxin 1-methyl-4-phenylpyridinium (MPP(+) ) on neuron-like PC12 cells, which is a well-accepted model of Parkinson's disease. Results showed an early increase in oxidants, which drives the modulation of c-Jun N-terminal kinase (JNK) and AKT/mammalian target of rapamycin (mTOR) pathways, mimicking peroxide treatment. However, the cell death found in neuronal PC12 cells treated with MPP(+) was not a caspase-associated apoptosis. Electron microscopic images illustrated autophagic cell death, which was confirmed by a Beclin-1 and ATG expression increase, accumulation of acidic vesicles, and rescue by an autophagy inhibitor. In conclusion, the boost in oxidants from MPP(+) treatment in neuronal PC12 is modulating both survival (AKT/mTOR) and death (JNK) pathways, which are the perpetrators of an autophagic cell death.


Asunto(s)
Autofagia/fisiología , MAP Quinasa Quinasa 4/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Western Blotting , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Estrés Oxidativo/fisiología , Células PC12 , Ratas , Transducción de Señal/fisiología
10.
Free Radic Res ; 45(11-12): 1333-41, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21923620

RESUMEN

Melatonin is an endogenous indolamine, classically known as a light/dark regulator. Besides classical functions, melatonin has also showed to have a wide range of antitumoral effects in numerous cancer experimental models. However, no definite mechanism has been described to explain the whole range of antineoplasic effects. Here we describe a dual effect of melatonin on intracellular redox state in relation to its antiproliferative vs cytotoxic actions in cancer cells. Thus, inhibition of proliferation correlates with a decrease on intracellular reactive oxygen species (ROS) and increase of antioxidant defences (antioxidant enzymes and intracellular gluthation,GSH levels), while induction of cell death correlates with an increase on intracellular ROS and decrease of antioxidant defences. Moreover, cell death can be prevented by other well-known antioxidants or can be increased by hydrogen peroxide. Thus, tumour cell fate will depend on the ability of melatonin to induce either an antioxidant environment--related to the antiproliferative effect or a prooxidant environment related to the cytotoxic effect.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Melatonina/farmacología , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Humanos , Oxidación-Reducción/efectos de los fármacos
11.
J Pineal Res ; 50(3): 345-55, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21392090

RESUMEN

Incorporation of new therapeutic agents remains as a major challenge for treatment of patients with malignant haematological disorders. Melatonin is an indolamine without relevant side effects. It has been shown previously to exhibit synergism with several chemotherapeutic drugs in Ewing sarcoma cells by potentiating the extrinsic pathway of apoptosis. It also sensitizes human glioma cells against TRAIL by increasing DR5 expression. Here, we report the induction of cell death by melatonin in several human malignant haematological cell lines through the activation of the extrinsic pathway of apoptosis. Such activation was mediated by the increase in the expression of the death receptors Fas, DR4 and DR5 and their ligands Fas L and TRAIL, with a remarkable rise in the expression of Fas and Fas L. The cytotoxic effect and the increase in Fas and Fas L were dependent on Akt activation. Results were corroborated in blasts from bone marrow and peripheral blood of acute myeloid leukaemia patients, where melatonin induced cell death and increased both Fas and Fas L expressions. We conclude that melatonin may be considered as a potential antileukaemic agent and its therapeutic use, either alone or in combination with current chemotherapeutic drugs, should be taken into consideration for further research.


Asunto(s)
Leucemia/metabolismo , Melatonina/farmacología , Receptores de Muerte Celular/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/genética , Caspasa 3/metabolismo , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Células HL-60 , Humanos , Receptores de Muerte Celular/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Células Tumorales Cultivadas , Receptor fas/genética , Receptor fas/metabolismo
12.
Cancer Lett ; 287(2): 216-23, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19632770

RESUMEN

Despite the common expression of death receptors, many types of cancer including gliomas are resistant to the death receptor ligand (TRAIL). Melatonin antitumoral actions have been extensively described, including oncostatic properties on several tumor types and improvement of chemotherapeutic regimens. Here, we found that melatonin effectively increase cell sensitivity to TRAIL-induced cell apoptosis in A172 and U87 human glioma cells. The effect seems to be related to a modulation of PKC activity which in turns decreases Akt activation leading to an increase in death receptor 5 (DR5) levels and a decrease in the antiapoptotic proteins survivin and bcl-2 levels.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Glioma/patología , Melatonina/farmacología , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Glioma/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Survivin
13.
J Pineal Res ; 48(1): 72-80, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20025643

RESUMEN

Ewing sarcoma, the second most frequent bone cancer type, affects mainly adolescents, who have a survival of 50% 5 yr after diagnosis. Current treatments include a combination of surgery, radiotherapy and chemotherapy, which present potential serious side effects. Melatonin, a natural molecule without relevant side effects, has been previously shown to induce cytotoxicity in SK-N-MC cells, a Ewing sarcoma cell line. Here, we found that there is a synergy in the antitumor effect when melatonin (50 mum-1 mm) is combined with vincristine at the concentration of 5-10 nm or with ifosfamide at the range of 100 mum-1 mm. This synergism is due to the potentiation of cell death, particularly to the potentiation of apoptosis, i.e., mainly the extrinsic apoptotic pathway. There is a significant increase in the activation of caspase-3, -8, -9 and Bid when melatonin is combined with vincristine or ifosfamide compared to the individual treatments. Finally, there is also a potentiation of the early free radical production, likely dependent on the extrinsic apoptosis pathway activation, when the drugs are combined with melatonin. Other proteins which are related to this pathway including mitogen-activated protein kinase or protein kinase B/Akt are not involved in apoptosis induced by these agents separately or when combined. The results shown here together with the facts that: (i) no relevant side effects have been reported for melatonin and (ii) melatonin has a cytoprotective effect on noncancer cells, opens the door for a new approach in the treatment of the Ewing sarcoma family of tumors.


Asunto(s)
Apoptosis/efectos de los fármacos , Melatonina/uso terapéutico , Sarcoma de Ewing/tratamiento farmacológico , Antineoplásicos Alquilantes/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Western Blotting , Caspasa 3/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Etopósido/uso terapéutico , Citometría de Flujo , Humanos , Ifosfamida/uso terapéutico , Vincristina/uso terapéutico
14.
J Neurochem ; 107(1): 127-40, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18665912

RESUMEN

Oxidative stress has been shown to mediate neuron damage in Parkinson's disease (PD). In the present report, we intend to clarify the intracellular pathways mediating dopaminergic neuron death after oxidative stress production using post-mitotic PC12 cells treated with the neurotoxin 6-hydroxydopamine (6-OHDA). The use of post-mitotic cells is crucial, because one of the suggested intracellular pathways implicated in neuron death relates to the re-entry of neurons (post-mitotic cells) in the cell cycle. We find that 6-OHDA sequentially increases intracellular oxidants, functional cell damage and caspase-3 activation, leading to cell death after 12 h of incubation. Prevention of cell damage by different antioxidants supports the implication of oxidative stress in the observed neurotoxicity. Oxidative stress-dependent phosphorylation of the MAPK JNK and oxidative stress-independent PKB/Akt dephosphorylation are involved in 6-OHDA neurotoxicity. Decrease in p21(WAF1/CIP1) and cyclin-D1 expression, disappearance of the non-phosphorylated band of retinoblastoma protein (pRb), and expression of proliferating cell nuclear antigen, not present in PC12 post-mitotic cells, suggest a re-entry of differentiated cells into cell cycle. Our results indicate that such a re-entry is mediated by oxidative stress and is involved in 6-OHDA-induced cell death. We conclude that at least three intracellular pathways are involved in 6-OHDA-induced cell death in differentiated PC12 cells: JNK activation, cell cycle progression (both oxidative stress-dependent), and Akt dephosphorylation (not related to the increase of oxidants); the three pathways are necessary for the cells to die, since blocking one of them is sufficient to keep the cells alive.


Asunto(s)
Dopamina/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Estrés Oxidativo/fisiología , Oxidopamina/toxicidad , Transducción de Señal/fisiología , Animales , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Ciclo Celular/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Diferenciación Celular/fisiología , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Genes cdc/efectos de los fármacos , Genes cdc/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Mitosis/fisiología , Degeneración Nerviosa/inducido químicamente , Neuronas/efectos de los fármacos , Neuronas/patología , Neurotoxinas , Células PC12 , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/fisiopatología , Simpaticolíticos
15.
J Pineal Res ; 43(3): 239-44, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17803520

RESUMEN

Classical anticancer therapies often are ineffective in patients with malignant glioma who have a survival of <1 year. Our previous studies showed a potent inhibitory effect of melatonin on glioma cell proliferation. This effect seems to be mediated by the well-known antioxidant properties of this molecule and the negative regulation of some intracellular effectors, such as the kinase Akt or the transcription factor nuclear factor (NF)-kappaB. Finally, protein kinase C (PKC) also seems to be implicated in this effect although the intracellular pathways involved have not been elucidated. In this study, we analyzed the role of PKC in the regulation by melatonin of intracellular effectors leading to inhibition of cell proliferation. Activation of PKC by incubation with triphorbol ester acetate (TPA) blocks the inhibitory effect of melatonin on Akt and NF-kappaB activity. Moreover, incubation with melatonin induces a decrease in p21 expression in these cells that is partially blocked by co-incubation with TPA. Taken together, these results suggest that melatonin's oncostatic effect on glioma cells is mediated, at least in part, by the inhibition of PKC activity which, in turn, results in Akt and NF-kappaB activity inhibition and modulation of cell cycle-related gene expression.


Asunto(s)
Glioma/metabolismo , Melatonina/metabolismo , Proteína Quinasa C/metabolismo , Animales , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Ésteres del Forbol/farmacología , Ratas
16.
Free Radic Biol Med ; 42(11): 1715-22, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17462539

RESUMEN

Tumor cells are able to survive and proliferate despite the higher-than-average level of reactive oxygen species (ROS) they exhibit. This is generally taken as a clue as to the implications of ROS in cell proliferation. In fact many mitogenic intracellular signaling pathways could be redox regulated, more particularly those involving tyrosine kinase receptors (RTK). In the present work we use N-acetylcysteine (NAC)-a well-known antioxidant molecule-to study the implications of cellular redox state on rat C6 glioma cell proliferation. NAC is shown to decrease glioma cell proliferation, inducing a cell cycle arrest in the G(0)/G(1) phase and markedly up-regulating p21 expression. A rapid, and glutathione-independent, decrease in intracellular oxidants was observed as well. NAC also lowers Akt activity, extracellular signal-regulated kinase 1/2, and the redox-sensitive transcription factor NF-kappaB, all of which are ROS related and seem to be in close connection with cell proliferation. NAC effects apparently relate to protein kinase C (PKC) activity because 100 nM TPA-a PKC activator-induces a partial blockage of the NAC antiproliferative effect. Bringing our results together, it seems that intracellular reduction of oxidants in C6 glioma cells can induce inhibition of cell proliferation by modulating RTK-related intracellular signaling pathways.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Neoplasias Encefálicas/metabolismo , Proliferación Celular/efectos de los fármacos , Glioma/metabolismo , Animales , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Regulación hacia Arriba
17.
J Neurochem ; 100(3): 736-46, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17263795

RESUMEN

The pineal hormone melatonin has neuroprotective effects in a large number of models of neurodegeneration. Melatonin crosses the blood-brain barrier, shows a decrease in its nocturnal peaks in blood with age that has been associated with the development of neurodegenerative disorders, and has been shown to be harmless at high concentrations. These properties make melatonin a potential therapeutic agent against neurodegenerative disorders but the pathways involved in such neuroprotective effects remain unknown. In the present report we study the intracellular pathways implicated in the complete neuroprotection provided by melatonin against glutamate-induced oxytosis in the HT22 mouse hippocampal cell line. Our results strongly suggest that melatonin prevents oxytosis through a direct antioxidant effect specifically targeted at the mitochondria. Firstly, none of the described transducers of melatonin signalling seems to be implicated in the neuroprotection provided by this indole. Secondly, melatonin does not prevent cytosolic GSH depletion-dependent increase in reactive oxygen species (ROS), but it totally prevents mitochondrial ROS production despite the fact that the latter is much higher than the former. And finally, there is a high correlation between the concentration at which melatonin and closely related indoles exert a direct antioxidant effect in vitro and a neuroprotective effect against glutamate-induced oxytosis.


Asunto(s)
Hipocampo/metabolismo , Melatonina/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Estrés Oxidativo/fisiología , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Ácido Glutámico/toxicidad , Glutatión/deficiencia , Hipocampo/efectos de los fármacos , Melatonina/farmacología , Ratones , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
18.
J Pineal Res ; 41(2): 130-5, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16879318

RESUMEN

Low concentrations (nanomolar) of melatonin had been previously shown to inhibit cell proliferation in several cancer cell lines as well as in experimental animal models. Additionally, cell growth inhibition and differentiation of prostate cancer cell lines by high concentrations (micromolar to millimolar) of melatonin have been recently reported. In the present paper, we show the induction of apoptosis by high doses of melatonin in the human neuroblastoma cell line SK-N-MC. We found accumulation of cells in the G2/M cell cycle phase and induction of cellular death, measured as lactate dehydrogenase (LDH) released into the culture medium, under millimolar concentration of melatonin. Apoptosis was evaluated using 4,6-diamidino-2-phenylindole staining, DNA gel electrophoresis, electron microscopy, and annexin V binding. Apoptosis progressed through the classical pathway, which involves caspase-3 activation. Cell death was dose and time-dependent; the lowest effective concentration of melatonin was 100 microm. Treatment with 1 mm melatonin for 6 days induced cell death in 75% of the cells. This novel finding shows that a nontoxic natural indoleamine may be potential therapy for some types of human neuroblastomas.


Asunto(s)
Apoptosis/efectos de los fármacos , Melatonina/farmacología , Caspasa 3 , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Humanos , L-Lactato Deshidrogenasa/metabolismo , Melatonina/administración & dosificación , Transducción de Señal
19.
Anat Rec A Discov Mol Cell Evol Biol ; 288(9): 1026-30, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16892423

RESUMEN

Tryptamine derivatives are a family of biogenic amines that have been suggested to be modulators of brain function at physiological concentrations. However, pharmacological concentrations of these amines display amphetamine-like properties, and they seem to play a role in brain disorders. Amphetamines induce autophagy in nerve cells, and this type of cell death has also been involved in neurodegenerative diseases. In the present work, we clearly demonstrate for the very first time that high concentrations of tryptamine (0.1-1 mM) induce autophagy in HT22 and SK-N-SH nerve cell lines and in primary cultures of astrocytes, glial cells being less sensitive than neurons. Ultrastructural cell morphology shows all of the typical hallmarks of autophagy. There is no nuclear chromatin condensation, endoplasmic reticulum and mitochondria are swollen, and a great number of double-membraned autophagosomes and residual bodies can be shown in the cytoplasm. Autophagosomes and residual bodies contain mitochondria, membranes, and vesicles and remain unabridged until the cell membrane is disrupted and the cell dies. The same results have been found when cells were incubated with high concentrations of 5-methoxytryptamine (0.1-1 mM). Our results establish a possible link between the role of tryptamine derivatives in brain disorders and the presence of autophagic cell death in these kinds of disorders.


Asunto(s)
Autofagia/efectos de los fármacos , Enfermedades Neurodegenerativas/etiología , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Triptaminas/toxicidad , Animales , Astrocitos/efectos de los fármacos , Astrocitos/ultraestructura , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Estructuras Citoplasmáticas/efectos de los fármacos , Estructuras Citoplasmáticas/ultraestructura , Relación Dosis-Respuesta a Droga , Humanos , Microscopía Electrónica de Transmisión , Enfermedades Neurodegenerativas/patología , Neuroglía/ultraestructura , Neuronas/ultraestructura , Ratas , Ratas Wistar , Serotonina/toxicidad , Serotoninérgicos/toxicidad
20.
Cancer Res ; 66(2): 1081-8, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16424044

RESUMEN

Melatonin is an indolamine mostly produced in the pineal gland, soluble in water, and highly lipophilic, which allows it to readily cross the blood-brain barrier. Melatonin possesses antioxidant properties and its long-term administration in rodents has not been found to cause noteworthy side effects. In the present work, we found that millimolar concentrations of this indolamine reduced cell growth of C6 glioma cells by 70% after 72 hours of treatment, inhibiting cell progression from G(1) to S phase of the cell cycle. Intraperitoneal administration of 15 mg/kg body weight of melatonin to rats previously injected in the flank with C6 glioma cells reduces tumor growth by 50% 2 weeks after the implant. Inhibition of cell growth does not depend on melatonin membrane receptor activation whereas it seemingly relates to the reduction of intracellular basal free radical levels by 30%. Increase of basal redox state of the cells and constitutive activation of tyrosine kinase receptor [receptor tyrosine kinase (RTK)] pathways, including the extracellular signal-regulated kinase 1/2 (ERK1/2) and the Akt and protein kinase C (PKC) signaling pathways, contribute to the progression of the gliomas leading to the constitutive activation of the redox-dependent survival transcription factor nuclear factor kappaB (NF-kappaB). The antioxidant effect of melatonin in C6 cells is associated to inhibition of NF-kappaB and Akt, but not of ERK1/2. The antiproliferative effect of the indolamine on these cells is partially abolished when coincubated with the PKC activator 12-O-tetradecanoylphorbol-13-acetate, thus indicating that the ability of melatonin to change cellular redox state may be inactivating the pathway RTK/PKC/Akt/NF-kappaB.


Asunto(s)
Antioxidantes/fisiología , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Glioma/patología , Melatonina/fisiología , Transducción de Señal , Animales , Ciclo Celular , Radicales Libres , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/biosíntesis , Oxidación-Reducción , Proteínas Proto-Oncogénicas c-akt/fisiología , Ratas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...