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.
Wiley Interdiscip Rev RNA ; 15(3): e1862, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38837618

RESUMEN

Cell death plays a crucial role in various physiological and pathological processes. Until recently, programmed cell death was mainly attributed to caspase-dependent apoptosis. However, emerging evidence suggests that caspase-independent cell death (CICD) mechanisms also contribute significantly to cellular demise. We and others have reported and functionally characterized numerous long noncoding RNAs (lncRNAs) that modulate caspase-dependent apoptotic pathways potentially in a pathway-dependent manner. However, the interplay between lncRNAs and CICD pathways has not been comprehensively documented. One major reason for this is that most CICD pathways have been recently discovered with some being partially characterized at the molecular level. In this review, we discuss the emerging evidence that implicates specific lncRNAs in the regulation and execution of CICD. We summarize the diverse mechanisms through which lncRNAs modulate different forms of CICD, including ferroptosis, necroptosis, cuproptosis, and others. Furthermore, we highlight the intricate regulatory networks involving lncRNAs, protein-coding genes, and signaling pathways that orchestrate CICD in health and disease. Understanding the molecular mechanisms and functional implications of lncRNAs in CICD may unravel novel therapeutic targets and diagnostic tools for various diseases, paving the way for innovative strategies in disease management and personalized medicine. This article is categorized under: RNA in Disease and Development > RNA in Disease.


Asunto(s)
Muerte Celular , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , Animales , Muerte Celular/genética , Caspasas/metabolismo , Caspasas/genética , Transducción de Señal , Apoptosis/genética
2.
Mol Microbiol ; 120(6): 845-873, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37818865

RESUMEN

Thermostable direct haemolysin (TDH) is the key virulence factor secreted by the human gastroenteric bacterial pathogen Vibrio parahaemolyticus. TDH is a membrane-damaging pore-forming toxin. It evokes potent cytotoxicity, the mechanism of which still remains under-explored. Here, we have elucidated the mechanistic details of cell death response elicited by TDH. Employing Caco-2 intestinal epithelial cells and THP-1 monocytic cells, we show that TDH induces some of the hallmark features of apoptosis-like programmed cell death. TDH triggers caspase-3 and 7 activations in the THP-1 cells, while caspase-7 activation is observed in the Caco-2 cells. Interestingly, TDH appears to induce caspase-independent cell death. Higher XIAP level and lower Smac/Diablo level upon TDH intoxication provide plausible explanation for the functional inability of caspases in the THP-1 cells, in particular. Further exploration reveals that mitochondria play a central role in the TDH-induced cell death. TDH triggers mitochondrial damage, resulting in the release of AIF and endonuclease G, responsible for the execution of caspase-independent cell death. Among the other critical mediators of cell death, ROS is found to play an important role in the THP-1 cells, while PARP-1 appears to play a critical role in the Caco-2 cells. Altogether, our work provides critical new insights into the mechanism of cell death induction by TDH, showing a common central theme of non-classical programmed cell death. Our study also unravels the interplay of crucial molecules in the underlying signalling processes. Our findings add valuable insights into the role of TDH in the context of the host-pathogen interaction processes.


Asunto(s)
Vibrio parahaemolyticus , Humanos , Células CACO-2 , Apoptosis , Caspasas
3.
Biomol Ther (Seoul) ; 31(5): 526-535, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37226044

RESUMEN

Breast cancer is the most common cancer and a frequent cause of cancer-related deaths among women wordlwide. As therapeutic strategies for breast cancer have limitations, novel chemotherapeutic reagents and treatment strategies are needed. In this study, we investigated the anti-cancer effect of synthetic homoisoflavane derivatives of cremastranone on breast cancer cells. Homoisoflavane derivatives, SH-17059 and SH-19021, reduced cell proliferation through G2/M cell cycle arrest and induced caspase-independent cell death. These compounds increased heme oxygenase-1 (HO-1) and 5-aminolevulinic acid synthase 1 (ALAS1), suggesting downregulation of heme. They also induced reactive oxygen species (ROS) generation and lipid peroxidation. Furthermore, they reduced expression of glutathione peroxidase 4 (GPX4). Therefore, we suggest that the SH-17059 and SH-19021 induced the caspase-independent cell death through the accumulation of iron from heme degradation, and the ferroptosis might be one of the potential candidates for caspase-independent cell death.

4.
Biol Pharm Bull ; 46(2): 177-186, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36724946

RESUMEN

Huntington's disease (HD) is a hereditary neurodegenerative disease that involves an expansion of the CAG repeats of the Huntingtin (HTT) gene, but the disease onset and progression do not necessarily correspond to the extent of CAG repeats. Decreased mitochondrial complex II activity has also been reported to be closely associated with disease pathogenesis. Here, we examined the mechanism of cell death induced by 3-nitropropionic acid (3-NP), a mitochondrial complex II inhibitor, using striatal cells (STHdhQ111 cells) derived from HD model mice with mutant HTT carrying the CAG repeat extended. Treatment with 3-NP (5 mM) enhanced cell death in STHdhQ111 compared to STHdhQ7 cells with normal HTT. Ferrostatin-1, an inhibitor of ferroptosis, and deferoxamine, an iron chelator, markedly inhibited 3-NP-induced cell death in both the STHdh cell lines. On the other hands, cell death was not abrogated by a broad-spectrum caspase inhibitor, Z-VAD-FMK, indicating that this cell death was caspase-independent. Cell death caused by 3-NP is suggested to be due to ferroptosis. Furthermore, 3-NP-induced cell death was markedly inhibited by GSK2795039, a reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) inhibitor, suggesting that cell death is mainly mediated by intracellular superoxide anion (O2-) production through NOX2. Furthermore, a mitochondria-targeted superoxide dismutase mimetic (Mito-TEMPO), partially inhibited 3-NP-induced cell death, suggesting that O2- production in the mitochondria is partially responsible for cell death. These results indicate that 3-NP-induced cell death in the STHdhQ111 cells is caspase-independent, non-apoptotic, and that ferroptotic cell death is mainly induced via NOX2 activation.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Ratones , Animales , Especies Reactivas de Oxígeno/metabolismo , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas del Tejido Nervioso/metabolismo , Caspasas/metabolismo , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo
5.
FEBS J ; 289(12): 3477-3488, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35029032

RESUMEN

Adult T-cell leukemia/lymphoma (ATL) develops after a long period of human T-cell leukemia virus (HTLV)-1 infection and is associated with host aging in addition to genetic abnormalities in HTLV-1 infected cells. SIRT1 is a histone deacetylase involved in cell cycle and apoptosis. We previously showed the high expression of SIRT1 protein in peripheral blood mononuclear cells from patients with ATL. There have been many reports that SIRT1 inhibitors show tumor-suppressive effects. On the other hand, SIRT1 activator SRT1720 induces the cell death of multiple myeloma and breast cancer cells. However, the effect of SRT1720 on ATL is unknown. This study aimed to evaluate the effect of SRT1720 on cell death in leukemic cell lines in vitro and freshly isolated ATL cells ex vivo and in an ATL in vivo mouse model. SRT1720 reduced cell viability in vitro and ex vivo. Additionally, SRT1720 increased the number of apoptotic cells, as shown by annexin V positive cells, cleaved poly (ADP-ribose) polymerase 1, cleaved caspase-3, and fragmented DNA. SRT1720 also induced mitochondrial outer membrane permeabilization with the generation of mitochondrial reactive oxygen species and autophagy. However, SIRT1 knockdown did not attenuate SRT1720-induced cell death in leukemic cell lines. Finally, SRT1720 treatment decreased the growth of human ATL tumor xenografts in immunodeficient mice. Our study shows that while SRT1720 does not target SIRT1, it induces cell death in ATL cells via apoptosis and autophagy and has antitumor activity.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos , Leucemia-Linfoma de Células T del Adulto , Animales , Apoptosis , Muerte Celular , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucocitos Mononucleares/metabolismo , Ratones , Sirtuina 1/genética , Sirtuina 1/metabolismo
6.
Chem Biol Interact ; 343: 109433, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33689707

RESUMEN

Pancreatic cancer is one of the most malignant cancers around the world. The co-occurrence of mutation in KRAS and p53 makes it highly aggressive, proliferative, metastatic, and resistant to apoptotic cell death. Therefore, there is a need to trigger an alternate mechanism of cancer cell death in apoptosis-resistant pancreatic cancer. Autophagic cell death could be an alternate viable option for treatment in such cases. Thus, the identification of small molecules as autophagy modulators with potent anticancer efficacy would be of great importance in pancreatic cancer. The present study investigates fluorinated thiazolidionol (FTZ) driven autophagy modulation, underlying mechanism, and regulation of critical sentinels of oncogenic signaling in pancreatic cancer cells. We identified that FTZ triggered autophagic cell death in pancreatic cancer cells, independent of apoptosis evidenced by an increase in cytoplasmic vacuoles formation, autophagy flux, LC3-II expression, and p62 degradation. Further, the crucial events of apoptosis i.e., Caspase-3 activation and PARP cleavage, were not observed, indicating the non-occurrence of apoptotic cell death. Moreover, FTZ was able to activate AMPK and suppress PI3k/Akt/mTOR as well as MEK/ERK, the key oncogenic signaling pathways in cancer cells. Furthermore, treatment with FTZ suppressed migration, invasion, and angiogenesis in pancreatic cancer cells. Studies in vivo revealed significant regression of tumors by FTZ in nude mice model. Overall, our study demonstrates that FTZ induces autophagic cell death in pancreatic cancer cells independent of apoptosis, which is accompanied by AMPK activation and suppression of critical sentinels of oncogenic signaling in pancreatic cancer cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/uso terapéutico , Muerte Celular Autofágica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Tiazoles/uso terapéutico , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones Desnudos , Tiazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
ChemMedChem ; 16(12): 1945-1955, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33682316

RESUMEN

In this work, we describe how stereochemically complex polycyclic compounds can be generated by applying a synthetic sequence comprising an intramolecular Ugi reaction followed by a Pictet-Spengler cyclization on steroid-derived scaffolds. The resulting compounds, which combine a fragment derived from a natural product and a scaffold not found in nature. are both structurally distinct and globally similar to natural products at the same time, and interrogate an alternative region of the chemical space. One of the new compounds showed significant antiproliferative activity on HepG2 cells through a caspase-independent cell-death mechanism, an appealing feature when new antitumor compounds are searched.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Caspasas/metabolismo , Isoquinolinas/farmacología , Piperazinas/farmacología , Esteroides/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Productos Biológicos/síntesis química , Productos Biológicos/química , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células Hep G2 , Humanos , Isoquinolinas/síntesis química , Isoquinolinas/química , Estructura Molecular , Piperazinas/síntesis química , Piperazinas/química , Estereoisomerismo , Esteroides/síntesis química , Esteroides/química
8.
Int J Radiat Biol ; 97(3): 394-400, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33320756

RESUMEN

PURPOSE: Caspases are common mediators of cell death. Evasion of cell death including apoptosis are considered to be hallmarks of cancer. A deeper understanding of the apoptotic cascade may aid improving cancer therapies. Our aim was to characterize the progression of cell death following UV-induced genotoxic injury in a defined cell culture model. MATERIALS AND METHODS: Hela cells were UV-irradiated with doses ranging from 0.1 to 60 mJ/cm2. Cells were counted and colony forming assays were performed with caspase inhibitors. RESULTS: In our model of HeLa cells, cells remain >90% viable until 6 hrs after UV radiation (UVR), but more than half of the cells are dead after 12 - 72 hrs after UVR. Within a dose range between 0.1 and 50 mJ/cm2, viability ranges roughly between 20 and 30%. The difference between the lowest dose applied (0.1 mJ/cm2) and the other doses applied is significant, with the exception of the next higher dose of 1 mJ/cm2. The activation of caspases precedes the cell death induction by several hrs. Caspase-9 starts to be activated at 1 hr after UVR followed by caspases 3, 6 and 7 which are fully active at 2 hrs after UVR while caspase-8 is fully active only 3 hrs after UVR. Most caspases are only weakly or not active at 0.1 mJ/cm2 after 3 hrs, but fully active at the same time point with increased radiation doses. PARP-1, a caspase substrate, is cleaved immediately after activation of the caspases. Colony formation activity of the tumor cells decreases exponentially after UVR dropping down to < 0.01% plating efficiency at a dose of 60 mJ/cm2. Interestingly, this drop in plating efficiency cannot be rescued by any of the two caspase inhibitors tested. CONCLUSIONS: UV-induced cell death in this model involves the activation of apoptosis-related caspases, but this activation seems to be dispensable for the execution of cell death. Further experiments should clarify which mechanisms of cell death are really necessary for the execution of this type of cell death.


Asunto(s)
Caspasas/metabolismo , Daño del ADN , Rayos Ultravioleta/efectos adversos , Supervivencia Celular/efectos de la radiación , Activación Enzimática , Células HeLa , Humanos
9.
Mol Metab ; 40: 101027, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32480041

RESUMEN

OBJECTIVES: Apoptosis-Inducing Factor (AIF) is a protein involved in mitochondrial electron transport chain assembly/stability and programmed cell death. The relevant role of this protein is underlined because mutations altering mitochondrial AIF properties result in acute pediatric mitochondriopathies and tumor metastasis. By generating an original AIF-deficient mouse strain, this study attempted to analyze, in a single paradigm, the cellular and developmental metabolic consequences of AIF loss and the subsequent oxidative phosphorylation (OXPHOS) dysfunction. METHODS: We developed a novel AIF-deficient mouse strain and assessed, using molecular and cell biology approaches, the cellular, embryonic, and adult mice phenotypic alterations. Additionally, we conducted ex vivo assays with primary and immortalized AIF knockout mouse embryonic fibroblasts (MEFs) to establish the cell death characteristics and the metabolic adaptive responses provoked by the mitochondrial electron transport chain (ETC) breakdown. RESULTS: AIF deficiency destabilized mitochondrial ETC and provoked supercomplex disorganization, mitochondrial transmembrane potential loss, and high generation of mitochondrial reactive oxygen species (ROS). AIF-/Y MEFs counterbalanced these OXPHOS alterations by mitochondrial network reorganization and a metabolic reprogramming toward anaerobic glycolysis illustrated by the AMPK phosphorylation at Thr172, the overexpression of the glucose transporter GLUT-4, the subsequent enhancement of glucose uptake, and the anaerobic lactate generation. A late phenotype was characterized by the activation of P53/P21-mediated senescence. Notably, approximately 2% of AIF-/Y MEFs diminished both mitochondrial mass and ROS levels and spontaneously proliferated. These cycling AIF-/Y MEFs were resistant to caspase-independent cell death inducers. The AIF-deficient mouse strain was embryonic lethal between E11.5 and E13.5 with energy loss, proliferation arrest, and increased apoptotic levels. Contrary to AIF-/Y MEFs, the AIF KO embryos were unable to reprogram their metabolism toward anaerobic glycolysis. Heterozygous AIF+/- females displayed progressive bone marrow, thymus, and spleen cellular loss. In addition, approximately 10% of AIF+/- females developed perinatal hydrocephaly characterized by brain development impairment, meningeal fibrosis, and medullar hemorrhages; those mice died 5 weeks after birth. AIF+/- with hydrocephaly exhibited loss of ciliated epithelium in the ependymal layer. This phenotype was triggered by the ROS excess. Accordingly, it was possible to diminish the occurrence of hydrocephalus AIF+/- females by supplying dams and newborns with an antioxidant in drinking water. CONCLUSIONS: In a single knockout model and at 3 different levels (cell, embryo, and adult mice) we demonstrated that by controlling the mitochondrial OXPHOS/metabolism, AIF is a key factor regulating cell differentiation and fate. Additionally, by providing new insights into the pathological consequences of mitochondrial OXPHOS dysfunction, our new findings pave the way for novel pharmacological strategies.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Animales , Apoptosis/fisiología , Caspasas/metabolismo , Respiración de la Célula , Femenino , Fibroblastos/metabolismo , Ingeniería Genética/métodos , Glucólisis/genética , Hidrocefalia/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/genética , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos/genética , Mitocondrias/metabolismo , Modelos Animales , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo
10.
Phytother Res ; 34(1): 173-183, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31515931

RESUMEN

We have investigated the potential cell death mechanism promoted by Coriolus versicolor fungus-derived protein-bound polysaccharides (PBPs) in melanoma cells. Knowing that melanogenesis has the potential to affect the tumor behavior and melanoma therapy outcome, the cytotoxic effects of PBPs were evaluated in human SKMel-188 melanoma cell line, whose phenotype, amelanotic versus pigmented, depends on the concentration of melanin precursors in the culture medium. Our results showed that inhibitory effect of PBPs (100 and 200 µg/ml) towards melanoma cells is inversely associated with the pigmentation level. This cytotoxicity induced in nonpigmented melanoma cells by PBPs was caspase-independent; however, it was accompanied by an increased intracellular reactive oxygen species (ROS) generation. The ROS production was controlled by c-Jun N-terminal kinase (JNK) because SP600125, a JNK inhibitor, significantly reduced ROS generation and protected cells against PBPs-induced death. We also found that PBPs-induced lactate dehydrogenase release in amelanotic melanoma cells was abolished by co-treatment with receptor-interacting serine/threonine-protein kinase 1 inhibitor, implying engagement of this kinase in PBPs-induced death pathway. The results suggest that PBPs induce an alternative programmed cell death, regulated by receptor-interacting protein-1 and ROS and that this process is modified by melanin content in melanoma cells. These findings are remarkable when considering the use of commercially available Coriolus versicolor by patients who suffer from melanoma cancer.


Asunto(s)
Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Melanoma/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Humanos , Polisacáridos/metabolismo , Especies Reactivas de Oxígeno
11.
Biochem Pharmacol ; 173: 113724, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31756327

RESUMEN

Earlier studies from our laboratory have demonstrated that Oxyresveratrol (OXY), a hydroxyl-substituted stilbene, exhibits potent inhibition of human melanoma cell proliferation. The present study defines a cytotoxic effect of OXY on the highly chemo-resistant, triple-negative human breast cancer cell line MDA-MB-231. OXY-mediated cell death resulted in accumulation of cells at the sub-G1 phase of the cell cycle, induced chromatin condensation, DNA fragmentation, phosphatidylserine externalization and PARP cleavage, indicative of apoptosis. Interestingly, morphology and cell viability studies with the pan-caspase inhibitor, QVD-OPH revealed that OXY-induced cell death was caspase-independent. Docking studies also showed that OXY can bind to the S1 site of caspase-3, and could also exert an inhibitory effect on this executioner caspase. The immunoblot analysis demonstrating the absence of caspase cleavage during cell death further confirmed these findings. OXY was also observed to induce the production of reactive oxygen species, which caused the depolarization of the mitochondrial membrane resulting in translocation of Apoptosis Inducing Factor (AIF) into the nucleus. Pretreatment of the cells with N-Acetyl Cysteine antioxidant prevented cell death resulting from OXY treatment. Thus, OXY initiates ROS-mediated, apoptosis-like cell death, involving mitochondrial membrane depolarization, translocation of AIF into the nucleus, and DNA fragmentation, resulting in caspase-independent cell death in MDA-MB-231 cells. The cytotoxicity manifested by OXY was also observed in 3D cell culture models and primary cells, thereby providing a basis for the utilization of OXY as a novel template for the future design of anticancer therapeutics.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Caspasas/metabolismo , Mitocondrias/efectos de los fármacos , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/metabolismo , Estilbenos/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Caspasa 3/química , Caspasa 3/metabolismo , Caspasas/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Simulación del Acoplamiento Molecular , Extractos Vegetales/química , Extractos Vegetales/metabolismo , Unión Proteica , Estilbenos/química , Estilbenos/metabolismo
12.
Cancers (Basel) ; 11(9)2019 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-31505859

RESUMEN

X-linked inhibitor of apoptosis protein (XIAP) is an emerging crucial therapeutic target in cancer. We report on the discovery and characterisation of small organic molecules from Piper genus plants exhibiting XIAP antagonism, namely erioquinol, a quinol substituted in the 4-position with an alkenyl group and the alkenylphenols eriopodols A-C. Another isolated compound was originally identified as gibbilimbol B. Erioquinol was the most potent inhibitor of human cancer cell viability when compared with gibbilimbol B and eriopodol A was listed as intermediate. Gibbilimbol B and eriopodol A induced apoptosis through mitochondrial permeabilisation and caspase activation while erioquinol acted on cell fate via caspase-independent/non-apoptotic mechanisms, likely involving mitochondrial dysfunctions and aberrant generation of reactive oxygen species. In silico modelling and molecular approaches suggested that all molecules inhibit XIAP by binding to XIAP-baculoviral IAP repeat domain. This demonstrates a novel aspect of XIAP as a key determinant of tumour control, at the molecular crossroad of caspase-dependent/independent cell death pathway and indicates molecular aspects to develop tumour-effective XIAP antagonists.

13.
Neuroscience ; 400: 72-84, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30625334

RESUMEN

Spino-cerebellar ataxia type 7 (SCA7) is a polyglutamine (polyQ) disorder characterized by neurodegeneration of the brain, cerebellum, and retina caused by a polyglutamine expansion in ataxin7. The presence of an expanded polyQ tract in a mutant protein is known to induce protein aggregation, cellular stress, toxicity, and finally cell death. However, the consequences of the presence of mutant ataxin7 in the retina and the mechanisms underlying photoreceptor degeneration remain poorly understood. In this study, we show that in a retinal SCA7 mouse model, polyQ ataxin7 induces stress within the retina and activates Muller cells. Moreover, unfolded protein response and autophagy are activated in SCA7 photoreceptors. We have also shown that the photoreceptor death does not involve a caspase-dependent apoptosis but instead involves apoptosis inducing factor (AIF) and Leukocyte Elastase Inhibitor (LEI/L-DNase II). When these two cell death effectors are downregulated by their siRNA, a significant reduction in photoreceptor death is observed. These results highlight the consequences of polyQ protein expression in the retina and the role of caspase-independent pathways involved in photoreceptor cell death.


Asunto(s)
Ataxina-7/metabolismo , Muerte Celular , Péptidos/metabolismo , Degeneración Retiniana/metabolismo , Ataxias Espinocerebelosas/metabolismo , Animales , Factor Inductor de la Apoptosis/metabolismo , Ataxina-7/genética , Calpaína/metabolismo , Caspasas/metabolismo , Catepsinas/metabolismo , Modelos Animales de Enfermedad , Endodesoxirribonucleasas/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/etiología , Transducción de Señal , Ataxias Espinocerebelosas/complicaciones , Estrés Fisiológico
14.
BMC Cancer ; 18(1): 791, 2018 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-30081901

RESUMEN

BACKGROUND: Sirtuin 2 (SIRT2) is a member of the sirtuin family, nicotinamide adenine dinucleotide+-dependent deacylases, which participates in modulation of cell cycle control, neurodegeneration, and tumorigenesis. SIRT2 expression increases in acute myeloid leukemia blasts. Downregulation of SIRT2 using siRNA causes apoptosis of HeLa cells. Therefore, selective inhibitors of SIRT2 are candidate therapeutic agents for cancer. Adult T-cell leukemia/lymphoma (ATL) is a T-cell malignancy that has a poor prognosis and develops after long-term infection with human T-cell leukemia virus (HTLV)-1. Sirtuin 1 inhibition has been shown to induce apoptosis and autophagy in HTLV-1-infected cell lines, whereas the effects of SIRT2 inhibition alone have not been elucidated. METHODS: We assessed the efficacy of our small molecule selective SIRT2 inhibitors NCO-90/141 to induce leukemic cell death. Cell viability was examined using the cell proliferation reagent Cell Count Reagent SF. Apoptotic cells were detected by annexin V-FITC and terminal deoxynucleotidyl transferase dUTP nick end labeling assays by flow cytometry. Caspase activity was detected using an APOPCYTO Intracellular Caspase Activity Detection Kit. The presence of autophagic vacuoles was assessed using a Cyto-ID Autophagy Detection Kit. RESULTS: Our novel small molecule SIRT2-specific inhibitors NCO-90/141 inhibited cell growth of leukemic cell lines including HTLV-1-transformed T-cells. NCO-90/141 induced apoptosis via caspase activation and mitochondrial superoxide generation in leukemic cell lines. However, a caspase inhibitor did not prevent this caspase-associated cell death. Interestingly, NCO-90/141 increased the LC3-II level together with autophagosome accumulation, indicating autophagic cell death. Thus, NCO-90/141 simultaneously caused apoptosis and autophagy. CONCLUSIONS: These results suggest that NCO-90/141 are highly effective against leukemic cells in caspase-dependent or -independent manners via autophagy, and they may have a novel therapeutic potential for treatment of leukemias including ATL.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Leucemia/tratamiento farmacológico , Sirtuina 2/antagonistas & inhibidores , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Células HL-60 , Humanos , Células Jurkat , Leucemia/enzimología , Leucemia/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/patología , Transducción de Señal/efectos de los fármacos , Sirtuina 2/metabolismo , Superóxidos/metabolismo
15.
BMC Cell Biol ; 19(1): 11, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973136

RESUMEN

BACKGROUND: Apoptosis, the most well-known type of programmed cell death, can induce in a paracrine manner a proliferative response in neighboring surviving cells called apoptosis-induced proliferation (AiP). While having obvious benefits when triggered in developmental processes, AiP is a serious obstacle in cancer therapy, where apoptosis is frequently induced by chemotherapy. Therefore, in this study, we evaluated the capacity of an alternative type of cell death, called caspase-independent cell death, to promote proliferation. RESULTS: Using a novel in vitro isogenic cellular model to trigger either apoptosis or caspase-independent cell death, we found that the later has no obvious compensatory proliferation effects on neighboring cells. CONCLUSIONS: This study enforces the idea that alternative types of cell death such as caspase-independent cell death could be considered to replace apoptosis in the context of cancer treatment.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Melanoma/enzimología , Melanoma/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Modelos Biológicos
16.
Biomed Pharmacother ; 103: 18-28, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29635124

RESUMEN

Phosphoethanolamine (PEA) is a fundamental precursor during the biosynthesis of cell membranes phospholipids. In the past few years, it has been described as a potential antitumor agent. In previous studies, we demonstrated that PEA showed antitumor properties in vitro and in vivo in a wide range of tumor cell lines. Herein, we showed that PEA possesses cytotoxic properties and notably revealed to induce caspase-independent cell death. Of interest, we provided evidence that PEA inhibits melanoma cells proliferation through the reduction of C-RAF. Molecular docking of PEA evidenced that this compound indeed fits satisfactory in the binding site located between the dimers of C-RAF protein with 107,01 Šand score of -29,62. Also, PEA arrested A2058 cells at G2/M phase in the cell cycle. Moreover, cell proliferation, migration and adhesion capacities of A2058 cells were also inhibited by PEA. Most importantly, PEA inhibited tumor growth of melanoma tumors and prolonged survival rate of mice. Also, PEA induced a significant immune response in a syngeneic metastatic melanoma model. Taken together, these data indicate that PEA is a promising candidate for future developments in cancer field.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Etanolaminas/farmacología , Melanoma/patología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Antineoplásicos/farmacología , Sitios de Unión , Puntos de Control del Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Etanolaminas/química , Humanos , Melanoma/enzimología , Melanoma/inmunología , Melanoma Experimental/enzimología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Metástasis de la Neoplasia , Fosforilación/efectos de los fármacos , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
18.
Vet Comp Oncol ; 16(1): 69-76, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28371355

RESUMEN

BACKGROUND: SRC kinase (SRC proto-oncogene, non-receptor tyrosine kinase) is a promising target for the treatment of solid cancers including human melanoma. Bosutinib (Bosu), a SRC inhibitor, has already been applied to the treatment of human chronic myelogenous leukemia and also has been assessed its safety in dogs. AIM: The aim of this study was to clarify a novel anti-tumour mechanism of Bosu in canine and human melanoma cells. MATERIALS AND METHODS: The canine and human melanoma cells were treated with Bosu and its effects were evaluated by the cell viability, the protein expression levels such as caspase-3 and LC3, Annexin V/Propidium iodide staining, and confocal immunostaining. RESULTS: Bosu induced the massive caspase-independent cell death, and blocked autophagy flux, which resulted from lysosomal dysfunction. Lysosomal dysfunction caused by Bosu was due to lysosomal membrane permeabilization (LMP), which resulted in the release of lysosomal hydrolases including cathepsin B. CONCLUSION: Our data suggest that Bosu induces the cell death through induction of LMP in melanoma cells and is a promising therapeutic agent for treatment of melanoma in both dogs and humans.


Asunto(s)
Compuestos de Anilina/farmacología , Muerte Celular/efectos de los fármacos , Lisosomas/efectos de los fármacos , Melanoma/tratamiento farmacológico , Nitrilos/farmacología , Quinolinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Western Blotting , Caspasas/metabolismo , Línea Celular Tumoral , Enfermedades de los Perros/tratamiento farmacológico , Perros , Melanoma/veterinaria , Microscopía Confocal , Proto-Oncogenes Mas
19.
Mol Biol Rep ; 44(4): 323-332, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28748410

RESUMEN

Recent reports on various cancer models demonstrate a great potential of cytosine deaminase/5-fluorocytosine suicide system in cancer therapy. However, this approach has limited success and its application to patients has not reached the desirable clinical significance. Accordingly, the improvement of this suicide system is an actively developing trend in gene therapy. The purpose of this study was to explore the cytotoxic effect observed after co-expression of hepatitis A virus 3C protease (3C) and yeast cytosine deaminase/uracil phosphoribosyltransferase fusion protein (FCU1) in a bicistronic vector. A set of mono- and bicistronic plasmid constructs was generated to provide individual or combined expression of 3C and FCU1. The constructs were introduced into HEK293 and HeLa cells, and target protein synthesis as well as the effect of 5-fluorocytosine on cell death and the time course of the cytotoxic effect was studied. The obtained vectors provide for the synthesis of target proteins in human cells. The expression of the genes in a bicistronic construct provide for the cytotoxic effect comparable to that observed after the expression of genes in monocistronic constructs. At the same time, co-expression of FCU1 and 3C recapitulated their cytotoxic effects. The combined effect of the killer and suicide genes was studied for the first time on human cells in vitro. The integration of different gene therapy systems inducing cell death (FCU1 and 3C genes) in a bicistronic construct allowed us to demonstrate that it does not interfere with the cytotoxic effect of each of them. A combination of cytotoxic genes in multicistronic vectors can be used to develop pluripotent gene therapy agents.


Asunto(s)
Cisteína Endopeptidasas/biosíntesis , Citosina Desaminasa/biosíntesis , Flucitosina/farmacología , Terapia Genética/métodos , Virus de la Hepatitis A Humana/enzimología , Pentosiltransferasa/biosíntesis , Proteínas Virales/biosíntesis , Proteasas Virales 3C , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Flucitosina/farmacocinética , Genes Transgénicos Suicidas , Vectores Genéticos , Células HEK293 , Células HeLa , Virus de la Hepatitis A Humana/metabolismo , Humanos , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Plásmidos/genética , Profármacos/farmacocinética , Profármacos/farmacología , Transducción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
BMC Cancer ; 17(1): 123, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28187758

RESUMEN

BACKGROUND: Photodynamic therapy (PDT) has proven to be a promising alternative to current cancer treatments, especially if combined with conventional approaches. The technique is based on the administration of a non-toxic photosensitizing agent to the patient with subsequent localized exposure to a light source of a specific wavelength, resulting in a cytotoxic response to oxidative damage. The present study intended to evaluate in vitro the type of induced death and the genotoxic and mutagenic effects of PDT alone and associated with cisplatin. METHODS: We used the cell lines SiHa (ATCC® HTB35™), C-33 A (ATCC® HTB31™) and HaCaT cells, all available at Dr. Christiane Soares' Lab. Photosensitizers were Photogem (PGPDT) and methylene blue (MBPDT), alone or combined with cisplatin. Cell death was accessed through Hoechst and Propidium iodide staining and caspase-3 activity. Genotoxicity and mutagenicity were accessed via flow cytometry with anti-gama-H2AX and micronuclei assay, respectively. Data were analyzed by one-way ANOVA with Tukey's posthoc test. RESULTS: Both MBPDT and PGPDT induced caspase-independent death, but MBPDT induced the morphology of typical necrosis, while PGPDT induced morphological alterations most similar to apoptosis. Cisplatin predominantly induced apoptosis, and the combined therapy induced variable rates of apoptosis- or necrosis-like phenotypes according to the cell line, but the percentage of dead cells was always higher than with monotherapies. MBPDT, either as monotherapy or in combination with cisplatin, was the unique therapy to induce significant damage to DNA (double strand breaks) in the three cell lines evaluated. However, there was no mutagenic potential observed for the damage induced by MBPDT, since the few cells that survived the treatment have lost their clonogenic capacity. CONCLUSIONS: Our results elicit the potential of combined therapy in diminishing the toxicity of antineoplastic drugs. Ultimately, photodynamic therapy mediated by either methylene blue or Photogem as monotherapy or in combination with cisplatin has low mutagenic potential, which supports its safe use in clinical practice for the treatment of cervical cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Cisplatino/farmacología , Luz , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular , Línea Celular Tumoral , Femenino , Histonas/metabolismo , Humanos , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Micronúcleos con Defecto Cromosómico/efectos de la radiación , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Fotoquimioterapia/métodos , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA