Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Immunity ; 38(4): 729-41, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23562161

RESUMEN

The therapeutic efficacy of anthracyclines relies on antitumor immune responses elicited by dying cancer cells. How chemotherapy-induced cell death leads to efficient antigen presentation to T cells, however, remains a conundrum. We found that intratumoral CD11c(+)CD11b(+)Ly6C(hi) cells, which displayed some characteristics of inflammatory dendritic cells and included granulomonocytic precursors, were crucial for anthracycline-induced anticancer immune responses. ATP released by dying cancer cells recruited myeloid cells into tumors and stimulated the local differentiation of CD11c(+)CD11b(+)Ly6C(hi) cells. Such cells efficiently engulfed tumor antigens in situ and presented them to T lymphocytes, thus vaccinating mice, upon adoptive transfer, against a challenge with cancer cells. Manipulations preventing tumor infiltration by CD11c(+)CD11b(+)Ly6C(hi) cells, such as the local overexpression of ectonucleotidases, the blockade of purinergic receptors, or the neutralization of CD11b, abolished the immune system-dependent antitumor activity of anthracyclines. Our results identify a subset of tumor-infiltrating leukocytes as therapy-relevant antigen-presenting cells.


Asunto(s)
Antraciclinas/administración & dosificación , Células Presentadoras de Antígenos/inmunología , Antineoplásicos/administración & dosificación , Células Dendríticas/inmunología , Neoplasias Experimentales/inmunología , Traslado Adoptivo , Animales , Antraciclinas/efectos adversos , Antígenos Ly/metabolismo , Antígenos de Neoplasias/inmunología , Antineoplásicos/efectos adversos , Apoptosis , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Precursoras de Granulocitos/inmunología , Inmunidad Celular , Ratones , Ratones Endogámicos C57BL , Células Precursoras de Monocitos y Macrófagos/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Nucleotidasas/metabolismo , Receptores Purinérgicos/metabolismo
2.
Proc Natl Acad Sci U S A ; 111(8): 3020-5, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24516128

RESUMEN

Tetraploidy constitutes a genomically metastable state that can lead to aneuploidy and genomic instability. Tetraploid cells are frequently found in preneoplastic lesions, including intestinal cancers arising due to the inactivation of the tumor suppressor adenomatous polyposis coli (APC). Using a phenotypic screen, we identified resveratrol as an agent that selectively reduces the fitness of tetraploid cells by slowing down their cell cycle progression and by stimulating the intrinsic pathway of apoptosis. Selective killing of tetraploid cells was observed for a series of additional agents that indirectly or directly stimulate AMP-activated protein kinase (AMPK) including salicylate, whose chemopreventive action has been established by epidemiological studies and clinical trials. Both resveratrol and salicylate reduced the formation of tetraploid or higher-order polyploid cells resulting from the culture of human colon carcinoma cell lines or primary mouse epithelial cells lacking tumor protein p53 (TP53, best known as p53) in the presence of antimitotic agents, as determined by cytofluorometric and videomicroscopic assays. Moreover, oral treatment with either resveratrol or aspirin, the prodrug of salicylate, repressed the accumulation of tetraploid intestinal epithelial cells in the Apc(Min/+) mouse model of colon cancer. Collectively, our results suggest that the chemopreventive action of resveratrol and aspirin involves the elimination of tetraploid cancer cell precursors.


Asunto(s)
Poliposis Adenomatosa del Colon/prevención & control , Aspirina/uso terapéutico , Muerte Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Estilbenos/uso terapéutico , Tetraploidía , Animales , Aspirina/farmacología , Línea Celular Tumoral , Células Epiteliales/química , Citometría de Flujo , Procesamiento de Imagen Asistido por Computador , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos C57BL , Microscopía por Video , Resveratrol , Estilbenos/farmacología
3.
EMBO J ; 30(24): 4908-20, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22081109

RESUMEN

Autophagic responses are coupled to the activation of the inhibitor of NF-κB kinase (IKK). Here, we report that the essential autophagy mediator Beclin 1 and TGFß-activated kinase 1 (TAK1)-binding proteins 2 and 3 (TAB2 and TAB3), two upstream activators of the TAK1-IKK signalling axis, constitutively interact with each other via their coiled-coil domains (CCDs). Upon autophagy induction, TAB2 and TAB3 dissociate from Beclin 1 and bind TAK1. Moreover, overexpression of TAB2 and TAB3 suppresses, while their depletion triggers, autophagy. The expression of the C-terminal domain of TAB2 or TAB3 or that of the CCD of Beclin 1 competitively disrupts the interaction between endogenous Beclin 1, TAB2 and TAB3, hence stimulating autophagy through a pathway that requires endogenous Beclin 1, TAK1 and IKK to be optimally efficient. These results point to the existence of an autophagy-stimulatory 'switch' whereby TAB2 and TAB3 abandon inhibitory interactions with Beclin 1 to engage in a stimulatory liaison with TAK1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Beclina-1 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Estructura Terciaria de Proteína , Técnicas del Sistema de Dos Híbridos
4.
EMBO J ; 29(7): 1272-84, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20186124

RESUMEN

Tetraploidy can constitute a metastable intermediate between normal diploidy and oncogenic aneuploidy. Here, we show that the absence of p53 is not only permissive for the survival but also for multipolar asymmetric divisions of tetraploid cells, which lead to the generation of aneuploid cells with a near-to-diploid chromosome content. Multipolar mitoses (which reduce the tetraploid genome to a sub-tetraploid state) are more frequent when p53 is downregulated and the product of the Mos oncogene is upregulated. Mos inhibits the coalescence of supernumerary centrosomes that allow for normal bipolar mitoses of tetraploid cells. In the absence of p53, Mos knockdown prevents multipolar mitoses and exerts genome-stabilizing effects. These results elucidate the mechanisms through which asymmetric cell division drives chromosomal instability in tetraploid cells.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Genes mos , Mitosis , Poliploidía , Proteína p53 Supresora de Tumor/metabolismo , Aneuploidia , Animales , Carcinoma/genética , Línea Celular Tumoral , Centrosoma/metabolismo , Inestabilidad Cromosómica , Neoplasias del Colon/genética , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Proteína p53 Supresora de Tumor/genética
5.
Eur J Med Chem ; 264: 115994, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38070431

RESUMEN

Because they hold together molecules by means of non-covalent interactions - relatively weak and thus, potentially reversible - the anionic calixarenes have become an interesting tool for efficiently binding a large range of ligands - from gases to large organic molecules. Being highly water soluble and conveniently biocompatible, they showed growing interest for many interdisciplinary fields, particularly in biology and medicine. Thanks to their intrinsic conical shape, they provide suitable platforms, from vesicles to bilayers. This is a valuable characteristic, as so they mimic the biologically functional architectures. The anionic calixarenes propose efficient alternatives for overcoming the limitations linked to drug delivery and bioavailability, as well as drug resistance along with limiting the undesirable side effects. Moreover, the dynamic non-covalent binding with the drugs enables predictable and on demand drug release, controlled by the stimuli present in the targeted environment. This particular feature instigated the use of these versatile, stimuli-responsive compounds for sensing biomarkers of diverse pathologies. The present review describes the recent achievements of the anionic calixarenes in the field of life science, from drug carriers to biomedical engineering, with a particular outlook on their applications for the diagnosis and treatment of different pathologies.


Asunto(s)
Calixarenos , Calixarenos/química , Sistemas de Liberación de Medicamentos , Portadores de Fármacos , Disponibilidad Biológica , Liberación de Fármacos
6.
Cell Death Dis ; 14(4): 238, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37015922

RESUMEN

Many anticancer agents induce apoptosis, mitotic catastrophe or cellular senescence. Here, we report the functional characterization of an experimental inducer of tumor necrosis factor (TNF)-independent necrosis, necrocide-1 (NC1). NC1 (but not its stereoisomer) killed a panel of human cancer cells (but not normal cells) at nanomolar concentrations and with a non-apoptotic, necrotic morphotype, both in vitro and in vivo. NC1-induced killing was not inhibited by caspase blockers, anti-apoptotic BCL2 overexpression or TNFα neutralization, suggesting that NC1 elicits a bona fide necrotic pathway. However, pharmacological or genetic inhibition of necroptosis, pyroptosis and ferroptosis failed to block NC1-mediated cell death. Instead, NC1 elicited reactive oxygen species (ROS) production by mitochondria, and elimination of mitochondrial DNA, quenching of mitochondrial ROS, as well as blockade of mitochondrial permeability transition with cyclosporine A, interfered with NC1-induced cell death. NC1 induced hallmarks of immunogenic cell death incurring calreticulin (CALR) exposure, ATP secretion and high mobility group box 1 (HMGB1) release. Taken together, these data identify a previously uncharacterized signaling cascade leading to an immunogenic variant of mitochondrion-regulated necrosis, supporting the notion that eliciting regulated necrosis may constitute a valid approach for anticancer therapy.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Especies Reactivas de Oxígeno/metabolismo , Necrosis , Apoptosis , Muerte Celular/fisiología , Antineoplásicos/farmacología , Neoplasias/genética
7.
Cancer Metastasis Rev ; 30(1): 61-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21249425

RESUMEN

The success of some chemo- and radiotherapeutic regimens relies on the induction of immunogenic tumor cell death and on the induction of an anticancer immune response. Cells succumbing to immunogenic cell death undergo specific changes in their surface characteristics and release pro-immunogenic factors according to a defined spatiotemporal pattern. This stimulates antigen presenting cells such as dendritic cells to efficiently take up tumor antigens, process them, and cross-prime cytotoxic T lymphocytes, thus eliciting a tumor-specific cognate immune response. Such a response can also target therapy-resistant tumor (stem) cells, thereby leading, at least in some instances, to tumor eradication. In this review, we shed some light on the molecular identity of the factors that are required for cell death to be perceived as immunogenic. We discuss the intriguing observations that the most abundant endoplasmic reticulum protein, calreticulin, the most abundant intracellular metabolite, ATP, and the most abundant non-histone chromatin-binding protein, HMGB1, can determine whether cell death is immunogenic as they appear on the surface or in the microenvironment of dying cells.


Asunto(s)
Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Muerte Celular/inmunología , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
8.
Bull Mem Acad R Med Belg ; 166(3-4): 130-8; discussion 139-40, 2011.
Artículo en Francés | MEDLINE | ID: mdl-22375493

RESUMEN

Immunogenic cell death, characterized by calreticulin exposure on the surface of the dying cell, release of the nuclear protein high mobility group box 1 (HMGB1), and release of ATP, enables stimulation of the immune system. We outlined the importance of this kind of cell death for the success of some anticancer chemotherapies. However, defects in the immunogenic cell death signalling pathway can lead to therapeutic failure, apparently because anticancer immune responses must contribute to the efficacy of chemotherapeutic regimens. These defects can be related to the therapy, the tumour cell, the host or the tumour-host interface. It is necessary to characterize these defects to restore and improve the efficacy of anticancer chemotherapies.


Asunto(s)
Antineoplásicos/farmacología , Muerte Celular/inmunología , Resistencia a Antineoplásicos/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Antineoplásicos/uso terapéutico , Humanos
9.
Apoptosis ; 14(4): 364-75, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19145485

RESUMEN

It is still enigmatic under which circumstances cellular demise induces an immune response or rather remains immunologically silent. Moreover, the question remains open under which circumstances apoptotic, autophagic or necrotic cells are immunogenic or tolerogenic. Although apoptosis appears to be morphologically homogenous, recent evidence suggests that the pre-apoptotic surface-exposure of calreticulin may dictate the immune response to tumor cells that succumb to anticancer treatments. Moreover, the release of high-mobility group box 1 (HMGB1) during late apoptosis and secondary necrosis contributes to efficient antigen presentation and cytotoxic T-cell activation because HMGB1 can bind to Toll like receptor 4 on dendritic cells, thereby stimulating optimal antigen processing. Cell death accompanied by autophagy also may facilitate cross priming events. Apoptosis, necrosis and autophagy are closely intertwined processes. Often, cells manifest autophagy before they undergo apoptosis or necrosis, and apoptosis is generally followed by secondary necrosis. Whereas apoptosis and necrosis irreversibly lead to cell death, autophagy can clear cells from stress factors and thus facilitate cellular survival. We surmise that the response to cellular stress like chemotherapy or ionizing irradiation, dictates the immunological response to dying cells and that this immune response in turn determines the clinical outcome of anticancer therapies. The purpose of this review is to summarize recent insights into the immunogenicity of dying tumor cells as a function of the cell death modality.


Asunto(s)
Apoptosis/inmunología , Calreticulina/inmunología , Inmunidad/inmunología , Neoplasias/inmunología , Animales , Autofagia , Muerte Celular/inmunología , Humanos , Modelos Biológicos , Necrosis , Neoplasias/patología
10.
Cancers (Basel) ; 11(1)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30641971

RESUMEN

Half of soft-tissue sarcomas are tumors with complex genomics, which display no specific genetic alterations and respond poorly to treatment. It is therefore necessary to find new therapeutic targets for these sarcomas. Despite genetic heterogeneity across samples, oncogenesis may be driven by common pathway alterations. Therefore, genomic and transcriptomic profiles of 106 sarcomas with complex genomics were analyzed to identify common pathways with altered genes. This brought out a gene belonging to the "cell cycle" biological pathway, RCBTB1 (RCC1 And BTB Domain Containing Protein 1), which is lost and downregulated in 62.5% of metastatic tumors against 34% of non-metastatic tumors. A retrospective study of three sarcoma cohorts revealed that low RCBTB1 expression is prognostic for metastatic progression, specifically in patients that received chemotherapy. In vitro and in vivo, RCBTB1 overexpression in leiomyosarcoma cells specifically sensitized to docetaxel-induced apoptosis. This was associated with increased mitotic rate in vitro and higher growth rate of xenografts. By contrast, RCBTB1 inhibition decreased cell proliferation and protected sarcoma cells from apoptosis induced by docetaxel. Collectively, these data evidenced that RCBTB1 is frequently deleted in sarcomas with complex genomics and that its downregulation is associated with a higher risk of developing metastasis for patients receiving chemotherapy, likely due to their higher resistance to docetaxel.

11.
Cancer Treat Rev ; 31(5): 361-79, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15994016

RESUMEN

Uveal melanoma is the most frequent intra-ocular cancer. The recent development of new chromosome-related technologies have permitted the elucidation of both the cytogenetics and the natural history of this disease. Fifty to 60% of uveal melanomas are linked to a monosomy 3, which appears as an early and determinant event in tumor progression. Tumors with this anomaly have a very poor prognosis. Recent work suggests that this category of uveal melanoma represents a distinct pathologic entity from that associated with normal disomy 3. Chromosome 6 aberrations probably constitute a second entry point into the process of cancerogenesis, while gains in 8q seem to appear later in the natural history of uveal melanomas due to their higher frequency in larger tumors. Other anomalies will be reviewed. In spite of significant improvements in the local treatment of uveal melanoma, many patients die due to tumor metastasis. This disease is characterized by a constitutive chemoresistance whose typical multidrug resistance phenotype (MDR) is particularly complex since different combinations of several resistance proteins are simultaneously produced. Regulation of the expression of these proteins is a research priority, increasingly so as gene therapy-dependent chemosensitization strategies expand. Therefore, the development and improvement of methods to determine the chemoresistance profile become a crucial objective today in the therapeutic strategies against uveal melanoma.


Asunto(s)
Aberraciones Cromosómicas , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Terapia Genética , Melanoma/genética , Melanoma/terapia , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/terapia , Antineoplásicos/farmacología , Aberraciones Cromosómicas/efectos de los fármacos , Humanos , Melanoma/tratamiento farmacológico , Monosomía , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Neoplasias de la Úvea/tratamiento farmacológico
12.
Semin Oncol ; 32(6): 583-90, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16338424

RESUMEN

The multidrug resistance (MDR) phenotype of cancers has generated a large amount of research, owing to its constant fatal clinical outcome. Many studies have focused on the discovery of chemomodulators; however, in spite of this huge effort, the side effects that these products induce, and their additive toxicity when used in the presence of anticancer drugs, have led to the disaffection of the pharmaceutical industry and possibly slowed down research in pharmacological modulation. New tools developed using molecular biology techniques have opened the way for gene therapy and given birth to new therapeutic hopes. However, these discoveries and especially their clinical applications have slowed due to a lack of knowledge of the systems that finely regulate the MDR genes. This weakness explains why, to date, no general review has focused on the possibilities of gene therapy of MDR derived form the strategic options now available. Based on molecular foundations and recent fundamental discoveries, we seek to inform clinicians of the therapeutic hopes for chemoresistant tumors brought about by potent and specific new tools such as transcriptional decoys, interfering RNAs, etc. After describing the causes and mechanisms of MDR, we critically review these new strategies and their corresponding clinical trials.


Asunto(s)
Resistencia a Múltiples Medicamentos , Genes MDR , Terapia Genética , Ensayos Clínicos como Asunto , Humanos , Neoplasias/genética , Neoplasias/terapia , Fenotipo , Interferencia de ARN
13.
Oncoimmunology ; 3(7): e944047, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25610726

RESUMEN

There is ample experimental and clinical evidence that chemotherapies are more efficient if they succeed in (re)activating immune surveillance, hence triggering a long-term immune response against residual tumor cells. Most of the preclinical evidence supporting this notion has been obtained with transplantable cancers, for which it has been shown that chemotherapy-induced autophagy in cancer cells is mandatory for the recruitment of myeloid cells into the tumor bed and the subsequent T lymphocyte-mediated reduction in tumor growth. Here, we characterized the chemotherapeutic response of melanomas caused by 4-hydroxy-tamoxifen-induced expression of the Cre recombinase in melanocytes that results in the activation of oncogenic Braf together with the inactivation of the tumor suppressor Pten, as well as the optional inactivation of the essential autophagy gene Atg7. Systemic chemotherapy with the anthracycline Mitoxantrone (MTX) reduced the growth of autophagy-competent melanomas (genotype: BrafCa/+; Ptenfl/fl; Atg7+/+), yet failed to affect the progression of autophagy-deficient melanomas (genotype: BrafCa/+; Ptenfl/fl; Atg7fl/fl). The growth-inhibitory effect of MTX on autophagy-competent melanomas was abolished by the combined depletion of CD4+ or CD8+ T lymphocytes. In conclusion, it appears that the success of chemotherapy against "spontaneous," genetically induced cancers is governed by the same rules as those applicable to transplantable tumors.

14.
Oncoimmunology ; 3: e28276, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25050202

RESUMEN

Cis-diamminedichloridoplatinum(II) (CDDP), commonly referred to as cisplatin, is a chemotherapeutic drug used for the treatment of a wide range of solid cancers. CDDP is a relatively poor inducer of immunogenic cell death (ICD), a cell death modality that converts dying cells into a tumor vaccine, stimulating an immune response against residual cancer cells that permits long-lasting immunity and a corresponding reduction in tumor growth. The incapacity of CDDP to trigger ICD is at least partially due to its failure to stimulate the premortem endoplasmic reticulum (ER)-stress response required for the externalization of the "eat-me" signal calreticulin (CRT) on the surface of dying cancer cells. Here, we developed a murine cancer cell line genetically modified to express the ER resident protein reticulon-1c (Rtn-1c) by virtue of tetracycline induction and showed that enforced Rtn-1c expression combined with CDDP treatment promoted CRT externalization to the surface of cancer cells. In contrast to single agent treatments, the tetracycline-mediated Rtn-1c induction combined with CDDP chemotherapy stimulated ICD as measured by the capacity of dying tumor cells, inoculated into syngenic immunocompetent mice, to mount an immune response to tumor re-challenge 1 week later. More importantly, established tumors, forced to constitutively express Rtn-1c in vivo by continuous treatment with tetracycline, became responsive to CDDP and exhibited a corresponding reduction in the rate of tumor growth. The combined therapeutic effects of Rtn-1c induction with CDDP treatment was only detected in the context of an intact immune system and not in nu/nu mice lacking thymus-dependent T lymphocytes. Altogether, these results indicate that the artificial or "synthetic" induction of immunogenic cell death by genetic manipulation of the ER-stress response can improve the efficacy of chemotherapy with CDDP by stimulating anticancer immunity.

15.
Oncoimmunology ; 3: e28473, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25050214

RESUMEN

Immunogenic cell death (ICD) inducers can be defined as agents that exert cytotoxic effects while stimulating an immune response against dead cell-associated antigens. When initiated by anthracyclines, ICD is accompanied by stereotyped molecular changes, including the pre-apoptotic exposure of calreticulin (CRT) on the cell surface, the lysosomal secretion of ATP during the blebbing phase of apoptosis, and the release of high mobility group box 1 (HMGB1) from dead cells. By means of genetically engineered human osteosarcoma U2OS cells, we screened the 879 anticancer compounds of the National Cancer Institute (NCI) Mechanistic Diversity Set for their ability to promote all these hallmarks of ICD in vitro. In line with previous findings from our group, several cardiac glycosides exhibit a robust propensity to elicit the major manifestations of ICD in cultured neoplastic cells. This screen pointed to septacidin, an antibiotic produced by Streptomyces fibriatus, as a novel putative inducer of ICD. In low-throughput validation experiments, septacidin promoted CRT exposure, ATP secretion and HGMB1 release from both U2OS cells and murine fibrosarcoma MCA205 cells. Moreover, septacidin-killed MCA205 cells protected immunocompetent mice against a re-challenge with living cancer cells of the same type. Finally, the antineoplastic effects of septacidin on established murine tumors were entirely dependent on T lymphocytes. Altogether, these results underscore the suitability of the high-throughput screening system described here for the identification of novel ICD inducers.

16.
Methods Mol Biol ; 965: 121-42, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23296654

RESUMEN

Cellular senescence, which can be defined as a stress response preventing the propagation of cells that have accumulated potentially oncogenic alterations, is invariably associated with a permanent cell cycle arrest. Such an irreversible blockage is mainly mediated by the persistent upregulation of one or more cyclin-dependent kinase inhibitors (CKIs), including (though not limited to) p16( INK4A ) and p21( CIP1 ) and p27( KIP1 ). CKIs operate by binding to cyclin-dependent kinases (CDKs), de facto inhibiting their enzymatic activity. Here, we provide an immunoblotting-based method for the detection and quantification of CKIs in vitro and ex vivo, together with a set of guidelines for the interpretation of results.


Asunto(s)
Puntos de Control del Ciclo Celular , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Immunoblotting/métodos , Senescencia Celular , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/aislamiento & purificación , Electroforesis , Regulación de la Expresión Génica , Células HeLa , Humanos
17.
Methods Mol Biol ; 1004: 43-56, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23733568

RESUMEN

During necrosis and following some instances of apoptosis (in particular in the absence of a proficient phagocytic system), the nonhistone chromatin component high-mobility group box 1 (HMGB1) is released in the extracellular space. In vivo, extracellular HMGB1 can bind Toll-like receptor 4 on the surface of dendritic cells, de facto operating as a danger-associated molecular pattern and alarming the organism to the presence of stressful conditions. Recent results indicate that the release of HMGB1 is one of the key features for cell death to be perceived as immunogenic, i.e., to be capable of triggering a cognate immune response in vivo. Thus, only anticancer agents that-among other features-allow for the release of HMGB1 as they induce cell death are expected to stimulate anticancer immune responses. To investigate the immunogenic potential of conventional anticancer agents and novel cell death inducers on a high-throughput scale, we engineered human osteosarcoma U2OS cells to express HMGB1 fused at the N-terminus of the green fluorescent protein (GFP). Coupled to fluorescence microscopy workstations for automated image acquisition and analysis, this HMGB1-GFP-based biosensor is amenable for the identification of potential inducers of immunogenic cell death among large chemical libraries.


Asunto(s)
Técnicas Biosensibles/métodos , Proteína HMGB1/análisis , Proteína HMGB1/metabolismo , Muerte Celular , Células Cultivadas , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía Fluorescente , Proteínas Recombinantes de Fusión/metabolismo
18.
Oncoimmunology ; 2(6): e24568, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23894718

RESUMEN

Tumor cells succumb to chemotherapy while releasing ATP. We have found that extracellular ATP attracts dendritic cell (DC) precursors into the tumor bed, facilitates their permanence in the proximity of dying cells and promotes their differentiation into mature DCs endowed with the capacity of presenting tumor-associated antigens.

19.
Oncoimmunology ; 1(3): 393-395, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22737627

RESUMEN

The efficacy of antineoplastic chemotherapies with anthracyclines or oxaliplatin relies on the induction of immunogenic cell death, thus provoking an anticancer immune response. Recently, we observed that overexpression of CD39, an ATP-degrading enzyme expressed on the cell surface, abolishes the immunogenicity of cell death, thus rendering cancers resistant against chemotherapy.

20.
Autophagy ; 8(3): 413-5, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22361584

RESUMEN

One particular strategy to render anticancer therapies efficient consists of converting the patient's own tumor cells into therapeutic vaccines, via the induction of immunogenic cell death (ICD). One of the hallmarks of ICD dwells in the active release of ATP by cells committed to undergo, but not yet having succumbed to, apoptosis. We observed that the knockdown of essential autophagy-related genes (ATG3, ATG5, ATG7 and BECN1) abolishes the pre-apoptotic secretion of ATP by several human and murine cancer cell lines undergoing ICD. Accordingly, autophagy-competent, but not autophagy-deficient, tumor cells treated with ICD inducers in vitro could induce a tumor-specific immune response in vivo. Cancer cell lines stably depleted of ATG5 or ATG7 normally generate tumors in vivo, and such autophagy-deficient neoplasms, upon systemic treatment with ICD inducers, exhibit the same levels of apoptosis (as monitored by nuclear shrinkage and caspase-3 activation) and necrosis (as determined by following the kinetics of HMGB1 release) as their autophagy-proficient counterparts. However, autophagy-incompetent cancers fail to release ATP, to recruit immune effectors into the tumor bed and to respond to chemotherapy in conditions in which autophagy-competent tumors do so. The intratumoral administration of ecto-ATPase inhibitors increases extracellular ATP concentrations, re-establishes the therapy-induced recruitment of dendritic cells and T cells into the tumor bed, and restores the chemotherapeutic response of autophagy-deficient cancers. Altogether, these results suggest that autophagy-incompetent tumor cells escape from chemotherapy-induced (and perhaps natural?) immunosurveillance because they are unable to release ATP.


Asunto(s)
Antineoplásicos/uso terapéutico , Autofagia/inmunología , Neoplasias/inmunología , Neoplasias/patología , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA