Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Intervalo de año de publicación
1.
Mol Oncol ; 18(3): 620-640, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38098337

RESUMEN

The small GTPase Rac1 (Ras-related C3 botulinum toxin substrate 1) has been implicated in cancer progression and in the poor prognosis of various types of tumors. Rac1 SUMOylation occurs during epithelial-mesenchymal transition (EMT), and it is required for tumor cell migration and invasion. Here we identify POTEE (POTE Ankyrin domain family member E) as a novel Rac1-SUMO1 effector involved in breast cancer malignancy that controls invadopodium formation through the activation of Rac1-SUMO1. POTEE activates Rac1 in the invadopodium by recruiting TRIO-GEF (triple functional domain protein), and it induces tumor cell proliferation and metastasis in vitro and in vivo. We found that the co-localization of POTEE with Rac1 is correlated with more aggressive breast cancer subtypes. Given its role in tumor dissemination, the leading cause of cancer-related deaths, POTEE could represent a potential therapeutic target for these types of cancer.


Asunto(s)
Neoplasias de la Mama , Podosomas , Humanos , Femenino , Transducción de Señal , Podosomas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Movimiento Celular , Línea Celular Tumoral
2.
Cancers (Basel) ; 13(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34771470

RESUMEN

BACKGROUND: Tribbles pseudokinase 3 (TRIB3) has been proposed to both promote and restrict cancer generation and progression. However, the precise mechanisms that determine this dual role of TRIB3 in cancer remain to be understood. In this study we aimed to investigate the role of TRIB3 in luminal breast cancer, the most frequent subtype of this malignancy. METHODS: We genetically manipulated TRIB3 expression in a panel of luminal breast cancer cell lines and analyzed its impact on cell proliferation, and the phosphorylation, levels, or subcellular localization of TRIB3 and other protein regulators of key signaling pathways in luminal breast cancer. We also analyzed TRIB3 protein expression in samples from luminal breast cancer patients and performed bioinformatic analyses in public datasets. RESULTS: TRIB3 enhanced the proliferation and AKT phosphorylation in luminal A (HER2-) but decreased them in luminal B (HER2+) breast cancer cell lines. TRIB3 negatively regulated the stability of HER2 in luminal B breast cancer cell lines. TRIB3 expression was associated with increased disease-free survival and a better response to therapy in luminal breast cancer patients. CONCLUSIONS: Our findings support the exploration of TRIB3 as a potential biomarker and therapeutic target in luminal breast cancer.

3.
Nature ; 592(7856): 799-803, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33854232

RESUMEN

Mammalian development, adult tissue homeostasis and the avoidance of severe diseases including cancer require a properly orchestrated cell cycle, as well as error-free genome maintenance. The key cell-fate decision to replicate the genome is controlled by two major signalling pathways that act in parallel-the MYC pathway and the cyclin D-cyclin-dependent kinase (CDK)-retinoblastoma protein (RB) pathway1,2. Both MYC and the cyclin D-CDK-RB axis are commonly deregulated in cancer, and this is associated with increased genomic instability. The autophagic tumour-suppressor protein AMBRA1 has been linked to the control of cell proliferation, but the underlying molecular mechanisms remain poorly understood. Here we show that AMBRA1 is an upstream master regulator of the transition from G1 to S phase and thereby prevents replication stress. Using a combination of cell and molecular approaches and in vivo models, we reveal that AMBRA1 regulates the abundance of D-type cyclins by mediating their degradation. Furthermore, by controlling the transition from G1 to S phase, AMBRA1 helps to maintain genomic integrity during DNA replication, which counteracts developmental abnormalities and tumour growth. Finally, we identify the CHK1 kinase as a potential therapeutic target in AMBRA1-deficient tumours. These results advance our understanding of the control of replication-phase entry and genomic integrity, and identify the AMBRA1-cyclin D pathway as a crucial cell-cycle-regulatory mechanism that is deeply interconnected with genomic stability in embryonic development and tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ciclina D/metabolismo , Inestabilidad Genómica , Fase S , Animales , Línea Celular , Proliferación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Replicación del ADN , Regulación del Desarrollo de la Expresión Génica , Genes Supresores de Tumor , Humanos , Ratones , Ratones Noqueados , Mutaciones Letales Sintéticas
4.
Cancer Res ; 80(23): 5216-5230, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33023950

RESUMEN

SNAI2 overexpression appears to be associated with poor prognosis in breast cancer, yet it remains unclear in which breast cancer subtypes this occurs. Here we show that excess SNAI2 is associated with a poor prognosis of luminal B HER2+/ERBB2+ breast cancers in which SNAI2 expression in the stroma but not the epithelium correlates with tumor proliferation. To determine how stromal SNAI2 might influence HER2+ tumor behavior, Snai2-deficient mice were crossed with a mouse line carrying the ErbB2/Neu protooncogene to generate HER2+/ERBB2+ breast cancer. Tumors generated in this model expressed SNAI2 in the stroma but not the epithelium, allowing for the role of stromal SNAI2 to be studied without interference from the epithelial compartment. The absence of SNAI2 in the stroma of HER2+/ERBB2+ tumors is associated with: (i) lower levels of cyclin D1 (CCND1) and reduced tumor epithelium proliferation; (ii) higher levels of AKT and a lower incidence of metastasis; (iii) lower levels of angiopoietin-2 (ANGPT2), and more necrosis. Together, these results indicate that the loss of SNAI2 in cancer-associated fibroblasts limits the production of some cytokines, which influences AKT/ERK tumor signaling and subsequent proliferative and metastatic capacity of ERBB2+ breast cancer cells. Accordingly, SNAI2 expression in the stroma enhanced the tumorigenicity of luminal B HER2+/ERBB2+ breast cancers. This work emphasizes the importance of stromal SNAI2 in breast cancer progression and patients' prognosis. SIGNIFICANCE: Stromal SNAI2 expression enhances the tumorigenicity of luminal B HER2+ breast cancers and can identify a subset of patients with poor prognosis, making SNAI2 a potential therapeutic target for this disease. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/23/5216/F1.large.jpg.


Asunto(s)
Neoplasias de la Mama/patología , Receptor ErbB-2/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Células del Estroma/patología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Noqueados , Receptor ErbB-2/genética , Factores de Transcripción de la Familia Snail/genética , Células del Estroma/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Theranostics ; 10(11): 5120-5136, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32308772

RESUMEN

Glioblastoma (GBM) is one of the most aggressive forms of cancer. It has been proposed that the presence within these tumors of a population of cells with stem-like features termed Glioma Initiating Cells (GICs) is responsible for the relapses that take place in the patients with this disease. Targeting this cell population is therefore an issue of great therapeutic interest in neuro-oncology. We had previously found that the neurotrophic factor MIDKINE (MDK) promotes resistance to glioma cell death. The main objective of this work is therefore investigating the role of MDK in the regulation of GICs. Methods: Assays of gene and protein expression, self-renewal capacity, autophagy and apoptosis in cultures of GICs derived from GBM samples subjected to different treatments. Analysis of the growth of GICs-derived xenografts generated in mice upon blockade of the MDK and its receptor the ALK receptor tyrosine kinase (ALK) upon exposure to different treatments. Results: Genetic or pharmacological inhibition of MDK or ALK decreases the self-renewal and tumorigenic capacity of GICs via the autophagic degradation of the transcription factor SOX9. Blockade of the MDK/ALK axis in combination with temozolomide depletes the population of GICs in vitro and has a potent anticancer activity in xenografts derived from GICs. Conclusions: The MDK/ALK axis regulates the self-renewal capacity of GICs by controlling the autophagic degradation of the transcription factor SOX9. Inhibition of the MDK/ALK axis may be a therapeutic strategy to target GICs in GBM patients.


Asunto(s)
Quinasa de Linfoma Anaplásico/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Midkina/metabolismo , Células Madre Neoplásicas/metabolismo , Temozolomida/farmacología , Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Animales , Antineoplásicos Alquilantes/farmacología , Autofagia/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular , Femenino , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Ratones , Ratones Desnudos , Midkina/antagonistas & inhibidores , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Exp Med ; 217(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32219437

RESUMEN

Gene dosage is a key defining factor to understand cancer pathogenesis and progression, which requires the development of experimental models that aid better deconstruction of the disease. Here, we model an aggressive form of prostate cancer and show the unconventional association of LKB1 dosage to prostate tumorigenesis. Whereas loss of Lkb1 alone in the murine prostate epithelium was inconsequential for tumorigenesis, its combination with an oncogenic insult, illustrated by Pten heterozygosity, elicited lethal metastatic prostate cancer. Despite the low frequency of LKB1 deletion in patients, this event was significantly enriched in lung metastasis. Modeling the role of LKB1 in cellular systems revealed that the residual activity retained in a reported kinase-dead form, LKB1K78I, was sufficient to hamper tumor aggressiveness and metastatic dissemination. Our data suggest that prostate cells can function normally with low activity of LKB1, whereas its complete absence influences prostate cancer pathogenesis and dissemination.


Asunto(s)
Neoplasias de la Próstata/enzimología , Proteínas Serina-Treonina Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Epitelio/enzimología , Epitelio/patología , Células HEK293 , Heterocigoto , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas Mutantes/metabolismo , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/metabolismo , Próstata/enzimología , Próstata/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/metabolismo
7.
J Cell Sci ; 132(20)2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31578236

RESUMEN

Post-translational modifications directly control protein activity and, thus, they represent an important means to regulate the responses of cells to different stimuli. Protein SUMOylation has recently been recognised as one such modification, and it has been associated with various diseases, including different types of cancer. However, the precise way that changes in SUMOylation influence the tumorigenic properties of cells remains to be fully clarified. Here, we show that blocking the SUMO pathway by depleting SUMO1 and UBC9, or by exposure to ginkgolic acid C15:1 or 2-D08 (two different SUMOylation inhibitors), induces cell death, also inhibiting the invasiveness of tumour cells. Indeed, diminishing the formation of SUMO1 complexes induces autophagy-mediated cancer cell death through increasing the expression of Tribbles pseudokinase 3 (TRIB3). Moreover, we found that blocking the SUMO pathway inhibits tumour cell invasion by decreasing RAC1 SUMOylation. These findings shed new light on the mechanisms by which SUMO1 modifications regulate the survival, and the migratory and invasive capacity of tumour cells, potentially establishing the bases to develop novel anti-cancer treatments based on the inhibition of SUMOylation.


Asunto(s)
Muerte Celular Autofágica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteína SUMO-1/metabolismo , Sumoilación , Proteína de Unión al GTP rac1/metabolismo , Humanos , Células MCF-7 , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Proteína SUMO-1/genética , Proteína de Unión al GTP rac1/genética
8.
Biochem Pharmacol ; 157: 266-274, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30195736

RESUMEN

Glioblastoma multiforme (GBM) is the most frequent and aggressive type of brain tumor due, at least in part, to its poor response to current anticancer treatments. These features could be explained, at least partially, by the presence within the tumor mass of a small population of cells termed Glioma Initiating Cells (GICs) that has been proposed to be responsible for the relapses occurring in this disease. Thus, the development of novel therapeutic approaches (and specifically those targeting the population of GICs) is urgently needed to improve the survival of the patients suffering this devastating disease. Previous observations by our group and others have shown that Δ9-Tetrahydrocannabinol (THC, the main active ingredient of marijuana) and other cannabinoids including cannabidiol (CBD) exert antitumoral actions in several animal models of cancer, including gliomas. We also found that the administration of THC (or of THC + CBD at a 1:1 ratio) in combination with temozolomide (TMZ), the benchmark agent for the treatment of GBM, synergistically reduces the growth of glioma xenografts. In this work we investigated the effect of the combination of TMZ and THC:CBD mixtures containing different ratios of the two cannabinoids in preclinical glioma models, including those derived from GICs. Our findings show that TMZ + THC:CBD combinations containing a higher proportion of CDB (but not TMZ + CBD alone) produce a similar antitumoral effect as the administration of TMZ together with THC and CBD at a 1:1 ratio in xenografts generated with glioma cell lines. In addition, we also found that the administration of TMZ + THC:CBD at a 1:1 ratio reduced the growth of orthotopic xenografts generated with GICs derived from GBM patients and enhanced the survival of the animals bearing these intracranial xenografts. Remarkably, the antitumoral effect observed in GICs-derived xenografts was stronger when TMZ was administered together with cannabinoid combinations containing a higher proportion of CBD. These findings support the notion that the administration of TMZ together with THC:CBD combinations - and specifically those containing a higher proportion of CBD - may be therapeutically explored to target the population of GICs in GBM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Cannabidiol/uso terapéutico , Dronabinol/uso terapéutico , Glioblastoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Temozolomida/uso terapéutico , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Femenino , Glioblastoma/patología , Humanos , Masculino , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Biochem Pharmacol ; 157: 275-284, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30125556

RESUMEN

Glioblastoma multiforme (GBM) is the most frequent and aggressive form of brain cancer. These features are explained at least in part by the high resistance exhibited by these tumors to current anticancer therapies. Thus, the development of novel therapeutic approaches is urgently needed to improve the survival of the patients suffering this devastating disease. Δ9-Tetrahydrocannabinol (THC, the major active ingredient of marijuana), and other cannabinoids have been shown to exert antitumoral actions in animal models of cancer, including glioma. The mechanism of these anticancer actions relies, at least in part, on the ability of these compounds to stimulate autophagy-mediated apoptosis in tumor cells. Previous observations from our group demonstrated that local administration of THC (or of THC + CBD at a 1:1 ratio, a mixture that resembles the composition of the cannabinoid-based medicine Sativex®) in combination with Temozolomide, the benchmark agent for the treatment of GBM, synergistically reduces the growth of glioma xenografts. With the aim of optimizing the possible clinical utilization of cannabinoids in anti-GBM therapies, in this work we explored the anticancer efficacy of the systemic administration of cannabinoids in combination with TMZ in preclinical models of glioma. Our results show that oral administration of Sativex-like extracts (containing THC and CBD at a 1:1 ratio) in combination with TMZ produces a strong antitumoral effect in both subcutaneous and intracranial glioma cell-derived tumor xenografts. In contrast, combined administration of Sativex-like and BCNU (another alkylating agent used for the treatment of GBM which share structural similarities with the TMZ) did not show a stronger effect than individual treatments. Altogether, our findings support the notion that the combined administration of TMZ and oral cannabinoids could be therapeutically exploited for the management of GBM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Cannabidiol/uso terapéutico , Dronabinol/uso terapéutico , Glioma/tratamiento farmacológico , Temozolomida/uso terapéutico , Administración Oral , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Encefálicas/patología , Cannabidiol/administración & dosificación , Carmustina/uso terapéutico , Línea Celular Tumoral , Dronabinol/administración & dosificación , Glioma/patología , Xenoinjertos , Humanos , Masculino , Ratones Desnudos , Temozolomida/administración & dosificación
10.
Med. hist ; 38(2): 4-21, 2018. ilus
Artículo en Español | IBECS | ID: ibc-176821

RESUMEN

El siglo XIX fue un periodo en el que se produjo un creciente interés por los venenos y los crímenes por envenenamiento a pesar de no ser formas habituales de homicidio. La nueva toxicología pretendía ofrecer herramientas para combatir este tipo de crímenes. Sin embargo, fueron precisamente los debates surgidos durante los procesos judiciales, los que ayudaron a configurar la toxicología del siglo XIX. Alejados de las pautas ofrecidas en los manuales y ante la necesidad de mostrar a un juez carente de formación en estas materias, la presencia o ausencia del veneno, los toxicólogos del siglo XIX pusieron en juego todas sus estrategias para vencer a otros expertos y convencer a los profanos. A mediados de 1844 se produjo en Madrid un caso de envenenamiento que llamó la atención tanto de la prensa médica como de la prensa periódica española. Dos factores contribuyeron a su popularidad: las fechas en las que se produjo (tan solo un año después de la creación de la cátedra de Medicina Legal en las Facultades de Madrid y Barcelona); y la participación como peritos de algunos de los personajes españoles más influyentes en la medicina legal y toxicología española como es el caso de Pere Mata i Fontanet (1811-1877). Pere Mata desempeñó una labor importante en los tres terrenos que contribuyeron decisivamente a la homogeneidad de la comunidad de toxicólogos: la formación universitaria, la literatura académica y la organización profesional. El análisis de un caso de envenenamiento como el que se desarrolla en este trabajo, permite considerar muchas de las cuestiones relacionadas con la toxicología en el siglo XIX: la constitución de una nueva disciplina académica, la creación de una comunidad de expertos, las controversias públicas y la gestión de las pruebas periciales en los tribunales


The XIXth century saw a growing interest in poisons and crimes by poison although these are not usual ways of murder. New technology aspired to offer tools to combat this type of crimes. However, it was precisely the debates that arose during trials that helped to configure XIXth century toxicology. Far from the guidelines offered in manuals and facing the need to demonstrate to a judge, lacking In training in these subjects the presence or absence of the poison, XIXth century toxicologists used all of their strategies to beat other experts and convince the layperson. In the middle of 1844 there was a case of poisoning in Madrid that caught the attention of both the medical press and the Spanish newspapers. Two factors contributed to its popularity: the date that it happened 8only a year after the creation of the chair of Forensic Medicine at the faculties in Madrid and Barcelona); and the participation as experts of some of the most influential Spaniards in forensic medicine and Spanish toxicology such as Pere Mata I Fontanet (1811-1877). Pere Mata carried out important work in the three fields which decisively contributed to the homogeneity of the community of toxicologists: university training, academic literature and the professional organization. The analysis of a case of poisoning as the one developed in this work permits the consideration of many issues related totoxicology in the XIXth century: the constitution of a new academic subject, the creation of a community of experts, public controversies and the management of expert evidence at trials


Asunto(s)
Historia del Siglo XIX , Crimen/historia , Intoxicación/historia , Intoxicación/mortalidad , Alcaloides Opiáceos/historia , Alcaloides Opiáceos/toxicidad , Toxicología Forense/historia , Opio/toxicidad , Alcaloides/historia , Alcaloides/toxicidad , Toxicología/historia , Toxicología/legislación & jurisprudencia
13.
Autophagy ; 12(11): 2213-2229, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27635674

RESUMEN

Autophagy is considered primarily a cell survival process, although it can also lead to cell death. However, the factors that dictate the shift between these 2 opposite outcomes remain largely unknown. In this work, we used Δ9-tetrahydrocannabinol (THC, the main active component of marijuana, a compound that triggers autophagy-mediated cancer cell death) and nutrient deprivation (an autophagic stimulus that triggers cytoprotective autophagy) to investigate the precise molecular mechanisms responsible for the activation of cytotoxic autophagy in cancer cells. By using a wide array of experimental approaches we show that THC (but not nutrient deprivation) increases the dihydroceramide:ceramide ratio in the endoplasmic reticulum of glioma cells, and this alteration is directed to autophagosomes and autolysosomes to promote lysosomal membrane permeabilization, cathepsin release and the subsequent activation of apoptotic cell death. These findings pave the way to clarify the regulatory mechanisms that determine the selective activation of autophagy-mediated cancer cell death.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Ceramidas/farmacología , Lisosomas/metabolismo , Neoplasias/patología , Transporte Biológico/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dronabinol/farmacología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestructura , Lisosomas/efectos de los fármacos , Lisosomas/ultraestructura , Modelos Biológicos , Permeabilidad , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Esfingolípidos/biosíntesis
14.
Nat Cell Biol ; 18(6): 645-656, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27214280

RESUMEN

Cellular transformation and cancer progression is accompanied by changes in the metabolic landscape. Master co-regulators of metabolism orchestrate the modulation of multiple metabolic pathways through transcriptional programs, and hence constitute a probabilistically parsimonious mechanism for general metabolic rewiring. Here we show that the transcriptional co-activator peroxisome proliferator-activated receptor gamma co-activator 1α (PGC1α) suppresses prostate cancer progression and metastasis. A metabolic co-regulator data mining analysis unveiled that PGC1α is downregulated in prostate cancer and associated with disease progression. Using genetically engineered mouse models and xenografts, we demonstrated that PGC1α opposes prostate cancer progression and metastasis. Mechanistically, the use of integrative metabolomics and transcriptomics revealed that PGC1α activates an oestrogen-related receptor alpha (ERRα)-dependent transcriptional program to elicit a catabolic state and metastasis suppression. Importantly, a signature based on the PGC1α-ERRα pathway exhibited prognostic potential in prostate cancer, thus uncovering the relevance of monitoring and manipulating this pathway for prostate cancer stratification and treatment.


Asunto(s)
Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Ratones , Metástasis de la Neoplasia/patología , Neoplasias de la Próstata/patología , Receptores de Estrógenos/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
15.
Neuropharmacology ; 108: 345-52, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27179908

RESUMEN

Cannabinoid CB1 receptor, the molecular target of endocannabinoids and cannabis active components, is one of the most abundant metabotropic receptors in the brain. Cannabis is widely used for both recreational and medicinal purposes. Despite the ever-growing fundamental roles of microRNAs in the brain, the possible molecular connections between the CB1 receptor and microRNAs are surprisingly unknown. Here, by using reporter gene constructs that express interaction sequences for microRNAs in human SH-SY5Y neuroblastoma cells, we show that CB1 receptor activation enhances the expression of several microRNAs, including let-7d. This was confirmed by measuring hsa-let-7d expression levels. Accordingly, knocking-down CB1 receptor in zebrafish reduced dre-let-7d levels, and knocking-out CB1 receptor in mice decreased mmu-let-7d levels in the cortex, striatum and hippocampus. Conversely, knocking-down let-7d increased CB1 receptor mRNA expression in zebrafish, SH-SY5Y cells and primary striatal neurons. Likewise, in primary striatal neurons chronically exposed to a cannabinoid or opioid agonist, a let-7d-inhibiting sequence facilitated not only cannabinoid or opioid signaling but also cannabinoid/opioid cross-signaling. Taken together, these findings provide the first evidence for a bidirectional link between the CB1 receptor and a microRNA, namely let-7d, and thus unveil a new player in the complex process of cannabinoid action.


Asunto(s)
Cannabinoides/biosíntesis , MicroARNs/biosíntesis , Receptor Cannabinoide CB1/biosíntesis , Animales , Canfanos/farmacología , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Pez Cebra
16.
Clin Cancer Res ; 22(10): 2508-19, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26671995

RESUMEN

PURPOSE: ABTL0812 is a novel first-in-class, small molecule which showed antiproliferative effect on tumor cells in phenotypic assays. Here we describe the mechanism of action of this antitumor drug, which is currently in clinical development. EXPERIMENTAL DESIGN: We investigated the effect of ABTL0812 on cancer cell death, proliferation, and modulation of intracellular signaling pathways, using human lung (A549) and pancreatic (MiaPaCa-2) cancer cells and tumor xenografts. To identify cellular targets, we performed in silico high-throughput screening comparing ABTL0812 chemical structure against ChEMBL15 database. RESULTS: ABTL0812 inhibited Akt/mTORC1 axis, resulting in impaired cancer cell proliferation and autophagy-mediated cell death. In silico screening led us to identify PPARs, PPARα and PPARγ as the cellular targets of ABTL0812. We showed that ABTL0812 activates both PPAR receptors, resulting in upregulation of Tribbles-3 pseudokinase (TRIB3) gene expression. Upregulated TRIB3 binds cellular Akt, preventing its activation by upstream kinases, resulting in Akt inhibition and suppression of the Akt/mTORC1 axis. Pharmacologic inhibition of PPARα/γ or TRIB3 silencing prevented ABTL0812-induced cell death. ABTL0812 treatment induced Akt inhibition in cancer cells, tumor xenografts, and peripheral blood mononuclear cells from patients enrolled in phase I/Ib first-in-human clinical trial. CONCLUSIONS: ABTL0812 has a unique and novel mechanism of action, that defines a new and drugable cellular route that links PPARs to Akt/mTORC1 axis, where TRIB3 pseudokinase plays a central role. Activation of this route (PPARα/γ-TRIB3-Akt-mTORC1) leads to autophagy-mediated cancer cell death. Given the low toxicity and high tolerability of ABTL0812, our results support further development of ABTL0812 as a promising anticancer therapy. Clin Cancer Res; 22(10); 2508-19. ©2015 AACR.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Ratas , Transducción de Señal/efectos de los fármacos
17.
Artículo en Inglés | MEDLINE | ID: mdl-26071989

RESUMEN

It is well-established that cannabinoids exert palliative effects on some cancer-associated symptoms. In addition evidences obtained during the last fifteen years support that these compounds can reduce tumor growth in animal models of cancer. Cannabinoids have been shown to activate an ER-stress related pathway that leads to the stimulation of autophagy-mediated cancer cell death. In addition, cannabinoids inhibit tumor angiogenesis and decrease cancer cell migration. The mechanisms of resistance to cannabinoid anticancer action as well as the possible strategies to develop cannabinoid-based combinational therapies to fight cancer have also started to be explored. In this review we will summarize these observations (that have already helped to set the bases for the development of the first clinical studies to investigate the potential clinical benefit of using cannabinoids in anticancer therapies) and will discuss the possible future avenues of research in this area.


Asunto(s)
Antineoplásicos/uso terapéutico , Cannabinoides/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Cannabinoides/farmacología , Ensayos Clínicos como Asunto , Endocannabinoides/metabolismo , Humanos , Neoplasias/metabolismo
18.
Biochem Soc Trans ; 43(5): 1122-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26517935

RESUMEN

Tribbles pseudokinase 3 (TRIB3) belongs to the tribbles family of pseudokinases. In this article, we summarize several observation obtained by our laboratories supporting that TRIB3 plays a crucial role in the anti-cancer activity of cannabinoids (a novel family of potential anti-cancer agents derived from marijuana) and that TRIB3 genetic inactivation enhances cancer generation and progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Apoptosis/genética , Apoptosis/fisiología , Autofagia/genética , Autofagia/fisiología , Proteínas de Ciclo Celular/genética , Endocannabinoides/metabolismo , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/patología , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/genética , Transducción de Señal/genética
20.
J Invest Dermatol ; 135(6): 1629-1637, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25674907

RESUMEN

Although the global incidence of cutaneous melanoma is increasing, survival rates for patients with metastatic disease remain <10%. Novel treatment strategies are therefore urgently required, particularly for patients bearing BRAF/NRAS wild-type tumors. Targeting autophagy is a means to promote cancer cell death in chemotherapy-resistant tumors, and the aim of this study was to test the hypothesis that cannabinoids promote autophagy-dependent apoptosis in melanoma. Treatment with Δ(9)-Tetrahydrocannabinol (THC) resulted in the activation of autophagy, loss of cell viability, and activation of apoptosis, whereas cotreatment with chloroquine or knockdown of Atg7, but not Beclin-1 or Ambra1, prevented THC-induced autophagy and cell death in vitro. Administration of Sativex-like (a laboratory preparation comprising equal amounts of THC and cannabidiol (CBD)) to mice bearing BRAF wild-type melanoma xenografts substantially inhibited melanoma viability, proliferation, and tumor growth paralleled by an increase in autophagy and apoptosis compared with standard single-agent temozolomide. Collectively, our findings suggest that THC activates noncanonical autophagy-mediated apoptosis of melanoma cells, suggesting that cytotoxic autophagy induction with Sativex warrants clinical evaluation for metastatic disease.


Asunto(s)
Autofagia , Cannabinoides/química , Melanoma/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Cannabidiol , Cannabinol/química , Muerte Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Dacarbazina/análogos & derivados , Dacarbazina/química , Dronabinol/química , Combinación de Medicamentos , Humanos , Masculino , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Desnudos , Microscopía Confocal , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias/metabolismo , Extractos Vegetales/química , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo , Temozolomida , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...