Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nature ; 594(7863): 436-441, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34079128

RESUMEN

A delicate equilibrium of WNT agonists and antagonists in the intestinal stem cell (ISC) niche is critical to maintaining the ISC compartment, as it accommodates the rapid renewal of the gut lining. Disruption of this balance by mutations in the tumour suppressor gene APC, which are found in approximately 80% of all human colon cancers, leads to unrestrained activation of the WNT pathway1,2. It has previously been established that Apc-mutant cells have a competitive advantage over wild-type ISCs3. Consequently, Apc-mutant ISCs frequently outcompete all wild-type stem cells within a crypt, thereby reaching clonal fixation in the tissue and initiating cancer formation. However, whether the increased relative fitness of Apc-mutant ISCs involves only cell-intrinsic features or whether Apc mutants are actively involved in the elimination of their wild-type neighbours remains unresolved. Here we show that Apc-mutant ISCs function as bona fide supercompetitors by secreting WNT antagonists, thereby inducing differentiation of neighbouring wild-type ISCs. Lithium chloride prevented the expansion of Apc-mutant clones and the formation of adenomas by rendering wild-type ISCs insensitive to WNT antagonists through downstream activation of WNT by inhibition of GSK3ß. Our work suggests that boosting the fitness of healthy cells to limit the expansion of pre-malignant clones may be a powerful strategy to limit the formation of cancers in high-risk individuals.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Competencia Celular , Genes APC , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Mutación , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Animales , Diferenciación Celular/genética , Femenino , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Neoplasias Intestinales/metabolismo , Cloruro de Litio/farmacología , Masculino , Ratones , Organoides/citología , Organoides/metabolismo , Organoides/patología , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo
2.
Mol Cell ; 49(3): 524-35, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23273978

RESUMEN

Binding within or nearby target genes involved in cell proliferation and survival enables the p53 tumor suppressor gene to regulate their transcription and cell-cycle progression. Using genome-wide chromatin-binding profiles, we describe binding of p53 also to regions located distantly from any known p53 target gene. Interestingly, many of these regions possess conserved p53-binding sites and all known hallmarks of enhancer regions. We demonstrate that these p53-bound enhancer regions (p53BERs) indeed contain enhancer activity and interact intrachromosomally with multiple neighboring genes to convey long-distance p53-dependent transcription regulation. Furthermore, p53BERs produce, in a p53-dependent manner, enhancer RNAs (eRNAs) that are required for efficient transcriptional enhancement of interacting target genes and induction of a p53-dependent cell-cycle arrest. Thus, our results ascribe transcription enhancement activity to p53 with the capacity to regulate multiple genes from a single genomic binding site. Moreover, eRNA production from p53BERs is required for efficient p53 transcription enhancement.


Asunto(s)
Elementos de Facilitación Genéticos , ARN/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control del Ciclo Celular/genética , Cromatina/metabolismo , Cromosomas Humanos/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes , Humanos , Células MCF-7 , Modelos Genéticos , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/metabolismo
3.
Cancer Cell Int ; 20(1): 578, 2020 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-33292279

RESUMEN

BACKGROUND: Cancer results from the accumulation of mutations leading to the acquisition of cancer promoting characteristics such as increased proliferation and resistance to cell death. In colorectal cancer, an early mutation leading to such features usually occurs in the APC or CTNNB1 genes, thereby activating Wnt signalling. However, substantial phenotypic differences between cancers originating within the same organ, such as molecular subtypes, are not fully reflected by differences in mutations. Indeed, the phenotype seems to result from a complex interplay between the cell-intrinsic features and the acquired mutations, which is difficult to disentangle when established tumours are studied. METHODS: We use a 3D in vitro organoid model to study the early phase of colorectal cancer development. From three different murine intestinal locations we grow organoids. These are transformed to resemble adenomas after Wnt activation through lentiviral transduction with a stable form of ß-Catenin. The gene expression before and after Wnt activation is compared within each intestinal origin and across the three locations using RNA sequencing. To validate and generalize our findings, we use gene expression data from patients. RESULTS: In reaction to Wnt activation we observe downregulation of location specific genes and differentiation markers. A similar effect is seen in patient data, where genes with significant differential expression between the normal left and right colon are downregulated in the cancer samples. Furthermore, the signature of Wnt target genes differs between the three intestinal locations in the organoids. The location specific Wnt signatures are dominated by genes which have been lowly expressed in the tissue of origin, and are the targets of transcription factors that are activated following enhanced Wnt signalling. CONCLUSION: We observed that the region-specific cell identity has a substantial effect on the reaction to Wnt activation in a simple intestinal adenoma model. These findings provide a way forward in resolving the distinct biology between left- and right-sided human colon cancers with potential clinical relevance.

4.
Proc Natl Acad Sci U S A ; 113(5): E587-96, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26768848

RESUMEN

Complex interactions between DNA herpesviruses and host factors determine the establishment of a life-long asymptomatic latent infection. The lymphotropic Epstein-Barr virus (EBV) seems to avoid recognition by innate sensors despite massive transcription of immunostimulatory small RNAs (EBV-EBERs). Here we demonstrate that in latently infected B cells, EBER1 transcripts interact with the lupus antigen (La) ribonucleoprotein, avoiding cytoplasmic RNA sensors. However, in coculture experiments we observed that latent-infected cells trigger antiviral immunity in dendritic cells (DCs) through selective release and transfer of RNA via exosomes. In ex vivo tonsillar cultures, we observed that EBER1-loaded exosomes are preferentially captured and internalized by human plasmacytoid DCs (pDCs) that express the TIM1 phosphatidylserine receptor, a known viral- and exosomal target. Using an EBER-deficient EBV strain, enzymatic removal of 5'ppp, in vitro transcripts, and coculture experiments, we established that 5'pppEBER1 transfer via exosomes drives antiviral immunity in nonpermissive DCs. Lupus erythematosus patients suffer from elevated EBV load and activated antiviral immunity, in particular in skin lesions that are infiltrated with pDCs. We detected high levels of EBER1 RNA in such skin lesions, as well as EBV-microRNAs, but no intact EBV-DNA, linking non-cell-autonomous EBER1 presence with skin inflammation in predisposed individuals. Collectively, our studies indicate that virus-modified exosomes have a physiological role in the host-pathogen stand-off and may promote inflammatory disease.


Asunto(s)
Células Dendríticas/virología , Infecciones por Virus de Epstein-Barr/genética , Exosomas/metabolismo , ARN Viral/metabolismo , Transporte Biológico , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/genética , Humanos , Proteoma
5.
STAR Protoc ; 4(4): 102588, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37773752

RESUMEN

Long non-coding RNAs (lncRNAs) are emerging as key regulators in the initiation, growth, and progression of cancer. High-throughput CRISPR-based techniques systematically assess the function of genes or regulatory elements present in the human genome. Here, we present a protocol for identifying essential lncRNAs in cancer using CRISPRi-based dropout screens. We describe steps to select target sites, design guide RNAs, and generate CRISPRi cell lines. We then detail the execution and analysis of CRISPRi-based dropout screens.


Asunto(s)
Neoplasias , ARN Largo no Codificante , Humanos , Sistemas CRISPR-Cas/genética , ARN Guía de Sistemas CRISPR-Cas , ARN Largo no Codificante/genética , Neoplasias/diagnóstico , Neoplasias/genética , Genoma Humano
6.
EMBO Mol Med ; 14(12): e16194, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36321561

RESUMEN

The majority of colorectal cancers (CRCs) present with early mutations in tumor suppressor gene APC. APC mutations result in oncogenic activation of the Wnt pathway, which is associated with hyperproliferation, cytoskeletal remodeling, and a global increase in mRNA translation. To compensate for the increased biosynthetic demand, cancer cells critically depend on protein chaperones to maintain proteostasis, although their function in CRC remains largely unexplored. In order to investigate the role of molecular chaperones in driving CRC initiation, we captured the transcriptomic profiles of murine wild type and Apc-mutant organoids during active transformation. We discovered a strong transcriptional upregulation of Hspb1, which encodes small heat shock protein 25 (HSP25). We reveal an indispensable role for HSP25 in facilitating Apc-driven transformation, using both in vitro organoid cultures and mouse models, and demonstrate that chemical inhibition of HSP25 using brivudine reduces the development of premalignant adenomas. These findings uncover a hitherto unknown vulnerability in intestinal transformation that could be exploited for the development of chemopreventive strategies in high-risk individuals.


Asunto(s)
Transcriptoma , Animales , Ratones , Regulación hacia Arriba
7.
Cancers (Basel) ; 12(12)2020 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-33352769

RESUMEN

Maintenance of the intestinal epithelium is dependent on the control of stem cell (SC) proliferation and differentiation. The fine regulation of these cellular processes requires a complex dynamic interplay between several signaling pathways, including Wnt, Notch, Hippo, EGF, Ephrin, and BMP/TGF-ß. During the initiation and progression of colorectal cancer (CRC), key events, such as oncogenic mutations, influence these signaling pathways, and tilt the homeostatic balance towards proliferation and dedifferentiation. Therapeutic strategies to specifically target these deregulated signaling pathways are of particular interest. However, systemic blocking or activation of these pathways poses major risks for normal stem cell function and tissue homeostasis. Interestingly, long non-coding RNAs (lncRNAs) have recently emerged as potent regulators of key cellular processes often deregulated in cancer. Because of their exceptional tissue and tumor specificity, these regulatory RNAs represent attractive targets for cancer therapy. Here, we discuss how lncRNAs participate in the maintenance of intestinal homeostasis and how they can contribute to the deregulation of each signaling pathway in CRC. Finally, we describe currently available molecular tools to develop lncRNA-targeted cancer therapies.

8.
Crit Rev Oncol Hematol ; 139: 125-127, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30890387

RESUMEN

Exosome-mediated transfer of regulatory RNAs is a key feature that enables cancer cells to shape a tumor-promoting environment. Cancers growing in the bone can use this communication modality to disrupt the homeostatic balance between bone forming and bone resorbing cells, which results in the release of bone-embedded factors supporting cancer growth and progression. Long noncoding RNAs (lncRNAs) are potent regulators of cell fate determination with exceptional cell- and tissue-specificity that are secreted by cancer cells via exosomes. In multiple myeloma (MM), the exosomal transfer of the lncRNA RUNX2-AS1 specifically inhibits the osteogenic differentiation capacity of mesenchymal stem cells (MSC) by repressing the master regulator of bone formation RUNX2. Detailed studies into the role of exosomal lncRNA transfer in the bone microenvironment in vivo might constitute the basis for the development of novel therapeutic strategies for tumor-associated bone lesions.


Asunto(s)
Neoplasias Óseas/patología , Exosomas/metabolismo , Células Madre Mesenquimatosas/patología , ARN Largo no Codificante/genética , Microambiente Tumoral , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Exosomas/genética , Humanos , Células Madre Mesenquimatosas/metabolismo
9.
Nat Cell Biol ; 20(10): 1193-1202, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30177776

RESUMEN

Solid malignancies have been speculated to depend on cancer stem cells (CSCs) for expansion and relapse after therapy. Here we report on quantitative analyses of lineage tracing data from primary colon cancer xenograft tissue to assess CSC functionality in a human solid malignancy. The temporally obtained clone size distribution data support a model in which stem cell function in established cancers is not intrinsically, but is entirely spatiotemporally orchestrated. Functional stem cells that drive tumour expansion predominantly reside at the tumour edge, close to cancer-associated fibroblasts. Hence, stem cell properties change in time depending on the cell location. Furthermore, although chemotherapy enriches for cells with a CSC phenotype, in this context functional stem cell properties are also fully defined by the microenvironment. To conclude, we identified osteopontin as a key cancer-associated fibroblast-produced factor that drives in situ clonogenicity in colon cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Proliferación Celular/genética , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Oxaliplatino/administración & dosificación , Tamoxifeno/administración & dosificación , Microambiente Tumoral/genética
10.
Clin Cancer Res ; 23(14): 3721-3733, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28053020

RESUMEN

Purpose: Human osteosarcoma is a genetically heterogeneous bone malignancy with poor prognosis despite the employment of aggressive chemotherapy regimens. Because druggable driver mutations have not been established, dissecting the interactions between osteosarcoma cells and supporting stroma may provide insights into novel therapeutic targets.Experimental Design: By using a bioluminescent orthotopic xenograft mouse model of osteosarcoma, we evaluated the effect of tumor extracellular vesicle (EV)-educated mesenchymal stem cells (TEMSC) on osteosarcoma progression. Characterization and functional studies were designed to assess the mechanisms underlying MSC education. Independent series of tissue specimens were analyzed to corroborate the preclinical findings, and the composition of patient serum EVs was analyzed after isolation with size-exclusion chromatography.Results: We show that EVs secreted by highly malignant osteosarcoma cells selectively incorporate a membrane-associated form of TGFß, which induces proinflammatory IL6 production by MSCs. TEMSCs promote tumor growth, accompanied with intratumor STAT3 activation and lung metastasis formation, which was not observed with control MSCs. Importantly, intravenous administration of the anti-IL6 receptor antibody tocilizumab abrogated the tumor-promoting effects of TEMSCs. RNA-seq analysis of human osteosarcoma tissues revealed a distinct TGFß-induced prometastatic gene signature. Tissue microarray immunostaining indicated active STAT3 signaling in human osteosarcoma, consistent with the observations in TEMSC-treated mice. Finally, we isolated pure populations of EVs from serum and demonstrated that circulating levels of EV-associated TGFß are increased in osteosarcoma patients.Conclusions: Collectively, our findings suggest that TEMSCs promote osteosarcoma progression and provide the basis for testing IL6- and TGFß-blocking agents as new therapeutic options for osteosarcoma patients. Clin Cancer Res; 23(14); 3721-33. ©2017 AACR.


Asunto(s)
Interleucina-6/genética , Neoplasias Pulmonares/genética , Osteosarcoma/genética , Factor de Transcripción STAT3/genética , Factor de Crecimiento Transformador beta/genética , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/genética , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Transducción de Señal/genética , Análisis de Matrices Tisulares
11.
Stem Cell Investig ; 3: 22, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27488310

RESUMEN

Intestinal stem cells (ISCs) are adult multipotent cells essential for the maintenance of intestinal epithelial homeostasis. Wnt signaling activity ensures that the pool of ISCs at the basis of the intestinal crypts is preserved. Dysregulation of the Wnt pathway is often observed in cancer and supports malignant progression. Chiacchiera and colleagues recently demonstrated the implication of the polycomb complex PRC1 in the regulation of the Wnt pathway in adult ISCs. The authors show that PRC1 maintains intestinal homeostasis by repressing the expression of ZICs, a family of transcription factors inactivating the ß-catenin/TCF complex. Importantly, interfering with PRC1 activity completely inhibits the formation of Wnt-dependent tumors. These findings reveal a new layer of epigenetic regulation of the Wnt pathway and open novel opportunities for cancer stem cell targeted therapy.

12.
Expert Opin Biol Ther ; 15(5): 723-34, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25819025

RESUMEN

INTRODUCTION: Regulation of gene expression involves a variety of mechanisms driven by a complex regulatory network of factors. Control of transcription is an important step in gene expression regulation, which integrates the function of cis-acting and trans-acting elements. Among cis-regulatory elements, enhancer RNA (eRNA)-producing domains recently emerged as widespread and potent regulators of transcription and cell fate decision. Thus, manipulation of eRNA levels becomes a novel and appealing avenue for the design of new therapeutic treatments. AREAS COVERED: In this review, we focus on eRNA-producing domains. We describe mechanisms involved in their cell-type specific selection and activation as well as their epigenetic features. In addition, we present their function and the growing evidences of their deregulation in human diseases. Finally, we discuss eRNAs as potential therapeutic targets. EXPERT OPINION: As key factors in the control of transcription, eRNAs appear to possess a great potential for the establishment of new therapy options. However, thorough testing as well as providing the genetic toolbox to target eRNAs will be needed to fully assess the practical and clinical possibilities.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Marcación de Gen/tendencias , ARN/genética , Animales , Marcación de Gen/métodos , Humanos , ARN/metabolismo , Transcripción Genética/genética
13.
Nat Commun ; 6: 6520, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25813522

RESUMEN

p53 binds enhancers to regulate key target genes. Here, we globally mapped p53-regulated enhancers by looking at enhancer RNA (eRNA) production. Intriguingly, while many p53-induced enhancers contained p53-binding sites, most did not. As long non-coding RNAs (lncRNAs) are prominent regulators of chromatin dynamics, we hypothesized that p53-induced lncRNAs contribute to the activation of enhancers by p53. Among p53-induced lncRNAs, we identified LED and demonstrate that its suppression attenuates p53 function. Chromatin-binding and eRNA expression analyses show that LED associates with and activates strong enhancers. One prominent target of LED was located at an enhancer region within CDKN1A gene, a potent p53-responsive cell cycle inhibitor. LED knockdown reduces CDKN1A enhancer induction and activity, and cell cycle arrest following p53 activation. Finally, promoter-associated hypermethylation analysis shows silencing of LED in human tumours. Thus, our study identifies a new layer of complexity in the p53 pathway and suggests its dysregulation in cancer.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Mama/genética , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Proteína p53 Supresora de Tumor/genética , Inmunoprecipitación de Cromatina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Elementos de Facilitación Genéticos , Femenino , Humanos , Hibridación Fluorescente in Situ , Células MCF-7 , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN
14.
Cell Res ; 23(10): 1151-2, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23877407

RESUMEN

Recently, various studies shed light on the functional significance of enhancer RNAs. Two recent studies published in Nature by Li et al. and Lam et al. highlight the importance of these newly characterized RNA molecules and their key role in controlling transcriptional programs.


Asunto(s)
Regulación hacia Abajo/genética , Elementos de Facilitación Genéticos/genética , Estrógenos/farmacología , Macrófagos/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , ARN no Traducido/genética , Transcripción Genética/genética , Activación Transcripcional/efectos de los fármacos , Animales , Humanos
15.
Nat Commun ; 2: 513, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22027593

RESUMEN

MicroRNAs (miRNAs) interact with 3'-untranslated regions of messenger RNAs to restrict expression of most protein-coding genes during normal development and cancer. RNA-binding proteins (RBPs) can control the biogenesis, stability and activity of miRNAs. Here we identify RBM38 in a genetic screen for RBPs whose expression controls miRNA access to target mRNAs. RBM38 is induced by p53 and its ability to modulate miRNA-mediated repression is required for proper p53 function. In contrast, RBM38 shows lower propensity to block the action of the p53-controlled miR-34a on SIRT1. Target selectivity is determined by the interaction of RBM38 with uridine-rich regions near miRNA target sequences. Furthermore, in large cohorts of human breast cancer, reduced RBM38 expression by promoter hypermethylation correlates with wild-type p53 status. Thus, our results indicate a novel layer of p53 gene regulation, which is required for its tumour suppressive function.


Asunto(s)
MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regiones no Traducidas 3' , Ciclo Celular , Línea Celular Tumoral , Metilación de ADN , Citometría de Flujo , Humanos , Espectroscopía de Resonancia Magnética , Microscopía Fluorescente , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
J Steroid Biochem Mol Biol ; 114(3-5): 174-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19429448

RESUMEN

Androgens play a major role in the growth and survival of primary prostate tumors. The molecular mechanisms involved in prostate cancer progression are not fully understood but genes that are regulated by androgens clearly influence this process. We searched for new androgen-regulated genes using the Affymetrix GeneChip Human Genome U95 Set in the androgen-sensitive LNCaP prostate cancer cell line. Analysis of gene expression profiles revealed that myosin light chain kinase (MLCK) mRNA levels were markedly down-regulated by the synthetic androgen R1881. The microarray data were confirmed by ribonuclease protection assays. RNA and protein analyses revealed that LNCaP cells express both long (non-muscle) and short (smooth muscle) isoforms, and that both isoforms are down-regulated by androgens. Taken together, these data identify MLCK as a novel downstream target of the androgen signalling pathway in prostate cells.


Asunto(s)
Andrógenos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Perfilación de la Expresión Génica , Quinasa de Cadena Ligera de Miosina/genética , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metribolona/farmacología , Neoplasias de la Próstata/patología , Isoformas de Proteínas/genética , ARN Mensajero/análisis , Congéneres de la Testosterona/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA