Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Genes Dev ; 32(15-16): 1020-1034, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30068703

RESUMEN

RNA-binding proteins (RBPs) are expressed broadly during both development and malignant transformation, yet their mechanistic roles in epithelial homeostasis or as drivers of tumor initiation and progression are incompletely understood. Here we describe a novel interplay between RBPs LIN28B and IMP1 in intestinal epithelial cells. Ribosome profiling and RNA sequencing identified IMP1 as a principle node for gene expression regulation downstream from LIN28B In vitro and in vivo data demonstrate that epithelial IMP1 loss increases expression of WNT target genes and enhances LIN28B-mediated intestinal tumorigenesis, which was reversed when we overexpressed IMP1 independently in vivo. Furthermore, IMP1 loss in wild-type or LIN28B-overexpressing mice enhances the regenerative response to irradiation. Together, our data provide new evidence for the opposing effects of the LIN28B-IMP1 axis on post-transcriptional regulation of canonical WNT signaling, with implications in intestinal homeostasis, regeneration and tumorigenesis.


Asunto(s)
Carcinogénesis , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Proteínas de Unión al ARN/metabolismo , Regulón , Vía de Señalización Wnt , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Humanos , Mucosa Intestinal/fisiología , Ratones , Ratones Transgénicos , Oncogenes , Biosíntesis de Proteínas , Proteínas de Unión al ARN/fisiología , Regeneración , Células Madre/metabolismo
2.
Nature ; 545(7653): 238-242, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28467820

RESUMEN

The canonical Wnt/ß-catenin signalling pathway governs diverse developmental, homeostatic and pathological processes. Palmitoylated Wnt ligands engage cell-surface frizzled (FZD) receptors and LRP5 and LRP6 co-receptors, enabling ß-catenin nuclear translocation and TCF/LEF-dependent gene transactivation. Mutations in Wnt downstream signalling components have revealed diverse functions thought to be carried out by Wnt ligands themselves. However, redundancy between the 19 mammalian Wnt proteins and 10 FZD receptors and Wnt hydrophobicity have made it difficult to attribute these functions directly to Wnt ligands. For example, individual mutations in Wnt ligands have not revealed homeostatic phenotypes in the intestinal epithelium-an archetypal canonical, Wnt pathway-dependent, rapidly self-renewing tissue, the regeneration of which is fueled by proliferative crypt Lgr5+ intestinal stem cells (ISCs). R-spondin ligands (RSPO1-RSPO4) engage distinct LGR4-LGR6, RNF43 and ZNRF3 receptor classes, markedly potentiate canonical Wnt/ß-catenin signalling, and induce intestinal organoid growth in vitro and Lgr5+ ISCs in vivo. However, the interchangeability, functional cooperation and relative contributions of Wnt versus RSPO ligands to in vivo canonical Wnt signalling and ISC biology remain unknown. Here we identify the functional roles of Wnt and RSPO ligands in the intestinal crypt stem-cell niche. We show that the default fate of Lgr5+ ISCs is to differentiate, unless both RSPO and Wnt ligands are present. However, gain-of-function studies using RSPO ligands and a new non-lipidated Wnt analogue reveal that these ligands have qualitatively distinct, non-interchangeable roles in ISCs. Wnt proteins are unable to induce Lgr5+ ISC self-renewal, but instead confer a basal competency by maintaining RSPO receptor expression that enables RSPO ligands to actively drive and specify the extent of stem-cell expansion. This functionally non-equivalent yet cooperative interaction between Wnt and RSPO ligands establishes a molecular precedent for regulation of mammalian stem cells by distinct priming and self-renewal factors, with broad implications for precise control of tissue regeneration.


Asunto(s)
Autorrenovación de las Células , Intestinos/citología , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Células Madre/metabolismo , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , Animales , Linaje de la Célula , Proliferación Celular , Femenino , Humanos , Ligandos , Masculino , Ratones , Organoides/citología , Organoides/crecimiento & desarrollo , Análisis de la Célula Individual , Nicho de Células Madre , Transcriptoma , Ubiquitina-Proteína Ligasas/metabolismo , beta Catenina/metabolismo
3.
J Biol Chem ; 292(7): 2586-2600, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28053090

RESUMEN

Gut microbiota play an important role in regulating the development of the host immune system, metabolic rate, and at times, disease pathogenesis. The factors and mechanisms that mediate interactions between microbiota and the intestinal epithelium are not fully understood. We provide novel evidence that microbiota may control intestinal epithelial stem cell (IESC) proliferation in part through microRNAs (miRNAs). We demonstrate that miRNA profiles differ dramatically across functionally distinct cell types of the mouse jejunal intestinal epithelium and that miRNAs respond to microbiota in a highly cell type-specific manner. Importantly, we also show that miRNAs in IESCs are more prominently regulated by microbiota compared with miRNAs in any other intestinal epithelial cell subtype. We identify miR-375 as one miRNA that is significantly suppressed by the presence of microbiota in IESCs. Using a novel method to knockdown gene and miRNA expression ex vivo enteroids, we demonstrate that we can knock down gene expression in Lgr5+ IESCs. Furthermore, when we knock down miR-375 in IESCs, we observe significantly increased proliferative capacity. Understanding the mechanisms by which microbiota regulate miRNA expression in IESCs and other intestinal epithelial cell subtypes will elucidate a critical molecular network that controls intestinal homeostasis and, given the heightened interest in miRNA-based therapies, may offer novel therapeutic strategies in the treatment of gastrointestinal diseases associated with altered IESC function.


Asunto(s)
Mucosa Intestinal/metabolismo , Células Madre/microbiología , Transcriptoma , Animales , Femenino , Vida Libre de Gérmenes , Proteínas Fluorescentes Verdes/genética , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Factor de Transcripción SOX9/genética , Células Madre/citología
4.
J Biol Chem ; 291(31): 15975-84, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27261459

RESUMEN

Proliferation and differentiation of intestinal epithelial cells (IECs) occur in part through precise regulation of key transcription factors, such as SOX9. MicroRNAs (miRNAs) have emerged as prominent fine-tuners of transcription factor expression and activity. We hypothesized that miRNAs, in part through the regulation of SOX9, may mediate IEC homeostasis. Bioinformatic analyses of the SOX9 3'-UTR revealed highly conserved target sites for nine different miRNAs. Of these, only the miR-30 family members were both robustly and variably expressed across functionally distinct cell types of the murine jejunal epithelium. Inhibition of miR-30 using complementary locked nucleic acids (LNA30bcd) in both human IECs and human colorectal adenocarcinoma-derived Caco-2 cells resulted in significant up-regulation of SOX9 mRNA but, interestingly, significant down-regulation of SOX9 protein. To gain mechanistic insight into this non-intuitive finding, we performed RNA sequencing on LNA30bcd-treated human IECs and found 2440 significantly increased genes and 2651 significantly decreased genes across three time points. The up-regulated genes are highly enriched for both predicted miR-30 targets, as well as genes in the ubiquitin-proteasome pathway. Chemical suppression of the proteasome rescued the effect of LNA30bcd on SOX9 protein levels, indicating that the regulation of SOX9 protein by miR-30 is largely indirect through the proteasome pathway. Inhibition of the miR-30 family led to significantly reduced IEC proliferation and a dramatic increase in markers of enterocyte differentiation. This in-depth analysis of a complex miRNA regulatory program in intestinal epithelial cell models provides novel evidence that the miR-30 family likely plays an important role in IEC homeostasis.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Enterocitos/metabolismo , Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Factor de Transcripción SOX9/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células CACO-2 , Enterocitos/citología , Humanos , Masculino , Ratones , Ratones Mutantes , MicroARNs/genética , Factor de Transcripción SOX9/genética , Ubiquitina-Proteína Ligasas/genética
5.
J Physiol ; 594(17): 4837-47, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27581568

RESUMEN

Wnt signalling is involved in multiple aspects of embryonic development and adult tissue homeostasis, notably via controlling cellular proliferation and differentiation. Wnt signalling is subject to stringent positive and negative regulation to promote proper development and homeostasis yet avoid aberrant growth. Such multi-layer regulation includes post-translational modification and processing of Wnt proteins themselves, R-spondin (Rspo) amplification of Wnt signalling, diverse receptor families, and intracellular and extracellular antagonists and destruction and transcription complexes. In the gastrointestinal tract, Wnt signalling is crucial for development and renewal of the intestinal epithelium. Intestinal stem cells (ISCs) undergo symmetric division and neutral drift dynamics to renew the intestinal epithelium. Sources of Wnts and Wnt amplifers such as R-spondins are beginning to be elucidated as well as their functional contribution to intestinal homeostasis. In this review we focus on regulation of ISCs and intestinal homeostasis by the Wnt/Rspo pathway, the potential cellular sources of Wnt signalling regulators and highlight potential future areas of study.


Asunto(s)
Intestinos/citología , Células Madre/fisiología , Vía de Señalización Wnt , Animales , Intestinos/fisiología , Regeneración
6.
FASEB J ; 29(7): 2828-42, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25837582

RESUMEN

Insulin-like growth factor 1 (IGF1) has potent trophic effects on normal or injured intestinal epithelium, but specific effects on intestinal stem cells (ISCs) are undefined. We used Sox9-enhanced green fluorescent protein (EGFP) reporter mice that permit analyses of both actively cycling ISCs (Sox9-EGFP(Low)) and reserve/facultative ISCs (Sox9-EGFP(High)) to study IGF1 action on ISCs in normal intestine or during crypt regeneration after high-dose radiation-induced injury. We hypothesized that IGF1 differentially regulates proliferation and gene expression in actively cycling and reserve/facultative ISCs. IGF1 was delivered for 5 days using subcutaneously implanted mini-pumps in uninjured mice or after 14 Gy abdominal radiation. ISC numbers, proliferation, and transcriptome were assessed. IGF1 increased epithelial growth in nonirradiated mice and enhanced crypt regeneration after radiation. In uninjured and regenerating intestines, IGF1 increased total numbers of Sox9-EGFP(Low) ISCs and percentage of these cells in M-phase. IGF1 increased percentages of Sox9-EGFP(High) ISCs in S-phase but did not expand this population. Microarray revealed that IGF1 activated distinct gene expression signatures in the 2 Sox9-EGFP ISC populations. In vitro IGF1 enhanced enteroid formation by Sox9-EGFP(High) facultative ISCs but not Sox9-EGFP(Low) actively cycling ISCs. Our data provide new evidence that IGF1 activates 2 ISC populations via distinct regulatory pathways to promote growth of normal intestinal epithelium and crypt regeneration after irradiation.


Asunto(s)
Células Madre Adultas/clasificación , Factor I del Crecimiento Similar a la Insulina/fisiología , Intestino Delgado/citología , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/fisiología , Animales , Ciclo Celular , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/fisiología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/fisiología , Ratones , Ratones Transgénicos , Células Madre Multipotentes/clasificación , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/fisiopatología , Receptor IGF Tipo 1/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Regeneración/efectos de los fármacos , Regeneración/fisiología , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 309(7): G578-89, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26251471

RESUMEN

Current views suggest that apoptosis eliminates genetically damaged cells that may otherwise form tumors. Prior human studies link elevated insulin and reduced apoptosis to risk of colorectal adenomas. We hypothesized that hyperinsulinemia associated with obesity would lead to reduced colon epithelial cell (CEC) apoptosis after radiation and that this effect would be altered by deletion of the insulin-like growth factor (IGF) 1 receptor (IGF1R) or the insulin receptor (IR). Mice with villin-Cre-mediated IGF1R or IR deletion in CECs and floxed littermates were fed a high-fat diet to induce obesity and hyperinsulinemia or control low-fat chow. Mice were exposed to 5-Gy abdominal radiation to induce DNA damage and euthanized 4 h later for evaluation of apoptosis by localization of cleaved caspase-3. Obese mice exhibited decreased apoptosis of genetically damaged CECs. IGF1R deletion did not affect CEC apoptosis in lean or obese animals. In contrast, IR loss increased CEC apoptosis in both diet groups but did not prevent antiapoptotic effects of obesity. Levels of p53 protein were significantly reduced in CECs of obese mice with intact IR but increased in both lean and obese mice without IR. Levels of mRNAs encoding proapoptotic Perp and the cell cycle inhibitor Cdkn1b/p27 were reduced in CECs of obese mice and increased in lean mice lacking IR. Together, our studies provide novel evidence for antiapoptotic roles of obesity and IR, but not IGF1R, in colonic epithelium after DNA damage. However, neither IR nor IGF1R deletion prevented a reduction in radiation-induced CEC apoptosis during obesity and hyperinsulinemia.


Asunto(s)
Apoptosis/efectos de la radiación , Colon/patología , Mucosa Intestinal/metabolismo , Obesidad/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Western Blotting , Caspasa 3 , Colon/metabolismo , Inmunohistoquímica , Masculino , Ratones , Traumatismos Experimentales por Radiación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética
8.
Am J Physiol Gastrointest Liver Physiol ; 308(2): G100-11, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25394660

RESUMEN

The insulin receptor (IR) regulates nutrient uptake and utilization in multiple organs, but its role in the intestinal epithelium is not defined. This study developed a mouse model with villin-Cre (VC) recombinase-mediated intestinal epithelial cell (IEC)-specific IR deletion (VC-IR(Δ/Δ)) and littermate controls with floxed, but intact, IR (IR(fl/fl)) to define in vivo roles of IEC-IR in mice fed chow or high-fat diet (HFD). We hypothesized that loss of IEC-IR would alter intestinal growth, biomarkers of intestinal epithelial stem cells (IESC) or other lineages, body weight, adiposity, and glucose or lipid handling. In lean, chow-fed mice, IEC-IR deletion did not affect body or fat mass, plasma glucose, or IEC proliferation. In chow-fed VC-IR(Δ/Δ) mice, mRNA levels of the Paneth cell marker lysozyme (Lyz) were decreased, but markers of other differentiated lineages were unchanged. During HFD-induced obesity, IR(fl/fl) and VC-IR(Δ/Δ) mice exhibited similar increases in body and fat mass, plasma insulin, mRNAs encoding several lipid-handling proteins, a decrease in Paneth cell number, and impaired glucose tolerance. In IR(fl/fl) mice, HFD-induced obesity increased circulating cholesterol; numbers of chromogranin A (CHGA)-positive enteroendocrine cells (EEC); and mRNAs encoding Chga, glucose-dependent insulinotrophic peptide (Gip), glucagon (Gcg), Lyz, IESC biomarkers, and the enterocyte cholesterol transporter Scarb1. All these effects were attenuated or lost in VC-IR(Δ/Δ) mice. These results demonstrate that IEC-IR is not required for normal growth of the intestinal epithelium in lean adult mice. However, our findings provide novel evidence that, during HFD-induced obesity, IEC-IR contributes to increases in EEC, plasma cholesterol, and increased expression of Scarb1 or IESC-, EEC-, and Paneth cell-derived mRNAs.


Asunto(s)
Colesterol/metabolismo , Dieta Alta en Grasa , Células Enteroendocrinas/metabolismo , Intestinos/patología , Células de Paneth/metabolismo , Receptor de Insulina/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Polipéptido Inhibidor Gástrico/metabolismo , Insulina/sangre , Mucosa Intestinal/metabolismo , Ratones , Ratones Transgénicos , Obesidad/metabolismo , ARN Mensajero/metabolismo
9.
J Cell Sci ; 126(Pt 24): 5645-56, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24127567

RESUMEN

Despite evidence for the impact of insulin on intestinal epithelial physiology and pathophysiology, the expression patterns, roles, and regulation of insulin receptor (IR) and IR isoforms in the intestinal epithelium are not well characterized. IR-A is thought to mediate the proliferative effects of insulin or insulin growth factors (IGFs) in fetal or cancer cells. IR-B is considered to be the metabolic receptor for insulin in specialized tissues. This study used a novel Sox9-EGFP reporter mouse that permits isolation of intestinal epithelial stem cells (IESCs), progenitors, enteroendocrine cells and differentiated lineages, the Apc(Min/+) mouse model of precancerous adenoma and normal human intestinal and colorectal cancer (CRC) cell lines. We tested the hypothesis that there is differential expression of IR-A or IR-B in stem and tumor cells versus differentiated intestinal epithelial cells (IECs) and that IR-B impacts cell proliferation. Our findings provide evidence that IR-B expression is significantly lower in highly proliferative IESCs and progenitor cells versus post-mitotic, differentiated IECs and in subconfluent and undifferentiated versus differentiated Caco-2 cells. IR-B is also reduced in Apc(Min/+) tumors and highly tumorigenic CRC cells. These differences in IR-B were accompanied by altered levels of mRNAs encoding muscleblind-like 2 (MBNL2), a known regulator of IR alternative splicing. Forced IR-B expression in subconfluent and undifferentiated Caco-2 cells reduced proliferation and increased biomarkers of differentiation. Our findings indicate that the impact of insulin on different cell types in the intestinal epithelium might differ depending on relative IR-B IR-A expression levels and provide new evidence for the roles of IR-B to limit proliferation of CRC cells.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/metabolismo , Receptor de Insulina/metabolismo , Células Madre/metabolismo , Animales , Células CACO-2 , Diferenciación Celular , Replicación del ADN , Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Ratones , Fenotipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor de Insulina/genética , Transducción de Señal , Proteína de la Zonula Occludens-1/metabolismo , beta Catenina/metabolismo
10.
Cell Rep ; 42(11): 113355, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37922313

RESUMEN

Somatic copy number gains are pervasive across cancer types, yet their roles in oncogenesis are insufficiently evaluated. This inadequacy is partly due to copy gains spanning large chromosomal regions, obscuring causal loci. Here, we employed organoid modeling to evaluate candidate oncogenic loci identified via integrative computational analysis of extreme copy gains overlapping with extreme expression dysregulation in The Cancer Genome Atlas. Subsets of "outlier" candidates were contextually screened as tissue-specific cDNA lentiviral libraries within cognate esophagus, oral cavity, colon, stomach, pancreas, and lung organoids bearing initial oncogenic mutations. Iterative analysis nominated the kinase DYRK2 at 12q15 as an amplified head and neck squamous carcinoma oncogene in p53-/- oral mucosal organoids. Similarly, FGF3, amplified at 11q13 in 41% of esophageal squamous carcinomas, promoted p53-/- esophageal organoid growth reversible by small molecule and soluble receptor antagonism of FGFRs. Our studies establish organoid-based contextual screening of candidate genomic drivers, enabling functional evaluation during early tumorigenesis.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/genética , Oncogenes , Transformación Celular Neoplásica/genética , Neoplasias/genética , Carcinogénesis/genética , Amplificación de Genes
11.
Am J Physiol Gastrointest Liver Physiol ; 302(10): G1111-32, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22361729

RESUMEN

Recent identification of intestinal epithelial stem cell (ISC) markers and development of ISC reporter mice permit visualization and isolation of regenerating ISCs after radiation to define their functional and molecular phenotypes. Previous studies in uninjured intestine of Sox9-EGFP reporter mice demonstrate that ISCs express low levels of Sox9-EGFP (Sox9-EGFP Low), whereas enteroendocrine cells (EEC) express high levels of Sox9-EGFP (Sox9-EGFP High). We hypothesized that Sox9-EGFP Low ISCs would expand after radiation, exhibit enhanced proliferative capacities, and adopt a distinct gene expression profile associated with rapid proliferation. Sox9-EGFP mice were given 14 Gy abdominal radiation and studied between days 3 and 9 postradiation. Radiation-induced changes in number, growth, and transcriptome of the different Sox9-EGFP cell populations were determined by histology, flow cytometry, in vitro culture assays, and microarray. Microarray confirmed that nonirradiated Sox9-EGFP Low cells are enriched for Lgr5 mRNA and mRNAs enriched in Lgr5-ISCs and identified additional putative ISC markers. Sox9-EGFP High cells were enriched for EEC markers, as well as Bmi1 and Hopx, which are putative markers of quiescent ISCs. Irradiation caused complete crypt loss, followed by expansion and hyperproliferation of Sox9-EGFP Low cells. From nonirradiated intestine, only Sox9-EGFP Low cells exhibited ISC characteristics of forming organoids in culture, whereas during regeneration both Sox9-EGFP Low and High cells formed organoids. Microarray demonstrated that regenerating Sox9-EGFP High cells exhibited transcriptomic changes linked to p53-signaling and ISC-like functions including DNA repair and reduced oxidative metabolism. These findings support a model in which Sox9-EGFP Low cells represent active ISCs, Sox9-EGFP High cells contain radiation-activatable cells with ISC characteristics, and both participate in crypt regeneration.


Asunto(s)
Proteínas Fluorescentes Verdes/biosíntesis , Yeyuno/efectos de la radiación , Factor de Transcripción SOX9/biosíntesis , Células Madre/efectos de la radiación , Animales , Proliferación Celular/efectos de la radiación , Células Cultivadas , Expresión Génica/efectos de la radiación , Proteínas de Homeodominio/biosíntesis , Yeyuno/metabolismo , Ratones , Ratones Transgénicos , Proteínas Nucleares/biosíntesis , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Regeneración/efectos de la radiación , Proteínas Represoras/biosíntesis
12.
Cancer Discov ; 11(6): 1562-1581, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33451982

RESUMEN

Mutations in ARID1A rank among the most common molecular aberrations in human cancer. However, oncogenic consequences of ARID1A mutation in human cells remain poorly defined due to lack of forward genetic models. Here, CRISPR/Cas9-mediated ARID1A knockout (KO) in primary TP53-/- human gastric organoids induced morphologic dysplasia, tumorigenicity, and mucinous differentiation. Genetic WNT/ß-catenin activation rescued mucinous differentiation, but not hyperproliferation, suggesting alternative pathways of ARID1A KO-mediated transformation. ARID1A mutation induced transcriptional regulatory modules characteristic of microsatellite instability and Epstein-Barr virus-associated subtype human gastric cancer, including FOXM1-associated mitotic genes and BIRC5/survivin. Convergently, high-throughput compound screening indicated selective vulnerability of ARID1A-deficient organoids to inhibition of BIRC5/survivin, functionally implicating this pathway as an essential mediator of ARID1A KO-dependent early-stage gastric tumorigenesis. Overall, we define distinct pathways downstream of oncogenic ARID1A mutation, with nonessential WNT-inhibited mucinous differentiation in parallel with essential transcriptional FOXM1/BIRC5-stimulated proliferation, illustrating the general utility of organoid-based forward genetic cancer analysis in human cells. SIGNIFICANCE: We establish the first human forward genetic modeling of a commonly mutated tumor suppressor gene, ARID1A. Our study integrates diverse modalities including CRISPR/Cas9 genome editing, organoid culture, systems biology, and small-molecule screening to derive novel insights into early transformation mechanisms of ARID1A-deficient gastric cancers.See related commentary by Zafra and Dow, p. 1327.This article is highlighted in the In This Issue feature, p. 1307.


Asunto(s)
Sistemas CRISPR-Cas , Transformación Celular Neoplásica , Proteínas de Unión al ADN/genética , Neoplasias Gástricas/genética , Factores de Transcripción/genética , Humanos , Modelos Biológicos , Mutación
13.
Cell Rep ; 26(9): 2509-2520.e4, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30811997

RESUMEN

Human enteroids-epithelial spheroids derived from primary gastrointestinal tissue-are a promising model to study pathogen-epithelial interactions. However, accessing the apical enteroid surface is challenging because it is enclosed within the spheroid. We developed a technique to reverse enteroid polarity such that the apical surface everts to face the media. Apical-out enteroids maintain proper polarity and barrier function, differentiate into the major intestinal epithelial cell (IEC) types, and exhibit polarized absorption of nutrients. We used this model to study host-pathogen interactions and identified distinct polarity-specific patterns of infection by invasive enteropathogens. Salmonella enterica serovar Typhimurium targets IEC apical surfaces for invasion via cytoskeletal rearrangements, and Listeria monocytogenes, which binds to basolateral receptors, invade apical surfaces at sites of cell extrusion. Despite different modes of entry, both pathogens exit the epithelium within apically extruding enteroid cells. This model will enable further examination of IECs in health and disease.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/microbiología , Interacciones Huésped-Patógeno , Mucosa Intestinal/citología , Técnicas de Cultivo de Célula , Diferenciación Celular , Polaridad Celular , Células Epiteliales/metabolismo , Ácidos Grasos/metabolismo , Humanos , Listeria monocytogenes/fisiología , Modelos Biológicos , Salmonella typhimurium/fisiología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Esferoides Celulares/microbiología
14.
Trends Cell Biol ; 28(12): 1062-1078, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30195922

RESUMEN

The intestinal epithelium is a rapidly renewing cellular compartment. This constant regeneration is a hallmark of intestinal homeostasis and requires a tightly regulated balance between intestinal stem cell (ISC) proliferation and differentiation. Since intestinal epithelial cells directly contact pathogenic environmental factors that continuously challenge their integrity, ISCs must also actively divide to facilitate regeneration and repair. Understanding niche adaptations that maintain ISC activity during homeostatic renewal and injury-induced intestinal regeneration is therefore a major and ongoing focus for stem cell biology. Here, we review recent concepts and propose an active interconversion of the ISC niche between homeostasis and injury-adaptive states that is superimposed upon an equally dynamic equilibrium between active and reserve ISC populations.


Asunto(s)
Homeostasis , Mucosa Intestinal/metabolismo , Nicho de Células Madre , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Mucosa Intestinal/patología
15.
Aging (Albany NY) ; 9(8): 1898-1915, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28854151

RESUMEN

Intestinal epithelial stem cells (IESCs) are critical to maintain intestinal epithelial function and homeostasis. We tested the hypothesis that aging promotes IESC dysfunction using old (18-22 months) and young (2-4 month) Sox9-EGFP IESC reporter mice. Different levels of Sox9-EGFP permit analyses of active IESC (Sox9-EGFPLow), activatable reserve IESC and enteroendocrine cells (Sox9-EGFPHigh), Sox9-EGFPSublow progenitors, and Sox9-EGFPNegative differentiated lineages. Crypt-villus morphology, cellular composition and apoptosis were measured by histology. IESC function was assessed by crypt culture, and proliferation by flow cytometry and histology. Main findings were confirmed in Lgr5-EGFP and Lgr5-LacZ mice. Aging-associated gene expression changes were analyzed by Fluidigm mRNA profiling. Crypts culture from old mice yielded fewer and less complex enteroids. Histology revealed increased villus height and Paneth cells per crypt in old mice. Old mice showed increased numbers and hyperproliferation of Sox9-EGFPLow IESC and Sox9-EGFPHigh cells. Cleaved caspase-3 staining demonstrated increased apoptotic cells in crypts and villi of old mice. Gene expression profiling revealed aging-associated changes in mRNAs associated with cell cycle, oxidative stress and apoptosis specifically in IESC. These findings provide new, direct evidence for aging associated IESC dysfunction, and define potential biomarkers and targets for translational studies to assess and maintain IESC function during aging.


Asunto(s)
Envejecimiento/patología , Proliferación Celular , Células Epiteliales/patología , Mucosa Intestinal/patología , Yeyuno/patología , Células Madre/patología , Factores de Edad , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Apoptosis , Ciclo Celular , Linaje de la Célula , Enterocitos/metabolismo , Enterocitos/patología , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Células Caliciformes/metabolismo , Células Caliciformes/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Yeyuno/metabolismo , Operón Lac , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Estrés Oxidativo , Células de Paneth/metabolismo , Células de Paneth/patología , Fenotipo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Esferoides Celulares , Células Madre/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos
16.
J Clin Invest ; 127(2): 593-607, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-28094771

RESUMEN

Orphan GPCRs provide an opportunity to identify potential pharmacological targets, yet their expression patterns and physiological functions remain challenging to elucidate. Here, we have used a genetically engineered knockin reporter mouse to map the expression pattern of the Gpr182 during development and adulthood. We observed that Gpr182 is expressed at the crypt base throughout the small intestine, where it is enriched in crypt base columnar stem cells, one of the most active stem cell populations in the body. Gpr182 knockdown had no effect on homeostatic intestinal proliferation in vivo, but led to marked increases in proliferation during intestinal regeneration following irradiation-induced injury. In the ApcMin mouse model, which forms spontaneous intestinal adenomas, reductions in Gpr182 led to more adenomas and decreased survival. Loss of Gpr182 enhanced organoid growth efficiency ex vivo in an EGF-dependent manner. Gpr182 reduction led to increased activation of ERK1/2 in basal and challenge models, demonstrating a potential role for this orphan GPCR in regulating the proliferative capacity of the intestine. Importantly, GPR182 expression was profoundly reduced in numerous human carcinomas, including colon adenocarcinoma. Together, these results implicate Gpr182 as a negative regulator of intestinal MAPK signaling-induced proliferation, particularly during regeneration and adenoma formation.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Proliferación Celular , Intestino Delgado/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias Experimentales/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Intestino Delgado/patología , Ratones , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Receptores Acoplados a Proteínas G/genética
17.
Nat Med ; 23(4): 450-460, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28288111

RESUMEN

Although blood-brain barrier (BBB) compromise is central to the etiology of diverse central nervous system (CNS) disorders, endothelial receptor proteins that control BBB function are poorly defined. The endothelial G-protein-coupled receptor (GPCR) Gpr124 has been reported to be required for normal forebrain angiogenesis and BBB function in mouse embryos, but the role of this receptor in adult animals is unknown. Here Gpr124 conditional knockout (CKO) in the endothelia of adult mice did not affect homeostatic BBB integrity, but resulted in BBB disruption and microvascular hemorrhage in mouse models of both ischemic stroke and glioblastoma, accompanied by reduced cerebrovascular canonical Wnt-ß-catenin signaling. Constitutive activation of Wnt-ß-catenin signaling fully corrected the BBB disruption and hemorrhage defects of Gpr124-CKO mice, with rescue of the endothelial gene tight junction, pericyte coverage and extracellular-matrix deficits. We thus identify Gpr124 as an endothelial GPCR specifically required for endothelial Wnt signaling and BBB integrity under pathological conditions in adult mice. This finding implicates Gpr124 as a potential therapeutic target for human CNS disorders characterized by BBB disruption.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Glioblastoma/genética , Infarto de la Arteria Cerebral Media/genética , Hemorragias Intracraneales/genética , Receptores Acoplados a Proteínas G/genética , Uniones Estrechas/metabolismo , Animales , Barrera Hematoencefálica/ultraestructura , Modelos Animales de Enfermedad , Células Endoteliales/ultraestructura , Matriz Extracelular/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glioblastoma/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Hemorragias Intracraneales/metabolismo , Ratones , Ratones Noqueados , Microscopía Electrónica , Microvasos , Pericitos/ultraestructura , Reacción en Cadena en Tiempo Real de la Polimerasa , Uniones Estrechas/ultraestructura , Vía de Señalización Wnt
18.
Endocrinology ; 155(9): 3302-14, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24914941

RESUMEN

Nutrient intake regulates intestinal epithelial mass and crypt proliferation. Recent findings in model organisms and rodents indicate nutrient restriction impacts intestinal stem cells (ISC). Little is known about the impact of diet-induced obesity (DIO), a model of excess nutrient intake on ISC. We used a Sox9-EGFP reporter mouse to test the hypothesis that an adaptive response to DIO or associated hyperinsulinemia involves expansion and hyperproliferation of ISC. The Sox9-EGFP reporter mouse allows study and isolation of ISC, progenitors, and differentiated lineages based on different Sox9-EGFP expression levels. Sox9-EGFP mice were fed a high-fat diet for 20 weeks to induce DIO and compared with littermates fed low-fat rodent chow. Histology, fluorescence activated cell sorting, and mRNA analyses measured impact of DIO on jejunal crypt-villus morphometry, numbers, and proliferation of different Sox9-EGFP cell populations and gene expression. An in vitro culture assay directly assessed functional capacity of isolated ISC. DIO mice exhibited significant increases in body weight, plasma glucose, insulin, and insulin-like growth factor 1 (IGF1) levels and intestinal Igf1 mRNA. DIO mice had increased villus height and crypt density but decreased intestinal length and decreased numbers of Paneth and goblet cells. In vivo, DIO resulted in a selective expansion of Sox9-EGFP(Low) ISC and percentage of ISC in S-phase. ISC expansion significantly correlated with plasma insulin levels. In vitro, isolated ISC from DIO mice formed fewer enteroids in standard 3D Matrigel culture compared to controls, indicating impaired ISC function. This decreased enteroid formation in isolated ISC from DIO mice was rescued by exogenous insulin, IGF1, or both. We conclude that DIO induces specific increases in ISC and ISC hyperproliferation in vivo. However, isolated ISC from DIO mice have impaired intrinsic survival and growth in vitro that can be rescued by exogenous insulin or IGF1.


Asunto(s)
Proliferación Celular , Dieta Alta en Grasa/efectos adversos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Intestinos/citología , Obesidad/metabolismo , Células Madre/citología , Animales , Femenino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Transgénicos , Obesidad/genética , Obesidad/fisiopatología , Transducción de Señal , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA