Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Syst Biol ; 13(4): 927, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28455349

RESUMEN

The intestinal epithelium is the fastest regenerative tissue in the body, fueled by fast-cycling stem cells. The number and identity of these dividing and migrating stem cells are maintained by a mosaic pattern at the base of the crypt. How the underlying regulatory scheme manages this dynamic stem cell niche is not entirely clear. We stimulated intestinal organoids with Notch ligands and inhibitors and discovered that intestinal stem cells employ a positive feedback mechanism via direct Notch binding to the second intron of the Notch1 gene. Inactivation of the positive feedback by CRISPR/Cas9 mutation of the binding sequence alters the mosaic stem cell niche pattern and hinders regeneration in organoids. Dynamical system analysis and agent-based multiscale stochastic modeling suggest that the positive feedback enhances the robustness of Notch-mediated niche patterning. This study highlights the importance of feedback mechanisms in spatiotemporal control of the stem cell niche.


Asunto(s)
Retroalimentación Fisiológica , Intestinos/citología , Receptor Notch1/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Sitios de Unión , Autorrenovación de las Células , Humanos , Mucosa Intestinal/metabolismo , Ratones , Mutación , Organoides/metabolismo , Receptor Notch1/química , Transducción de Señal , Nicho de Células Madre , Procesos Estocásticos , Biología de Sistemas/métodos
2.
Genes Dis ; 8(2): 203-214, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33997167

RESUMEN

Colorectal cancer is a leading cause of cancer deaths. Most colorectal cancer patients eventually develop chemoresistance to the current standard-of-care therapies. Here, we used patient-derived colorectal cancer organoids to demonstrate that resistant tumor cells undergo significant chromatin changes in response to oxaliplatin treatment. Integrated transcriptomic and chromatin accessibility analyses using ATAC-Seq and RNA-Seq identified a group of genes associated with significantly increased chromatin accessibility and upregulated gene expression. CRISPR/Cas9 silencing of fibroblast growth factor receptor 1 (FGFR1) and oxytocin receptor (OXTR) helped overcome oxaliplatin resistance. Similarly, treatment with oxaliplatin in combination with an FGFR1 inhibitor (PD166866) or an antagonist of OXTR (L-368,899) suppressed chemoresistant organoids. However, oxaliplatin treatment did not activate either FGFR1 or OXTR expression in another resistant organoid, suggesting that chromatin accessibility changes are patient-specific. The use of patient-derived cancer organoids in combination with transcriptomic and chromatin profiling may lead to precision treatments to overcome chemoresistance in colorectal cancer.

3.
PLoS One ; 14(1): e0209179, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30640896

RESUMEN

Certain organs are capable of containing the replication of various types of viruses. In the liver, infection of Hepatitis B virus (HBV), the etiological factor of Hepatitis B and hepatocellular carcinoma (HCC), often remains asymptomatic and leads to a chronic carrier state. Here we investigated how hepatocytes contain HBV replication and promote their own survival by orchestrating a translational defense mechanism via the stress-sensitive SUMO-2/3-specific peptidase SENP3. We found that SENP3 expression level decreased in HBV-infected hepatocytes in various models including HepG2-NTCP cell lines and a humanized mouse model. Downregulation of SENP3 reduced HBV replication and boosted host protein translation. We also discovered that IQGAP2, a Ras GTPase-activating-like protein, is a key substrate for SENP3-mediated de-SUMOylation. Downregulation of SENP3 in HBV infected cells facilitated IQGAP2 SUMOylation and degradation, which leads to suppression of HBV gene expression and restoration of global translation of host genes via modulation of AKT phosphorylation. Thus, The SENP3-IQGAP2 de-SUMOylation axis is a host defense mechanism of hepatocytes that restores host protein translation and suppresses HBV gene expression.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Virus de la Hepatitis B/fisiología , Hepatocitos/metabolismo , Hepatocitos/virología , Interacciones Microbiota-Huesped/fisiología , Animales , Cisteína Endopeptidasas/genética , Regulación hacia Abajo , Regulación Viral de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Hep G2 , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Interacciones Microbiota-Huesped/genética , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especificidad por Sustrato , Sumoilación , Replicación Viral/fisiología , Proteínas Activadoras de ras GTPasa/antagonistas & inhibidores , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
4.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 5022-5025, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30441469

RESUMEN

Classically, the Wnt/ß-catenin and Notch /Delta signaling pathways were thought to operate through separate mechanisms, performing distinct roles in tissue patterning. However, it has been shown that b-catenin activates transcription of Hesl, a signaling intermediate in the Notch /Delta pathway that controls its lateral inhibition mechanism. To investigate this non-canonical crosstalk mechanism, a new gene circuit, integrating the two pathways, is proposed and simulated in two-cell and multi-cell environments. This model also captures both Paneth cell- mediated and mesenchymal Wnt production. The simulations verify that the gene circuit is temporally bistable and capable of forming a pattern on a multi-cell grid. Last, the model exhibits a bifurcation based on the steady state concentration of Wnt and the relative amount of control b-catenin has over the Hesl promoter, providing a possible mechanism to explain why a homogeneous population of transit amplifying cells is observed directly above the more diverse stem niche.


Asunto(s)
Redes Reguladoras de Genes , Receptores Notch/genética , Factor de Transcripción HES-1/metabolismo , Vía de Señalización Wnt , Simulación por Computador , Humanos , Modelos Biológicos , Células de Paneth/fisiología , Regiones Promotoras Genéticas
5.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 2378-2381, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30440885

RESUMEN

Organoids are three-dimensional cell cultures that mimic organ functions and structures. The organoid model has been developed as a versatile in vitro platform for stem cell biology and diseases modeling. Tumor organoids are shown to share ~ 90% of genetic mutations with biopsies from same patients. However, it's not clear whether tumor organoids recapitulate the cellular heterogeneity observed in patient tumors. Here, we used single-cell RNA-Seq to investigate the transcriptomics of tumor organoids derived from human colorectal tumors, and applied machine learning methods to unbiasedly cluster subtypes in tumor organoids. Computational analysis reveals cancer heterogeneity sustained in tumor organoids, and the subtypes in organoids displayed high diversity. Furthermore, we treated the tumor organoids with a first-line cancer drug, Oxaliplatin, and investigated drug response in single-cell scale. Diversity of tumor cell populations in organoids were significantly perturbed by drug treatment. Single-cell analysis detected the depletion of chemosensitive subgroups and emergence of new drug tolerant subgroups after drug treatment. Our study suggests that the organoid model is capable of recapitulating clinical heterogeneity and its evolution in response to chemotherapy.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Organoides/metabolismo , Oxaliplatino/farmacología , Análisis de la Célula Individual , Transcriptoma , Técnicas de Cultivo de Célula , Humanos , Organoides/efectos de los fármacos
6.
Elife ; 72018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29629872

RESUMEN

Notch signalling maintains stem cell regeneration at the mouse intestinal crypt base and balances the absorptive and secretory lineages in the upper crypt and villus. Here we report the role of Fringe family of glycosyltransferases in modulating Notch activity in the two compartments. At the crypt base, RFNG is enriched in the Paneth cells and increases cell surface expression of DLL1 and DLL4. This promotes Notch activity in the neighbouring Lgr5+ stem cells assisting their self-renewal. Expressed by various secretory cells in the upper crypt and villus, LFNG promotes DLL surface expression and suppresses the secretory lineage . Hence, in the intestinal epithelium, Fringes are present in the ligand-presenting 'sender' secretory cells and promote Notch activity in the neighbouring 'receiver' cells. Fringes thereby provide for targeted modulation of Notch activity and thus the cell fate in the stem cell zone, or the upper crypt and villus.


Asunto(s)
Homeostasis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Intestinos/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Células Madre/citología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Glucosiltransferasas , Glicosiltransferasas , Péptidos y Proteínas de Señalización Intercelular/genética , Intestinos/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas/fisiología , Receptores Acoplados a Proteínas G/fisiología , Receptores Notch/genética , Transducción de Señal , Células Madre/metabolismo
7.
Cancer Res ; 76(11): 3411-21, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27197180

RESUMEN

Colorectal cancer cells with stem-like properties, referred to as colon cancer-initiating cells (CCIC), have high tumorigenic potential. While CCIC can differentiate to promote cellular heterogeneity, it remains unclear whether CCIC within a tumor contain distinct subpopulations. Here, we describe the co-existence of fast- and slow-cycling CCIC, which can undergo asymmetric division to generate each other, highlighting CCIC plasticity and interconvertibility. Fast-cycling CCIC express markers, such as LGR5 and CD133, rely on MYC for their proliferation, whereas slow-cycling CCIC express markers, such as BMI1 and hTERT, are independent of MYC. NOTCH signaling promotes asymmetric cell fate, regulating the balance between these two populations. Overall, our results illuminate the basis for CCIC heterogeneity and plasticity by defining a direct interconversion mechanism between slow- and fast-cycling CCIC. Cancer Res; 76(11); 3411-21. ©2016 AACR.


Asunto(s)
Diferenciación Celular , Neoplasias del Colon/patología , Células Madre Neoplásicas/patología , Receptores Notch/metabolismo , Antígeno AC133/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Neoplasias del Colon/metabolismo , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Telomerasa/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Sci Rep ; 6: 26069, 2016 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-27181744

RESUMEN

Rapidly cycling LGR5+ intestinal stem cells (ISCs) located at the base of crypts are the primary driver of regeneration. Additionally, BMI1 expression is correlated with a slow cycling pool of ISCs located at +4 position. While previous reports have shown interconversion between these two populations following tissue injury, we provide evidence that NOTCH signaling regulates the balance between these two populations and promotes asymmetric division as a mechanism for interconversion in the mouse intestine. In both in vitro and in vivo models, NOTCH suppression reduces the ratio of BMI1+/LGR5+ ISCs while NOTCH stimulation increases this ratio. Furthermore, NOTCH signaling can activate asymmetric division after intestinal inflammation. Overall, these data provide insights into ISC plasticity, demonstrating a direct interconversion mechanism between slow- and fast-cycling ISCs.


Asunto(s)
Células Madre Adultas/fisiología , Intestinos/citología , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Notch/metabolismo , Animales , División Celular , Plasticidad de la Célula , Transdiferenciación Celular , Células Cultivadas , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Acoplados a Proteínas G/genética , Regeneración , Transducción de Señal
9.
Cell Stem Cell ; 18(2): 189-202, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26849305

RESUMEN

Emerging evidence suggests that microRNAs can initiate asymmetric division, but whether microRNA and protein cell fate determinants coordinate with each other remains unclear. Here, we show that miR-34a directly suppresses Numb in early-stage colon cancer stem cells (CCSCs), forming an incoherent feedforward loop (IFFL) targeting Notch to separate stem and non-stem cell fates robustly. Perturbation of the IFFL leads to a new intermediate cell population with plastic and ambiguous identity. Lgr5+ mouse intestinal/colon stem cells (ISCs) predominantly undergo symmetric division but turn on asymmetric division to curb the number of ISCs when proinflammatory response causes excessive proliferation. Deletion of miR-34a inhibits asymmetric division and exacerbates Lgr5+ ISC proliferation under such stress. Collectively, our data indicate that microRNA and protein cell fate determinants coordinate to enhance robustness of cell fate decision, and they provide a safeguard mechanism against stem cell proliferation induced by inflammation or oncogenic mutation.


Asunto(s)
División Celular Asimétrica , Inflamación/patología , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Animales , División Celular Asimétrica/efectos de los fármacos , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Receptores Notch/metabolismo , Estrés Fisiológico/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
10.
Nat Commun ; 6: 6879, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25872451

RESUMEN

As patient survival drops precipitously from early-stage cancers to late-stage and metastatic cancers, microRNAs that promote relapse and metastasis can serve as prognostic and predictive markers as well as therapeutic targets for chemoprevention. Here we show that miR-1269a promotes colorectal cancer (CRC) metastasis and forms a positive feedback loop with TGF-ß signalling. miR-1269a is upregulated in late-stage CRCs, and long-term monitoring of 100 stage II CRC patients revealed that miR-1269a expression in their surgically removed primary tumours is strongly associated with risk of CRC relapse and metastasis. Consistent with clinical observations, miR-1269a significantly increases the ability of CRC cells to invade and metastasize in vivo. TGF-ß activates miR-1269 via Sox4, while miR-1269a enhances TGF-ß signalling by targeting Smad7 and HOXD10, hence forming a positive feedback loop. Our findings suggest that miR-1269a is a potential marker to inform adjuvant chemotherapy decisions for CRC patients and a potential therapeutic target to deter metastasis.


Asunto(s)
Neoplasias Colorrectales/genética , Retroalimentación Fisiológica , MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Técnica del Anticuerpo Fluorescente , Células HCT116 , Células HT29 , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , MicroARNs/metabolismo , Persona de Mediana Edad , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Trasplante de Neoplasias , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción SOXC/metabolismo , Proteína smad7/metabolismo , Factores de Transcripción/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA