Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
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.
J Bacteriol ; 194(3): 617-26, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22123249

RESUMEN

Lateral gene transfer (LGT) is essential for generating between-strain genomic recombinants of Chlamydia trachomatis to facilitate the organism's evolution. Because there is no reliable laboratory-based gene transfer system for C. trachomatis, in vitro generation of recombinants from antibiotic-resistant strains is being used to study LGT. However, selection pressures imposed on in vitro recombinants likely affect statistical properties of recombination relative to naturally occurring clinical recombinants, including prevalence at particular loci. We examined multiple loci for 16 in vitro-derived recombinants of ofloxacin- and rifampin-resistant L(1) and D strains, respectively, grown with both antibiotics, and compared these with the same sequenced loci among 11 clinical recombinants. Breakpoints and recombination frequency were examined using phylogenetics, bioinformatics, and statistics. In vitro and clinical isolates clustered perfectly into two groups, without misclassification, using Ward's minimum variance based on breakpoint data. As expected, gyrA (confers ofloxacin resistance) and rpoB (confers rifampin resistance) had significantly more breakpoints among in vitro recombinants than among clinical recombinants (P < 0.0001 and P = 0.02, respectively, using the Wilcoxon rank sum test). Unexpectedly, trpA also had significantly more breakpoints for in vitro recombinants (P < 0.0001). There was also significant selection at other loci. The strongest bias was for ompA in strain D (P = 3.3 × 10(-8)). Our results indicate that the in vitro model differs statistically from natural recombination events. Additional genomic studies are needed to determine the factors responsible for the observed selection biases at unexpected loci and whether these are important for LGT to inform approaches for genetically manipulating C. trachomatis.


Asunto(s)
Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/genética , Farmacorresistencia Bacteriana , Recombinación Genética , Antibacterianos/farmacología , Secuencia de Bases , Chlamydia trachomatis/clasificación , Chlamydia trachomatis/efectos de los fármacos , Chlamydia trachomatis/metabolismo , Ingeniería Genética , Humanos , Datos de Secuencia Molecular , Filogenia
3.
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
4.
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.
Nat Med ; 23(7): 878-884, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28628110

RESUMEN

With the goal of modeling human disease of the large intestine, we sought to develop an effective protocol for deriving colonic organoids (COs) from differentiated human embryonic stem cells (hESCs) or induced pluripotent stem cells (iPSCs). Extensive gene and immunohistochemical profiling confirmed that the derived COs represent colon rather than small intestine, containing stem cells, transit-amplifying cells, and the expected spectrum of differentiated cells, including goblet and endocrine cells. We applied this strategy to iPSCs derived from patients with familial adenomatous polyposis (FAP-iPSCs) harboring germline mutations in the WNT-signaling-pathway-regulator gene encoding APC, and we generated COs that exhibit enhanced WNT activity and increased epithelial cell proliferation, which we used as a platform for drug testing. Two potential compounds, XAV939 and rapamycin, decreased proliferation in FAP-COs, but also affected cell proliferation in wild-type COs, which thus limits their therapeutic application. By contrast, we found that geneticin, a ribosome-binding antibiotic with translational 'read-through' activity, efficiently targeted abnormal WNT activity and restored normal proliferation specifically in APC-mutant FAP-COs. These studies provide an efficient strategy for deriving human COs, which can be used in disease modeling and drug discovery for colorectal disease.


Asunto(s)
Adenoma/genética , Poliposis Adenomatosa del Colon/genética , Antibióticos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Neoplasias Colorrectales/genética , Células Madre Embrionarias Humanas , Organoides/efectos de los fármacos , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Western Blotting , Diferenciación Celular , Colon/citología , Colon/metabolismo , Neoplasias Colorrectales/patología , Ensayos de Selección de Medicamentos Antitumorales , Células Enteroendocrinas/citología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Gentamicinas/farmacología , Mutación de Línea Germinal , Células Caliciformes/citología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas , Microscopía Confocal , Mutación , Organoides/citología , Organoides/metabolismo , Organoides/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Sirolimus/farmacología , Vía de Señalización Wnt
7.
J Vis Exp ; (111)2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27285214

RESUMEN

Primary intestinal organoids are a valuable model system that has the potential to significantly impact the field of mucosal immunology. However, the complexities of the organoid growth characteristics carry significant caveats for the investigator. Specifically, the growth patterns of each individual organoid are highly variable and create a heterogeneous population of epithelial cells in culture. With such caveats, common tissue culture practices cannot be simply applied to the organoid system due to the complexity of the cellular structure. Counting and plating based solely on cell number, which is common for individually separated cells, such as cell lines, is not a reliable method for organoids unless some normalization technique is applied. Normalizing to total protein content is made complex due to the resident protein matrix. These characteristics in terms of cell number, shape and cell type should be taken into consideration when evaluating secreted contents from the organoid mass. This protocol has been generated to outline a simple procedure to culture and treat small intestinal organoids with microbial pathogens and pathogen associated molecular patterns (PAMPs). It also emphasizes the normalization techniques that should be applied when protein analysis are conducted after such a challenge.


Asunto(s)
Intestino Delgado/metabolismo , Organoides , Receptores de Reconocimiento de Patrones , Animales , Intestino Delgado/microbiología , Ratones , Técnicas de Cultivo de Tejidos
8.
Elife ; 52016 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-27077950

RESUMEN

The roles of long non-coding RNAs (lncRNAs) in regulating cancer and stem cells are being increasingly appreciated. Its diverse mechanisms provide the regulatory network with a bigger repertoire to increase complexity. Here we report a novel LncRNA, Lnc34a, that is enriched in colon cancer stem cells (CCSCs) and initiates asymmetric division by directly targeting the microRNA miR-34a to cause its spatial imbalance. Lnc34a recruits Dnmt3a via PHB2 and HDAC1 to methylate and deacetylate the miR-34a promoter simultaneously, hence epigenetically silencing miR-34a expression independent of its upstream regulator, p53. Lnc34a levels affect CCSC self-renewal and colorectal cancer (CRC) growth in xenograft models. Lnc34a is upregulated in late-stage CRCs, contributing to epigenetic miR-34a silencing and CRC proliferation. The fact that lncRNA targets microRNA highlights the regulatory complexity of non-coding RNAs (ncRNAs), which occupy the bulk of the genome.


Asunto(s)
División Celular , Neoplasias del Colon/patología , Regulación de la Expresión Génica , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Células Madre/fisiología , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Epigénesis Genética , Silenciador del Gen , Histona Desacetilasa 1/metabolismo , Humanos , Prohibitinas , Regiones Promotoras Genéticas
9.
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
10.
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
11.
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
12.
Int J Proteomics ; 2013: 791985, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23710360

RESUMEN

The receptor tyrosine kinase ErbB2 is a breast cancer biomarker whose posttranslational modifications (PTMs) are a key indicator of its activation. Quantifying the expression and PTMs of biomarkers such as ErbB2 by selected reaction monitoring (SRM) mass spectrometry has several limitations, including minimal coverage and extensive assay development time. Therefore, we assessed the utility of two high resolution, full scan mass spectrometry approaches, MS1 Filtering and SWATH MS2, for targeted ErbB2 proteomics. Endogenous ErbB2 immunoprecipitated from SK-BR-3 cells was in-gel digested with trypsin, chymotrypsin, Asp-N, or trypsin plus Asp-N in triplicate. Data-dependent acquisition with an AB SCIEX TripleTOF 5600 and MS1 Filtering data processing was used to assess peptide and PTM coverage as well as the reproducibility of enzyme digestion. Data-independent acquisition (SWATH) was also performed for MS2 quantitation. MS1 Filtering and SWATH MS2 allow quantitation of all detected analytes after acquisition, enabling the use of multiple proteases for quantitative assessment of target proteins. Combining high resolution proteomics with multiprotease digestion enabled quantitative mapping of ErbB2 with excellent reproducibility, improved amino acid sequence and PTM coverage, and decreased assay development time compared to typical SRM assays. These results demonstrate that high resolution quantitative proteomic approaches are an effective tool for targeted biomarker quantitation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA