Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.739
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(13): 3165-3186, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38906093

RESUMEN

Patterned morphologies, such as segments, spirals, stripes, and spots, frequently emerge during embryogenesis through self-organized coordination between cells. Yet, complex patterns also emerge in adults, suggesting that the capacity for spontaneous self-organization is a ubiquitous property of biological tissues. We review current knowledge on the principles and mechanisms of self-organized patterning in embryonic tissues and explore how these principles and mechanisms apply to adult tissues that exhibit features of patterning. We discuss how and why spontaneous pattern generation is integral to homeostasis and healing of tissues, illustrating it with examples from regenerative biology. We examine how aberrant self-organization underlies diverse pathological states, including inflammatory skin disorders and tumors. Lastly, we posit that based on such blueprints, targeted engineering of pattern-driving molecular circuits can be leveraged for synthetic biology and the generation of organoids with intricate patterns.


Asunto(s)
Tipificación del Cuerpo , Humanos , Animales , Homeostasis , Desarrollo Embrionario , Organoides/metabolismo
2.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38701782

RESUMEN

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Asunto(s)
Epigénesis Genética , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Vaina de Mielina/metabolismo , Humanos , Ratones , Remielinización/efectos de los fármacos , Oligodendroglía/metabolismo , Sistema Nervioso Central/metabolismo , Ratones Endogámicos C57BL , Rejuvenecimiento , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Organoides/metabolismo , Organoides/efectos de los fármacos , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/genética , Diferenciación Celular/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Masculino , Regeneración/efectos de los fármacos , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología
3.
Cell ; 186(3): 513-527.e19, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36657441

RESUMEN

Axial development of mammals involves coordinated morphogenetic events, including axial elongation, somitogenesis, and neural tube formation. To gain insight into the signals controlling the dynamics of human axial morphogenesis, we generated axially elongating organoids by inducing anteroposterior symmetry breaking of spatially coupled epithelial cysts derived from human pluripotent stem cells. Each organoid was composed of a neural tube flanked by presomitic mesoderm sequentially segmented into somites. Periodic activation of the somite differentiation gene MESP2 coincided in space and time with anteriorly traveling segmentation clock waves in the presomitic mesoderm of the organoids, recapitulating critical aspects of somitogenesis. Timed perturbations demonstrated that FGF and WNT signaling play distinct roles in axial elongation and somitogenesis, and that FGF signaling gradients drive segmentation clock waves. By generating and perturbing organoids that robustly recapitulate the architecture of multiple axial tissues in human embryos, this work offers a means to dissect mechanisms underlying human embryogenesis.


Asunto(s)
Desarrollo Embrionario , Mesodermo , Somitos , Animales , Humanos , Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Mamíferos/genética , Mesodermo/fisiología , Morfogénesis , Vía de Señalización Wnt , Organoides/metabolismo
4.
Cell ; 185(20): 3770-3788.e27, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36179669

RESUMEN

Realizing the full utility of brain organoids to study human development requires understanding whether organoids precisely replicate endogenous cellular and molecular events, particularly since acquisition of cell identity in organoids can be impaired by abnormal metabolic states. We present a comprehensive single-cell transcriptomic, epigenetic, and spatial atlas of human cortical organoid development, comprising over 610,000 cells, from generation of neural progenitors through production of differentiated neuronal and glial subtypes. We show that processes of cellular diversification correlate closely to endogenous ones, irrespective of metabolic state, empowering the use of this atlas to study human fate specification. We define longitudinal molecular trajectories of cortical cell types during organoid development, identify genes with predicted human-specific roles in lineage establishment, and uncover early transcriptional diversity of human callosal neurons. The findings validate this comprehensive atlas of human corticogenesis in vitro as a resource to prime investigation into the mechanisms of human cortical development.


Asunto(s)
Corteza Cerebral , Organoides , Diferenciación Celular , Corteza Cerebral/metabolismo , Humanos , Neurogénesis , Neuronas , Organoides/metabolismo
5.
Cell ; 184(8): 2084-2102.e19, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33765444

RESUMEN

The human brain has undergone rapid expansion since humans diverged from other great apes, but the mechanism of this human-specific enlargement is still unknown. Here, we use cerebral organoids derived from human, gorilla, and chimpanzee cells to study developmental mechanisms driving evolutionary brain expansion. We find that neuroepithelial differentiation is a protracted process in apes, involving a previously unrecognized transition state characterized by a change in cell shape. Furthermore, we show that human organoids are larger due to a delay in this transition, associated with differences in interkinetic nuclear migration and cell cycle length. Comparative RNA sequencing (RNA-seq) reveals differences in expression dynamics of cell morphogenesis factors, including ZEB2, a known epithelial-mesenchymal transition regulator. We show that ZEB2 promotes neuroepithelial transition, and its manipulation and downstream signaling leads to acquisition of nonhuman ape architecture in the human context and vice versa, establishing an important role for neuroepithelial cell shape in human brain expansion.


Asunto(s)
Evolución Biológica , Encéfalo/citología , Forma de la Célula/fisiología , Animales , Encéfalo/metabolismo , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Transición Epitelial-Mesenquimal/genética , Expresión Génica , Gorilla gorilla , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Organoides/citología , Organoides/metabolismo , Pan troglodytes , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo
6.
Cell ; 184(17): 4547-4563.e17, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34314701

RESUMEN

Frontotemporal dementia (FTD) because of MAPT mutation causes pathological accumulation of tau and glutamatergic cortical neuronal death by unknown mechanisms. We used human induced pluripotent stem cell (iPSC)-derived cerebral organoids expressing tau-V337M and isogenic corrected controls to discover early alterations because of the mutation that precede neurodegeneration. At 2 months, mutant organoids show upregulated expression of MAPT, glutamatergic signaling pathways, and regulators, including the RNA-binding protein ELAVL4, and increased stress granules. Over the following 4 months, mutant organoids accumulate splicing changes, disruption of autophagy function, and build-up of tau and P-tau-S396. By 6 months, tau-V337M organoids show specific loss of glutamatergic neurons as seen in individuals with FTD. Mutant neurons are susceptible to glutamate toxicity, which can be rescued pharmacologically by the PIKFYVE kinase inhibitor apilimod. Our results demonstrate a sequence of events that precede neurodegeneration, revealing molecular pathways associated with glutamate signaling as potential targets for therapeutic intervention in FTD.


Asunto(s)
Cerebro/patología , Proteína 4 Similar a ELAV/genética , Ácido Glutámico/metabolismo , Mutación/genética , Neuronas/patología , Organoides/metabolismo , Empalme del ARN/genética , Proteínas tau/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Biomarcadores/metabolismo , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Muerte Celular/efectos de los fármacos , Línea Celular , Humanos , Hidrazonas/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Morfolinas/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Organoides/efectos de los fármacos , Organoides/ultraestructura , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología , Empalme del ARN/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Gránulos de Estrés/efectos de los fármacos , Gránulos de Estrés/metabolismo , Sinapsis/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
7.
Cell ; 181(6): 1291-1306.e19, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32407674

RESUMEN

Enteroendocrine cells (EECs) sense intestinal content and release hormones to regulate gastrointestinal activity, systemic metabolism, and food intake. Little is known about the molecular make-up of human EEC subtypes and the regulated secretion of individual hormones. Here, we describe an organoid-based platform for functional studies of human EECs. EEC formation is induced in vitro by transient expression of NEUROG3. A set of gut organoids was engineered in which the major hormones are fluorescently tagged. A single-cell mRNA atlas was generated for the different EEC subtypes, and their secreted products were recorded by mass-spectrometry. We note key differences to murine EECs, including hormones, sensory receptors, and transcription factors. Notably, several hormone-like molecules were identified. Inter-EEC communication is exemplified by secretin-induced GLP-1 secretion. Indeed, individual EEC subtypes carry receptors for various EEC hormones. This study provides a rich resource to study human EEC development and function.


Asunto(s)
Células Enteroendocrinas/metabolismo , ARN Mensajero/genética , Células Cultivadas , Hormonas Gastrointestinales/genética , Tracto Gastrointestinal/metabolismo , Péptido 1 Similar al Glucagón/genética , Humanos , Organoides/metabolismo , Factores de Transcripción/genética , Transcriptoma/genética
8.
Cell ; 180(2): 233-247.e21, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31978343

RESUMEN

Wnt dependency and Lgr5 expression define multiple mammalian epithelial stem cell types. Under defined growth factor conditions, such adult stem cells (ASCs) grow as 3D organoids that recapitulate essential features of the pertinent epithelium. Here, we establish long-term expanding venom gland organoids from several snake species. The newly assembled transcriptome of the Cape coral snake reveals that organoids express high levels of toxin transcripts. Single-cell RNA sequencing of both organoids and primary tissue identifies distinct venom-expressing cell types as well as proliferative cells expressing homologs of known mammalian stem cell markers. A hard-wired regional heterogeneity in the expression of individual venom components is maintained in organoid cultures. Harvested venom peptides reflect crude venom composition and display biological activity. This study extends organoid technology to reptilian tissues and describes an experimentally tractable model system representing the snake venom gland.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Organoides/crecimiento & desarrollo , Venenos de Serpiente/metabolismo , Células Madre Adultas/metabolismo , Animales , Serpientes de Coral/metabolismo , Perfilación de la Expresión Génica/métodos , Organoides/metabolismo , Glándulas Salivales/metabolismo , Venenos de Serpiente/genética , Serpientes/genética , Serpientes/crecimiento & desarrollo , Células Madre/metabolismo , Toxinas Biológicas/genética , Transcriptoma/genética
9.
Cell ; 180(1): 188-204.e22, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31883794

RESUMEN

Glioblastomas exhibit vast inter- and intra-tumoral heterogeneity, complicating the development of effective therapeutic strategies. Current in vitro models are limited in preserving the cellular and mutational diversity of parental tumors and require a prolonged generation time. Here, we report methods for generating and biobanking patient-derived glioblastoma organoids (GBOs) that recapitulate the histological features, cellular diversity, gene expression, and mutational profiles of their corresponding parental tumors. GBOs can be generated quickly with high reliability and exhibit rapid, aggressive infiltration when transplanted into adult rodent brains. We further demonstrate the utility of GBOs to test personalized therapies by correlating GBO mutational profiles with responses to specific drugs and by modeling chimeric antigen receptor T cell immunotherapy. Our studies show that GBOs maintain many key features of glioblastomas and can be rapidly deployed to investigate patient-specific treatment strategies. Additionally, our live biobank establishes a rich resource for basic and translational glioblastoma research.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Glioblastoma/metabolismo , Organoides/crecimiento & desarrollo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Bancos de Muestras Biológicas , Femenino , Glioblastoma/genética , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Modelos Biológicos , Organoides/metabolismo , Reproducibilidad de los Resultados , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
10.
Cell ; 182(6): 1623-1640.e34, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32946783

RESUMEN

Human organoids recapitulating the cell-type diversity and function of their target organ are valuable for basic and translational research. We developed light-sensitive human retinal organoids with multiple nuclear and synaptic layers and functional synapses. We sequenced the RNA of 285,441 single cells from these organoids at seven developmental time points and from the periphery, fovea, pigment epithelium and choroid of light-responsive adult human retinas, and performed histochemistry. Cell types in organoids matured in vitro to a stable "developed" state at a rate similar to human retina development in vivo. Transcriptomes of organoid cell types converged toward the transcriptomes of adult peripheral retinal cell types. Expression of disease-associated genes was cell-type-specific in adult retina, and cell-type specificity was retained in organoids. We implicate unexpected cell types in diseases such as macular degeneration. This resource identifies cellular targets for studying disease mechanisms in organoids and for targeted repair in human retinas.


Asunto(s)
Diferenciación Celular/genética , Organoides/citología , Organoides/metabolismo , Retina/citología , Retina/metabolismo , Análisis de la Célula Individual/métodos , Sinapsis/fisiología , Transcriptoma/genética , Técnicas de Cultivo de Célula/métodos , Línea Celular , Electrofisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Hibridación in Situ , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Microscopía Electrónica , Familia de Multigenes , Naftoquinonas , Organoides/efectos de la radiación , Organoides/ultraestructura , Retina/patología , Retina/efectos de la radiación
11.
Cell ; 180(6): 1198-1211.e19, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32200801

RESUMEN

It has generally proven challenging to produce functional ß cells in vitro. Here, we describe a previously unidentified protein C receptor positive (Procr+) cell population in adult mouse pancreas through single-cell RNA sequencing (scRNA-seq). The cells reside in islets, do not express differentiation markers, and feature epithelial-to-mesenchymal transition characteristics. By genetic lineage tracing, Procr+ islet cells undergo clonal expansion and generate all four endocrine cell types during adult homeostasis. Sorted Procr+ cells, representing ∼1% of islet cells, can robustly form islet-like organoids when cultured at clonal density. Exponential expansion can be maintained over long periods by serial passaging, while differentiation can be induced at any time point in culture. ß cells dominate in differentiated islet organoids, while α, δ, and PP cells occur at lower frequencies. The organoids are glucose-responsive and insulin-secreting. Upon transplantation in diabetic mice, these organoids reverse disease. These findings demonstrate that the adult mouse pancreatic islet contains a population of Procr+ endocrine progenitors.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Receptor de Proteína C Endotelial/metabolismo , Islotes Pancreáticos/citología , Animales , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Islotes Pancreáticos/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Desnudos , Organoides/crecimiento & desarrollo , Organoides/metabolismo , Páncreas/citología , Páncreas/metabolismo , Proteína C/metabolismo , Células Madre/citología
12.
Cell ; 176(4): 743-756.e17, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30735633

RESUMEN

Direct comparisons of human and non-human primate brains can reveal molecular pathways underlying remarkable specializations of the human brain. However, chimpanzee tissue is inaccessible during neocortical neurogenesis when differences in brain size first appear. To identify human-specific features of cortical development, we leveraged recent innovations that permit generating pluripotent stem cell-derived cerebral organoids from chimpanzee. Despite metabolic differences, organoid models preserve gene regulatory networks related to primary cell types and developmental processes. We further identified 261 differentially expressed genes in human compared to both chimpanzee organoids and macaque cortex, enriched for recent gene duplications, and including multiple regulators of PI3K-AKT-mTOR signaling. We observed increased activation of this pathway in human radial glia, dependent on two receptors upregulated specifically in human: INSR and ITGB8. Our findings establish a platform for systematic analysis of molecular changes contributing to human brain development and evolution.


Asunto(s)
Corteza Cerebral/citología , Organoides/metabolismo , Animales , Evolución Biológica , Encéfalo/citología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Corteza Cerebral/metabolismo , Redes Reguladoras de Genes/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Macaca , Neurogénesis/genética , Organoides/crecimiento & desarrollo , Pan troglodytes , Células Madre Pluripotentes/citología , Análisis de la Célula Individual , Especificidad de la Especie , Transcriptoma/genética
13.
Cell ; 176(5): 1098-1112.e18, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30794774

RESUMEN

Increased levels of intestinal bile acids (BAs) are a risk factor for colorectal cancer (CRC). Here, we show that the convergence of dietary factors (high-fat diet) and dysregulated WNT signaling (APC mutation) alters BA profiles to drive malignant transformations in Lgr5-expressing (Lgr5+) cancer stem cells and promote an adenoma-to-adenocarcinoma progression. Mechanistically, we show that BAs that antagonize intestinal farnesoid X receptor (FXR) function, including tauro-ß-muricholic acid (T-ßMCA) and deoxycholic acid (DCA), induce proliferation and DNA damage in Lgr5+ cells. Conversely, selective activation of intestinal FXR can restrict abnormal Lgr5+ cell growth and curtail CRC progression. This unexpected role for FXR in coordinating intestinal self-renewal with BA levels implicates FXR as a potential therapeutic target for CRC.


Asunto(s)
Neoplasias Intestinales/metabolismo , Células Madre Neoplásicas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Línea Celular , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Ácido Desoxicólico/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Intestinales/genética , Intestinos , Hígado , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/fisiología , Organoides/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Factores de Riesgo , Transducción de Señal , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/metabolismo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología
14.
Cell ; 175(6): 1591-1606.e19, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30500538

RESUMEN

The mammalian liver possesses a remarkable regenerative ability. Two modes of damage response have been described: (1) The "oval cell" response emanates from the biliary tree when all hepatocytes are affected by chronic liver disease. (2) A massive, proliferative response of mature hepatocytes occurs upon acute liver damage such as partial hepatectomy (PHx). While the oval cell response has been captured in vitro by growing organoids from cholangiocytes, the hepatocyte proliferative response has not been recapitulated in culture. Here, we describe the establishment of a long-term 3D organoid culture system for mouse and human primary hepatocytes. Organoids can be established from single hepatocytes and grown for multiple months, while retaining key morphological, functional and gene expression features. Transcriptional profiles of the organoids resemble those of proliferating hepatocytes after PHx. Human hepatocyte organoids proliferate extensively after engraftment into mice and thus recapitulate the proliferative damage-response of hepatocytes.


Asunto(s)
Proliferación Celular , Hepatocitos/metabolismo , Organoides/metabolismo , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Hepatocitos/citología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Organoides/citología , Células Madre/citología , Células Madre/metabolismo , Factores de Tiempo
15.
Cell ; 173(2): 515-528.e17, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29625057

RESUMEN

Bladder cancer is the fifth most prevalent cancer in the U.S., yet is understudied, and few laboratory models exist that reflect the biology of the human disease. Here, we describe a biobank of patient-derived organoid lines that recapitulates the histopathological and molecular diversity of human bladder cancer. Organoid lines can be established efficiently from patient biopsies acquired before and after disease recurrence and are interconvertible with orthotopic xenografts. Notably, organoid lines often retain parental tumor heterogeneity and exhibit a spectrum of genomic changes that are consistent with tumor evolution in culture. Analyses of drug response using bladder tumor organoids show partial correlations with mutational profiles, as well as changes associated with treatment resistance, and specific responses can be validated using xenografts in vivo. Our studies indicate that patient-derived bladder tumor organoids represent a faithful model system for studying tumor evolution and treatment response in the context of precision cancer medicine.


Asunto(s)
Neoplasias de la Vejiga Urinaria/patología , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Variaciones en el Número de Copia de ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Mutación , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Medicina de Precisión , Trasplante Heterólogo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo
16.
Cell ; 174(4): 856-869.e17, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30096312

RESUMEN

Recent sequencing analyses have shed light on heterogeneous patterns of genomic aberrations in human gastric cancers (GCs). To explore how individual genetic events translate into cancer phenotypes, we established a biological library consisting of genetically engineered gastric organoids carrying various GC mutations and 37 patient-derived organoid lines, including rare genomically stable GCs. Phenotype analyses of GC organoids revealed divergent genetic and epigenetic routes to gain Wnt and R-spondin niche independency. An unbiased phenotype-based genetic screening identified a significant association between CDH1/TP53 compound mutations and the R-spondin independency that was functionally validated by CRISPR-based knockout. Xenografting of GC organoids further established the feasibility of Wnt-targeting therapy for Wnt-dependent GCs. Our results collectively demonstrate that multifaceted genetic abnormalities render human GCs independent of the stem cell niche and highlight the validity of the genotype-phenotype screening strategy in gaining deeper understanding of human cancers.


Asunto(s)
Adenocarcinoma/patología , Organoides/patología , Neoplasias Gástricas/patología , Estómago/patología , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Antígenos CD/genética , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Cadherinas/genética , Carcinogénesis , Proliferación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Organoides/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Trombospondinas/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteínas Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cell ; 175(5): 1307-1320.e22, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30392957

RESUMEN

In the small intestine, a niche of accessory cell types supports the generation of mature epithelial cell types from intestinal stem cells (ISCs). It is unclear, however, if and how immune cells in the niche affect ISC fate or the balance between self-renewal and differentiation. Here, we use single-cell RNA sequencing (scRNA-seq) to identify MHC class II (MHCII) machinery enrichment in two subsets of Lgr5+ ISCs. We show that MHCII+ Lgr5+ ISCs are non-conventional antigen-presenting cells in co-cultures with CD4+ T helper (Th) cells. Stimulation of intestinal organoids with key Th cytokines affects Lgr5+ ISC renewal and differentiation in opposing ways: pro-inflammatory signals promote differentiation, while regulatory cells and cytokines reduce it. In vivo genetic perturbation of Th cells or MHCII expression on Lgr5+ ISCs impacts epithelial cell differentiation and IEC fate during infection. These interactions between Th cells and Lgr5+ ISCs, thus, orchestrate tissue-wide responses to external signals.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Interleucina-10/metabolismo , Células Madre/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Citocinas/farmacología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Sistema Inmunológico/metabolismo , Intestinos/citología , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Salmonella enterica/patogenicidad , Células Madre/metabolismo , Linfocitos T Colaboradores-Inductores/citología
18.
Cell ; 171(4): 877-889.e17, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28965759

RESUMEN

N6-methyladenosine (m6A), installed by the Mettl3/Mettl14 methyltransferase complex, is the most prevalent internal mRNA modification. Whether m6A regulates mammalian brain development is unknown. Here, we show that m6A depletion by Mettl14 knockout in embryonic mouse brains prolongs the cell cycle of radial glia cells and extends cortical neurogenesis into postnatal stages. m6A depletion by Mettl3 knockdown also leads to a prolonged cell cycle and maintenance of radial glia cells. m6A sequencing of embryonic mouse cortex reveals enrichment of mRNAs related to transcription factors, neurogenesis, the cell cycle, and neuronal differentiation, and m6A tagging promotes their decay. Further analysis uncovers previously unappreciated transcriptional prepatterning in cortical neural stem cells. m6A signaling also regulates human cortical neurogenesis in forebrain organoids. Comparison of m6A-mRNA landscapes between mouse and human cortical neurogenesis reveals enrichment of human-specific m6A tagging of transcripts related to brain-disorder risk genes. Our study identifies an epitranscriptomic mechanism in heightened transcriptional coordination during mammalian cortical neurogenesis.


Asunto(s)
Neurogénesis , Prosencéfalo/embriología , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Animales , Ciclo Celular , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Organoides/metabolismo , Prosencéfalo/citología , Prosencéfalo/metabolismo , Estabilidad del ARN
19.
Cell ; 170(5): 875-888.e20, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28757253

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal human malignancies, owing in part to its propensity for metastasis. Here, we used an organoid culture system to investigate how transcription and the enhancer landscape become altered during discrete stages of disease progression in a PDA mouse model. This approach revealed that the metastatic transition is accompanied by massive and recurrent alterations in enhancer activity. We implicate the pioneer factor FOXA1 as a driver of enhancer activation in this system, a mechanism that renders PDA cells more invasive and less anchorage-dependent for growth in vitro, as well as more metastatic in vivo. In this context, FOXA1-dependent enhancer reprogramming activates a transcriptional program of embryonic foregut endoderm. Collectively, our study implicates enhancer reprogramming, FOXA1 upregulation, and a retrograde developmental transition in PDA metastasis.


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epigenómica , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Organoides/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología
20.
Cell ; 169(6): 985-999, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575679

RESUMEN

The WNT signal transduction cascade is a main regulator of development throughout the animal kingdom. Wnts are also key drivers of most types of tissue stem cells in adult mammals. Unsurprisingly, mutated Wnt pathway components are causative to multiple growth-related pathologies and to cancer. Here, we describe the core Wnt/ß-catenin signaling pathway, how it controls stem cells, and contributes to disease. Finally, we discuss strategies for Wnt-based therapies.


Asunto(s)
Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Anomalías Congénitas/metabolismo , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Organoides/metabolismo , Células Madre/metabolismo , Proteínas Wnt/química , Proteínas Wnt/genética , Vía de Señalización Wnt/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA