Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Cell ; 176(5): 1158-1173.e16, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30712869

RESUMEN

Homeostatic regulation of the intestinal enteroendocrine lineage hierarchy is a poorly understood process. We resolved transcriptional changes during enteroendocrine differentiation in real time at single-cell level using a novel knockin allele of Neurog3, the master regulator gene briefly expressed at the onset of enteroendocrine specification. A bi-fluorescent reporter, Neurog3Chrono, measures time from the onset of enteroendocrine differentiation and enables precise positioning of single-cell transcriptomes along an absolute time axis. This approach yielded a definitive description of the enteroendocrine hierarchy and its sub-lineages, uncovered differential kinetics between sub-lineages, and revealed time-dependent hormonal plasticity in enterochromaffin and L cells. The time-resolved map of transcriptional changes predicted multiple novel molecular regulators. Nine of these were validated by conditional knockout in mice or CRISPR modification in intestinal organoids. Six novel candidate regulators (Sox4, Rfx6, Tox3, Myt1, Runx1t1, and Zcchc12) yielded specific enteroendocrine phenotypes. Our time-resolved single-cell transcriptional map presents a rich resource to unravel enteroendocrine differentiation.


Asunto(s)
Linaje de la Célula/genética , Células Enteroendocrinas/metabolismo , Perfilación de la Expresión Génica/métodos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Linaje de la Célula/fisiología , Células Enteroendocrinas/fisiología , Colorantes Fluorescentes , Proteínas de Homeodominio/genética , Mucosa Intestinal/citología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Imagen Óptica/métodos , Organoides , Fenotipo , Análisis de la Célula Individual/métodos , Células Madre , Factores de Transcripción/genética , Transcriptoma/genética
4.
Cell ; 148(1-2): 33-45, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22265400

RESUMEN

Lineage tracing is the identification of all progeny of a single cell. Although its origins date back to developmental biology of invertebrates in the 19(th) century, lineage tracing is now an essential tool for studying stem cell properties in adult mammalian tissues. Lineage tracing provides a powerful means of understanding tissue development, homeostasis, and disease, especially when it is combined with experimental manipulation of signals regulating cell-fate decisions. Recently, the combination of inducible recombinases, multicolor reporter constructs, and live-cell imaging has provided unprecedented insights into stem cell biology. Here we discuss the different experimental strategies currently available for lineage tracing, their associated caveats, and new opportunities to integrate lineage tracing with the monitoring of intracellular signaling pathways.


Asunto(s)
Linaje de la Célula , Biología Evolutiva/métodos , Desarrollo Embrionario , Animales , Biología Evolutiva/historia , Genes Reporteros , Marcadores Genéticos , Historia del Siglo XIX , Humanos , Invertebrados/embriología , Recombinación Genética , Coloración y Etiquetado , Vertebrados/embriología
5.
Blood ; 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39172759

RESUMEN

Extramedullary disease (EMD) is a high-risk feature of multiple myeloma (MM) and remains a poor prognostic factor even in the era of novel immunotherapies. Here we applied spatial transcriptomics (tomo-seq [n=2] and 10X Visium [n=12]), and single-cell RNA sequencing (scRNAseq [n=3]) to a set of 14 EMD biopsies to dissect the three-dimensional architecture of tumor cells and their microenvironment. Overall, the infiltrating immune and stromal cells showed both intra- and inter-patient variation with no uniform distribution over the lesion. We observed substantial heterogeneity at the copy number level within plasma cells, including the emergence of new subclones in circumscribed areas of the tumor, consistent with genomic instability. We further identified spatial expression differences of GPRC5D and TNFRSF17, two important antigens for bispecific antibody therapy. EMD masses were infiltrated by various immune cells, including T-cells. Notably, exhausted TIM3+/PD-1+ T-cells diffusely co-localized with MM cells, whereas functional and activated CD8+ T-cells showed a focal infiltration pattern along with M1 macrophages in otherwise tumor-free regions. This segregation of fit and exhausted T-cells was resolved in the case of response to T-cell engaging bispecific antibodies. MM cells and microenvironment cells were embedded in a complex network that influenced immune activation and angiogenesis, and oxidative phosphorylation represented the major metabolic program within EMD lesions. In summary, spatial transcriptomics has revealed a multicellular ecosystem in EMD with checkpoint inhibition and dual targeting as potential new therapeutic avenues.

6.
Semin Cell Dev Biol ; 144: 20-30, 2023 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-36127261

RESUMEN

Adult stem cells are responsible for homoeostasis and regeneration of epithelial tissues. Stem cell function is regulated by both cell autonomous mechanisms as well as the niche. Deregulated stem cell function contributes to diseases such as cancer. Epithelial organoid cultures generated from tissue-resident adult stem cells have allowed unprecedented insights into the biology of epithelial tissues. The subsequent adaptation of organoid technology enabled the modelling of the communication of stem cells with their cellular and non-cellular niche as well as diseases. Starting from its first model described in 2009, the murine small intestinal organoid, we discuss here how epithelial organoid cultures have been become a prime in vitro research tool for cell and developmental biology, bioengineering, and biomedicine in the last decade.


Asunto(s)
Células Madre Adultas , Organoides , Animales , Ratones , Células Madre , Epitelio , Células de Paneth , Células Epiteliales
7.
Proc Natl Acad Sci U S A ; 116(29): 14630-14638, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31253707

RESUMEN

Mammalian epidermal stem cells maintain homeostasis of the skin epidermis and contribute to its regeneration throughout adult life. While 2D mouse epidermal stem cell cultures have been established decades ago, a long-term, feeder cell- and serum-free culture system recapitulating murine epidermal architecture has not been available. Here we describe an epidermal organoid culture system that allows long-term, genetically stable expansion of adult epidermal stem cells. Our epidermal expansion media combines atypically high calcium concentrations, activation of cAMP, FGF, and R-spondin signaling with inhibition of bone morphogenetic protein (BMP) signaling. Organoids are established robustly from adult mouse skin and expand over at least 6 mo, while maintaining the basal-apical organization of the mouse interfollicular epidermis. The system represents a powerful tool to study epidermal homeostasis and disease in vitro.


Asunto(s)
Células Madre Adultas/fisiología , Técnicas de Cultivo de Célula/métodos , Epidermis/fisiología , Queratinocitos/fisiología , Organoides/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Acoplados a Proteínas G/genética , Factores de Tiempo
8.
Proc Natl Acad Sci U S A ; 115(52): E12245-E12254, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30530645

RESUMEN

The significance of cardiac stem cell (CSC) populations for cardiac regeneration remains disputed. Here, we apply the most direct definition of stem cell function (the ability to replace lost tissue through cell division) to interrogate the existence of CSCs. By single-cell mRNA sequencing and genetic lineage tracing using two Ki67 knockin mouse models, we map all proliferating cells and their progeny in homoeostatic and regenerating murine hearts. Cycling cardiomyocytes were only robustly observed in the early postnatal growth phase, while cycling cells in homoeostatic and damaged adult myocardium represented various noncardiomyocyte cell types. Proliferative postdamage fibroblasts expressing follistatin-like protein 1 (FSTL1) closely resemble neonatal cardiac fibroblasts and form the fibrotic scar. Genetic deletion of Fstl1 in cardiac fibroblasts results in postdamage cardiac rupture. We find no evidence for the existence of a quiescent CSC population, for transdifferentiation of other cell types toward cardiomyocytes, or for proliferation of significant numbers of cardiomyocytes in response to cardiac injury.


Asunto(s)
Proliferación Celular , Lesiones Cardíacas/fisiopatología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas Relacionadas con la Folistatina/genética , Proteínas Relacionadas con la Folistatina/metabolismo , Lesiones Cardíacas/genética , Lesiones Cardíacas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Embarazo , Células Madre/citología , Células Madre/metabolismo
9.
Circ Res ; 121(12): 1346-1359, 2017 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-29018034

RESUMEN

RATIONALE: Arrhythmogenic cardiomyopathy is caused primarily by mutations in genes encoding desmosome proteins. Ventricular arrhythmias are the cardinal and typically early manifestations, whereas myocardial fibroadiposis is the pathological hallmark. Homozygous DSP (desmoplakin) and JUP (junction protein plakoglobin) mutations are responsible for a subset of patients with arrhythmogenic cardiomyopathy who exhibit cardiac arrhythmias and dysfunction, palmoplanter keratosis, and hair abnormalities (cardiocutaneous syndromes). OBJECTIVE: To determine phenotypic consequences of deletion of Dsp in a subset of cells common to the heart and skin. METHODS AND RESULTS: Expression of CSPG4 (chondroitin sulfate proteoglycan 4) was detected in epidermal keratinocytes and the cardiac conduction system. CSPG4pos cells constituted ≈5.6±3.3% of the nonmyocyte cells in the mouse heart. Inducible postnatal deletion of Dsp under the transcriptional control of the Cspg4 locus led to ventricular arrhythmias, atrial fibrillation, atrioventricular conduction defects, and death by 4 months of age. Cardiac arrhythmias occurred early and in the absence of cardiac dysfunction and excess cardiac fibroadipocytes, as in human arrhythmogenic cardiomyopathy. The mice exhibited palmoplantar keratosis and progressive alopecia, leading to alopecia totalis, associated with accelerated proliferation and impaired terminal differentiation of keratinocytes. The phenotype is similar to human cardiocutaneous syndromes caused by homozygous mutations in DSP. CONCLUSIONS: Deletion of Dsp under the transcriptional regulation of the CSPG4 locus led to lethal cardiac arrhythmias in the absence of cardiac dysfunction or fibroadiposis, palmoplantar keratosis, and alopecia, resembling the human cardiocutaneous syndromes. The findings offer a cellular basis for early cardiac arrhythmias in patients with arrhythmogenic cardiomyopathy and cardiocutaneous syndromes.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/metabolismo , Desmoplaquinas/metabolismo , Queratosis/metabolismo , Fenotipo , Animales , Antígenos/genética , Displasia Ventricular Derecha Arritmogénica/genética , Células Cultivadas , Desmoplaquinas/genética , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Queratosis/genética , Ratones , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteoglicanos/genética , Síndrome
10.
Nature ; 504(7479): 277-281, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24336287

RESUMEN

Fibroblasts are the major mesenchymal cell type in connective tissue and deposit the collagen and elastic fibres of the extracellular matrix (ECM). Even within a single tissue, fibroblasts exhibit considerable functional diversity, but it is not known whether this reflects the existence of a differentiation hierarchy or is a response to different environmental factors. Here we show, using transplantation assays and lineage tracing in mice, that the fibroblasts of skin connective tissue arise from two distinct lineages. One forms the upper dermis, including the dermal papilla that regulates hair growth and the arrector pili muscle, which controls piloerection. The other forms the lower dermis, including the reticular fibroblasts that synthesize the bulk of the fibrillar ECM, and the preadipocytes and adipocytes of the hypodermis. The upper lineage is required for hair follicle formation. In wounded adult skin, the initial wave of dermal repair is mediated by the lower lineage and upper dermal fibroblasts are recruited only during re-epithelialization. Epidermal ß-catenin activation stimulates the expansion of the upper dermal lineage, rendering wounds permissive for hair follicle formation. Our findings explain why wounding is linked to formation of ECM-rich scar tissue that lacks hair follicles. They also form a platform for discovering fibroblast lineages in other tissues and for examining fibroblast changes in ageing and disease.


Asunto(s)
Linaje de la Célula , Fibroblastos/citología , Piel/citología , Piel/crecimiento & desarrollo , Cicatrización de Heridas/fisiología , Adipocitos/citología , Adipocitos/metabolismo , Animales , Dermis/anatomía & histología , Dermis/citología , Dermis/embriología , Dermis/crecimiento & desarrollo , Femenino , Fibroblastos/trasplante , Folículo Piloso/citología , Folículo Piloso/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Músculo Liso/citología , Músculo Liso/metabolismo , Piel/anatomía & histología , Piel/embriología , beta Catenina/metabolismo
11.
Proc Natl Acad Sci U S A ; 113(42): 11859-11864, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27708166

RESUMEN

Current mouse models for colorectal cancer often differ significantly from human colon cancer, being largely restricted to the small intestine. Here, we aim to develop a colon-specific inducible mouse model that can faithfully recapitulate human colon cancer initiation and progression. Carbonic anhydrase I (Car1) is a gene expressed uniquely in colonic epithelial cells. We generated a colon-specific inducible Car1CreER knock-in (KI) mouse with broad Cre activity in epithelial cells of the proximal colon and cecum. Deletion of the tumor suppressor gene Apc using the Car1CreER KI caused tumor formation in the cecum but did not yield adenomas in the proximal colon. Mutation of both Apc and Kras yielded microadenomas in both the cecum and the proximal colon, which progressed to macroadenomas with significant morbidity. Aggressive carcinomas with some invasion into lymph nodes developed upon combined induction of oncogenic mutations of Apc, Kras, p53, and Smad4 Importantly, no adenomas were observed in the small intestine. Additionally, we observed tumors from differentiated Car1-expressing cells with Apc/Kras mutations, suggesting that a top-down model of intestinal tumorigenesis can occur with multiple mutations. Our results establish the Car1CreER KI as a valuable mouse model to study colon-specific tumorigenesis and metastasis as well as cancer-cell-of-origin questions.


Asunto(s)
Neoplasias del Colon/etiología , Regulación de la Expresión Génica , Integrasas/genética , Ratones Transgénicos , Adenoma/etiología , Adenoma/metabolismo , Adenoma/patología , Animales , Biomarcadores de Tumor , Anhidrasa Carbónica I/genética , Anhidrasa Carbónica I/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Activación Enzimática , Técnicas de Sustitución del Gen , Marcación de Gen , Genes APC , Genes ras , Sitios Genéticos , Humanos , Inmunohistoquímica , Integrasas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Mutación , Especificidad de Órganos/genética , Investigación
12.
Dev Biol ; 428(2): 273-282, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28526587

RESUMEN

Adult stem cells self-renew and replenish differentiated cells in various organs and tissues throughout a mammal's life. Over the last 25 years an ever-growing body of knowledge has unraveled the essential regulation of adult mammalian epithelia by the canonical Wnt signaling with its key intracellular effector ß-catenin. In this review, we discuss the principles of the signaling pathway and its role in adult epithelial stem cells of the intestine and skin during homeostasis and tumorigenesis. We further highlight the research that led to the identification of new stem cell markers and methods to study adult stem cells ex vivo.


Asunto(s)
Células Madre Adultas/metabolismo , Células Epiteliales/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Animales , Autorrenovación de las Células/genética , Autorrenovación de las Células/fisiología , Células Epidérmicas , Epidermis/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Modelos Biológicos , Vía de Señalización Wnt/genética
13.
Vet Dermatol ; 29(5): 375-e126, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29963730

RESUMEN

BACKGROUND: Perturbations of epidermal and follicular homeostasis have been attributed to a variety of skin diseases affecting dogs. The availability of an in vitro system to investigate these diseases is important to understand underlying pathomechanisms. OBJECTIVES: To establish an accurate and reliable in vitro 3D system of canine keratinocyte organoids to lay the basis for studying functional defects in interfollicular epidermis (IFE) and hair follicle (HF) morphogenesis, reconstitution and differentiation that lead to alopecic and epidermal diseases. ANIMALS: Skin biopsies were obtained from freshly euthanized dogs of different breeds with no skin abnormalities. METHODS: Cells derived from microdissected IFE and HFs were seeded in Matrigel and keratinocyte organoids were grown and characterized using immunohistochemistry, RT-qPCR and RNA sequencing. RESULTS: Both organoid lines develop into a basal IFE-like cell type. Gene and protein expression analysis revealed high mRNA and protein levels of keratins 5 and 14, IFE differentiation markers and intercellular molecules. Key markers of HF stem cells were lacking. Withdrawal of growth factors resulted in upregulation of markers such as KRT16, Involucrin, KRT17 and SOX9, showing the potential of the organoids to develop towards more differentiated tissue. CONCLUSION AND CLINICAL IMPORTANCE: Our 3D in vitro culture system provides the basis to explore epidermal function, to investigate the culture conditions necessary for the development of organoids with a HF signature and to address cutaneous disorders in dogs. However, for induction of HF signatures or hair growth, addition of different growth factors or co-culture with dermal papilla will be required.


Asunto(s)
Perros/anatomía & histología , Queratinocitos/citología , Técnicas de Cultivo de Órganos/veterinaria , Animales , Biopsia/veterinaria , Células Cultivadas/citología , Células Epidérmicas , Queratinocitos/patología , Técnicas de Cultivo de Órganos/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Análisis de Secuencia de ARN/veterinaria , Piel/citología , Piel/patología , Enfermedades de la Piel/patología , Enfermedades de la Piel/veterinaria
15.
Dev Biol ; 382(1): 15-26, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23948231

RESUMEN

In postnatal skin the transcription factor Sox2 is expressed in the dermal papilla (DP) of guard/awl/auchene hair follicles and by mechanosensory Merkel cells in the touch domes of guard hairs. To investigate the consequences of Sox2 ablation in skin we deleted Sox2 in DP cells via Blimp1Cre and in Merkel cells via K14Cre. Loss of Sox2 from the DP did not inhibit hair follicle morphogenesis or establishment of the dermis and hypodermis. However, Sox2 expression in the DP was necessary for postnatal maintenance of awl/auchene hair follicles. Deletion of Sox2 via K14Cre resulted in a decreased number of Merkel cells but had no effect on other epithelial compartments or on the dermis. The reduced number of Merkel cells did not affect the number or patterning of guard hairs, nerve density or the interaction of nerve cells with the touch domes. We conclude that Sox2 is a marker of two distinct lineages in the skin and regulates the number of differentiated cells in the case of the Merkel cell lineage and hair follicle type in the case of the DP.


Asunto(s)
Linaje de la Célula , Factores de Transcripción SOXB1/metabolismo , Piel/citología , Animales , Animales Recién Nacidos , Tipificación del Cuerpo , Recuento de Células , Dermis/citología , Dermis/metabolismo , Células Epidérmicas , Epidermis/metabolismo , Eliminación de Gen , Folículo Piloso/citología , Folículo Piloso/metabolismo , Homeostasis , Integrasas/metabolismo , Queratina-14/metabolismo , Células de Merkel/citología , Células de Merkel/metabolismo , Ratones , Morfogénesis , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Piel/inervación , Piel/metabolismo , Piel/ultraestructura , Sinapsis/metabolismo , Sinapsis/ultraestructura , Factores de Transcripción/metabolismo , Vibrisas/citología
16.
Development ; 138(23): 5189-99, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22031549

RESUMEN

Hair follicle formation depends on reciprocal epidermal-dermal interactions and occurs during skin development, but not in adult life. This suggests that the properties of dermal fibroblasts change during postnatal development. To examine this, we used a PdgfraEGFP mouse line to isolate GFP-positive fibroblasts from neonatal skin, adult telogen and anagen skin and adult skin in which ectopic hair follicles had been induced by transgenic epidermal activation of ß-catenin (EF skin). We also isolated epidermal cells from each mouse. The gene expression profile of EF epidermis was most similar to that of anagen epidermis, consistent with activation of ß-catenin signalling. By contrast, adult dermis with ectopic hair follicles more closely resembled neonatal dermis than adult telogen or anagen dermis. In particular, genes associated with mitosis were upregulated and extracellular matrix-associated genes were downregulated in neonatal and EF fibroblasts. We confirmed that sustained epidermal ß-catenin activation stimulated fibroblasts to proliferate to reach the high cell density of neonatal skin. In addition, the extracellular matrix was comprehensively remodelled, with mature collagen being replaced by collagen subtypes normally present only in developing skin. The changes in proliferation and extracellular matrix composition originated from a specific subpopulation of fibroblasts located beneath the sebaceous gland. Our results show that adult dermis is an unexpectedly plastic tissue that can be reprogrammed to acquire the molecular, cellular and structural characteristics of neonatal dermis in response to cues from the overlying epidermis.


Asunto(s)
Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Transducción de Señal/fisiología , Piel/citología , Factores de Edad , Animales , Proliferación Celular , Matriz Extracelular/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Técnicas Histológicas , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , beta Catenina/metabolismo
17.
Adv Sci (Weinh) ; 11(16): e2303379, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38380561

RESUMEN

Patient-Derived Organoids (PDO) and Xenografts (PDX) are the current gold standards for patient-derived models of cancer (PDMC). Nevertheless, how patient tumor cells evolve in these models and the impact on drug response remains unclear. Herein, the transcriptomic and chromatin accessibility landscapes of matched colorectal cancer (CRC) PDO, PDX, PDO-derived PDX (PDOX), and original patient tumors (PT) are compared. Two major remodeling axes are discovered. The first axis delineates PDMC from PT, and the second axis distinguishes PDX and PDO. PDOX are more similar to PDX than PDO, indicating the growth environment is a driving force for chromatin adaptation. Transcription factors (TF) that differentially bind to open chromatins between matched PDO and PDOX are identified. Among them, KLF14 and EGR2 footprints are enriched in PDOX relative to matched PDO, and silencing of KLF14 or EGR2 promoted tumor growth. Furthermore, EPHA4, a shared downstream target gene of KLF14 and EGR2, altered tumor sensitivity to MEK inhibitor treatment. Altogether, patient-derived CRC cells undergo both common and distinct chromatin remodeling in PDO and PDX/PDOX, driven largely by their respective microenvironments, which results in differences in growth and drug sensitivity and needs to be taken into consideration when interpreting their ability to predict clinical outcome.


Asunto(s)
Ensamble y Desensamble de Cromatina , Neoplasias Colorrectales , Organoides , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Humanos , Ensamble y Desensamble de Cromatina/genética , Ratones , Animales , Organoides/metabolismo , Modelos Animales de Enfermedad
18.
Front Cell Dev Biol ; 11: 1223032, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37849736

RESUMEN

Loss of intestinal epithelial barrier function is a hallmark in digestive tract inflammation. The detailed mechanisms remain unclear due to the lack of suitable cell-based models in barrier research. Here we performed a detailed functional characterization of human intestinal organoid cultures under different conditions with the aim to suggest an optimized ex-vivo model to further analyse inflammation-induced intestinal epithelial barrier dysfunction. Differentiated Caco2 cells as a traditional model for intestinal epithelial barrier research displayed mature barrier functions which were reduced after challenge with cytomix (TNFα, IFN-γ, IL-1ß) to mimic inflammatory conditions. Human intestinal organoids grown in culture medium were highly proliferative, displayed high levels of LGR5 with overall low rates of intercellular adhesion and immature barrier function resembling conditions usually found in intestinal crypts. WNT-depletion resulted in the differentiation of intestinal organoids with reduced LGR5 levels and upregulation of markers representing the presence of all cell types present along the crypt-villus axis. This was paralleled by barrier maturation with junctional proteins regularly distributed at the cell borders. Application of cytomix in immature human intestinal organoid cultures resulted in reduced barrier function that was accompanied with cell fragmentation, cell death and overall loss of junctional proteins, demonstrating a high susceptibility of the organoid culture to inflammatory stimuli. In differentiated organoid cultures, cytomix induced a hierarchical sequence of changes beginning with loss of cell adhesion, redistribution of junctional proteins from the cell border, protein degradation which was accompanied by loss of epithelial barrier function. Cell viability was observed to decrease with time but was preserved when initial barrier changes were evident. In summary, differentiated intestinal organoid cultures represent an optimized human ex-vivo model which allows a comprehensive reflection to the situation observed in patients with intestinal inflammation. Our data suggest a hierarchical sequence of inflammation-induced intestinal barrier dysfunction starting with loss of intercellular adhesion, followed by redistribution and loss of junctional proteins resulting in reduced barrier function with consecutive epithelial death.

19.
Nat Commun ; 14(1): 4631, 2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37532704

RESUMEN

Plasmodium falciparum (Pf) parasite development in liver represents the initial step of the life-cycle in the human host after a Pf-infected mosquito bite. While an attractive stage for life-cycle interruption, understanding of parasite-hepatocyte interaction is inadequate due to limitations of existing in vitro models. We explore the suitability of hepatocyte organoids (HepOrgs) for Pf-development and show that these cells permitted parasite invasion, differentiation and maturation of different Pf strains. Single-cell messenger RNA sequencing (scRNAseq) of Pf-infected HepOrg cells has identified 80 Pf-transcripts upregulated on day 5 post-infection. Transcriptional profile changes are found involving distinct metabolic pathways in hepatocytes with Scavenger Receptor B1 (SR-B1) transcripts highly upregulated. A novel functional involvement in schizont maturation is confirmed in fresh primary hepatocytes. Thus, HepOrgs provide a strong foundation for a versatile in vitro model for Pf liver-stages accommodating basic biological studies and accelerated clinical development of novel tools for malaria control.


Asunto(s)
Malaria Falciparum , Malaria , Humanos , Plasmodium falciparum/genética , Hígado/metabolismo , Hepatocitos/metabolismo , Malaria/parasitología , Organoides/metabolismo , Malaria Falciparum/parasitología
20.
Nat Biotechnol ; 41(1): 60-69, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35879361

RESUMEN

Extending the success of cellular immunotherapies against blood cancers to the realm of solid tumors will require improved in vitro models that reveal therapeutic modes of action at the molecular level. Here we describe a system, called BEHAV3D, developed to study the dynamic interactions of immune cells and patient cancer organoids by means of imaging and transcriptomics. We apply BEHAV3D to live-track >150,000 engineered T cells cultured with patient-derived, solid-tumor organoids, identifying a 'super engager' behavioral cluster comprising T cells with potent serial killing capacity. Among other T cell concepts we also study cancer metabolome-sensing engineered T cells (TEGs) and detect behavior-specific gene signatures that include a group of 27 genes with no previously described T cell function that are expressed by super engager killer TEGs. We further show that type I interferon can prime resistant organoids for TEG-mediated killing. BEHAV3D is a promising tool for the characterization of behavioral-phenotypic heterogeneity of cellular immunotherapies and may support the optimization of personalized solid-tumor-targeting cell therapies.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Neoplasias/genética , Neoplasias/terapia , Inmunoterapia/métodos , Organoides/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA