Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Exp Clin Cancer Res ; 43(1): 116, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637831

RESUMEN

BACKGROUND: Protein arginine methyltransferase 6 (PRMT6) plays a crucial role in various pathophysiological processes and diseases. Glioblastoma (GBM; WHO Grade 4 glioma) is the most common and lethal primary brain tumor in adults, with a prognosis that is extremely poor, despite being less common than other systemic malignancies. Our current research finds PRMT6 upregulated in GBM, enhancing tumor malignancy. Yet, the specifics of PRMT6's regulatory processes and potential molecular mechanisms in GBM remain largely unexplored. METHODS: PRMT6's expression and prognostic significance in GBM were assessed using glioma public databases, immunohistochemistry (IHC), and immunoblotting. Scratch and Transwell assays examined GBM cell migration and invasion. Immunoblotting evaluated the expression of epithelial-mesenchymal transition (EMT) and Wnt-ß-catenin pathway-related proteins. Dual-luciferase reporter assays and ChIP-qPCR assessed the regulatory relationship between PRMT6 and YTHDF2. An in situ tumor model in nude mice evaluated in vivo conditions. RESULTS: Bioinformatics analysis indicates high expression of PRMT6 and YTHDF2 in GBM, correlating with poor prognosis. Functional experiments show PRMT6 and YTHDF2 promote GBM migration, invasion, and EMT. Mechanistic experiments reveal PRMT6 and CDK9 co-regulate YTHDF2 expression. YTHDF2 binds and promotes the degradation of negative regulators APC and GSK3ß mRNA of the Wnt-ß-catenin pathway, activating it and consequently enhancing GBM malignancy. CONCLUSIONS: Our results demonstrate the PRMT6-YTHDF2-Wnt-ß-Catenin axis promotes GBM migration, invasion, and EMT in vitro and in vivo, potentially serving as a therapeutic target for GBM.


Asunto(s)
Glioblastoma , Glioma , Animales , Ratones , Glioblastoma/patología , beta Catenina/genética , beta Catenina/metabolismo , Activación Transcripcional , Ratones Desnudos , Línea Celular Tumoral , Factores de Transcripción/metabolismo , Glioma/patología , Vía de Señalización Wnt , Transición Epitelial-Mesenquimal/genética , Proliferación Celular/genética , Movimiento Celular , Regulación Neoplásica de la Expresión Génica
2.
Dev Cell ; 58(19): 1996-2010.e6, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37557173

RESUMEN

Hepatocytes, the liver's predominant cells, perform numerous essential biological functions. However, crucial events and regulators during hepatocyte maturation require in-depth investigation. In this study, we performed single-cell RNA sequencing (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq) to explore the precise hepatocyte development process in mice. We defined three maturation stages of postnatal hepatocytes, each of which establishes specific metabolic functions and exhibits distinct proliferation rates. Hepatic zonation is gradually formed during hepatocyte maturation. Hepatocytes or their nuclei with distinct ploidies exhibit zonation preferences in distribution and asynchrony in maturation. Moreover, by combining gene regulatory network analysis with in vivo genetic manipulation, we identified critical maturation- and zonation-related transcription factors. This study not only delineates the comprehensive transcriptomic profiles of hepatocyte maturation but also presents a paradigm to identify genes that function in the development of hepatocyte maturation and zonation by combining genetic manipulation and measurement of coordinates in a single-cell developmental trajectory.

3.
Dev Cell ; 58(18): 1688-1700.e6, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37490911

RESUMEN

The effectiveness of multiomics analyses in defining cell differentiation pathways during development is ambiguous. During liver development, hepatoblasts follow a default or directed pathway to differentiate into hepatocytes or cholangiocytes, respectively, and this provides a practical model to address this issue. Our study discovered that promoter-associated histone modifications and chromatin accessibility dynamics, rather than enhancer-associated histone modifications, effectively delineated the "default vs. directed" process of hepatoblast differentiation. Histone H3K27me3 on bivalent promoters is associated with this asymmetric differentiation strategy in mice and humans. We demonstrated that Ezh2 and Jmjd3 exert opposing regulatory roles in hepatoblast-cholangiocyte differentiation. Additionally, active enhancers, regulated by P300, correlate with the development of both hepatocytes and cholangiocytes. This research proposes a model highlighting the division of labor between promoters and enhancers, with promoter-associated chromatin modifications governing the "default vs. directed" differentiation mode of hepatoblasts, whereas enhancer-associated modifications primarily dictate the progressive development processes of hepatobiliary lineages.


Asunto(s)
Epigenómica , Hepatocitos , Ratones , Humanos , Animales , Diferenciación Celular , Hepatocitos/metabolismo , Histonas/genética , Histonas/metabolismo , Cromatina/genética , Cromatina/metabolismo
4.
Life Sci Alliance ; 6(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37037595

RESUMEN

Enhancer of zeste homolog 2 (EZH2) is an important transcriptional regulator in development that catalyzes H3K27me3. The role of EZH2 in epicardial development is still unknown. In this study, we show that EZH2 is expressed in epicardial cells during both human and mouse heart development. Ezh2 epicardial deletion resulted in impaired epicardial cell migration, myocardial hypoplasia, and defective coronary plexus development, leading to embryonic lethality. By using RNA sequencing, we identified that EZH2 controls the transcription of tissue inhibitor of metalloproteinase 3 (TIMP3) in epicardial cells during heart development. Loss-of-function studies revealed that EZH2 promotes epicardial cell migration by suppressing TIMP3 expression. We also found that epicardial Ezh2 deficiency-induced TIMP3 up-regulation leads to extracellular matrix reconstruction in the embryonic myocardium by mass spectrometry. In conclusion, our results demonstrate that EZH2 is required for epicardial cell migration because it blocks Timp3 transcription, which is vital for heart development. Our study provides new insight into the function of EZH2 in cell migration and epicardial development.


Asunto(s)
Movimiento Celular , Proteína Potenciadora del Homólogo Zeste 2 , Corazón , Animales , Humanos , Ratones , Movimiento Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Corazón/crecimiento & desarrollo
5.
Int J Gen Med ; 15: 4399-4407, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35502183

RESUMEN

Background: The homeobox gene 5 (HOXB5) encodes a transcription factor that regulates the embryonic development of the central nervous system. Notably, its expression pattern and prognostic role in glioma remain unelucidated. Methods: This study identified the relationship between HOXB5 and glioma by investigating HOXB5 expression data from The Cancer Genome Atlas and the Genotype Tissue Expression databases and validating the obtained data using the Chinese Glioma Genome Atlas database. Western blots were used to identify HOXB5 expression levels in glioma cells and clinical samples. Kaplan-Meier and multivariate Cox regression analyses were performed to assess the prognostic value of HOXB5. The key functions and signaling pathways related to HOXB5 were analyzed using GO, KEGG, and GSEA. Immune infiltration was calculated using the microenvironment cell populations-counter, estimate the proportion of immune and cancer, and ESTIMATE algorithms. Results: The expression of HOXB5 was upregulated in glioma and generally increased with malignancy. HOXB5 was an independent prognostic factor for glioma patients. A nomogram was further built that integrated HOXB5, and it showed stratifying prediction accuracy and efficiency. HOXB5 was associated with the regulation of cell growth, endothelial cell growth, and the IL-6/JAK-STAT3 pathway, and was determined to possibly promote stomatal specimen enrichment and angiogenesis. Conclusion: HOXB5 protein is overexpressed in glioma and might serve as a good predictive factor of this disease.

6.
Cell Res ; 32(1): 38-53, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34341490

RESUMEN

Limited knowledge of cellular and molecular mechanisms underlying hematopoietic stem cell and multipotent progenitor (HSC/MPP) expansion within their native niche has impeded the application of stem cell-based therapies for hematological malignancies. Here, we constructed a spatiotemporal transcriptome map of mouse fetal liver (FL) as a platform for hypothesis generation and subsequent experimental validation of novel regulatory mechanisms. Single-cell transcriptomics revealed three transcriptionally heterogeneous HSC/MPP subsets, among which a CD93-enriched subset exhibited enhanced stem cell properties. Moreover, by employing integrative analysis of single-cell and spatial transcriptomics, we identified novel HSC/MPP 'pocket-like' units (HSC PLUS), composed of niche cells (hepatoblasts, stromal cells, endothelial cells, and macrophages) and enriched with growth factors. Unexpectedly, macrophages showed an 11-fold enrichment in the HSC PLUS. Functionally, macrophage-HSC/MPP co-culture assay and candidate molecule testing, respectively, validated the supportive role of macrophages and growth factors (MDK, PTN, and IGFBP5) in HSC/MPP expansion. Finally, cross-species analysis and functional validation showed conserved cell-cell interactions and expansion mechanisms but divergent transcriptome signatures between mouse and human FL HSCs/MPPs. Taken together, these results provide an essential resource for understanding HSC/MPP development in FL, and novel insight into functional HSC/MPP expansion ex vivo.


Asunto(s)
Células Endoteliales , Transcriptoma , Animales , Hematopoyesis/genética , Células Madre Hematopoyéticas , Hígado , Ratones
7.
Cancer Cell Int ; 21(1): 633, 2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34838021

RESUMEN

BACKGROUND: Current treatment options for glioma are limited, and the prognosis of patients with glioma is poor as the available drugs show low therapeutic efficacy. Furthermore, the molecular mechanisms associated with glioma remain poorly understood. METTL1 mainly catalyzes the formation of N(7)-methylguanine at position 46 of the transfer RNA sequence, thereby regulating the translation process. However, the role of METTL1 in glioma has not been studied to date. The purpose of this study was to analyze the expression and prognosis of METTL1 in glioma, and to explore the potential analysis mechanism. METHODS: Data from five publicly available databases were used to analyze METTL1 expression across different tumor types and its differential expression between carcinoma and adjacent normal tissues. The expression of METTL1 in glioma was further validated using real-time polymerase chain reaction and immunohistochemistry. Meanwhile, siRNA was used to knockdown METTL1 in U87 glioma cells, and the resultant effect on glioma proliferation was verified using the Cell Counting Kit 8 (CCK8) assay. Furthermore, a nomogram was constructed to predict the association between METTL1 expression and the survival rate of patients with glioma. RESULTS: METTL1 expression increased with increasing glioma grades and was significantly higher in glioma than in adjacent noncancerous tissues. In addition, high expression of METTL1 promoted cell proliferation. Moreover, METTL1 expression was associated with common clinical risk factors and was significantly associated with the prognosis and survival of patients with glioma. Univariate and multivariate Cox regression analyses revealed that METTL1 expression may be used as an independent prognostic risk factor for glioma. Furthermore, results of functional enrichment and pathway analyses indicate that the mechanism of METTL1 in glioma is potentially related to the MAPK signaling pathway. CONCLUSIONS: High METTL1 expression is significantly associated with poor prognosis of patients with glioma and may represent a valuable independent risk factor. In addition, high expression of METTL1 promotes glioma proliferation and may regulate mitogen-activated protein kinase (MAPK) signaling pathway. Thus, METTL1 may be a potential biomarker for glioma. Further investigations are warranted to explore its clinical use.

9.
Cell Res ; 31(8): 886-903, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33692492

RESUMEN

The pancreatic islet contains multiple hormone+ endocrine lineages (α, ß, δ, PP and ε cells), but the developmental processes that underlie endocrinogenesis are poorly understood. Here, we generated novel mouse lines and combined them with various genetic tools to enrich all types of hormone+ cells for well-based deep single-cell RNA sequencing (scRNA-seq), and gene coexpression networks were extracted from the generated data for the optimization of high-throughput droplet-based scRNA-seq analyses. These analyses defined an entire endocrinogenesis pathway in which different states of endocrine progenitor (EP) cells sequentially differentiate into specific endocrine lineages in mice. Subpopulations of the EP cells at the final stage (EP4early and EP4late) show different potentials for distinct endocrine lineages. ε cells and an intermediate cell population were identified as distinct progenitors that independently generate both α and PP cells. Single-cell analyses were also performed to delineate the human pancreatic endocrinogenesis process. Although the developmental trajectory of pancreatic lineages is generally conserved between humans and mice, clear interspecies differences, including differences in the proportions of cell types and the regulatory networks associated with the differentiation of specific lineages, have been detected. Our findings support a model in which sequential transient progenitor cell states determine the differentiation of multiple cell lineages and provide a blueprint for directing the generation of pancreatic islets in vitro.


Asunto(s)
Islotes Pancreáticos , Páncreas , Animales , Diferenciación Celular , Linaje de la Célula , Humanos , Ratones , Células Madre
10.
Cell Res ; 31(3): 326-344, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33106598

RESUMEN

Defining the precise regionalization of specified definitive endoderm progenitors is critical for understanding the mechanisms underlying the generation and regeneration of respiratory and digestive organs, yet the patterning of endoderm progenitors remains unresolved, particularly in humans. We performed single-cell RNA sequencing on endoderm cells during the early somitogenesis stages in mice and humans. We developed molecular criteria to define four major endoderm regions (foregut, lip of anterior intestinal portal, midgut, and hindgut) and their developmental pathways. We identified the cell subpopulations in each region and their spatial distributions and characterized key molecular features along the body axes. Dorsal and ventral pancreatic progenitors appear to originate from the midgut population and follow distinct pathways to develop into an identical cell type. Finally, we described the generally conserved endoderm patterning in humans and clear differences in dorsal cell distribution between species. Our study comprehensively defines single-cell endoderm patterning and provides novel insights into the spatiotemporal process that drives establishment of early endoderm domains.


Asunto(s)
Tipificación del Cuerpo/genética , Embrión de Mamíferos/citología , Endodermo/citología , Intestinos/citología , Labio/citología , Animales , Células Cultivadas , Femenino , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , RNA-Seq/métodos , Análisis de la Célula Individual/métodos
11.
Cell Rep ; 33(10): 108455, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33296648

RESUMEN

The ever-increasing therapeutic and pharmaceutical demand for liver cells calls for systems that enable mass production of hepatic cells. Here we describe a large-scale suspension system that uses human endoderm stem cells (hEnSCs) as precursors to generate functional and transplantable hepatocytes (E-heps) or cholangiocytes (E-chos). hEnSC-derived hepatic populations are characterized by single-cell transcriptomic analyses and compared with hESC-derived counterparts, in-vitro-maintained or -expanded primary hepatocytes and adult cells, which reveals that hepatic differentiation of hEnSCs recapitulates in vivo development and that the heterogeneities of the resultant populations can be manipulated by regulating the EGF and MAPK signaling pathways. Functional assessments demonstrate that E-heps and E-chos possess properties comparable with adult counterparts and that, when transplanted intraperitoneally, encapsulated E-heps were able to rescue rats with acute liver failure. Our study lays the foundation for cell-based therapeutic agents and in vitro applications for liver diseases.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Endodermo/citología , Hepatocitos/citología , Células Madre Embrionarias Humanas/citología , Conductos Biliares/citología , Conductos Biliares/metabolismo , Diferenciación Celular/fisiología , Endodermo/metabolismo , Endodermo/trasplante , Células Epiteliales/citología , Células Epiteliales/metabolismo , Hepatocitos/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/trasplante , Humanos , Hígado/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante
12.
Biomed Res Int ; 2020: 1346340, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32775408

RESUMEN

PURPOSE: In our study, we aimed to screen the risk factors that affect overall survival (OS) and cancer-specific survival (CSS) in adult glioma patients and to develop and evaluate nomograms. METHODS: Primary high-grade gliomas patients being retrieved from the surveillance, epidemiology and end results (SEER) database, between 2004 and 2015, then they randomly assigned to a training group and a validation group. Univariate and multivariate Cox analysis models were used to choose the variables significantly correlated with the prognosis of high-grade glioma patients. And these variables were used to construct the nomograms. Next, concordance index (C-index), calibration plot and receiver operating characteristics (ROCs) curve were used to evaluate the accuracy of the nomogram model. In addition, the decision curve analysis (DCA) was used to analyze the benefit of nomogram and prognostic indicators commonly used in clinical practice. RESULTS: A total of 6395 confirmed glioma patients were selected from the SEER database, divided into training set (n =3166) and validation set (n =3229). Age at diagnosis, tumor grade, tumor size, histological type, surgical type, radiotherapy and chemotherapy were screened out by Cox analysis model. For OS nomogram, the C-index of the training set was 0.741 (95% CI: 0.751-0.731), and the validation set was 0.738 (95% CI: 0.748-0.728). For CSS nomogram, the C-index of the training set was 0.739 (95% CI: 0.749-0.729), and the validation set was 0.738 (95% CI: 0.748-0.728). The net benefit and net reduction in inverventions of nomograms in the decision curve analysis (DCA) was higher than histological type. CONCLUSIONS: We developed nomograms to predict 3- and 5-year OS rates and 3- and 5-year CSS rates in adult high-grade glioma patients. Both the training set and the validation set showed good calibration and validation, indicating the clinical applicability of the nomogram and good predictive results.


Asunto(s)
Glioma/patología , Adulto , Factores de Edad , Anciano , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias/métodos , Nomogramas , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , Estudios Retrospectivos , Factores de Riesgo , Programa de VERF , Tasa de Supervivencia
13.
Cell Res ; 30(12): 1109-1126, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32690901

RESUMEN

During embryogenesis, the liver is the site of hepatogenesis and hematopoiesis and contains many cell lineages derived from the endoderm and mesoderm. However, the characteristics and developmental programs of many of these cell lineages remain unclear, especially in humans. Here, we performed single-cell RNA sequencing of whole human and mouse fetal livers throughout development. We identified four cell lineage families of endoderm-derived, erythroid, non-erythroid hematopoietic, and mesoderm-derived non-hematopoietic cells, and defined the developmental pathways of the major cell lineage families. In both humans and mice, we identified novel markers of hepatic lineages and an ID3+ subpopulation of hepatoblasts as well as verified that hepatoblast differentiation follows the "default-directed" model. Additionally, we found that human but not mouse fetal hepatocytes display heterogeneity associated with expression of metabolism-related genes. We described the developmental process of erythroid progenitor cells during human and mouse hematopoiesis. Moreover, despite the general conservation of cell differentiation programs between species, we observed different cell lineage compositions during hematopoiesis in the human and mouse fetal livers. Taken together, these results reveal the dynamic cell landscape of fetal liver development and illustrate the similarities and differences in liver development between species, providing an extensive resource for inducing various liver cell lineages in vitro.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Hígado/citología , Organogénesis/genética , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Biomarcadores/metabolismo , Eritrocitos/citología , Eritropoyesis/genética , Feto/citología , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hígado/embriología , Mesodermo/citología , Ratones , Análisis de Componente Principal , ARN/metabolismo , Procesos Estocásticos
14.
Development ; 147(7)2020 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-32280064

RESUMEN

Understanding the mechanisms that underlie the generation and regeneration of ß cells is crucial for developing treatments for diabetes. However, traditional research methods, which are based on populations of cells, have limitations for defining the precise processes of ß-cell differentiation and trans-differentiation, and the associated regulatory mechanisms. The recent development of single-cell technologies has enabled re-examination of these processes at a single-cell resolution to uncover intermediate cell states, cellular heterogeneity and molecular trajectories of cell fate specification. Here, we review recent advances in understanding ß-cell generation and regeneration, in vivo and in vitro, from single-cell technologies, which could provide insights for optimization of diabetes therapy strategies.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Regeneración/fisiología , Análisis de la Célula Individual/métodos , Animales , Rastreo Celular/métodos , Rastreo Celular/tendencias , Humanos , Páncreas/citología , Páncreas/fisiología , Análisis de la Célula Individual/tendencias
15.
Mol Metab ; 37: 100982, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32247924

RESUMEN

OBJECTIVES: The streptozotocin (STZ) model is widely used in diabetes research. However, the cellular and molecular states of pancreatic endocrine cells in this model remain unclear. This study explored the molecular characteristics of islet cells treated with STZ and re-evaluated ß-cell dysfunction and regeneration in the STZ model. METHODS: We performed single-cell RNA sequencing of pancreatic endocrine cells from STZ-treated mice. High-quality sequencing data from 2,999 cells were used to identify clusters via Louvain clustering analysis. Principal component analysis (PCA), t-distributed stochastic neighbor embedding (t-SNE), uniform manifold approximation and projection (UMAP), force-directed layout (FDL), and differential expression analysis were used to define the heterogeneity and transcriptomic changes in islet cells. In addition, qPCR and immunofluorescence staining were used to confirm findings from the sequencing data. RESULTS: Untreated ß-cells were divided into two populations at the transcriptomic level, a large high-Glut2 expression (Glut2high) population and a small low-Glut2 expression (Glut2low) population. At the transcriptomic level, Glut2low ß-cells in adult mice did not represent a developmentally immature state, although a fraction of genes associated with ß-cell maturation and function were downregulated in Glut2low cells. After a single high-dose STZ treatment, most Glut2high cells were killed, but Glut2low cells survived and over time changed to a distinct cell state. We did not observe conversion of Glut2low to Glut2high ß-cells up to 9 months after STZ treatment. In addition, we did not detect transcriptomic changes in the non-ß endocrine cells or a direct trans-differentiation pathway from the α-cell lineage to the ß-cell lineage in the STZ model. CONCLUSIONS: We identified the heterogeneity of ß-cells in both physiological and pathological conditions. However, we did not observe conversion of Glut2low to Glut2high ß-cells, transcriptomic changes in the non-ß endocrine cells, or direct trans-differentiation from the α-cell lineage to the ß-cell lineage in the STZ model. Our results clearly define the states of islet cells treated with STZ and allow us to re-evaluate the STZ model widely used in diabetes studies.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatología , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Células Secretoras de Glucagón/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/fisiología , Masculino , Ratones , Ratones Transgénicos , Análisis de la Célula Individual/métodos , Estreptozocina/farmacología , Transcriptoma/genética
16.
EMBO J ; 38(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30737258

RESUMEN

The generation of terminally differentiated cell lineages during organogenesis requires multiple, coordinated cell fate choice steps. However, this process has not been clearly delineated, especially in complex solid organs such as the pancreas. Here, we performed single-cell RNA-sequencing in pancreatic cells sorted from multiple genetically modified reporter mouse strains at embryonic stages E9.5-E17.5. We deciphered the developmental trajectories and regulatory strategies of the exocrine and endocrine pancreatic lineages as well as intermediate progenitor populations along the developmental pathways. Notably, we discovered previously undefined programs representing the earliest events in islet α- and ß-cell lineage allocation as well as the developmental pathway of the "first wave" of α-cell generation. Furthermore, we demonstrated that repressing ERK pathway activity is essential for inducing both α- and ß-lineage differentiation. This study provides key insights into the regulatory mechanisms underlying cell fate choice and stepwise cell fate commitment and can be used as a resource to guide the induction of functional islet lineage cells from stem cells in vitro.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Organogénesis , Páncreas/metabolismo , Análisis de la Célula Individual/métodos , Animales , Femenino , Ratones , Páncreas/citología
17.
Semin Cell Dev Biol ; 92: 63-76, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30193996

RESUMEN

The liver is an indispensable organ for metabolism and drug detoxification. The liver consists of endoderm-derived hepatobiliary lineages and various mesoderm-derived cells, and interacts with the surrounding tissues and organs through the ventral mesentery. Liver development, from hepatic specification to liver maturation, requires close interactions with mesoderm-derived cells, such as mesothelial cells, hepatic stellate cells, mesenchymal cells, liver sinusoidal endothelial cells and hematopoietic cells. These cells affect liver development through precise signaling events and even direct physical contact. Through the use of new techniques, emerging studies have recently led to a deeper understanding of liver development and its related mechanisms, especially the roles of mesodermal cells in liver development. Based on these developments, the current protocols for in vitro hepatocyte-like cell induction and liver-like tissue construction have been optimized and are of great importance for the treatment of liver diseases. Here, we review the roles of mesoderm-derived cells in the processes of liver development, hepatocyte-like cell induction and liver-like tissue construction.


Asunto(s)
Hígado/embriología , Mesodermo/metabolismo , Animales , Humanos , Mesodermo/citología , Ratones
18.
J Vis Exp ; (139)2018 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-30320740

RESUMEN

Pancreatic endocrine cells, which are clustered in islets, regulate blood glucose stability and energy metabolism. The distinct cell types in islets, including insulin-secreting ß cells, are differentiated from common endocrine progenitors during the embryonic stage. Immature endocrine cells expand via cell proliferation and mature during a long postnatal developmental period. However, the mechanisms underlying these processes are not clearly defined. Single-cell RNA-sequencing is a promising approach for the characterization of distinct cell populations and tracing cell lineage differentiation pathways. Here, we describe a method for the single-cell RNA-sequencing of isolated pancreatic ß cells from embryonic, neonatal and postnatal pancreases.


Asunto(s)
Células Endocrinas/metabolismo , Perfilación de la Expresión Génica/métodos , Células Secretoras de Insulina/citología , Análisis de la Célula Individual/métodos , Animales , Diferenciación Celular , Proliferación Celular , Glucosa/metabolismo , Insulina/metabolismo , Ratones , Análisis de Secuencia de ARN
19.
EMBO Rep ; 19(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30065074

RESUMEN

The pancreas of vertebrates is separately derived from both the dorsal and ventral endodermal domains. However, the difference between these two programs has been unclear. Here, using a pancreatic determination gene, Pdx1, driven GFP transgenic mouse strain, we identified Pdx1-GFP highly expressing cells (Pdx1high) and Pdx1-GFP lowly expressing cells (Pdx1low) in both embryonic dorsal Pdx1-expressing region (DPR) and ventral Pdx1-expressing region (VPR). We analyzed the transcriptomes of single Pdx1low and Pdx1high cells from the DPR and VPR. In the VPR, Pdx1low cells have an intermediate progenitor identity and can generate hepatoblasts, extrahepatobiliary cells, and Pdx1high pancreatic progenitor cells. In the DPR, Pdx1high cells are directly specified as pancreatic progenitors, whereas Pdx1low cells are precocious endocrine cells. Therefore, our study defines distinct road maps for dorsal and ventral pancreatic progenitor specification. The findings provide guidance for optimization of current ß-cell induction protocols by following the in vivo dorsal pancreatic specification program.


Asunto(s)
Proteínas de Homeodominio/genética , Páncreas/crecimiento & desarrollo , Células Madre/metabolismo , Transactivadores/genética , Transcriptoma/genética , Animales , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Transgénicos/genética , Páncreas/embriología , Páncreas/metabolismo , Análisis de la Célula Individual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...