Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nature ; 572(7771): 660-664, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31435013

RESUMEN

Implantation is a milestone event during mammalian embryogenesis. Implantation failure is a considerable cause of early pregnancy loss in humans1. Owing to the difficulty of obtaining human embryos early after implantation in vivo, it remains unclear how the gene regulatory network and epigenetic mechanisms control the implantation process. Here, by combining an in vitro culture system for the development human embryos after implantation and single-cell multi-omics sequencing technologies, more than 8,000 individual cells from 65 human peri-implantation embryos were systematically analysed. Unsupervised dimensionality reduction and clustering algorithms of the transcriptome data show stepwise implantation routes for the epiblast, primitive endoderm and trophectoderm lineages, suggesting robust preparation for the proper establishment of a mother-to-offspring connection during implantation. Female embryos showed initiation of random X chromosome inactivation based on analysis of parental allele-specific expression of X-chromosome-linked genes during implantation. Notably, using single-cell triple omics sequencing analysis, the re-methylation of the genome in cells from the primitive endoderm lineage was shown to be much slower than in cells of both epiblast and trophectoderm lineages during the implantation process, which indicates that there are distinct re-establishment features in the DNA methylome of the epiblast and primitive endoderm-even though both lineages are derived from the inner cell mass. Collectively, our work provides insights into the complex molecular mechanisms that regulate the implantation of human embryos, and helps to advance future efforts to understanding early embryonic development and reproductive medicine.


Asunto(s)
Metilación de ADN , Desarrollo Embrionario/genética , Epigenoma , Transcriptoma/genética , Linaje de la Célula/genética , Cromosomas Humanos X/genética , Variaciones en el Número de Copia de ADN/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , RNA-Seq , Análisis de la Célula Individual , Inactivación del Cromosoma X/genética
2.
PLoS Biol ; 19(5): e3001229, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34003819

RESUMEN

DNA methylation, chromatin accessibility, and gene expression represent different levels information in biological process, but a comprehensive multiomics analysis of the mammalian heart is lacking. Here, we applied nucleosome occupancy and methylome sequencing, which detected DNA methylation and chromatin accessibility simultaneously, as well as RNA-seq, for multiomics analysis of the 4 chambers of adult and fetal human hearts, and adult mouse hearts. Our results showed conserved region-specific patterns in the mammalian heart at transcriptome and DNA methylation level. Adult and fetal human hearts showed distinct features in DNA methylome, chromatin accessibility, and transcriptome. Novel long noncoding RNAs were identified in the human heart, and the gene expression profiles of major cardiovascular diseases associated genes were displayed. Furthermore, cross-species comparisons revealed human-specific and mouse-specific differentially expressed genes between the atria and ventricles. We also reported the relationship among multiomics and found there was a bell-shaped relationship between gene-body methylation and expression in the human heart. In general, our study provided comprehensive spatiotemporal and evolutionary insights into the regulation of gene expression in the heart.


Asunto(s)
Corazón/crecimiento & desarrollo , Corazón/fisiología , Animales , Cromatina/metabolismo , Islas de CpG/genética , ADN/genética , Metilación de ADN/genética , Epigénesis Genética/genética , Epigenómica/métodos , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Ventrículos Cardíacos/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Ratones , Nucleosomas/metabolismo , Especificidad de Órganos/genética , ARN Largo no Codificante/metabolismo , Especificidad de la Especie , Transcriptoma/genética
3.
PLoS Biol ; 17(7): e3000365, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31269016

RESUMEN

The developmental pathway of the neural retina (NR) and retinal pigment epithelium (RPE) has been revealed by extensive research in mice. However, the molecular mechanisms underlying the development of the human NR and RPE, as well as the interactions between these two tissues, have not been well defined. Here, we analyzed 2,421 individual cells from human fetal NR and RPE using single-cell RNA sequencing (RNA-seq) technique and revealed the tightly regulated spatiotemporal gene expression network of human retinal cells. We identified major cell classes of human fetal retina and potential crucial transcription factors for each cell class. We dissected the dynamic expression patterns of visual cycle- and ligand-receptor interaction-related genes in the RPE and NR. Moreover, we provided a map of disease-related genes for human fetal retinal cells and highlighted the importance of retinal progenitor cells as potential targets of inherited retinal diseases. Our findings captured the key in vivo features of the development of the human NR and RPE and offered insightful clues for further functional studies.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma , Adulto , Ciclo Celular/genética , Células Cultivadas , Análisis por Conglomerados , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Humanos , Retina/citología , Retina/embriología , Enfermedades de la Retina/genética , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/embriología
4.
Gut ; 69(7): 1283-1293, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31744909

RESUMEN

OBJECTIVE: Familial adenomatous polyposis (FAP) is characterised by the development of hundreds to thousands of adenomas at different evolutionary stages in the colon and rectum that will inevitably progress to adenocarcinomas if left untreated. Here, we investigated the genetic alterations and transcriptomic transitions from precancerous adenoma to carcinoma. DESIGN: Whole-exome sequencing, whole-genome sequencing and single-cell RNA sequencing were performed on matched adjacent normal tissues, multiregionally sampled adenomas at different stages and carcinomas from six patients with FAP and one patient with MUTYH-associated polyposis (n=56 exomes, n=56 genomes and n=8,757 single cells). Genomic alterations (including copy number alterations and somatic mutations), clonal architectures and transcriptome dynamics during adenocarcinoma carcinogenesis were comprehensively investigated. RESULTS: Genomic evolutionary analysis showed that adjacent lesions from the same patient with FAP can originate from the same cancer-primed cell. In addition, the tricarboxylic acid cycle pathway was strongly repressed in adenomas and was then slightly alleviated in carcinomas. Cells from the 'normal' colon epithelium of patients with FAP already showed metabolic reprogramming compared with cells from the normal colon epithelium of patients with sporadic colorectal cancer. CONCLUSIONS: The process described in the previously reported field cancerisation model also occurs in patients with FAP and can contribute to the formation of adjacent lesions in patients with FAP. Reprogramming of carbohydrate metabolism has already occurred at the precancerous adenoma stage. Our study provides an accurate picture of the genomic and transcriptomic landscapes during the initiation and progression of carcinogenesis, especially during the transition from adenoma to carcinoma.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Carcinogénesis/genética , Poliposis Adenomatosa del Colon/metabolismo , Carcinogénesis/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Redes y Vías Metabólicas/genética , Linaje , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Secuenciación del Exoma , Secuenciación Completa del Genoma
5.
Protein Cell ; 15(4): 285-304, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37345888

RESUMEN

Colorectal cancer (CRC) is a highly heterogeneous cancer and exploring novel therapeutic options is a pressing issue that needs to be addressed. Here, we established human CRC tumor-derived organoids that well represent both morphological and molecular heterogeneities of original tumors. To efficiently identify repurposed drugs for CRC, we developed a robust organoid-based drug screening system. By combining the repurposed drug library and computation-based drug prediction, 335 drugs were tested and 34 drugs with anti-CRC effects were identified. More importantly, we conducted a detailed transcriptome analysis of drug responses and divided the drug response signatures into five representative patterns: differentiation induction, growth inhibition, metabolism inhibition, immune response promotion, and cell cycle inhibition. The anticancer activities of drug candidates were further validated in the established patient-derived organoids-based xenograft (PDOX) system in vivo. We found that fedratinib, trametinib, and bortezomib exhibited effective anticancer effects. Furthermore, the concordance and discordance of drug response signatures between organoids in vitro and pairwise PDOX in vivo were evaluated. Our study offers an innovative approach for drug discovery, and the representative transcriptome features of drug responses provide valuable resources for developing novel clinical treatments for CRC.


Asunto(s)
Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Detección Precoz del Cáncer , Organoides/patología
6.
Sci Rep ; 14(1): 9906, 2024 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689033

RESUMEN

CUL4B, a crucial scaffolding protein in the largest E3 ubiquitin ligase complex CRL4B, is involved in a broad range of physiological and pathological processes. While previous research has shown that CUL4B participates in maintaining intestinal homeostasis and function, its involvement in facilitating intestinal recovery following ionizing radiation (IR) damage has not been fully elucidated. Here, we utilized in vivo and in vitro models to decipher the role of CUL4B in intestinal repair after IR-injury. Our findings demonstrated that prior to radiation exposure, CUL4B inhibited the ubiquitination modification of PSME3, which led to the accumulation of PSME3 and subsequent negative regulation of p53-mediated apoptosis. In contrast, after radiation, CUL4B dissociated from PSME3 and translocated into the nucleus at phosphorylated histones H2A (γH2AX) foci, thereby impeding DNA damage repair and augmenting p53-mediated apoptosis through inhibition of BRCA1 phosphorylation and RAD51. Our study elucidated the dynamic role of CUL4B in the repair of radiation-induced intestinal damage and uncovered novel molecular mechanisms underlying the repair process, suggesting a potential therapeutic strategy of intestinal damage after radiation therapy for cancers.


Asunto(s)
Apoptosis , Proteínas Cullin , Intestinos , Regeneración , Proteína p53 Supresora de Tumor , Animales , Humanos , Ratones , Apoptosis/efectos de la radiación , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Proteínas Cullin/metabolismo , Proteínas Cullin/genética , Daño del ADN , Reparación del ADN , Histonas/metabolismo , Intestinos/efectos de la radiación , Intestinos/patología , Ratones Endogámicos C57BL , Fosforilación/efectos de la radiación , Recombinasa Rad51/metabolismo , Radiación Ionizante , Regeneración/efectos de la radiación , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación
7.
Cell Stem Cell ; 30(3): 283-299.e9, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36787740

RESUMEN

Stem cell-independent reprogramming of differentiated cells has recently been identified as an important paradigm for repairing injured tissues. Following periportal injury, mature hepatocytes re-activate reprogramming/progenitor-related genes (RRGs) and dedifferentiate into liver progenitor-like cells (LPLCs) in both mice and humans, which contribute remarkably to regeneration. However, it remains unknown which and how external factors trigger hepatocyte reprogramming. Here, by employing single-cell transcriptional profiling and lineage-specific deletion tools, we uncovered that periportal-specific LPLC formation was initiated by regionally activated Kupffer cells but not peripheral monocyte-derived macrophages. Unexpectedly, using in vivo screening, the proinflammatory factor IL-6 was identified as the niche signal repurposed for RRG induction via STAT3 activation, which drove RRG expression through binding to their pre-accessible enhancers. Notably, RRGs were activated through injury-specific rather than liver embryogenesis-related enhancers. Collectively, these findings depict an injury-specific niche signal and the inflammation-mediated transcription in driving the conversion of hepatocytes into a progenitor phenotype.


Asunto(s)
Interleucina-6 , Macrófagos del Hígado , Animales , Humanos , Ratones , Diferenciación Celular , Hepatocitos/metabolismo , Interleucina-6/metabolismo , Macrófagos del Hígado/fisiología , Hígado , Regeneración Hepática/fisiología
8.
Genome Biol ; 23(1): 106, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35484598

RESUMEN

BACKGROUND: Patient-derived organoid culture is a powerful system for studying the molecular mechanisms of cancers, especially colorectal cancer (CRC), one of the most prevalent cancers worldwide. There are two main types of 3D culture methods for colonic cells, but the similarities and differences between gene expression patterns in different culture media remain largely unexplored. RESULTS: Here, we establish patient-derived organoids from colorectal cancer patients and perform single-cell RNA-Seq for pairwise samples from seven patients for both organoids and their corresponding tumor and normal tissues in vivo. We find that organoids derived from tumor tissues faithfully recapitulate the main gene expression signatures of cancer cells in vivo. On the other hand, organoids derived from normal tissues exhibited some tumor-like features at the whole transcriptome level but retained normal genomic features, such as CNVs, point mutations, and normal global DNA methylation levels, for both cultural media. More importantly, we show that conditioned medium outperforms chemical-defined medium in long-term culture of tumor epithelial cells. Finally, we mutually exchange the culture medium for the organoids and find that after interchanging the medium, the organoid cells basically maintain the transcriptome characteristics of the original medium. CONCLUSIONS: Our work gives a thorough evaluation of both the cultural conditions and the biological features of organoids of CRC patients.


Asunto(s)
Neoplasias Colorrectales , Organoides , Neoplasias Colorrectales/patología , Metilación de ADN , Humanos , Organoides/metabolismo , Organoides/patología , Análisis de Secuencia de ARN
9.
Genome Med ; 14(1): 93, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35974387

RESUMEN

BACKGROUND: Colorectal cancer (CRC) ranks as the second-leading cause of cancer-related death worldwide with metastases being the main cause of cancer-related death. Here, we investigated the genomic and transcriptomic alterations in matching adjacent normal tissues, primary tumors, and metastatic tumors of CRC patients. METHODS: We performed whole genome sequencing (WGS), multi-region whole exome sequencing (WES), simultaneous single-cell RNA-Seq, and single-cell targeted cDNA Sanger sequencing on matching adjacent normal tissues, primary tumors, and metastatic tumors from 12 metastatic colorectal cancer patients (n=84 for genomes, n=81 for exomes, n=9120 for single cells). Patient-derived tumor organoids were used to estimate the anti-tumor effects of a PPAR inhibitor, and self-renewal and differentiation ability of stem cell-like tumor cells. RESULTS: We found that the PPAR signaling pathway was prevalently and aberrantly activated in CRC tumors. Blocking of PPAR pathway both suppressed the growth and promoted the apoptosis of CRC organoids in vitro, indicating that aberrant activation of the PPAR signaling pathway plays a critical role in CRC tumorigenesis. Using matched samples from the same patient, distinct origins of the metastasized tumors between lymph node and liver were revealed, which was further verified by both copy number variation and mitochondrial mutation profiles at single-cell resolution. By combining single-cell RNA-Seq and single-cell point mutation identification by targeted cDNA Sanger sequencing, we revealed important phenotypic differences between cancer cells with and without critical point mutations (KRAS and TP53) in the same patient in vivo at single-cell resolution. CONCLUSIONS: Our data provides deep insights into how driver mutations interfere with the transcriptomic state of cancer cells in vivo at a single-cell resolution. Our findings offer novel knowledge on metastatic mechanisms as well as potential markers and therapeutic targets for CRC diagnosis and therapy. The high-precision single-cell RNA-seq dataset of matched adjacent normal tissues, primary tumors, and metastases from CRCs may serve as a rich resource for further studies.


Asunto(s)
Neoplasias Colorrectales , Metástasis de la Neoplasia , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Variaciones en el Número de Copia de ADN , ADN Complementario , Genómica , Humanos , Mutación , Receptores Activados del Proliferador del Peroxisoma/antagonistas & inhibidores , Receptores Activados del Proliferador del Peroxisoma/genética , Transcriptoma
10.
Cell Discov ; 8(1): 92, 2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36104333

RESUMEN

Small bowel adenocarcinomas (SBAs) are rare malignant tumors with a high mortality rate, and their molecular characteristics are still largely unexplored. Here we performed single-cell RNA sequencing for tumor samples from 12 SBA patients and predicted drug candidates for SBA. We identified four prevalent subtypes of malignant cells with distinct signatures including cell cycle program, mitochondria program, metabolism program and epithelial-mesenchymal transition (EMT) program. The progression relationships of these four subtypes of malignant cells were also revealed, which started from the cell cycle program, through the mitochondria program and then progressing into either the metabolism program or the EMT program. Importantly, ligand-receptor interaction pairs were found to be specifically enriched in pairs of EMT-program malignant cells and highly exhausted CD8+ T cells, suggesting that cancer cell subpopulations with EMT features may contribute most to the exhaustion of T cells. We also showed that the duodenal subtype of SBA exhibited molecular features more similar to gastric cancer whereas jejunal subtype of SBA more similar to colorectal cancer. Especially, we predicted specific drugs for SBA based on differential gene expression signatures between malignant cells and normal epithelial cells of SBA, and verified more potent inhibitory effects of volasertib and tozasertib for SBA cancer cells than conventional drugs of SBA at the same concentration, which provides new clues for treatments of SBA. In summary, our study provides a blueprint of the molecular signatures of both tumor cells and tumor microenvironment cells in SBA and reveals potential targets and drug candidates for its clinical treatments.

11.
Cancer Res ; 82(21): 3903-3916, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-35969151

RESUMEN

High-grade serous cancer (HGSC) is the most common subtype of ovarian cancer. HGSC is highly aggressive with poor patient outcomes, and a deeper understanding of HGSC tumorigenesis could help guide future treatment development. To systematically characterize the underlying pathologic mechanisms and intratumoral heterogeneity in human HGSC, we used an optimized single-cell multiomics sequencing technology to simultaneously analyze somatic copy-number alterations (SCNA), DNA methylation, chromatin accessibility, and transcriptome in individual cancer cells. Genes associated with interferon signaling, metallothioneins, and metabolism were commonly upregulated in ovarian cancer cells. Integrated multiomics analyses revealed that upregulation of interferon signaling and metallothioneins was influenced by both demethylation of their promoters and hypomethylation of satellites and LINE1, and potential key transcription factors regulating glycolysis using chromatin accessibility data were uncovered. In addition, gene expression and DNA methylation displayed similar patterns in matched primary and abdominal metastatic tumor cells of the same genetic lineage, suggesting that metastatic cells potentially preexist in the subclones of primary tumors. Finally, the lineages of cancer cells with higher residual DNA methylation levels and upregulated expression of CCN1 and HSP90AA1 presented greater metastatic potential. This study characterizes the critical genetic, epigenetic, and transcriptomic features and their mutual regulatory relationships in ovarian cancer, providing valuable resources for identifying new molecular mechanisms and potential therapeutic targets for HGSC. SIGNIFICANCE: Integrated analysis of multiomic changes and epigenetic regulation in high-grade serous ovarian cancer provides insights into the molecular characteristics of this disease, which could help improve diagnosis and treatment.


Asunto(s)
Cistadenocarcinoma Seroso , Neoplasias Ováricas , Humanos , Femenino , Cistadenocarcinoma Seroso/patología , Epigénesis Genética , Carcinoma Epitelial de Ovario/genética , Neoplasias Ováricas/patología , Cromatina , Interferones/metabolismo
12.
Cell Rep ; 34(9): 108788, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33657375

RESUMEN

The heterogeneity and molecular characteristics of progenitor cells, especially glial progenitors, in the developing human cerebral cortex remain elusive. Here, we find that EGFR expression begins to sharply increase after gestational week (GW) 20, which corresponds to the beginning stages of human gliogenesis. In addition, EGFR+ cells are mainly distributed in the germinal zone and frequently colocalize with the stemness marker SOX2 during this period. Then, by performing single-cell RNA sequencing on these EGFR+ cells, we successfully enriched and characterized various glial- and neuronal-lineage progenitor cells and validated their phenotypes in fixed slices. Notably, we identified two subgroups with molecular characteristics similar to those of astrocytes, and the immunostaining results show that these cells are mainly distributed in the outer subventricular zone and might originate from the outer radial glial cells. In short, the EGFR-sorting strategy and molecular signatures in the diverse lineages provide insights into human glial development.


Asunto(s)
Linaje de la Célula , Corteza Cerebral/fisiología , Células-Madre Neurales/fisiología , Neurogénesis , Neuroglía/fisiología , Neuronas/fisiología , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Bases de Datos Genéticas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Humanos , Ratones , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/genética , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Fenotipo , RNA-Seq , Análisis de la Célula Individual , Transcripción Genética
13.
Genome Biol ; 19(1): 31, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540203

RESUMEN

BACKGROUND: Organogenesis is crucial for proper organ formation during mammalian embryonic development. However, the similarities and shared features between different organs and the cellular heterogeneity during this process at single-cell resolution remain elusive. RESULTS: We perform single-cell RNA sequencing analysis of 1916 individual cells from eight organs and tissues of E9.5 to E11.5 mouse embryos, namely, the forebrain, hindbrain, skin, heart, somite, lung, liver, and intestine. Based on the regulatory activities rather than the expression patterns, all cells analyzed can be well classified into four major groups with epithelial, mesodermal, hematopoietic, and neuronal identities. For different organs within the same group, the similarities and differences of their features and developmental paths are revealed and reconstructed. CONCLUSIONS: We identify mutual interactions between epithelial and mesenchymal cells and detect epithelial cells with prevalent mesenchymal features during organogenesis, which are similar to the features of intermediate epithelial/mesenchymal cells during tumorigenesis. The comprehensive transcriptome at single-cell resolution profiled in our study paves the way for future mechanistic studies of the gene-regulatory networks governing mammalian organogenesis.


Asunto(s)
Epitelio/metabolismo , Mesodermo/metabolismo , Organogénesis/genética , Transcriptoma , Animales , Células Epiteliales/metabolismo , Epitelio/embriología , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Mesodermo/embriología , Ratones , Neuronas/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual
14.
Science ; 362(6418): 1060-1063, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30498128

RESUMEN

Although genomic instability, epigenetic abnormality, and gene expression dysregulation are hallmarks of colorectal cancer, these features have not been simultaneously analyzed at single-cell resolution. Using optimized single-cell multiomics sequencing together with multiregional sampling of the primary tumor and lymphatic and distant metastases, we developed insights beyond intratumoral heterogeneity. Genome-wide DNA methylation levels were relatively consistent within a single genetic sublineage. The genome-wide DNA demethylation patterns of cancer cells were consistent in all 10 patients whose DNA we sequenced. The cancer cells' DNA demethylation degrees clearly correlated with the densities of the heterochromatin-associated histone modification H3K9me3 of normal tissue and those of repetitive element long interspersed nuclear element 1. Our work demonstrates the feasibility of reconstructing genetic lineages and tracing their epigenomic and transcriptomic dynamics with single-cell multiomics sequencing.


Asunto(s)
Neoplasias Colorrectales/genética , Epigenómica/métodos , Análisis de la Célula Individual/métodos , Metilación de ADN , Femenino , Estudio de Asociación del Genoma Completo , Histonas , Humanos , Masculino , Análisis de Secuencia de ADN
15.
Nat Cell Biol ; 20(10): 1227, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30038257

RESUMEN

In the PDF version of this Resource originally published, on the author list the superscript number 9 to indicate Rui Wang was an equally contributing author was missing owing to a technical error. This has now been amended. In addition, the authors wish to replace Supplementary Table 2, as in the original version Group 1 immune cells were mis-classified into Group 46 immune cells, resulting in three groups of immune cells where there should have been four. Supplementary Table 2 has now been replaced.

16.
Nat Cell Biol ; 20(6): 721-734, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29802404

RESUMEN

The development of the digestive tract is critical for proper food digestion and nutrient absorption. Here, we analyse the main organs of the digestive tract, including the oesophagus, stomach, small intestine and large intestine, from human embryos between 6 and 25 weeks of gestation as well as the large intestine from adults using single-cell RNA-seq analyses. In total, 5,227 individual cells are analysed and 40 cell types clearly identified. Their crucial biological features, including developmental processes, signalling pathways, cell cycle, nutrient digestion and absorption metabolism, and transcription factor networks, are systematically revealed. Moreover, the differentiation and maturation processes of the large intestine are thoroughly investigated by comparing the corresponding transcriptome profiles between embryonic and adult stages. Our work offers a rich resource for investigating the gene regulation networks of the human fetal digestive tract and adult large intestine at single-cell resolution.


Asunto(s)
Células Epiteliales/fisiología , Tracto Gastrointestinal/fisiología , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma , Adulto , Factores de Edad , Diferenciación Celular/genética , Proliferación Celular/genética , Células Epiteliales/metabolismo , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/metabolismo , Marcadores Genéticos , Genotipo , Edad Gestacional , Humanos , Morfogénesis , Fenotipo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA